1
|
Tomatis C, Ferrer MF, Aquila S, Thomas PD, Arrías PN, Ferrelli L, Pidre M, Romanowski V, Carrera Silva EA, Gómez RM. Baculovirus surface display of a chimeric E-NS1 protein of YFV protects against YFV infection. Vaccine 2024; 42:126045. [PMID: 38852036 DOI: 10.1016/j.vaccine.2024.06.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/10/2024]
Abstract
Yellow fever (YF) is a disease caused by the homonymous flavivirus that can be prevented by a vaccine containing attenuated viruses. Since some individuals cannot receive this vaccine, the development of alternatives is desirable. Here, we developed a recombinant baculovirus (rBV) surface display platform utilizing a chimeric E-NS1 protein as a vaccine candidate. A pBacPAK9 vector containing the baculoviral GP64 signal peptide, the YFV prM, E, NS1 and the ectodomain of VSV-G sequences was synthesized. This transfer plasmid and the bAcGOZA bacmid were cotransfected into Sf9 cells, and an rBV-E-NS1 was obtained, which was characterized by PCR, WB, IFI and FACS analysis. Mice immunized with rBV-E-NS1 elicited a specific humoral and cellular immune response and were protected after YFV infection. In summary, we have developed an rBV that expresses YFV major antigen proteins on its surface, which opens new alternatives that can be tested in a mouse model.
Collapse
Affiliation(s)
- Carla Tomatis
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular (IBBM), CONICET-UNLP, La Plata, Buenos Aires, Argentina; Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental (IMEX), Academia Nacional de Medicina, Ciudad de Buenos Aires, Argentina
| | - María Florencia Ferrer
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular (IBBM), CONICET-UNLP, La Plata, Buenos Aires, Argentina
| | - Silvia Aquila
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular (IBBM), CONICET-UNLP, La Plata, Buenos Aires, Argentina
| | - Pablo Daniel Thomas
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular (IBBM), CONICET-UNLP, La Plata, Buenos Aires, Argentina
| | - Paula Nazarena Arrías
- Laboratorio de Virología Molecular, Instituto de Biotecnología y Biología Molecular (IBBM), CONICET-UNLP, La Plata, Buenos Aires, Argentina
| | - Leticia Ferrelli
- Laboratorio de Virología Molecular, Instituto de Biotecnología y Biología Molecular (IBBM), CONICET-UNLP, La Plata, Buenos Aires, Argentina
| | - Matías Pidre
- Laboratorio de Virología Molecular, Instituto de Biotecnología y Biología Molecular (IBBM), CONICET-UNLP, La Plata, Buenos Aires, Argentina
| | - Victor Romanowski
- Laboratorio de Virología Molecular, Instituto de Biotecnología y Biología Molecular (IBBM), CONICET-UNLP, La Plata, Buenos Aires, Argentina
| | - Eugenio Antonio Carrera Silva
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental (IMEX), Academia Nacional de Medicina, Ciudad de Buenos Aires, Argentina.
| | - Ricardo Martín Gómez
- Laboratorio de Patogénesis Viral, Instituto de Biotecnología y Biología Molecular (IBBM), CONICET-UNLP, La Plata, Buenos Aires, Argentina.
| |
Collapse
|
2
|
Lafoux B, Baillet N, Picard C, Fourcaud G, Borges-Cardoso V, Reynard S, Journeaux A, Germain C, Perthame E, Mateo M, Hortion J, Carnec X, Pietrosemoli N, Moroso M, Lacroix O, Jourjon O, Barron S, Vallve A, Duthey A, Jacquot F, Barrot L, Dirheimer M, Raoul H, Nougier C, Baize S. Hemostasis defects underlying the hemorrhagic syndrome caused by mammarenaviruses in a cynomolgus macaque model. Blood 2023; 142:2092-2104. [PMID: 37699247 DOI: 10.1182/blood.2023020351] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 07/18/2023] [Accepted: 08/15/2023] [Indexed: 09/14/2023] Open
Abstract
Viral hemorrhagic fevers (HF) are a group of acute febrile diseases with high mortality rates. Although hemostatic dysfunction appears to be a major determinant of the severity of the disease, it is still unclear what pathogenic mechanisms lead to it. In clinical studies it is found that arenaviruses, such as Lassa, Machupo, and Guanarito viruses cause HF that vary in symptoms and biological alterations. In this study we aimed to characterize the hemostatic dysfunction induced by arenaviral HF to determine its implication in the severity of the disease and to elucidate the origin of this syndrome. We found that lethal infection with Machupo, Guanarito, and Lassa viruses is associated with cutaneomucosal, cerebral, digestive, and pulmonary hemorrhages. The affected animals developed a severe alteration of the coagulation system, which was concomitant with acute hepatitis, minor deficit of hepatic factor synthesis, presence of a plasmatic inhibitor of coagulation, and dysfunction of the fibrinolytic system. Despite signs of increased vascular permeability, endothelial cell infection was not a determinant factor of the hemorrhagic syndrome. There were also alterations of the primary hemostasis during lethal infection, with moderate to severe thrombocytopenia and platelet dysfunction. Finally, we show that lethal infection is accompanied by a reduced hematopoietic potential of the bone marrow. This study provides an unprecedented characterization of the hemostasis defects induced by several highly pathogenic arenaviruses.
Collapse
Affiliation(s)
- Blaise Lafoux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Nicolas Baillet
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Caroline Picard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Gustave Fourcaud
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Virginie Borges-Cardoso
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Stéphanie Reynard
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Alexandra Journeaux
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Clara Germain
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Emeline Perthame
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Mathieu Mateo
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Jimmy Hortion
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Xavier Carnec
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| | - Natalia Pietrosemoli
- Institut Pasteur, Université Paris Cité, Bioinformatics and Biostatistics Hub, Paris, France
| | - Marie Moroso
- Laboratoire P4 INSERM-Jean Mérieux, INSERM US003, Lyon, France
| | - Orianne Lacroix
- Laboratoire P4 INSERM-Jean Mérieux, INSERM US003, Lyon, France
| | - Ophélie Jourjon
- Laboratoire P4 INSERM-Jean Mérieux, INSERM US003, Lyon, France
| | - Stéphane Barron
- Laboratoire P4 INSERM-Jean Mérieux, INSERM US003, Lyon, France
| | - Audrey Vallve
- Laboratoire P4 INSERM-Jean Mérieux, INSERM US003, Lyon, France
| | - Aurélie Duthey
- Laboratoire P4 INSERM-Jean Mérieux, INSERM US003, Lyon, France
| | | | - Laura Barrot
- Laboratoire P4 INSERM-Jean Mérieux, INSERM US003, Lyon, France
| | - Manon Dirheimer
- Laboratoire P4 INSERM-Jean Mérieux, INSERM US003, Lyon, France
| | - Hervé Raoul
- Laboratoire P4 INSERM-Jean Mérieux, INSERM US003, Lyon, France
| | - Christophe Nougier
- Service d'hématologie Biologique, Centre de Pathologie et Biologie Est, Groupement Hospitalier Est, Hospices Civils de Lyon, Bron, France
| | - Sylvain Baize
- Unité de Biologie des Infections Virales Emergentes, Institut Pasteur, Lyon, France
- Centre International de Recherche en Infectiologie, Université de Lyon, INSERM U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, Centre National de la Recherche Scientifique UMR5308, Lyon, France
| |
Collapse
|
3
|
Carestia A, Godin LC, Jenne CN. Step up to the platelet: Role of platelets in inflammation and infection. Thromb Res 2023; 231:182-194. [PMID: 36307228 DOI: 10.1016/j.thromres.2022.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/28/2022] [Accepted: 10/03/2022] [Indexed: 11/18/2022]
Abstract
Platelets are anucleated cells derived from megakaryocytes that are primarily responsible for hemostasis. However, in recent years, these cytoplasts have become increasingly recognized as immune cells, able to detect, interact with, and kill pathogens. As platelets are involved in both immunity and coagulation, they have a central role in immunothrombosis, a physiological process in which immune cells induce the formation of microthrombi to both prevent the spread of pathogens, and to help facilitate clearance. In this review, we will highlight the role of platelets as key players in the inflammatory and innate immune response against bacterial and viral infection, including direct and indirect interactions with pathogens and other immune cells.
Collapse
Affiliation(s)
- Agostina Carestia
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| | - Laura C Godin
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| | - Craig N Jenne
- Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Canada.
| |
Collapse
|
4
|
Rodríguez CS, Charó N, Tatti S, Gómez RM, D’Atri LP, Schattner M. Regulation of megakaryo/thrombopoiesis by endosomal toll-like receptor 7 and 8 activation of CD34 + cells in a viral infection model. Res Pract Thromb Haemost 2023; 7:100184. [PMID: 37538496 PMCID: PMC10394566 DOI: 10.1016/j.rpth.2023.100184] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 08/05/2023] Open
Abstract
Background CD34+ cells, megakaryocytes (MKs), and platelets express toll-like receptors (TLRs) that enable these cells to amplify the host innate immune response. However, the role of TLR7/TLR8 activation in megakaryopoiesis has not yet been investigated. Objectives We evaluated the effect of coxsackievirus B3 (CVB3) and synthetic TLR7/TLR8 agonists on the development of human MKs and production of platelets. Methods CD34+ cells from human umbilical cord were inoculated with CVB3 or stimulated with synthetic TLR7/TLR8 agonists and then cultured in the presence of thrombopoietin. Results CD34+ cells, MK progenitor cells, and mature MKs expressed TLR7 and TLR8, and exposure to CVB3 resulted in productive infection, as determined by the presence of viral infectious particles in culture supernatants. Cell expansion, differentiation into MKs, MK maturation, and platelet biogenesis were significantly reduced in CD34+-infected cultures. The reduction in MK growth was not due to an alteration in cellular proliferation but was accompanied by an increase in cellular apoptosis and pyroptosis. Impairment of MK generation and maturation of viable cells were also associated with decreased expression of transcription factors involved in these processes. These effects were completely abrogated by TLR7 but not TLR8 antagonists and mimicked by TLR7 but not TLR8 agonists. CVB3 infection of CD34+ cells increased the immunophenotype of MKs characterized as CD148+/CD48+ or CD41+/CD53+ cells. Conclusion These data suggest a novel role of TLR7 in megakaryo/thrombopoiesis that may contribute to a better understanding of the molecular basis underlying thrombocytopenia and the immunologic role of MKs in viral infection processes.
Collapse
Affiliation(s)
- Camila Sofía Rodríguez
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, IMEX-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Nancy Charó
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, IMEX-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | | | - Ricardo Martín Gómez
- Laboratory of Animal Viruses, Institute of Biotechnology and Molecular Biology, UNLP-CONICET, La Plata, Argentina
| | - Lina Paola D’Atri
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, IMEX-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis and Immunobiology of Inflammation, IMEX-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| |
Collapse
|
5
|
Activation of Most Toll-Like Receptors in Whole Human Blood Attenuates Platelet Deposition on Collagen under Flow. J Immunol Res 2023; 2023:1884439. [PMID: 36703865 PMCID: PMC9873445 DOI: 10.1155/2023/1884439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 12/16/2022] [Accepted: 12/22/2022] [Indexed: 01/19/2023] Open
Abstract
Platelets have toll-like receptors (TLRs); however, their function in thrombosis or hemostasis under flow conditions is not fully known. Thrombin-inhibited anticoagulated whole blood was treated with various TLR agonists and then perfused over fibrillar collagen using microfluidic assay at venous wall shear rate (100 s-1). Platelet deposition was imaged with fluorescent anti-CD61. For perfusion of whole blood without TLR agonist addition, platelets rapidly accumulated on collagen and eventually occluded the microchannels. Interestingly, most of the tested TLR agonists (Pam3CKS4, MALP-2, polyinosinic-polycytidylic acid HMW, imiquimod, and CpG oligodeoxynucleotides) strongly reduced platelet deposition on collagen, while only the TLR4 agonist endotoxin lipopolysaccharide (LPS) enhanced deposition. Following 90 sec of deposition under flow of untreated blood, the addition of various TLR-7 agonists (imiquimod, vesatolimod, and GSK2245035) all caused immediate blockade of further platelet deposition. Since TLR signaling can activate nuclear factor-kappaB (NF-κB), the IKK-inhibitor (IKK inhibitor VII) and NF-κB inhibitor (Bay 11-7082) were tested. The IKK/NF-κB inhibitors strongly inhibited platelet deposition under flow. Furthermore, addition of Pam3CSK4 (TLR1/2 ligand), MALP-2 (TLR2/6 ligand), and Imquimod (TLR7 ligand) reduced phosphotidylserine (PS) exposure. Activation of TLR1/2, TLR2/6, TLR3, TLR7, and TLR9 in whole blood reduced platelet deposition under flow on collagen; however, LPS (major Gram negative bacterial pathogenic component) activation of LTR4 was clearly prothrombotic.
Collapse
|
6
|
Tang X, Xu Q, Yang S, Huang X, Wang L, Huang F, Luo J, Zhou X, Wu A, Mei Q, Zhao C, Wu J. Toll-like Receptors and Thrombopoiesis. Int J Mol Sci 2023; 24:ijms24021010. [PMID: 36674552 PMCID: PMC9864288 DOI: 10.3390/ijms24021010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2022] [Revised: 12/27/2022] [Accepted: 01/03/2023] [Indexed: 01/06/2023] Open
Abstract
Platelets are the second most abundant blood component after red blood cells and can participate in a variety of physiological and pathological functions. Beyond its traditional role in hemostasis and thrombosis, it also plays an indispensable role in inflammatory diseases. However, thrombocytopenia is a common hematologic problem in the clinic, and it presents a proportional relationship with the fatality of many diseases. Therefore, the prevention and treatment of thrombocytopenia is of great importance. The expression of Toll-like receptors (TLRs) is one of the most relevant characteristics of thrombopoiesis and the platelet inflammatory function. We know that the TLR family is found on the surface or inside almost all cells, where they perform many immune functions. Of those, TLR2 and TLR4 are the main stress-inducing members and play an integral role in inflammatory diseases and platelet production and function. Therefore, the aim of this review is to present and discuss the relationship between platelets, inflammation and the TLR family and extend recent research on the influence of the TLR2 and TLR4 pathways and the regulation of platelet production and function. Reviewing the interaction between TLRs and platelets in inflammation may be a research direction or program for the treatment of thrombocytopenia-related and inflammatory-related diseases.
Collapse
Affiliation(s)
- Xiaoqin Tang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Qian Xu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Shuo Yang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Xinwu Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
| | - Long Wang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Feihong Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Jiesi Luo
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Xiaogang Zhou
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Anguo Wu
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Qibing Mei
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
| | - Chunling Zhao
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Correspondence: (C.Z.); (J.W.); Tel.: +86-186-8307-3667 (C.Z.); +86-139-8241-6641 (J.W.)
| | - Jianming Wu
- Department of Physiology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- Institute of Cardiovascular Research, the Key Laboratory of Medical Electrophysiology, Ministry of Education of China, Luzhou 646000, China
- Correspondence: (C.Z.); (J.W.); Tel.: +86-186-8307-3667 (C.Z.); +86-139-8241-6641 (J.W.)
| |
Collapse
|
7
|
Garcia C, Compagnon B, Poëtte M, Gratacap MP, Lapébie FX, Voisin S, Minville V, Payrastre B, Vardon-Bounes F, Ribes A. Platelet Versus Megakaryocyte: Who Is the Real Bandleader of Thromboinflammation in Sepsis? Cells 2022; 11:1507. [PMID: 35563812 PMCID: PMC9104300 DOI: 10.3390/cells11091507] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/27/2022] [Accepted: 04/28/2022] [Indexed: 11/24/2022] Open
Abstract
Platelets are mainly known for their key role in hemostasis and thrombosis. However, studies over the last two decades have shown their strong implication in mechanisms associated with inflammation, thrombosis, and the immune system in various neoplastic, inflammatory, autoimmune, and infectious diseases. During sepsis, platelets amplify the recruitment and activation of innate immune cells at the site of infection and contribute to the elimination of pathogens. In certain conditions, these mechanisms can lead to thromboinflammation resulting in severe organ dysfunction. Here, we discuss the interactions of platelets with leukocytes, neutrophil extracellular traps (NETs), and endothelial cells during sepsis. The intrinsic properties of platelets that generate an inflammatory signal through the NOD-like receptor family, pyrin domain-containing 3 (NLRP3) inflammasome are discussed. As an example of immunothrombosis, the implication of platelets in vaccine-induced immune thrombotic thrombocytopenia is documented. Finally, we discuss the role of megakaryocytes (MKs) in thromboinflammation and their adaptive responses.
Collapse
Affiliation(s)
- Cédric Garcia
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| | - Baptiste Compagnon
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Michaël Poëtte
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Marie-Pierre Gratacap
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| | - François-Xavier Lapébie
- Service de Médecine Vasculaire, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Sophie Voisin
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
| | - Vincent Minville
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Bernard Payrastre
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| | - Fanny Vardon-Bounes
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
- Pôle Anesthésie-Réanimation, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France;
| | - Agnès Ribes
- Laboratoire d’Hématologie, Centre Hospitalier Universitaire de Toulouse, 31059 Toulouse, France; (C.G.); (S.V.); (B.P.)
- Institut des Maladies Métaboliques et Cardiovasculaires, Inserm UMR1297 and Université Toulouse 3, 31024 Toulouse, France; (B.C.); (M.P.); (M.-P.G.); (F.V.-B.)
| |
Collapse
|
8
|
Schrottmaier WC, Schmuckenschlager A, Pirabe A, Assinger A. Platelets in Viral Infections - Brave Soldiers or Trojan Horses. Front Immunol 2022; 13:856713. [PMID: 35419008 PMCID: PMC9001014 DOI: 10.3389/fimmu.2022.856713] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/03/2022] [Indexed: 11/13/2022] Open
Abstract
Viral infections are often associated with platelet activation and haemostatic complications. In line, low platelet counts represent a hallmark for poor prognosis in many infectious diseases. The underlying cause of platelet dysfunction in viral infections is multifaceted and complex. While some viruses directly interact with platelets and/or megakaryocytes to modulate their function, also immune and inflammatory responses directly and indirectly favour platelet activation. Platelet activation results in increased platelet consumption and degradation, which contributes to thrombocytopenia in these patients. The role of platelets is often bi-phasic. Initial platelet hyper-activation is followed by a state of platelet exhaustion and/or hypo-responsiveness, which together with low platelet counts promotes bleeding events. Thereby infectious diseases not only increase the thrombotic but also the bleeding risk or both, which represents a most dreaded clinical complication. Treatment options in these patients are limited and new therapeutic strategies are urgently needed to prevent adverse outcome. This review summarizes the current literature on platelet-virus interactions and their impact on viral pathologies and discusses potential intervention strategies. As pandemics and concomitant haemostatic dysregulations will remain a recurrent threat, understanding the role of platelets in viral infections represents a timely and pivotal challenge.
Collapse
Affiliation(s)
- Waltraud C Schrottmaier
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anna Schmuckenschlager
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Anita Pirabe
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| | - Alice Assinger
- Institute of Vascular Biology and Thrombosis Research, Centre of Physiology and Pharmacology, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
9
|
Menter DG, Afshar-Kharghan V, Shen JP, Martch SL, Maitra A, Kopetz S, Honn KV, Sood AK. Of vascular defense, hemostasis, cancer, and platelet biology: an evolutionary perspective. Cancer Metastasis Rev 2022; 41:147-172. [PMID: 35022962 PMCID: PMC8754476 DOI: 10.1007/s10555-022-10019-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/04/2022] [Indexed: 01/08/2023]
Abstract
We have established considerable expertise in studying the role of platelets in cancer biology. From this expertise, we were keen to recognize the numerous venous-, arterial-, microvascular-, and macrovascular thrombotic events and immunologic disorders are caused by severe, acute-respiratory-syndrome coronavirus 2 (SARS-CoV-2) infections. With this offering, we explore the evolutionary connections that place platelets at the center of hemostasis, immunity, and adaptive phylogeny. Coevolutionary changes have also occurred in vertebrate viruses and their vertebrate hosts that reflect their respective evolutionary interactions. As mammals adapted from aquatic to terrestrial life and the heavy blood loss associated with placentalization-based live birth, platelets evolved phylogenetically from thrombocytes toward higher megakaryocyte-blebbing-based production rates and the lack of nuclei. With no nuclei and robust RNA synthesis, this adaptation may have influenced viral replication to become less efficient after virus particles are engulfed. Human platelets express numerous receptors that bind viral particles, which developed from archetypal origins to initiate aggregation and exocytic-release of thrombo-, immuno-, angiogenic-, growth-, and repair-stimulatory granule contents. Whether by direct, evolutionary, selective pressure, or not, these responses may help to contain virus spread, attract immune cells for eradication, and stimulate angiogenesis, growth, and wound repair after viral damage. Because mammalian and marsupial platelets became smaller and more plate-like their biophysical properties improved in function, which facilitated distribution near vessel walls in fluid-shear fields. This adaptation increased the probability that platelets could then interact with and engulf shedding virus particles. Platelets also generate circulating microvesicles that increase membrane surface-area encounters and mark viral targets. In order to match virus-production rates, billions of platelets are generated and turned over per day to continually provide active defenses and adaptation to suppress the spectrum of evolving threats like SARS-CoV-2.
Collapse
Affiliation(s)
- David G Menter
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Vahid Afshar-Kharghan
- Division of Internal Medicine, Benign Hematology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - John Paul Shen
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephanie L Martch
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Anirban Maitra
- Department of Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Scott Kopetz
- Department of GI Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kenneth V Honn
- Department of Pathology, Bioactive Lipids Research Program, Wayne State University, 5101 Cass Ave. 430 Chemistry, Detroit, MI, 48202, USA
- Department of Pathology, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
- Cancer Biology Division, Wayne State University School of Medicine, 431 Chemistry Bldg, Detroit, MI, 48202, USA
| | - Anil K Sood
- Department of Gynecologic Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Center for RNA Interference and Non-Coding RNA, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- Department of Cancer Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
10
|
Lahon A, Arya RP, Banerjea AC. Dengue Virus Dysregulates Master Transcription Factors and PI3K/AKT/mTOR Signaling Pathway in Megakaryocytes. Front Cell Infect Microbiol 2021; 11:715208. [PMID: 34513730 PMCID: PMC8427595 DOI: 10.3389/fcimb.2021.715208] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 08/05/2021] [Indexed: 01/27/2023] Open
Abstract
Dengue virus (DENV) infection can cause either self-limited dengue fever or hemorrhagic complications. Low platelet count is one of the manifestations of dengue fever. Megakaryocytes are the sole producers of platelets. However, the role of both host and viral factors in megakaryocyte development, maturation, and platelet production is largely unknown in DENV infection. PI3K/AKT/mTOR pathway plays a significant role in cell survival, maturation, and megakaryocyte development. We were interested to check whether pathogenic insult can impact this pathway. We observed decreased expression of most of the major key molecules associated with the PI3K/AKT/mTOR pathway in DENV infected MEG-01 cells. In this study, the involvement of PI3K/AKT/mTOR pathway in megakaryocyte development and maturation was confirmed with the use of specific inhibitors in infected MEG-01 cells. Our results showed that direct pharmacologic inhibition of this pathway greatly impacted megakaryopoiesis associated molecule CD61 and some essential transcription factors (GATA-1, GATA-2, and NF-E2). Additionally, we observed apoptosis in megakaryocytes due to DENV infection. Our results may suggest that DENV impairs PI3K/AKT/mTOR axis and molecules involved in the development and maturation of megakaryocytes. It is imperative to investigate the role of these molecules in the context of megakaryopoiesis during DENV infection to better understand the pathways and mechanisms, which in turn might provide insights into the development of antiviral strategies.
Collapse
Affiliation(s)
- Anismrita Lahon
- Laboratory of Virology, National Institute of Immunology, New Delhi, India
| | - Ravi P Arya
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Akhil C Banerjea
- Laboratory of Virology, National Institute of Immunology, New Delhi, India.,Institute of Advanced Virology, Kerala, India
| |
Collapse
|
11
|
Dengue virus induces interferon-β by activating RNA sensing pathways in megakaryocytes. Immunol Lett 2021; 236:31-36. [PMID: 34111476 DOI: 10.1016/j.imlet.2021.06.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 05/28/2021] [Accepted: 06/03/2021] [Indexed: 11/21/2022]
Abstract
Activation of innate receptors in megakaryocytes (MKs) may affect the ability to produce functional platelets. Low platelet count is one of the clinical manifestations of dengue virus (DENV) infection. In MKs, the effect of innate receptors during DENV-infection is not well studied. Here we used MEG-01 cells to investigate DENV serotype 2 induced innate receptors in these cells. DENV RNA was estimated by qRT-PCR in the culture supernatant. The expression of innate receptors was determined by western blot and qPCR. DENV infection led to increased expression of RIG-I at 24 hrs post-infection (hpi) and MDA-5 at 48 and 72 hpi (p<0.05). However, no change in the expression of TLR3 at protein level was observed. Activation of MDA-5 resulted in increased expression of IFN-β and ISG-15 in DENV infected MEG-01 cells, which was further confirmed by MDA-5 siRNA treatment. Apart from inducing innate receptors, DENV significantly decreases the expression of CD61, an activation marker of megakaryocyteson MEG-01 cells as observed by flow cytometry analysis (p<0.01). Results from this study confirm that DENV infection activates the type-I interferon in megakaryocytes and may play a significant role in maturation and development.
Collapse
|
12
|
Xue L, Guo W, Li L, Ou S, Zhu T, Cai L, Ding W, Wu W. Metabolomic profiling identifies a novel mechanism for heat stroke‑related acute kidney injury. Mol Med Rep 2021; 23:241. [PMID: 33655337 PMCID: PMC7893796 DOI: 10.3892/mmr.2021.11880] [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: 04/13/2020] [Accepted: 08/20/2020] [Indexed: 11/27/2022] Open
Abstract
Heat stroke can induce a systemic inflammatory response, which may lead to multi‑organ dysfunction including acute kidney injury (AKI) and electrolyte disturbances. To investigate the pathogenesis of heat stroke (HS)‑related AKI, a mouse model of HS was induced by increasing the animal's core temperature to 41˚C. Blood samples obtained from the tail vein were used to measure plasma glucose and creatinine levels. Micro‑positron emission tomography‑computed tomography (micro‑PET/CT), H&E staining and transmission electron microscopy were conducted to examine metabolic and morphological changes in the mouse kidneys. Immunohistochemistry (IHC) and western blot analyses were performed to investigate the expression of apoptosis‑inducing factor mitochondria‑associated 2 (Aifm2), high‑mobility group box 1 (HMGB1) and receptor for advanced glycosylation end products (RAGE). Liquid chromatography‑mass spectrometry analysis was conducted to find differential metabolites and signaling pathways. The HS mouse model was built successfully, with significantly increased creatinine levels detected in the serum of HS mice compared with controls, whereas micro‑PET/CT revealed active metabolism in the whole body of HS mice. H&E and TUNEL staining revealed that the kidneys of HS mice exhibited signs of hemorrhage and apoptosis. IHC and western blotting demonstrated significant upregulation of Aifm2, HMGB1 and RAGE in response to HS. Finally, 136 differential metabolites were screened out, and enrichment of the 'biosynthesis of unsaturated fatty acids' pathway was detected. HS‑associated AKI is the renal manifestation of systemic inflammatory response syndrome, and may be triggered by the HMGB1/RAGE pathway. Metabolomics indicated increased adrenic acid, docosahexaenoic acid and eicosapentaenoic acid may serve as metabolic biomarkers for AKI in HS. The findings suggested that a correlation between the HMGB1/RAGE pathway and biosynthesis of unsaturated fatty acids may contribute to the progression of HS‑related AKI.
Collapse
Affiliation(s)
- Ling Xue
- Department of Urology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenli Guo
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Li Li
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Santao Ou
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Tingting Zhu
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Liang Cai
- Department of Nuclear Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Wenfei Ding
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Weihua Wu
- Department of Nephrology, Sichuan Clinical Research Center for Nephropathy, The Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| |
Collapse
|
13
|
Feng FE, Zhang GC, Liu FQ, He Y, Zhu XL, Liu X, Wang Y, Wang JZ, Fu HX, Chen YH, Han W, Chang YJ, Xu LP, Liu KY, Huang XJ, Zhang XH. HCMV modulates c-Mpl/IEX-1 pathway-mediated megakaryo/thrombopoiesis via PDGFRα and αvβ3 receptors after allo-HSCT. J Cell Physiol 2021; 236:6726-6741. [PMID: 33611789 DOI: 10.1002/jcp.30335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 02/05/2021] [Accepted: 02/09/2021] [Indexed: 11/08/2022]
Abstract
Thrombocytopenia is a common complication of human cytomegalovirus (HCMV) infection in immunocompromised hosts, which contributes to poor prognosis even in patients receiving antiviral treatment. Here, we investigated the megakaryo/thrombopoiesis process, including the involvement of the c-Mpl/IEX-1 pathway, after HCMV infection, identified receptors mediating the interaction between megakaryocytes (MKs) and HCMV, and explored novel therapeutic targets. Our data shows that HCMV directly infects megakaryocytes in patients with HCMV DNAemia and influences megakaryopoiesis via the c-Mpl/IEX-1 pathway throughout megakaryocyte maturation, apoptosis, and platelet generation in vivo and in vitro. After treatment with inhibitors of PDGFRα and αvβ3, the HCMV infection rate in MKs was significantly reduced, suggesting that IMC-3G3 and anti-αvβ3 are potential therapeutic alternatives for viral infection. In summary, our study proposes a possible mechanism and potential treatments for thrombocytopenia caused by HCMV infection and other viral diseases associated with abnormal hemostasis.
Collapse
Affiliation(s)
- Fei-Er Feng
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Gao-Chao Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Feng-Qi Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Yun He
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Xiao-Lu Zhu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Xiao Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Jing-Zhi Wang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Hai-Xia Fu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Yu-Hong Chen
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Wei Han
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Ying-Jun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Lan-Ping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Kai-Yan Liu
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Xiao-Jun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| | - Xiao-Hui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, Beijing, China.,National Clinical Research Center for Hematologic Disease, Beijing, China.,Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China.,Collaborative Innovation Centre of Hematology, Peking University, Beijing, China
| |
Collapse
|
14
|
Dib PRB, Quirino-Teixeira AC, Merij LB, Pinheiro MBM, Rozini SV, Andrade FB, Hottz ED. Innate immune receptors in platelets and platelet-leukocyte interactions. J Leukoc Biol 2020; 108:1157-1182. [PMID: 32779243 DOI: 10.1002/jlb.4mr0620-701r] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 06/11/2020] [Accepted: 06/28/2020] [Indexed: 12/14/2022] Open
Abstract
Platelets are chief cells in hemostasis. Apart from their hemostatic roles, platelets are major inflammatory effector cells that can influence both innate and adaptive immune responses. Activated platelets have thromboinflammatory functions linking hemostatic and immune responses in several physiological and pathological conditions. Among many ways in which platelets exert these functions, platelet expression of pattern recognition receptors (PRRs), including TLR, Nod-like receptor, and C-type lectin receptor families, plays major roles in sensing and responding to pathogen-associated or damage-associated molecular patterns (PAMPs and DAMPs, respectively). In this review, an increasing body of evidence is compiled showing the participation of platelet innate immune receptors, including PRRs, in infectious diseases, sterile inflammation, and cancer. How platelet recognition of endogenous DAMPs participates in sterile inflammatory diseases and thrombosis is discussed. In addition, platelet recognition of both PAMPs and DAMPs initiates platelet-mediated inflammation and vascular thrombosis in infectious diseases, including viral, bacterial, and parasite infections. The study also focuses on the involvement of innate immune receptors in platelet activation during cancer, and their contribution to tumor microenvironment development and metastasis. Finally, how innate immune receptors participate in platelet communication with leukocytes, modulating leukocyte-mediated inflammation and immune functions, is highlighted. These cell communication processes, including platelet-induced release of neutrophil extracellular traps, platelet Ag presentation to T-cells and platelet modulation of monocyte cytokine secretion are discussed in the context of infectious and sterile diseases of major concern in human health, including cardiovascular diseases, dengue, HIV infection, sepsis, and cancer.
Collapse
Affiliation(s)
- Paula Ribeiro Braga Dib
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil.,Laboratory of Immunology, Infectious Diseases and Obesity, Department of Parasitology, Microbiology and Immunology, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Anna Cecíllia Quirino-Teixeira
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Laura Botelho Merij
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Mariana Brandi Mendonça Pinheiro
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Stephane Vicente Rozini
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Fernanda Brandi Andrade
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Eugenio Damaceno Hottz
- Laboratory of Immunothrombosis, Department of Biochemistry, Institute of Biological Sciences, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
15
|
Mateer EJ, Maruyama J, Card GE, Paessler S, Huang C. Lassa Virus, but Not Highly Pathogenic New World Arenaviruses, Restricts Immunostimulatory Double-Stranded RNA Accumulation during Infection. J Virol 2020; 94:e02006-19. [PMID: 32051278 PMCID: PMC7163147 DOI: 10.1128/jvi.02006-19] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Accepted: 02/09/2020] [Indexed: 12/14/2022] Open
Abstract
The arenaviruses Lassa virus (LASV), Junín virus (JUNV), and Machupo virus (MACV) can cause severe and fatal diseases in humans. Although these pathogens are closely related, the host immune responses to these virus infections differ remarkably, with direct implications for viral pathogenesis. LASV infection is immunosuppressive, with a very low-level interferon response. In contrast, JUNV and MACV infections stimulate a robust interferon (IFN) response in a retinoic acid-inducible gene I (RIG-I)-dependent manner and readily activate protein kinase R (PKR), a known host double-stranded RNA (dsRNA) sensor. In response to infection with RNA viruses, host nonself RNA sensors recognize virus-derived dsRNA as danger signals and initiate innate immune responses. Arenavirus nucleoproteins (NPs) contain a highly conserved exoribonuclease (ExoN) motif, through which LASV NP has been shown to degrade virus-derived immunostimulatory dsRNA in biochemical assays. In this study, we for the first time present evidence that LASV restricts dsRNA accumulation during infection. Although JUNV and MACV NPs also have the ExoN motif, dsRNA readily accumulated in infected cells and often colocalized with dsRNA sensors. Moreover, LASV coinfection diminished the accumulation of dsRNA and the IFN response in JUNV-infected cells. The disruption of LASV NP ExoN with a mutation led to dsRNA accumulation and impaired LASV replication in minigenome systems. Importantly, both LASV NP and RNA polymerase L protein were required to diminish the accumulation of dsRNA and the IFN response in JUNV infection. For the first time, we discovered a collaboration between LASV NP ExoN and L protein in limiting dsRNA accumulation. Our new findings provide mechanistic insights into the differential host innate immune responses to highly pathogenic arenavirus infections.IMPORTANCE Arenavirus NPs contain a highly conserved DEDDh ExoN motif, through which LASV NP degrades virus-derived, immunostimulatory dsRNA in biochemical assays to eliminate the danger signal and inhibit the innate immune response. Nevertheless, the function of NP ExoN in arenavirus infection remains to be defined. In this study, we discovered that LASV potently restricts dsRNA accumulation during infection and minigenome replication. In contrast, although the NPs of JUNV and MACV also harbor the ExoN motif, dsRNA readily formed during JUNV and MACV infections, accompanied by IFN and PKR responses. Interestingly, LASV NP alone was not sufficient to limit dsRNA accumulation. Instead, both LASV NP and L protein were required to restrict immunostimulatory dsRNA accumulation. Our findings provide novel and important insights into the mechanism for the distinct innate immune response to these highly pathogenic arenaviruses and open new directions for future studies.
Collapse
Affiliation(s)
- Elizabeth J Mateer
- Department of Pathology, Galveston National Laboratory and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| | - Junki Maruyama
- Department of Pathology, Galveston National Laboratory and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| | - Galen E Card
- Department of Pathology, Galveston National Laboratory and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| | - Slobodan Paessler
- Department of Pathology, Galveston National Laboratory and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| | - Cheng Huang
- Department of Pathology, Galveston National Laboratory and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
16
|
Abstract
Platelets have a well-recognized role in hemostasis and thrombosis, and they are important amplifiers of inflammation and innate immune responses. The formation of DNA extracellular traps (ETs) is a complex cellular mechanism, which occurs in response to microbial infections and sterile inflammation, and results in the release of DNA complexed with histones and various granular proteins. ETs were first discovered in neutrophils (NETs); however, it is now accepted that other leukocytes, including eosinophils (EETs) and monocytes/macrophages (MoETs/METs), can also generate them. Moreover, several types of ETs have been described.Increasing evidence has demonstrated that platelets modulate the formation of ETs. This review summarizes recent findings about the physiopathological role of platelets in the formation of ETs during infection and future perspectives in the field.
Collapse
Affiliation(s)
- Ricardo M Gómez
- Laboratorio De Virus Animales, Instituto De Biotecnología Y Biología Molecular, CONICET-UNLP, La Plata, Argentina.,Global Viral Network, Baltimore, MD, USA
| | - Aída O López Ortiz
- Laboratorio De Virus Animales, Instituto De Biotecnología Y Biología Molecular, CONICET-UNLP, La Plata, Argentina.,Laboratorio De Trombosis Experimental, Instituto De Medicina Experimental, CONICET-ANM, Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratorio De Trombosis Experimental, Instituto De Medicina Experimental, CONICET-ANM, Buenos Aires, Argentina
| |
Collapse
|
17
|
D'Atri LP, Rodríguez CS, Miguel CP, Pozner RG, Ortiz Wilczyñski JM, Negrotto S, Carrera-Silva EA, Heller PG, Schattner M. Activation of toll-like receptors 2 and 4 on CD34 + cells increases human megakaryo/thrombopoiesis induced by thrombopoietin. J Thromb Haemost 2019; 17:2196-2210. [PMID: 31397069 DOI: 10.1111/jth.14605] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2019] [Accepted: 08/07/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUND Platelet Toll-like receptor (TLR)2/4 are key players in amplifying the host immune response; however, their role in human megakaryo/thrombopoiesis has not yet been defined. OBJECTIVES We evaluated whether Pam3CSK4 or lipopolysaccharide (LPS), TLR2/4 ligands respectively, modulate human megakaryocyte development and platelet production. METHODS CD34+ cells from human umbilical cord were stimulated with LPS or Pam3CSK4 with or without thrombopoietin (TPO). RESULTS CD34+ cells and megakaryocytes express TLR2 and TLR4 at both RNA and protein level; however, direct stimulation of CD34+ cells with LPS or Pam3CSK4 had no effect on cell growth. Interestingly, both TLR ligands markedly increased TPO-induced CD34+ cell proliferation, megakaryocyte number and maturity, proplatelet and platelet production when added at day 0. In contrast, this synergism was not observed when TLR agonists were added 7 days after TPO addition. Interleukin-6 (IL-6) release was observed upon CD34+ or megakaryocyte stimulation with LPS or Pam3CSK4 but not with TPO and this effect was potentiated in combination with TPO. The increased proliferation and IL-6 production induced by TPO + LPS or Pam3CSK4 were suppressed by TLR2/4 or IL-6 neutralizing antibodies, as well as by PI3K/AKT and nuclear factor-κB inhibitors. Additionally, increased proplatelet and platelet production were associated with enhanced nuclear translocation of nuclear factor-E2. Finally, the supernatants of CD34+ cells stimulated with TPO+LPS-induced CFU-M colonies. CONCLUSIONS Our data suggest that the activation of TLR2 and TLR4 in CD34+ cells and megakaryocytes in the presence of TPO may contribute to warrant platelet provision during infection episodes by an autocrine IL-6 loop triggered by PI3K/NF-κB axes.
Collapse
Affiliation(s)
- Lina Paola D'Atri
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Camila Sofía Rodríguez
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Carolina Paula Miguel
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Roberto Gabriel Pozner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Juan Manuel Ortiz Wilczyñski
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Soledad Negrotto
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Eugenio Antonio Carrera-Silva
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | - Paula Graciela Heller
- Institute of Medical Research Dr. Alfredo Lanari, School of Medicine, University of Buenos Aires, Buenos Aires, Argentina
- Department of Hematology Research, National Scientific and Technical Research Council (CONICET), University of Buenos Aires, Institute of Medical Research (IDIM), Buenos Aires, Argentina
| | - Mirta Schattner
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine-CONICET-National Academy of Medicine, Buenos Aires, Argentina
| |
Collapse
|
18
|
Dengue viruses infect human megakaryocytes, with probable clinical consequences. PLoS Negl Trop Dis 2019; 13:e0007837. [PMID: 31765380 PMCID: PMC6901235 DOI: 10.1371/journal.pntd.0007837] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 12/09/2019] [Accepted: 10/10/2019] [Indexed: 11/30/2022] Open
Abstract
One of the most important clinical signs of dengue virus infection is the reduction of white blood cells and platelets in human peripheral blood (leukopenia and thrombocytopenia, respectively), which may significantly impair the clearance of dengue virus by the immune system. The cause of thrombocytopenia and leukopenia during dengue infection is still unknown, but may be related to severe suppression of bone marrow populations including hematopoietic stem cells and megakaryocytes, the progenitors of white blood cells and platelets respectively. Here, we explored the possibility that bone marrow suppression, including ablation of megakaryocyte populations, is caused by dengue virus infection of megakaryocytes. We used three different models to measure dengue virus infection and replication: in vitro, in a human megakaryocyte cell line with viral receptors, ex vivo, in primary human megakaryocytes, and in vivo, in humanized mice. All three systems support dengue virus infection and replication, including virus strains from serotypes 1, 2, and 3, and clinical signs, in vivo; all assays showed viral RNA and/or infectious viruses 7–14 days post-infection. Although we saw no significant decrease in cell viability in vitro, there was significant depletion of mature megakaryocytes in vivo. We conclude that megakaryocytes can produce dengue viruses in the bone marrow niche, and a reduction of cell numbers may affect bone marrow homeostasis. Dengue virus is the most common cause of viral hemorrhagic fever in humans. Over half of the world’s population lives in an at risk area for dengue virus infection, and this number will continue to grow as climate change allows the mosquito vectors of dengue virus to expand their breeding ranges to more temperate climates. Currently, there are no specific treatments for dengue virus infection. Understanding how dengue virus causes hemorrhagic fever could inform the development of these much needed treatments. Populations of important immune system mediators, such as white blood cells and platelets, are significantly dysregulated during dengue virus infection. These cells originate in the bone marrow, which experiences significant suppression, including a complete ablation of megakaryocytes (platelet progenitor cells), during DENV infection. Here, we add to the knowledge on how dengue virus induces bone marrow suppression by investigating whether dengue virus infects human megakaryocytes. We discovered that dengue virus infects human megakaryocytes in vitro, ex vivo, and in vivo models of dengue virus infection; however, dengue virus infection does not appear to directly affect viability of human megakaryocytes. Future studies will investigate whether infected megakaryocytes are still able to perform their functions of producing platelets and maintaining bone marrow homeostasis.
Collapse
|
19
|
Ferrer MF, Thomas P, López Ortiz AO, Errasti AE, Charo N, Romanowski V, Gorgojo J, Rodriguez ME, Carrera Silva EA, Gómez RM. Junin Virus Triggers Macrophage Activation and Modulates Polarization According to Viral Strain Pathogenicity. Front Immunol 2019; 10:2499. [PMID: 31695702 PMCID: PMC6817498 DOI: 10.3389/fimmu.2019.02499] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/07/2019] [Indexed: 01/24/2023] Open
Abstract
The New World arenavirus Junin (JUNV) is the etiological agent of Argentine hemorrhagic fever (AHF). Previous studies of human macrophage infection by the Old-World arenaviruses Mopeia and Lassa showed that while the non-pathogenic Mopeia virus replicates and activates human macrophages, the pathogenic Lassa virus replicates but fails to activate human macrophages. Less is known in regard to the impact of New World arenavirus infection on the human macrophage immune response. Macrophage activation is critical for controlling infections but could also be usurped favoring immune evasion. Therefore, it is crucial to understand how the JUNV infection modulates macrophage plasticity to clarify its role in AHF pathogenesis. With this aim in mind, we compared infection with the attenuated Candid 1 (C#1) or the pathogenic P strains of the JUNV virus in human macrophage cultures. The results showed that both JUNV strains similarly replicated and induced morphological changes as early as 1 day post-infection. However, both strains differentially induced the expression of CD71, the receptor for cell entry, the activation and maturation molecules CD80, CD86, and HLA-DR and selectively modulated cytokine production. Higher levels of TNF-α, IL-10, and IL-12 were detected with C#1 strain, while the P strain induced only higher levels of IL-6. We also found that C#1 strain infection skewed macrophage polarization to M1, whereas the P strain shifted the response to an M2 phenotype. Interestingly, the MERTK receptor, that negatively regulates the immune response, was down-regulated by C#1 strain and up-regulated by P strain infection. Similarly, the target genes of MERTK activation, the cytokine suppressors SOCS1 and SOCS3, were also increased after P strain infection, in addition to IRF-1, that regulates type I IFN levels, which were higher with C#1 compared with P strain infection. Together, this differential activation/polarization pattern of macrophages elicited by P strain suggests a more evasive immune response and may have important implications in the pathogenesis of AHF and underpinning the development of new potential therapeutic strategies.
Collapse
Affiliation(s)
- María F Ferrer
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Pablo Thomas
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Aída O López Ortiz
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina.,Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Andrea E Errasti
- Facultad de Medicina, Instituto de Farmacologia, University of Buenos Aries, Buenos Aires, Argentina
| | - Nancy Charo
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Victor Romanowski
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina.,Global Viral Network, Baltimore, MD, United States
| | - Juan Gorgojo
- Centro de Investigación y Desarrollo en Fermentaciones Industriales, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - María E Rodriguez
- Centro de Investigación y Desarrollo en Fermentaciones Industriales, CONICET-Universidad Nacional de La Plata, La Plata, Argentina
| | - Eugenio A Carrera Silva
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, CONICET-Academia Nacional de Medicina, Buenos Aires, Argentina
| | - Ricardo M Gómez
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, CONICET-Universidad Nacional de La Plata, La Plata, Argentina.,Global Viral Network, Baltimore, MD, United States
| |
Collapse
|
20
|
Speranza E, Ruibal P, Port JR, Feng F, Burkhardt L, Grundhoff A, Günther S, Oestereich L, Hiscox JA, Connor JH, Muñoz-Fontela C. T-Cell Receptor Diversity and the Control of T-Cell Homeostasis Mark Ebola Virus Disease Survival in Humans. J Infect Dis 2019; 218:S508-S518. [PMID: 29986035 DOI: 10.1093/infdis/jiy352] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Differences in T-cell phenotype, particularly the expression of markers of T-cell homeostasis, have been observed in fatal and nonfatal Ebola virus disease (EVD). However, the relationship between these markers with T-cell function and virus clearance during EVD is poorly understood. To gain biological insight into the role of T cells during EVD, combined transcriptomics and T-cell receptor sequencing was used to profile blood samples from fatal and nonfatal EVD patients from the recent West African EVD epidemic. Fatal EVD was characterized by strong T-cell activation and increased abundance of T-cell inhibitory molecules. However, the early T-cell response was oligoclonal and did not result in viral clearance. In contrast, survivors mounted highly diverse T-cell responses, maintained low levels of T-cell inhibitors, and cleared Ebola virus. Our findings highlight the importance of T-cell immunity in surviving EVD and strengthen the foundation for further research on targeting of the dendritic cell-T cell interface for postexposure immunotherapy.
Collapse
Affiliation(s)
- Emily Speranza
- Department of Microbiology, Boston University School of Medicine, Boston MA.,Department of Bioinformatics Program, Boston University, Boston MA.,Department of National Emerging Infectious Diseases Laboratories, Boston University, Boston MA.,Department of Mathematics and Statistics, Boston University, Boston MA
| | - Paula Ruibal
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Julia R Port
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner SiteHamburg, Germany
| | - Feng Feng
- Department of Microbiology, Boston University School of Medicine, Boston MA.,Department of Mathematics and Statistics, Boston University, Boston MA
| | - Lia Burkhardt
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Adam Grundhoff
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Hamburg, Germany
| | - Stephan Günther
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner SiteHamburg, Germany
| | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner SiteHamburg, Germany
| | - Julian A Hiscox
- Institute for Infection and Global Health, University of Liverpool, United Kingdom.,Singapore Immunology Network, A*STAR, Singapore
| | - John H Connor
- Department of Microbiology, Boston University School of Medicine, Boston MA.,Department of Bioinformatics Program, Boston University, Boston MA.,Department of National Emerging Infectious Diseases Laboratories, Boston University, Boston MA.,Department of Mathematics and Statistics, Boston University, Boston MA
| | - César Muñoz-Fontela
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.,German Center for Infection Research (DZIF), Partner SiteHamburg, Germany
| |
Collapse
|
21
|
Differential Immune Responses to Hemorrhagic Fever-Causing Arenaviruses. Vaccines (Basel) 2019; 7:vaccines7040138. [PMID: 31581720 PMCID: PMC6963578 DOI: 10.3390/vaccines7040138] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Revised: 09/25/2019] [Accepted: 09/30/2019] [Indexed: 12/28/2022] Open
Abstract
The family Arenaviridae contains several pathogens of major clinical importance. The Old World (OW) arenavirus Lassa virus is endemic in West Africa and is estimated to cause up to 300,000 infections each year. The New World (NW) arenaviruses Junín and Machupo periodically cause hemorrhagic fever outbreaks in South America. While these arenaviruses are highly pathogenic in humans, recent evidence indicates that pathogenic OW and NW arenaviruses interact with the host immune system differently, which may have differential impacts on viral pathogenesis. Severe Lassa fever cases are characterized by profound immunosuppression. In contrast, pathogenic NW arenavirus infections are accompanied by elevated levels of Type I interferon and pro-inflammatory cytokines. This review aims to summarize recent findings about interactions of these pathogenic arenaviruses with the innate immune machinery and the subsequent effects on adaptive immunity, which may inform the development of vaccines and therapeutics against arenavirus infections.
Collapse
|
22
|
Hottz ED, Bozza FA, Bozza PT. Platelets in Immune Response to Virus and Immunopathology of Viral Infections. Front Med (Lausanne) 2018; 5:121. [PMID: 29761104 PMCID: PMC5936789 DOI: 10.3389/fmed.2018.00121] [Citation(s) in RCA: 134] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 04/12/2018] [Indexed: 01/04/2023] Open
Abstract
Platelets are essential effector cells in hemostasis. Aside from their role in coagulation, platelets are now recognized as major inflammatory cells with key roles in the innate and adaptive arms of the immune system. Activated platelets have key thromboinflammatory functions linking coagulation to immune responses in various infections, including in response to virus. Recent studies have revealed that platelets exhibit several pattern recognition receptors (PRR) including those from the toll-like receptor, NOD-like receptor, and C-type lectin receptor family and are first-line sentinels in detecting and responding to pathogens in the vasculature. Here, we review the main mechanisms of platelets interaction with viruses, including their ability to sustain viral infection and replication, their expression of specialized PRR, and activation of thromboinflammatory responses against viruses. Finally, we discuss the role of platelet-derived mediators and platelet interaction with vascular and immune cells in protective and pathophysiologic responses to dengue, influenza, and human immunodeficiency virus 1 infections.
Collapse
Affiliation(s)
- Eugenio D Hottz
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Departamento de Bioquimica, Universidade Federal de Juiz de Fora, Juiz de Fora, Brazil
| | - Fernando A Bozza
- Laboratório de Medicina Intensiva, Instituto Nacional de Infectologia Evandro Chagas, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil.,Instituto D'Or de Pesquisa e Ensino, Rio de Janeiro, Brazil
| | - Patrícia T Bozza
- Laboratório de Imunofarmacologia, Instituto Oswaldo Cruz, Fundação Oswaldo Cruz, Rio de Janeiro, Brazil
| |
Collapse
|
23
|
Highly Pathogenic New World Arenavirus Infection Activates the Pattern Recognition Receptor Protein Kinase R without Attenuating Virus Replication in Human Cells. J Virol 2017; 91:JVI.01090-17. [PMID: 28794024 DOI: 10.1128/jvi.01090-17] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023] Open
Abstract
The arenavirus family consists of several highly pathogenic viruses, including the Old World (OW) arenavirus Lassa fever virus (LASV) and the New World (NW) Junin virus (JUNV) and Machupo virus (MACV). Host response to infection by these pathogenic arenaviruses is distinct in many aspects. JUNV and MACV infections readily induce an interferon (IFN) response in human cells, while LASV infection usually triggers an undetectable or weak IFN response. JUNV induces an IFN response through RIG-I, suggesting that the host non-self RNA sensor readily detects JUNV viral RNAs (vRNAs) during infection and activates IFN response. Double-stranded-RNA (dsRNA)-activated protein kinase R (PKR) is another host non-self RNA sensor classically known for its vRNA recognition activity. Here we report that infection with NW arenaviruses JUNV and MACV, but not OW LASV, activated PKR, concomitant with elevated phosphorylation of the translation initiation factor α subunit of eukaryotic initiation factor 2 (eIF2α). Host protein synthesis was substantially suppressed in MACV- and JUNV-infected cells but was only marginally reduced in LASV-infected cells. Despite the antiviral activity known for PKR against many other viruses, the replication of JUNV and MACV was not impaired but was slightly augmented in wild-type (wt) cells compared to that in PKR-deficient cells, suggesting that PKR or PKR activation did not negatively affect JUNV and MACV infection. Additionally, we found an enhanced IFN response in JUNV- or MACV-infected PKR-deficient cells, which was inversely correlated with virus replication.IMPORTANCE The detection of viral RNA by host non-self RNA sensors, including RIG-I and MDA5, is critical to the initiation of the innate immune response to RNA virus infection. Among pathogenic arenaviruses, the OW LASV usually does not elicit an interferon response. However, the NW arenaviruses JUNV and MACV readily trigger an IFN response in a RIG-I-dependent manner. Here, we demonstrate for the first time that pathogenic NW arenaviruses JUNV and MACV, but not the OW arenavirus LASV, activated the dsRNA-dependent PKR, another host non-self RNA sensor, during infection. Interestingly, the replication of JUNV and MACV was not restricted but was rather slightly augmented in the presence of PKR. Our data provide new evidence for a distinct interplay between host non-self RNA sensors and pathogenic arenaviruses, which also provides insights into the pathogenesis of arenaviruses and may facilitate the design of vaccines and treatments against arenavirus-caused diseases.
Collapse
|
24
|
Abstract
Hemorrhagic fevers caused by viruses were identified in the late 1950s in South America. These viruses have existed in their hosts, the New World rodents, for millions of years. Their emergence as infectious agents in humans coincided with changes in the environment and farming practices that caused explosions in their host rodent populations. Zoonosis into humans likely occurs because the pathogenic New World arenaviruses use human transferrin receptor 1 to enter cells. The mortality rate after infection with these viruses is high, but the mechanism by which disease is induced is still not clear. Possibilities include direct effects of cellular infection or the induction of high levels of cytokines by infected sentinel cells of the immune system, leading to endothelia and thrombocyte dysfunction and neurological disease. Here we provide a review of the ecology and molecular and cellular biology of New World arenaviruses, as well as a discussion of the current animal models of infection. The development of animal models, coupled with an improved understanding of the infection pathway and host response, should lead to the discovery of new drugs for treating infections.
Collapse
Affiliation(s)
- Nicolás Sarute
- Department of Microbiology and Immunology, University of Illinois College of Medicine at Chicago, Chicago, Illinois 60612; ,
| | - Susan R Ross
- Department of Microbiology and Immunology, University of Illinois College of Medicine at Chicago, Chicago, Illinois 60612; ,
| |
Collapse
|
25
|
Abstract
Platelets have various roles in vascular biology and homeostasis. They are the first actor in primary haemostasis and play important roles in thrombosis pathogenesis, but they are also part of innate immunity, which initiates and accelerate many inflammatory conditions. In some contexts, their immune functions are protective, while in others they contribute to adverse inflammatory outcomes. Platelets express numerous receptors and contain hundreds of secretory molecules that are crucial for platelet functional responses. The capacity of platelets to produce and secrete cytokines, chemokines and related molecules, under the control of specific intracellular pathways, is intimately related to their key role in inflammation. They are also able to intervene in tissue regeneration and repair because they produce pro-angiogenic mediators. Due to this characteristic platelets are involved in cancer progression and spreading. In this review we discuss the complex role of platelets, which bridges haemostasis, inflammation and immune response both in physiological and pathological conditions.
Collapse
Affiliation(s)
- Maria Elisa Mancuso
- Angelo Bianchi Bonomi Haemophilia and Thrombosis Centre, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| | - Elena Santagostino
- Angelo Bianchi Bonomi Haemophilia and Thrombosis Centre, Fondazione IRCCS Ca' Granda, Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
26
|
Thachil J. Platelets and infections in the resource-limited countries with a focus on malaria and viral haemorrhagic fevers. Br J Haematol 2017; 177:960-970. [PMID: 28295179 DOI: 10.1111/bjh.14582] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Infections continue to cause a high incidence of mortality and morbidity in resource-poor nations. Although antimicrobial therapy has aided mostly in dealing with the pathogenic micro-organisms themselves, the collateral damage caused by the infections continue to cause many deaths. Intensive care support and manipulation of the hosts' abnormal response to the infection have helped to improve mortality in well-resourced countries. But, in those areas with limited resources, this is not yet the case and simpler methods of diagnosis and interventions are required. Thrombocytopenia is one of the most common manifestations in all these infections and may be used as an easily available prognostic indicator and marker for the severity of the infections. In this review, the relevance of platelets in infections in general, and specifically to tropical infections, malaria, and viral haemorrhagic fevers in the emerging countries is discussed. Better understanding of the pathophysiology and the role of platelets in particular in such conditions is likely to translate into better patient care and thus reduce morbidity and mortality.
Collapse
Affiliation(s)
- Jecko Thachil
- Department of Haematology, Manchester Royal Infirmary, Manchester, UK
| |
Collapse
|
27
|
Oliveira RADS, Silva MMCD, Calzavara-Silva CE, Silva AM, Cordeiro MT, Moura PMMFD, Baptista PN, Marques ETDA, Gil LHVG. Primary dengue haemorrhagic fever in patients from northeast of Brazil is associated with high levels of interferon-β during acute phase. Mem Inst Oswaldo Cruz 2017; 111:378-84. [PMID: 27223651 PMCID: PMC4909036 DOI: 10.1590/0074-02760150453] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2015] [Accepted: 04/29/2016] [Indexed: 01/05/2023] Open
Abstract
Dengue is an acute febrile disease caused by the mosquito-borne dengue virus (DENV) that according to clinical manifestations can be classified as asymptomatic, mild or severe dengue. Severe dengue cases have been associated with an unbalanced immune response characterised by an over secretion of inflammatory cytokines. In the present study we measured type I interferon (IFN-I) transcript and circulating levels in primary and secondary DENV infected patients. We observed that dengue fever (DF) and dengue haemorrhagic fever (DHF) patients express IFN-I differently. While DF and DHF patients express interferon-α similarly (52,71 ± 7,40 and 49,05 ± 7,70, respectively), IFN- β were associated with primary DHF patients. On the other hand, secondary DHF patients were not able to secrete large amounts of IFN- β which in turn may have influenced the high-level of viraemia. Our results suggest that, in patients from our cohort, infection by DENV serotype 3 elicits an innate response characterised by higher levels of IFN- β in the DHF patients with primary infection, which could contribute to control infection evidenced by the low-level of viraemia in these patients. The present findings may contribute to shed light in the role of innate immune response in dengue pathogenesis.
Collapse
Affiliation(s)
- Renato Antônio Dos Santos Oliveira
- Fundação Oswaldo Cruz, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife PE , Brasil, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife, PE, Brasil
| | - Mayara Marques Carneiro da Silva
- Fundação Oswaldo Cruz, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife PE , Brasil, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife, PE, Brasil
| | - Carlos Eduardo Calzavara-Silva
- Fundação Oswaldo Cruz, Fundação Oswaldo Cruz, Centro de Pesquisas René Rachou, Departamento de Imunologia, Laboratório de Imunologia Celular e Molecular, Belo Horizonte MG , Brasil, Fundação Oswaldo Cruz, Centro de Pesquisas René Rachou, Departamento de Imunologia, Laboratório de Imunologia Celular e Molecular, Belo Horizonte, MG, Brasil
| | - Ana Maria Silva
- Fundação Oswaldo Cruz, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife PE , Brasil, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife, PE, Brasil
| | - Marli Tenório Cordeiro
- Fundação Oswaldo Cruz, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife PE , Brasil, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife, PE, Brasil
| | - Patrícia Muniz Mendes Freire de Moura
- Universidade de Pernambuco, Universidade de Pernambuco, Instituto de Ciências Biológicas, Recife PE , Brasil, Universidade de Pernambuco, Instituto de Ciências Biológicas, Recife, PE, Brasil
| | - Paulo Neves Baptista
- Universidade de Pernambuco, Universidade de Pernambuco, Faculdade de Ciências Médicas, Recife PE , Brasil, Universidade de Pernambuco, Faculdade de Ciências Médicas, Recife, PE, Brasil
| | - Ernesto Torres de Azevedo Marques
- Fundação Oswaldo Cruz, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife PE , Brasil, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife, PE, Brasil.,University of Pittsburgh, Center for Vaccine Research, Department of Infectious Diseases and Microbiology, Pittsburgh PA , US, University of Pittsburgh, Center for Vaccine Research, Department of Infectious Diseases and Microbiology, Pittsburgh, PA, US
| | - Laura Helena Vega Gonzales Gil
- Fundação Oswaldo Cruz, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife PE , Brasil, Fundação Oswaldo Cruz, Centro de Pesquisas Aggeu Magalhães, Departamento de Virologia, Recife, PE, Brasil
| |
Collapse
|
28
|
Viringipurampeer IA, Metcalfe AL, Bashar AE, Sivak O, Yanai A, Mohammadi Z, Moritz OL, Gregory-Evans CY, Gregory-Evans K. NLRP3 inflammasome activation drives bystander cone photoreceptor cell death in a P23H rhodopsin model of retinal degeneration. Hum Mol Genet 2016; 25:1501-16. [PMID: 27008885 DOI: 10.1093/hmg/ddw029] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 02/01/2016] [Indexed: 12/11/2022] Open
Abstract
The molecular signaling leading to cell death in hereditary neurological diseases such as retinal degeneration is incompletely understood. Previous neuroprotective studies have focused on apoptotic pathways; however, incomplete suppression of cell death with apoptosis inhibitors suggests that other mechanisms are at play. Here, we report that different signaling pathways are activated in rod and cone photoreceptors in the P23H rhodopsin mutant rat, a model representing one of the commonest forms of retinal degeneration. Up-regulation of the RIP1/RIP3/DRP1 axis and markedly improved survival with necrostatin-1 treatment highlighted necroptosis as a major cell-death pathway in degenerating rod photoreceptors. Conversely, up-regulation of NLRP3 and caspase-1, expression of mature IL-1β and IL-18 and improved cell survival with N-acetylcysteine treatment suggested that inflammasome activation and pyroptosis was the major cause of cone cell death. This was confirmed by generation of the P23H mutation on an Nlrp3-deficient background, which preserved cone viability. Furthermore, Brilliant Blue G treatment inhibited inflammasome activation, indicating that the 'bystander cell death' phenomenon was mediated through the P2RX7 cell-surface receptor. Here, we identify a new pathway in cones for bystander cell death, a phenomenon important in development and disease in many biological systems. In other retinal degeneration models different cell-death pathways are activated, which suggests that the particular pathways that are triggered are to some extent genotype-specific. This also implies that neuroprotective strategies to limit retinal degeneration need to be customized; thus, different combinations of inhibitors will be needed to target the specific pathways in any given disease.
Collapse
Affiliation(s)
- Ishaq A Viringipurampeer
- Department of Ophthalmology and Visual Sciences, University of British Columbia, 2550 Willow Street, Vancouver, BC, Canada V5Z 3N9
| | - Andrew L Metcalfe
- Department of Ophthalmology and Visual Sciences, University of British Columbia, 2550 Willow Street, Vancouver, BC, Canada V5Z 3N9
| | - Abu E Bashar
- Department of Ophthalmology and Visual Sciences, University of British Columbia, 2550 Willow Street, Vancouver, BC, Canada V5Z 3N9
| | - Olena Sivak
- Department of Ophthalmology and Visual Sciences, University of British Columbia, 2550 Willow Street, Vancouver, BC, Canada V5Z 3N9
| | - Anat Yanai
- Department of Ophthalmology and Visual Sciences, University of British Columbia, 2550 Willow Street, Vancouver, BC, Canada V5Z 3N9
| | - Zeinabsadat Mohammadi
- Department of Ophthalmology and Visual Sciences, University of British Columbia, 2550 Willow Street, Vancouver, BC, Canada V5Z 3N9
| | - Orson L Moritz
- Department of Ophthalmology and Visual Sciences, University of British Columbia, 2550 Willow Street, Vancouver, BC, Canada V5Z 3N9
| | - Cheryl Y Gregory-Evans
- Department of Ophthalmology and Visual Sciences, University of British Columbia, 2550 Willow Street, Vancouver, BC, Canada V5Z 3N9
| | - Kevin Gregory-Evans
- Department of Ophthalmology and Visual Sciences, University of British Columbia, 2550 Willow Street, Vancouver, BC, Canada V5Z 3N9
| |
Collapse
|
29
|
CCL5 derived from platelets increases megakaryocyte proplatelet formation. Blood 2015; 127:921-6. [PMID: 26647394 DOI: 10.1182/blood-2015-05-644583] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2015] [Accepted: 11/25/2015] [Indexed: 02/07/2023] Open
Abstract
In times of physiological stress, platelet count can transiently rise. What initiates this reactive thrombocytosis is poorly understood. Intriguingly, we found that treating megakaryocytes (MKs) with the releasate from activated platelets increased proplatelet production by 47%. Platelets store inflammatory cytokines, including the chemokine ligand 5 (CCL5, RANTES); after TRAP activation, platelets release over 25 ng/mL CCL5. We hypothesized that CCL5 could regulate platelet production by binding to its receptor, CCR5, on MKs. Maraviroc (CCR5 antagonist) or CCL5 immunodepletion diminished 95% and 70% of the effect of platelet releasate, respectively, suggesting CCL5 derived from platelets is sufficient to drive increased platelet production through MK CCR5. MKs cultured with recombinant CCL5 increased proplatelet production by 50% and had significantly higher ploidy. Pretreating the MK cultures with maraviroc prior to exposure to CCL5 reversed the augmented proplatelet formation and ploidy, suggesting that CCL5 increases MK ploidy and proplatelet formation in a CCR5-dependent manner. Interrogation of the Akt signaling pathway suggested that CCL5/CCR5 may influence proplatelet production by suppressing apoptosis. In an in vivo murine acute colitis model, platelet count significantly correlated with inflammation whereas maraviroc treatment abolished this correlation. We propose that CCL5 signaling through CCR5 may increase platelet counts during physiological stress.
Collapse
|
30
|
Superinfection exclusion is absent during acute Junin virus infection of Vero and A549 cells. Sci Rep 2015; 5:15990. [PMID: 26549784 PMCID: PMC4637830 DOI: 10.1038/srep15990] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 10/07/2015] [Indexed: 11/08/2022] Open
Abstract
Many viruses have evolved strategies of so-called "superinfection exclusion" to prevent re-infection of a cell that the same virus has already infected. Although Old World arenavirus infection results in down-regulation of its viral receptor and thus superinfection exclusion, whether New World arenaviruses have evolved such a mechanism remains unclear. Here we show that acute infection by the New World Junin virus (JUNV) failed to down-regulate the transferrin receptor and did not induce superinfection exclusion. We observed that Vero cells infected by a first round of JUNV (Candid1 strain) preserve an ability to internalize new incoming JUNV particles that is comparable to that of non-infected cells. Moreover, we developed a dual infection assay with the wild-type Candid1 JUNV and a recombinant JUNV-GFP virus to discriminate between first and second infections at the transcriptional and translational levels. We found that Vero and A549 cells already infected by JUNV were fully competent to transcribe viral RNA from a second round of infection. Furthermore, flow cytometry analysis of viral protein expression indicated that viral translation was normal, regardless of whether cells were previously infected or not. We conclude that in acutely infected cells, Junin virus lacks a superinfection exclusion mechanism.
Collapse
|
31
|
Rivadeneyra L, Pozner RG, Meiss R, Fondevila C, Gómez RM, Schattner M. Poly (I:C) downregulates platelet production and function through type I interferon. Thromb Haemost 2015; 114:982-93. [PMID: 26134179 DOI: 10.1160/th14-11-0951] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Accepted: 05/24/2015] [Indexed: 12/21/2022]
Abstract
Thrombocytopenia is a frequent complication of viral infections; the underlying mechanisms appear to depend on the identity of the virus involved. Previous research, including reports from our group, indicates that as well as having antiviral activity type I interferons (IFN I) selectively downregulate platelet production. In this study we extended understanding of the role of endogenous IFN I in megakaryo/thrombopoiesis by evaluating platelet and megakaryocyte physiology in mice treated with polyinosinic:polycytidylic acid [poly (I:C)], a synthetic analogue of double-stranded RNA, Toll-like receptor-3 ligand and strong IFNβ inducer. Mice-treated with poly (I:C) showed thrombocytopaenia, an increase in mean platelet volume and abnormal haemostatic and inflammatory platelet-mediated functionality, indicated by decreased fibrinogen binding and platelet adhesion, prolonged tail bleeding times and impaired P-Selectin externalisation, RANTES release and thrombin-induced platelet-neutrophil aggregate formation. These changes were associated with an increase in size and an abnormal distribution of bone marrow megakaryocytes within the vascular niche and were directly correlated with the plasmatic and bone marrow IFNβ levels. All these effects were absent in genetically modified mice lacking the IFN I receptor. Our results suggest that IFN I is the central mediator of poly (I:C)-induced thrombocytopenia and platelet dysfunction and indicate that these abnormalities are due to changes in the last stages of megakaryocyte development. These data provide new evidence for the role of IFN I in megakaryocyte distribution in the bone marrow niches and its influence on thrombopoiesis and haemostasis.
Collapse
Affiliation(s)
| | | | | | | | | | - Mirta Schattner
- Mirta Schattner, Institute of Experimental Medicine, Pacheco de Melo 3081, Buenos Aires 1425, Argentina, Tel.: +5411 48073926, Fax: +5411 48050712,
| |
Collapse
|
32
|
D'Atri LP, Etulain J, Rivadeneyra L, Lapponi MJ, Centurion M, Cheng K, Yin H, Schattner M. Expression and functionality of Toll-like receptor 3 in the megakaryocytic lineage. J Thromb Haemost 2015; 13:839-50. [PMID: 25594115 PMCID: PMC4424134 DOI: 10.1111/jth.12842] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 01/04/2015] [Indexed: 02/04/2023]
Abstract
BACKGROUND In addition to their key role in hemostasis, platelets and megakaryocytes regulate immune and inflammatory responses, in part through their expression of Toll-like receptors (TLRs). Among the TLRs, TLR3 recognizes dsRNA associated with viral infection. Thrombocytopenia is a frequent complication of viral infection. However, the expression and functionality of TLR3 in megakaryocytes and platelets is not yet well understood. OBJECTIVE To study the expression and functionality of TLR3 in the megakaryocytic lineage. METHODS AND RESULTS RT-PCR, flow cytometric and immunofluorescence assays showed that TLR3 is expressed in CD34(+) cells, megakaryocytes, and platelets. Immunoblotting assays showed that stimulation of megakaryocytes with two synthetic agonists of TLR3, Poly(I:C) and Poly(A:U), activated the nuclear factor-κB (NF-κB), phosphoinositide 3-kinase (PI3K)/Akt, extracellular signal-related kinase (ERK)1/2 and p38 pathways. TLR3-megakaryocyte activation resulted in reduced platelet production in vitro and interferon-β release through the PI3K-Akt and NF-κB signaling pathways. TLR3 ligands potentiated the aggregation mediated by classic platelet agonists. This effect was also observed for ATP release, but not for P-selectin or CD40L membrane exposure, indicating that TLR3 activation was not involved in α-granule release. In addition, TLR3 agonists induced activation of the NF-κB, PI3K-Akt and ERK1/2 pathways in platelets. Reductions in platelet production and platelet fibrinogen binding mediated by Poly(I:C) or Poly(A:U) were prevented by the presence of an inhibitor of the TLR3-dsRNA complex. CONCLUSIONS Our findings indicate that functional TLR3 is expressed in CD34(+) cells, megakaryocytes, and platelets, and suggest a potential role for this receptor in the megakaryopoiesis/thrombopoiesis alterations that occur in viral infections.
Collapse
Affiliation(s)
- L P D'Atri
- Laboratory of Experimental Thrombosis, Institute of Experimental Medicine, CONICET-National Academy of Medicine, Buenos Aires, Argentina
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Highly Pathogenic New World and Old World Human Arenaviruses Induce Distinct Interferon Responses in Human Cells. J Virol 2015; 89:7079-88. [PMID: 25926656 DOI: 10.1128/jvi.00526-15] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2015] [Accepted: 04/21/2015] [Indexed: 12/20/2022] Open
Abstract
UNLABELLED The arenavirus family includes several important pathogens that cause severe and sometimes fatal diseases in humans. The highly pathogenic Old World (OW) arenavirus Lassa fever virus (LASV) is the causative agent of Lassa fever (LF) disease in humans. LASV infections in severe cases are generally immunosuppressive without stimulating interferon (IFN) induction, a proinflammatory response, or T cell activation. However, the host innate immune responses to highly pathogenic New World (NW) arenaviruses are not well understood. We have previously shown that the highly pathogenic NW arenavirus, Junin virus (JUNV), induced an IFN response in human A549 cells. Here, we report that Machupo virus (MACV), another highly pathogenic NW arenavirus, also induces an IFN response. Importantly, both pathogenic NW arenaviruses, in contrast to the OW highly pathogenic arenavirus LASV, readily elicited an IFN response in human primary dendritic cells and A549 cells. Coinfection experiments revealed that LASV could potently inhibit MACV-activated IFN responses even at 6 h after MACV infection, while the replication levels of MACV and LASV were not affected by virus coinfection. Our results clearly demonstrated that although all viruses studied herein are highly pathogenic to humans, the host IFN responses toward infections with the NW arenaviruses JUNV and MACV are quite different from responses to infections with the OW arenavirus LASV, a discovery that needs to be further investigated in relevant animal models. This finding might help us better understand various interplays between the host immune system and highly pathogenic arenaviruses as well as distinct mechanisms underlying viral pathogenesis. IMPORTANCE Infections of humans with the highly pathogenic OW LASV are accompanied by potent suppression of interferon or proinflammatory cytokine production. In contrast, infections with the highly pathogenic NW arenavirus JUNV are associated with high levels of IFNs and cytokines in severe and fatal cases. Arenaviruses initially target macrophages and dendritic cells, which are potent IFN/cytokine-producers. In human macrophages, JUNV reportedly does not trigger IFN responses. We here demonstrated that JUNV activated IFN responses in human dendritic cells. MACV, another highly pathogenic NW arenavirus, also activated IFN responses. LASV did not induce detectable IFN responses, in spite of higher replication levels, and blocked the MACV-triggered IFN response in a coinfection assay. Although these viruses are highly pathogenic to humans, our study highlights distinct innate immune responses to infections with the NW arenaviruses JUNV and MACV and to infection with the OW arenavirus LASV and provides important insights into the virus-host interaction and pathogenesis.
Collapse
|
34
|
Human Plasmacytoid Dendritic Cells Elicited Different Responses after Infection with Pathogenic and Nonpathogenic Junin Virus Strains. J Virol 2015; 89:7409-13. [PMID: 25926646 DOI: 10.1128/jvi.01014-15] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2015] [Accepted: 04/22/2015] [Indexed: 12/24/2022] Open
Abstract
The arenavirus Junin virus (JUNV) is the etiologic agent of Argentine hemorrhagic fever. We characterized the JUNV infection of human peripheral blood-derived plasmacytoid dendritic cells (hpDC), demonstrating that hpDC are susceptible to infection with the C#1 strain (attenuated) and even more susceptible to infection with the P (virulent) JUNV strain. However, hpDC elicited different responses in terms of viability, activation, maturation, and cytokine expression after infection with both JUNV strains.
Collapse
|
35
|
Chabert A, Hamzeh-Cognasse H, Pozzetto B, Cognasse F, Schattner M, Gomez RM, Garraud O. Human platelets and their capacity of binding viruses: meaning and challenges? BMC Immunol 2015; 16:26. [PMID: 25913718 PMCID: PMC4411926 DOI: 10.1186/s12865-015-0092-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 04/03/2015] [Indexed: 01/16/2023] Open
Abstract
Blood platelets are first aimed at ensuring primary hemostasis. Beyond this role, they have been acknowledged as having functions in the maintenance of the vascular arborescence and, more recently, as being also innate immune cells, devoted notably to the detection of danger signals, of which infectious ones. Platelets express pathogen recognition receptors that can sense bacterial and viral moieties. Besides, several molecules that bind epithelial or sub-endothelial molecules and, so forth, are involved in hemostasis, happen to be able to ligate viral determinants, making platelets capable of either binding viruses or even to be infected by some of them. Further, as platelets express both Fc-receptors for Ig and complement receptors, they also bind occasionally virus-Ig or virus-Ig-complement immune complexes. Interplays of viruses with platelets are very complex and viral infections often interfere with platelet number and functions. Through a few instances of viral infections, the present review aims at presenting some of the most important interactions from pathophysiological and clinical points of view, which are observed between human viruses and platelets.
Collapse
Affiliation(s)
- Adrien Chabert
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France.
| | | | - Bruno Pozzetto
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France. .,Service des Agents infectieux et d'Hygiène, CHU de Saint-Etienne, 42055, Saint-Etienne, France.
| | - Fabrice Cognasse
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France. .,EFS Auvergne-Loire, 42023, Saint-Etienne, France.
| | - Mirta Schattner
- Laboratorio de Trombosis Experimental, Instituto de Medicina Experimental, ANM-CONICET, Buenos Aires, Argentina.
| | - Ricardo M Gomez
- Laboratorio de Virus Animales, Instituto de Biotecnología y Biología Molecular, UNLP-CONICET, La Plata, Argentina.
| | - Olivier Garraud
- EA3064-GIMAP, Université de Lyon, 42023, Saint-Etienne, France. .,Institut National de la Transfusion Sanguine, 75015, Paris, France. .,INTS, 6 rue Alexandre-Cabanel, 75015, Paris, France.
| |
Collapse
|
36
|
Fan H, Zhao Y, Zhu JH, Song FC, Ye JH, Wang ZY, Le JW. Thrombocytopenia as a predictor of severe acute kidney injury in patients with heat stroke. Ren Fail 2015; 37:877-81. [PMID: 25774629 DOI: 10.3109/0886022x.2015.1022851] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023] Open
Abstract
BACKGROUND Abnormalities of blood system often occur several days before acute kidney injury (AKI) in patients with heat stroke (HS). We aimed to investigate the prevalence and prognostic value of the early hematological markers in patients with AKI induced by HS. METHODS In a retrospective cohort study, we analyzed the case records of 176 patients with HS and evaluated the hematological markers for early prediction and risk classification in the patients with AKI. RESULTS Of 176, 103 (58%) HS cases developed AKI, and men comprised more than half (75%) of the sample population. The nadir platelet count significantly correlated with the levels of peak serum creatinine (r = -0.608, p < 0.01) and blood urea nitrogen (r = -0.546, p < 0.01), and the length of hospital stay (r = -0.393, p < 0.01). The areas under the receiver operating characteristic curves (AU-ROC) indicated the prognostic accuracy of hematological markers, AU-ROC was significantly higher with the nadir platelet count than that with the admission platelet count (AU-ROC of the nadir platelet: 0.73; 95% CI: 0.67-0.82; vs. AU-ROC of the admission platelet: 0.67; 95% CI: 0.59-0.75; p < 0.01). Multiple logistic regression results indicated that the nadir platelet count (adjusted ORs: 37.92; 95% CI: 2.18-87.21; p < 0.01) was independent predictor of AKI in HS. CONCLUSION The high mortality observed in HS complicated with AKI, and among the various hematological parameters assessed, thrombocytopenia is associated with AKI induced by HS independently.
Collapse
Affiliation(s)
- Heng Fan
- a Department of Intensive Care Unit , Ningbo First Hospital , Ningbo , China
| | | | | | | | | | | | | |
Collapse
|
37
|
Connolly-Andersen AM, Sundberg E, Ahlm C, Hultdin J, Baudin M, Larsson J, Dunne E, Kenny D, Lindahl TL, Ramström S, Nilsson S. Increased Thrombopoiesis and Platelet Activation in Hantavirus-Infected Patients. J Infect Dis 2015; 212:1061-9. [PMID: 25762786 DOI: 10.1093/infdis/jiv161] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/06/2015] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Thrombocytopenia is a common finding during viral hemorrhagic fever, which includes hemorrhagic fever with renal syndrome (HFRS). The 2 main causes for thrombocytopenia are impaired thrombopoiesis and/or increased peripheral destruction of platelets. In addition, there is an increased intravascular coagulation risk during HFRS, which could be due to platelet activation. METHODS Thrombopoiesis was determined by quantification of platelet counts, thrombopoietin, immature platelet fraction, and mean platelet volume during HFRS. The in vivo platelet activation was determined by quantification of soluble P-selectin (sP-selectin) and glycoprotein VI (sGPVI). The function of circulating platelets was determined by ex vivo stimulation followed by flow cytometry analysis of platelet surface-bound fibrinogen and P-selectin exposure. Intravascular coagulation during disease was determined by scoring for disseminated intravascular coagulation (DIC) and recording thromboembolic complications. RESULTS The levels of thrombopoietin, immature platelet fraction, and mean platelet volume all indicate increased thrombopoiesis during HFRS. Circulating platelets had reduced ex vivo function during disease compared to follow-up. Most interestingly, we observed significantly increased in vivo platelet activation in HFRS patients with intravascular coagulation (DIC and thromboembolic complications) as shown by sP-selectin and sGPVI levels. CONCLUSIONS HFRS patients have increased thrombopoiesis and platelet activation, which contributes to intravascular coagulation.
Collapse
Affiliation(s)
| | - Erik Sundberg
- Infectious Diseases, Department of Clinical Microbiology Clinical Chemistry, Department of Medical Biosciences, Umeå University, Sweden
| | - Clas Ahlm
- Infectious Diseases, Department of Clinical Microbiology
| | - Johan Hultdin
- Clinical Chemistry, Department of Medical Biosciences, Umeå University, Sweden
| | - Maria Baudin
- Infectious Diseases, Department of Clinical Microbiology
| | - Johanna Larsson
- Clinical Chemistry, Department of Medical Biosciences, Umeå University, Sweden
| | - Eimear Dunne
- Clinical Research Centre, Royal College of Surgeons in Ireland, Dublin
| | - Dermot Kenny
- Clinical Research Centre, Royal College of Surgeons in Ireland, Dublin
| | - Tomas L Lindahl
- Department of Clinical and Experimental Medicine, Linköping University, Sweden
| | - Sofia Ramström
- Department of Clinical and Experimental Medicine, Linköping University, Sweden
| | - Sofie Nilsson
- Clinical Chemistry, Department of Medical Biosciences, Umeå University, Sweden
| |
Collapse
|
38
|
Assinger A. Platelets and infection - an emerging role of platelets in viral infection. Front Immunol 2014; 5:649. [PMID: 25566260 PMCID: PMC4270245 DOI: 10.3389/fimmu.2014.00649] [Citation(s) in RCA: 252] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2014] [Accepted: 12/05/2014] [Indexed: 12/23/2022] Open
Abstract
Platelets are anucleate blood cells that play a crucial role in the maintenance of hemostasis. While platelet activation and elevated platelet counts (thrombocytosis) are associated with increased risk of thrombotic complications, low platelet counts (thrombocytopenia) and several platelet function disorders increase the risk of bleeding. Over the last years, more and more evidence has emerged that platelets and their activation state can also modulate innate and adaptive immune responses and low platelet counts have been identified as a surrogate marker for poor prognosis in septic patients. Viral infections often coincide with platelet activation. Host inflammatory responses result in the release of platelet activating mediators and a pro-oxidative and pro-coagulant environment, which favors platelet activation. However, viruses can also directly interact with platelets and megakaryocytes and modulate their function. Furthermore, platelets can be activated by viral antigen-antibody complexes and in response to some viruses B-lymphocytes also generate anti-platelet antibodies. All these processes contributing to platelet activation result in increased platelet consumption and removal and often lead to thrombocytopenia, which is frequently observed during viral infection. However, virus-induced platelet activation does not only modulate platelet count but also shape immune responses. Platelets and their released products have been reported to directly and indirectly suppress infection and to support virus persistence in response to certain viruses, making platelets a double-edged sword during viral infections. This review aims to summarize the current knowledge on platelet interaction with different types of viruses, the viral impact on platelet activation, and platelet-mediated modulations of innate and adaptive immune responses.
Collapse
Affiliation(s)
- Alice Assinger
- Department of Physiology and Pharmacology, Medical University of Vienna , Vienna , Austria ; Department of Medicine, Center for Molecular Medicine, Karolinska University Hospital , Stockholm , Sweden
| |
Collapse
|
39
|
Abstract
Viral hemorrhagic fevers (VHF) are acute zoonotic diseases that, early on, seem to cause platelet destruction or dysfunction. Here we present the four major ways viruses affect platelet development and function and new evidence of molecular factors that are preferentially induced by the more pathogenic members of the families Flaviviridae, Bunyaviridae, Arenaviridae, and Filoviridae. A systematic search was performed through the main medical electronic databases using as parameters all current findings concerning platelets in VHF. Additionally, the review contains information from conference proceedings.
Collapse
Affiliation(s)
- Juan C. Zapata
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
- * E-mail:
| | - Dermot Cox
- Molecular and Cellular Therapeutics School of Pharmacy, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Maria S. Salvato
- Institute of Human Virology, University of Maryland School of Medicine, Baltimore, Maryland, United States of America
| |
Collapse
|
40
|
Huang C, Walker AG, Grant AM, Kolokoltsova OA, Yun NE, Seregin AV, Paessler S. Potent inhibition of Junín virus infection by interferon in murine cells. PLoS Negl Trop Dis 2014; 8:e2933. [PMID: 24901990 PMCID: PMC4046933 DOI: 10.1371/journal.pntd.0002933] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 04/28/2014] [Indexed: 11/18/2022] Open
Abstract
The new world arenavirus Junín virus (JUNV) is the causative agent of Argentine hemorrhagic fever, a lethal human infectious disease. Adult laboratory mice are generally resistant to peripheral infection by JUNV. The mechanism underlying the mouse resistance to JUNV infection is largely unknown. We have reported that interferon receptor knockout mice succumb to JUNV infection, indicating the critical role of interferon in restricting JUNV infection in mice. Here we report that the pathogenic and vaccine strains of JUNV were highly sensitive to interferon in murine primary cells. Treatment with low concentrations of interferon abrogated viral NP protein expression in murine cells. The replication of both JUNVs was enhanced in IRF3/IRF7 deficient cells. In addition, the vaccine strain of JUNV displayed impaired growth in primary murine cells. Our data suggested a direct and potent role of host interferon response in restricting JUNV replication in mice. The defect in viral growth for vaccine JUNV might also partially explain its attenuation in mice. The new world arenavirus Junín virus (JUNV) is the causative agent of a lethal human infectious disease, Argentine hemorrhagic fever. Laboratory mice are used as models to study many viral diseases. However, adult laboratory mice are generally resistant to JUNV infection. Interferons are early immune regulatory molecules that induce potent anti-viral status in host cells and activate host immune cells to counteract virus infection. The activity of interferons relies on their cell surface receptors. We have previously reported that mutant mice with defect in interferon receptors succumbed to challenge with JUNV, highlighting the critical role of interferon in restricting JUNV infection in mice. Here we further study the basis of mouse resistance to JUNV infection and report that the replication of both pathogenic JUNV and its vaccine strains are highly sensitive to type I IFN treatment in mouse cells. However, both strains replicate efficiently in Africa green monkey-derived Vero cells and human cells when treated with high doses of interferon. Additionally, the vaccine strain replicates less efficiently in mouse cells compared with the pathogenic strain, which might partially explain its attenuation in mice. Our new findings help better understand the JUNV-host interaction.
Collapse
Affiliation(s)
- Cheng Huang
- Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Aida G. Walker
- Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Ashley M. Grant
- Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Olga A. Kolokoltsova
- Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nadezhda E. Yun
- Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Alexey V. Seregin
- Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Slobodan Paessler
- Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail:
| |
Collapse
|
41
|
Type I interferon is a therapeutic target for virus-induced lethal vascular damage. Proc Natl Acad Sci U S A 2014; 111:8925-30. [PMID: 24889626 DOI: 10.1073/pnas.1408148111] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The outcome of a viral infection reflects the balance between virus virulence and host susceptibility. The clone 13 (Cl13) variant of lymphocytic choriomeningitis virus--a prototype of Old World arenaviruses closely related to Lassa fever virus--elicits in C57BL/6 and BALB/c mice abundant negative immunoregulatory molecules, associated with T-cell exhaustion, negligible T-cell-mediated injury, and high virus titers that persist. Conversely, here we report that in NZB mice, despite the efficient induction of immunoregulatory molecules and high viremia, Cl13 generated a robust cytotoxic T-cell response, resulting in thrombocytopenia, pulmonary endothelial cell loss, vascular leakage, and death within 6-8 d. These pathogenic events required type I IFN (IFN-I) signaling on nonhematopoietic cells and were completely abrogated by IFN-I receptor blockade. Thus, IFN-I may play a prominent role in hemorrhagic fevers and other acute virus infections associated with severe vascular pathology, and targeting IFN-I or downstream effector molecules may be an effective therapeutic approach.
Collapse
|
42
|
Lander HM, Grant AM, Albrecht T, Hill T, Peters CJ. Endothelial cell permeability and adherens junction disruption induced by junín virus infection. Am J Trop Med Hyg 2014; 90:993-1002. [PMID: 24710609 DOI: 10.4269/ajtmh.13-0382] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
Junín virus (JUNV) is endemic to the fertile Pampas of Argentina, maintained in nature by the rodent host Calomys musculinus, and the causative agent of Argentine hemorrhagic fever (AHF), which is characterized by vascular dysfunction and fluid distribution abnormalities. Clinical as well as experimental studies implicate involvement of the endothelium in the pathogenesis of AHF, although little is known of its role. JUNV has been shown to result in productive infection of endothelial cells (ECs) in vitro with no visible cytopathic effects. In this study, we show that direct JUNV infection of primary human ECs results in increased vascular permeability as measured by electric cell substrate impedance sensing and transwell permeability assays. We also show that EC adherens junctions are disrupted during virus infection, which may provide insight into the role of the endothelium in the pathogenesis of AHF and possibly, other viral hemorrhagic fevers.
Collapse
Affiliation(s)
- Heather M Lander
- Departments of Pathology and Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Ashley M Grant
- Departments of Pathology and Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Thomas Albrecht
- Departments of Pathology and Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Terence Hill
- Departments of Pathology and Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| | - Clarence J Peters
- Departments of Pathology and Microbiology and Immunology, University of Texas Medical Branch, Galveston, Texas
| |
Collapse
|
43
|
Platelet production proceeds independently of the intrinsic and extrinsic apoptosis pathways. Nat Commun 2014; 5:3455. [PMID: 24632563 DOI: 10.1038/ncomms4455] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 02/14/2014] [Indexed: 12/18/2022] Open
Abstract
BH3 mimetic drugs that target BCL-2 family pro-survival proteins to induce tumour cell apoptosis represent a new era in cancer therapy. Clinical trials of navitoclax (ABT-263, which targets BCL-2, BCL-XL and BCL-W) have shown great promise, but encountered dose-limiting thrombocytopenia. Recent work has demonstrated that this is due to the inhibition of BCL-XL, which is essential for platelet survival. These findings raise new questions about the established model of platelet shedding by megakaryocytes, which is thought to be an apoptotic process. Here we generate mice with megakaryocyte-specific deletions of the essential mediators of extrinsic (Caspase-8) and intrinsic (BAK/BAX) apoptosis. We show that megakaryocytes possess a Fas ligand-inducible extrinsic apoptosis pathway. However, Fas activation does not stimulate platelet production, rather, it triggers Caspase-8-mediated killing. Combined loss of Caspase-8/BAK/BAX does not impair thrombopoiesis, but can protect megakaryocytes from death in mice infected with lymphocytic choriomeningitis virus. Thus, apoptosis is dispensable for platelet biogenesis.
Collapse
|
44
|
Koma T, Huang C, Kolokoltsova OA, Brasier AR, Paessler S. Innate immune response to arenaviral infection: a focus on the highly pathogenic New World hemorrhagic arenaviruses. J Mol Biol 2013; 425:4893-903. [PMID: 24075870 PMCID: PMC3864108 DOI: 10.1016/j.jmb.2013.09.028] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Revised: 09/19/2013] [Accepted: 09/22/2013] [Indexed: 12/13/2022]
Abstract
Arenaviruses are enveloped, negative-stranded RNA viruses that belong to the family Arenaviridae. This diverse family can be further classified into OW (Old World) and NW (New World) arenaviruses based on their antigenicity, phylogeny, and geographical distribution. Many of the NW arenaviruses are highly pathogenic viruses that cause systemic human infections characterized by hemorrhagic fever and/or neurological manifestations, constituting public health problems in their endemic regions. NW arenavirus infection induces a variety of host innate immune responses, which could contribute to the viral pathogenesis and/or influence the final outcome of virus infection in vitro and in vivo. On the other hand, NW arenaviruses have also developed several strategies to counteract the host innate immune response. We will review current knowledge regarding the interplay between the host innate immune response and NW arenavirus infection in vitro and in vivo, with emphasis on viral-encoded proteins and their effect on the type I interferon response. NW arenaviruses induce a variety of host innate immune responses. The arenaviruses have several strategies to counteract host innate immune response. We review the interplay between host innate immunity and the arenavirus infections.
Collapse
Affiliation(s)
- Takaaki Koma
- Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX 77550, USA
| | | | | | | | | |
Collapse
|
45
|
Arenaviruses bite the "dust". Blood 2013; 121:868-9. [PMID: 23393016 DOI: 10.1182/blood-2012-05-430686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
|
46
|
Pathogenic mechanisms involved in the hematological alterations of arenavirus-induced hemorrhagic fevers. Viruses 2013; 5:340-51. [PMID: 23337384 PMCID: PMC3564124 DOI: 10.3390/v5010340] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 01/17/2013] [Accepted: 01/18/2013] [Indexed: 11/17/2022] Open
Abstract
Viral hemorrhagic fevers (VHFs) caused by arenaviruses are acute diseases characterized by fever, headache, general malaise, impaired cellular immunity, eventual neurologic involvement, and hemostatic alterations that may ultimately lead to shock and death. The causes of the bleeding are still poorly understood. However, it is generally accepted that these causes are associated to some degree with impaired hemostasis, endothelial cell dysfunction and low platelet counts or function. In this article, we present the current knowledge about the hematological alterations present in VHF induced by arenaviruses, including new aspects on the underlying pathogenic mechanisms.
Collapse
|
47
|
Wang M, Wang J, Wang T, Li J, Hui L, Ha X. Thrombocytopenia as a predictor of severe acute kidney injury in patients with Hantaan virus infections. PLoS One 2013; 8:e53236. [PMID: 23301047 PMCID: PMC3534714 DOI: 10.1371/journal.pone.0053236] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2012] [Accepted: 11/27/2012] [Indexed: 11/19/2022] Open
Abstract
Background Hematological abnormalities often occur several days before kidney injury in patients with hemorrhagic fever with renal syndrome (HFRS). We aimed to investigate the prevalence and prognostic value of the early hematological markers in patients with HFRS caused by Hantaan virus (HTNV) infection. Methods In a retrospective cohort study, we analyzed the case records of 112 patients with acute HTNV infection and evaluated the hematological markers for early prediction and risk stratification of HFRS patients with acute kidney injury (AKI). Results Of 112 patients analyzed, 66 (59%) developed severe AKI, defined as either receipt of acute dialysis or increased serum creatinine ≥354 µmol/L. The prognostic accuracy of hematological markers, as quantified by the area under the receiver-operating-characteristic curve (AUC), was highest with the nadir platelet count (AUC, 0.89; 95% CI, 0.83–0.95), as compared with the admission platelet count (AUC, 0.84; 95% CI, 0.77–0.92), and the admission and peak leukocyte counts. The nadir platelet count correlated moderately with the levels of peak blood urea nitrogen (r = –0.616) and serum creatinine (r = –0.589), the length of hospital stay (r = –0.599), and the number of dialysis sessions that each patient received during hospital stay (r = –0.625). By multivariate analysis, decreased nadir platelet count remained independently associated with the development of severe AKI (odds ratio, 27.57; 95% CI, 6.96–109.16; P<0.0001). Conclusions Thrombocytopenia, rather than leukocytosis, is independently associated with subsequent severe AKI among patients with acute HTNV infection.
Collapse
Affiliation(s)
- Meiliang Wang
- Center for Experimental Medicine, Lanzhou General Hospital, Lanzhou Military Command, Lanzhou, Gansu, China.
| | | | | | | | | | | |
Collapse
|
48
|
Paessler S, Walker DH. Pathogenesis of the viral hemorrhagic fevers. ANNUAL REVIEW OF PATHOLOGY-MECHANISMS OF DISEASE 2012; 8:411-40. [PMID: 23121052 DOI: 10.1146/annurev-pathol-020712-164041] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Four families of enveloped RNA viruses, filoviruses, flaviviruses, arenaviruses, and bunyaviruses, cause hemorrhagic fevers. These viruses are maintained in specific natural cycles involving nonhuman primates, bats, rodents, domestic ruminants, humans, mosquitoes, and ticks. Vascular instability varies from mild to fatal shock, and hemorrhage ranges from none to life threatening. The pathogenic mechanisms are extremely diverse and include deficiency of hepatic synthesis of coagulation factors owing to hepatocellular necrosis, cytokine storm, increased permeability by vascular endothelial growth factor, complement activation, and disseminated intravascular coagulation in one or more hemorrhagic fevers. The severity of disease caused by these agents varies tremendously; there are extremely high fatality rates in Ebola and Marburg hemorrhagic fevers, and asymptomatic infection predominates in yellow fever and dengue viral infections. Although ineffective immunity and high viral loads are characteristic of several viral hemorrhagic fevers, severe plasma leakage occurs at the time of viral clearance and defervescence in dengue hemorrhagic fever.
Collapse
Affiliation(s)
- Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA.
| | | |
Collapse
|
49
|
Grant A, Seregin A, Huang C, Kolokoltsova O, Brasier A, Peters C, Paessler S. Junín virus pathogenesis and virus replication. Viruses 2012. [PMID: 23202466 PMCID: PMC3497054 DOI: 10.3390/v4102317] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Junín virus, the etiological agent of Argentine hemorrhagic fever, causes significant morbidity and mortality. The virus is spread through the aerosolization of host rodent excreta and endemic to the humid pampas of Argentina. Recently, significant progress has been achieved with the development of new technologies (e.g. reverse genetics) that have expanded knowledge about the pathogenesis and viral replication of Junín virus. We will review the pathogenesis of Junín virus in various animal models and the role of innate and adaptive immunity during infection. We will highlight current research regarding the role of molecular biology of Junín virus in elucidating virus attenuation. We will also summarize current knowledge on Junín virus pathogenesis focusing on the recent development of vaccines and potential therapeutics.
Collapse
Affiliation(s)
- Ashley Grant
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (A.G.); (A.S.); (C.H.); (O.A.); (C.P.); (S.P.)
| | - Alexey Seregin
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (A.G.); (A.S.); (C.H.); (O.A.); (C.P.); (S.P.)
| | - Cheng Huang
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (A.G.); (A.S.); (C.H.); (O.A.); (C.P.); (S.P.)
| | - Olga Kolokoltsova
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (A.G.); (A.S.); (C.H.); (O.A.); (C.P.); (S.P.)
| | - Allan Brasier
- Institute for Translational Sciences, Department of Internal Medicine and Sealy Center for Molecular Medicine, University of Texas Medical Branch, Galveston, Texas;
| | - Clarence Peters
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (A.G.); (A.S.); (C.H.); (O.A.); (C.P.); (S.P.)
| | - Slobodan Paessler
- Department of Pathology, University of Texas Medical Branch, Galveston, TX 77555, USA; (A.G.); (A.S.); (C.H.); (O.A.); (C.P.); (S.P.)
- Author to whom correspondence should be addressed; ; Tel.: +1-409-266-6913; Fax: +1-409-747-0762
| |
Collapse
|
50
|
Huang C, Kolokoltsova OA, Yun NE, Seregin AV, Poussard AL, Walker AG, Brasier AR, Zhao Y, Tian B, de la Torre JC, Paessler S. Junín virus infection activates the type I interferon pathway in a RIG-I-dependent manner. PLoS Negl Trop Dis 2012; 6:e1659. [PMID: 22629479 PMCID: PMC3358329 DOI: 10.1371/journal.pntd.0001659] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2012] [Accepted: 04/11/2012] [Indexed: 12/21/2022] Open
Abstract
Junín virus (JUNV), an arenavirus, is the causative agent of Argentine hemorrhagic fever, an infectious human disease with 15-30% case fatality. The pathogenesis of AHF is still not well understood. Elevated levels of interferon and cytokines are reported in AHF patients, which might be correlated to the severity of the disease. However the innate immune response to JUNV infection has not been well evaluated. Previous studies have suggested that the virulent strain of JUNV does not induce IFN in human macrophages and monocytes, whereas the attenuated strain of JUNV was found to induce IFN response in murine macrophages via the TLR-2 signaling pathway. In this study, we investigated the interaction between JUNV and IFN pathway in human epithelial cells highly permissive to JUNV infection. We have determined the expression pattern of interferon-stimulated genes (ISGs) and IFN-β at both mRNA and protein levels during JUNV infection. Our results clearly indicate that JUNV infection activates the type I IFN response. STAT1 phosphorylation, a downstream marker of activation of IFN signaling pathway, was readily detected in JUNV infected IFN-competent cells. Our studies also demonstrated for the first time that RIG-I was required for IFN production during JUNV infection. IFN activation was detected during infection by either the virulent or attenuated vaccine strain of JUNV. Curiously, both virus strains were relatively insensitive to human IFN treatment. Our studies collectively indicated that JUNV infection could induce host type I IFN response and provided new insights into the interaction between JUNV and host innate immune system, which might be important in future studies on vaccine development and antiviral treatment.
Collapse
Affiliation(s)
- Cheng Huang
- Galveston National Laboratory, Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Olga A. Kolokoltsova
- Galveston National Laboratory, Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Nadezdha E. Yun
- Galveston National Laboratory, Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Alexey V. Seregin
- Galveston National Laboratory, Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Allison L. Poussard
- Galveston National Laboratory, Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Aida G. Walker
- Galveston National Laboratory, Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Allan R. Brasier
- Department of Internal Medicine and Sealy Center for Molecular Medicine, Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Yingxin Zhao
- Department of Internal Medicine and Sealy Center for Molecular Medicine, Institute for Translational Sciences, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Bing Tian
- Department of Internal Medicine, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Juan Carlos de la Torre
- Department of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, California, United States of America
| | - Slobodan Paessler
- Galveston National Laboratory, Department of Pathology and Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|