101
|
Sironen T, Sane J, Lokki ML, Meri S, Andersson LC, Hautala T, Kauma H, Vuorinen S, Rasmuson J, Evander M, Ahlm C, Vaheri A. Fatal Puumala Hantavirus Disease: Involvement of Complement Activation and Vascular Leakage in the Pathobiology. Open Forum Infect Dis 2017; 4:ofx229. [PMID: 29255728 PMCID: PMC5726462 DOI: 10.1093/ofid/ofx229] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Accepted: 10/19/2017] [Indexed: 12/13/2022] Open
Abstract
The case-fatality rate of hantavirus disease depends strongly on the causative hantavirus, ranging from 0.1% to 40%. However, the pathogenesis is not fully understood, and at present no licensed therapies exist. We describe fatal cases caused by Puumala hantavirus indicating involvement of complement activation and vascular leakage.
Collapse
Affiliation(s)
| | - Jussi Sane
- Medicum, University of Helsinki, Helsinki, Finland
| | | | - Seppo Meri
- Medicum, University of Helsinki, Helsinki, Finland
| | | | - Timo Hautala
- Department of Internal Medicine, Oulu University Hospital, Oulu, Finland
| | - Heikki Kauma
- Department of Internal Medicine, Oulu University Hospital, Oulu, Finland
| | | | - Johan Rasmuson
- Department of Clinical Microbiology, Infectious Diseases, Umeå University, Umeå, Sweden
| | - Magnus Evander
- Department of Clinical Microbiology, Virology, Umeå University, Umeå, Sweden
| | - Clas Ahlm
- Department of Clinical Microbiology, Infectious Diseases, Umeå University, Umeå, Sweden
| | - Antti Vaheri
- Medicum, University of Helsinki, Helsinki, Finland
| |
Collapse
|
102
|
Schönrich G, Raftery MJ. Exploring the Immunopathogenesis of Viral Hemorrhagic Fever in Mice with a Humanized Immune System. Front Immunol 2017; 8:1202. [PMID: 29018450 PMCID: PMC5622932 DOI: 10.3389/fimmu.2017.01202] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 09/11/2017] [Indexed: 01/23/2023] Open
Abstract
Viral hemorrhagic fever (VHF) as a disease entity was first codified in the 1930s by soviet scientists investigating patients suffering from hantavirus infection. The group of hemorrhagic fever viruses (HFVs) has since expanded to include members from at least four different virus families: Arenaviridae, Bunyaviridae, Filoviridae, and Flaviviridae, all enveloped single-stranded RNA viruses. After infection, the natural hosts of HFVs do not develop symptoms, whereas humans can be severely affected. This observation and other evidence from experimental data suggest that the human immune system plays a crucial role in VHF pathogenesis. For this reason mice with a human immune system, referred to here as humanized mice (humice), are valuable tools that provide insight into disease mechanisms and allow for preclinical testing of novel vaccinations approaches as well as antiviral agents. In this article, we review the impact of humice in VHF research.
Collapse
Affiliation(s)
- Günther Schönrich
- Institute of Medical Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Martin J Raftery
- Institute of Medical Virology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
103
|
Younan P, Iampietro M, Nishida A, Ramanathan P, Santos RI, Dutta M, Lubaki NM, Koup RA, Katze MG, Bukreyev A. Ebola Virus Binding to Tim-1 on T Lymphocytes Induces a Cytokine Storm. mBio 2017; 8:e00845-17. [PMID: 28951472 PMCID: PMC5615193 DOI: 10.1128/mbio.00845-17] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/17/2017] [Indexed: 12/14/2022] Open
Abstract
Ebola virus (EBOV) disease (EVD) results from an exacerbated immunological response that is highlighted by a burst in the production of inflammatory mediators known as a "cytokine storm." Previous reports have suggested that nonspecific activation of T lymphocytes may play a central role in this phenomenon. T-cell immunoglobulin and mucin domain-containing protein 1 (Tim-1) has recently been shown to interact with virion-associated phosphatidylserine to promote infection. Here, we demonstrate the central role of Tim-1 in EBOV pathogenesis, as Tim-1-/- mice exhibited increased survival rates and reduced disease severity; surprisingly, only a limited decrease in viremia was detected. Tim-1-/- mice exhibited a modified inflammatory response as evidenced by changes in serum cytokines and activation of T helper subsets. A series of in vitro assays based on the Tim-1 expression profile on T cells demonstrated that despite the apparent absence of detectable viral replication in T lymphocytes, EBOV directly binds to isolated T lymphocytes in a phosphatidylserine-Tim-1-dependent manner. Exposure to EBOV resulted in the rapid development of a CD4Hi CD3Low population, non-antigen-specific activation, and cytokine production. Transcriptome and Western blot analysis of EBOV-stimulated CD4+ T cells confirmed the induction of the Tim-1 signaling pathway. Furthermore, comparative analysis of transcriptome data and cytokine/chemokine analysis of supernatants highlight the similarities associated with EBOV-stimulated T cells and the onset of a cytokine storm. Flow cytometry revealed virtually exclusive binding and activation of central memory CD4+ T cells. These findings provide evidence for the role of Tim-1 in the induction of a cytokine storm phenomenon and the pathogenesis of EVD.IMPORTANCE Ebola virus infection is characterized by a massive release of inflammatory mediators, which has come to be known as a cytokine storm. The severity of the cytokine storm is consistently linked with fatal disease outcome. Previous findings have demonstrated that specific T-cell subsets are key contributors to the onset of a cytokine storm. In this study, we investigated the role of Tim-1, a T-cell-receptor-independent trigger of T-cell activation. We first demonstrated that Tim-1-knockout (KO) mice survive lethal Ebola virus challenge. We then used a series of in vitro assays to demonstrate that Ebola virus directly binds primary T cells in a Tim-1-phosphatidylserine-dependent manner. We noted that binding induces a cytokine storm-like phenomenon and that blocking Tim-1-phosphatidylserine interactions reduces viral binding, T-cell activation, and cytokine production. These findings highlight a previously unknown role of Tim-1 in the development of a cytokine storm and "immune paralysis."
Collapse
Affiliation(s)
- Patrick Younan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mathieu Iampietro
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Palaniappan Ramanathan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Rodrigo I Santos
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mukta Dutta
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Ndongala Michel Lubaki
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Washington National Primate Research Center, Seattle, Washington, USA
| | - Alexander Bukreyev
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
104
|
Younan P, Iampietro M, Nishida A, Ramanathan P, Santos RI, Dutta M, Lubaki NM, Koup RA, Katze MG, Bukreyev A. Ebola Virus Binding to Tim-1 on T Lymphocytes Induces a Cytokine Storm. mBio 2017; 8:e00845-17. [PMID: 28951472 DOI: 10.1128/mbio.00845-17] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Accepted: 08/17/2017] [Indexed: 12/12/2022] Open
Abstract
Ebola virus (EBOV) disease (EVD) results from an exacerbated immunological response that is highlighted by a burst in the production of inflammatory mediators known as a "cytokine storm." Previous reports have suggested that nonspecific activation of T lymphocytes may play a central role in this phenomenon. T-cell immunoglobulin and mucin domain-containing protein 1 (Tim-1) has recently been shown to interact with virion-associated phosphatidylserine to promote infection. Here, we demonstrate the central role of Tim-1 in EBOV pathogenesis, as Tim-1-/- mice exhibited increased survival rates and reduced disease severity; surprisingly, only a limited decrease in viremia was detected. Tim-1-/- mice exhibited a modified inflammatory response as evidenced by changes in serum cytokines and activation of T helper subsets. A series of in vitro assays based on the Tim-1 expression profile on T cells demonstrated that despite the apparent absence of detectable viral replication in T lymphocytes, EBOV directly binds to isolated T lymphocytes in a phosphatidylserine-Tim-1-dependent manner. Exposure to EBOV resulted in the rapid development of a CD4Hi CD3Low population, non-antigen-specific activation, and cytokine production. Transcriptome and Western blot analysis of EBOV-stimulated CD4+ T cells confirmed the induction of the Tim-1 signaling pathway. Furthermore, comparative analysis of transcriptome data and cytokine/chemokine analysis of supernatants highlight the similarities associated with EBOV-stimulated T cells and the onset of a cytokine storm. Flow cytometry revealed virtually exclusive binding and activation of central memory CD4+ T cells. These findings provide evidence for the role of Tim-1 in the induction of a cytokine storm phenomenon and the pathogenesis of EVD.IMPORTANCE Ebola virus infection is characterized by a massive release of inflammatory mediators, which has come to be known as a cytokine storm. The severity of the cytokine storm is consistently linked with fatal disease outcome. Previous findings have demonstrated that specific T-cell subsets are key contributors to the onset of a cytokine storm. In this study, we investigated the role of Tim-1, a T-cell-receptor-independent trigger of T-cell activation. We first demonstrated that Tim-1-knockout (KO) mice survive lethal Ebola virus challenge. We then used a series of in vitro assays to demonstrate that Ebola virus directly binds primary T cells in a Tim-1-phosphatidylserine-dependent manner. We noted that binding induces a cytokine storm-like phenomenon and that blocking Tim-1-phosphatidylserine interactions reduces viral binding, T-cell activation, and cytokine production. These findings highlight a previously unknown role of Tim-1 in the development of a cytokine storm and "immune paralysis."
Collapse
Affiliation(s)
- Patrick Younan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mathieu Iampietro
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Andrew Nishida
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Palaniappan Ramanathan
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Rodrigo I Santos
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Mukta Dutta
- Department of Microbiology, University of Washington, Seattle, Washington, USA
| | - Ndongala Michel Lubaki
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| | - Richard A Koup
- Immunology Laboratory, Vaccine Research Center, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Michael G Katze
- Department of Microbiology, University of Washington, Seattle, Washington, USA
- Washington National Primate Research Center, Seattle, Washington, USA
| | - Alexander Bukreyev
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, USA
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, USA
- Galveston National Laboratory, The University of Texas Medical Branch, Galveston, Texas, USA
- The University of Texas Medical Branch, Galveston, Texas, USA
| |
Collapse
|
105
|
Gibb R, Moses LM, Redding DW, Jones KE. Understanding the cryptic nature of Lassa fever in West Africa. Pathog Glob Health 2017; 111:276-288. [PMID: 28875769 PMCID: PMC5694855 DOI: 10.1080/20477724.2017.1369643] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Lassa fever (LF) is increasingly recognized by global health institutions as an important rodent-borne disease with severe impacts on some of West Africa's poorest communities. However, our knowledge of LF ecology, epidemiology and distribution is limited, which presents barriers to both short-term disease forecasting and prediction of long-term impacts of environmental change on Lassa virus (LASV) zoonotic transmission dynamics. Here, we synthesize current knowledge to show that extrapolations from past research have produced an incomplete picture of the incidence and distribution of LF, with negative consequences for policy planning, medical treatment and management interventions. Although the recent increase in LF case reports is likely due to improved surveillance, recent studies suggest that future socio-ecological changes in West Africa may drive increases in LF burden. Future research should focus on the geographical distribution and disease burden of LF, in order to improve its integration into public policy and disease control strategies.
Collapse
Affiliation(s)
- Rory Gibb
- Centre for Biodiversity and Environment Research, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Lina M. Moses
- Department of Global Community Health and Behavioral Sciences, Tulane University, New Orleans, LA, USA
| | - David W. Redding
- Centre for Biodiversity and Environment Research, Department of Genetics, Evolution and Environment, University College London, London, UK
| | - Kate E. Jones
- Centre for Biodiversity and Environment Research, Department of Genetics, Evolution and Environment, University College London, London, UK
- Institute of Zoology, Zoological Society of London, London, UK
| |
Collapse
|
106
|
Interferon-γ-Driven iNOS: A Molecular Pathway to Terminal Shock in Arenavirus Hemorrhagic Fever. Cell Host Microbe 2017; 22:354-365.e5. [DOI: 10.1016/j.chom.2017.07.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 05/11/2017] [Accepted: 07/11/2017] [Indexed: 01/21/2023]
|
107
|
Bixler SL, Duplantier AJ, Bavari S. Discovering Drugs for the Treatment of Ebola Virus. CURRENT TREATMENT OPTIONS IN INFECTIOUS DISEASES 2017; 9:299-317. [PMID: 28890666 PMCID: PMC5570806 DOI: 10.1007/s40506-017-0130-z] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Purpose of review Ebola virus, a member of the Filoviridae family, is a causative agent of severe viral hemorrhagic fever in humans. Over the past 40 years, the virus has been linked to several high mortality outbreaks in Africa with the recent West African outbreak resulting in over 11,000 deaths. This review provides a summary of the status of the drug discovery and development process for therapeutics for Ebola virus disease, with a focus on the strategies being used and the challenges facing each stage of the process. Recent findings Despite the wealth of in vitro efficacy data, preclinical data in animal models, and human clinical data, no therapeutics have been approved for the treatment of Ebola virus disease. However, several promising candidates, such as ZMapp and GS-5734, have advanced into ongoing clinical trials. Summary The gravity of the 2014-2016 outbreak spurred a heightened effort to identify and develop new treatments for Ebola virus disease, including small molecules, immunotherapeutics, host factors, and clinical disease management options. Disclaimer Opinions, interpretations, conclusions, and recommendations are those of the authors and are not necessarily endoresed by the U.S. Army.
Collapse
Affiliation(s)
- Sandra L Bixler
- United States Army Medical Research Institute of Infectious Diseases, 1425 Porter St, Frederick, MD 21702 USA
| | - Allen J Duplantier
- United States Army Medical Research Institute of Infectious Diseases, 1425 Porter St, Frederick, MD 21702 USA
| | - Sina Bavari
- United States Army Medical Research Institute of Infectious Diseases, 1425 Porter St, Frederick, MD 21702 USA
| |
Collapse
|
108
|
Vijayan AL, Vanimaya, Ravindran S, Saikant R, Lakshmi S, Kartik R, G M. Procalcitonin: a promising diagnostic marker for sepsis and antibiotic therapy. J Intensive Care 2017; 5:51. [PMID: 28794881 PMCID: PMC5543591 DOI: 10.1186/s40560-017-0246-8] [Citation(s) in RCA: 257] [Impact Index Per Article: 32.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2017] [Accepted: 07/21/2017] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Sepsis is a global healthcare problem, characterized by whole body inflammation in response to microbial infection, which leads to organ dysfunction. It is becoming a frequent complication in hospitalized patients. Early and differential diagnosis of sepsis is needed critically to avoid unnecessary usage of antimicrobial agents and for proper antibiotic treatments through the screening of biomarkers that sustains with diagnostic significance. MAIN BODY OF ABSTRACT Current targeting conventional markers (C-reactive protein, white blood cell, tumour necrosis factor-α, interleukins, etc.) are non-specific for diagnosing sepsis. Procalcitonin (PCT), a member of the calcitonin super family could be a critical tool for the diagnosis of sepsis. But to distinguish between bacterial versus viral infections, procalcitonin alone may not be effective. Rapid elevation in the concentration of procalcitonin and other newly emerging biomarkers during an infection and its correlation with severity of illness makes it an ideal biomarker for bacterial infection. Beside this, the procalcitonin levels can be used for monitoring response to antimicrobial therapy, diagnosis of secondary inflammations, diagnosis of renal involvement in paediatric urinary tract infection, etc. The present article summarizes the relevance of procalcitonin in the diagnosis of sepsis and how it can be useful in determining the therapeutic approaches. CONCLUSION Further studies are needed to better understand the application of PCT in the diagnosis of sepsis, differentiating between microbial and non-microbial infection cases and determining the therapeutic approaches for sepsis.
Collapse
Affiliation(s)
- Ashitha L. Vijayan
- Diagnostic Products Division, Corporate R&D Centre, HLL Lifecare Limited, Akkulam, Sreekariyam (P.O), Trivandrum, Kerala India
| | - Vanimaya
- Diagnostic Products Division, Corporate R&D Centre, HLL Lifecare Limited, Akkulam, Sreekariyam (P.O), Trivandrum, Kerala India
| | - Shilpa Ravindran
- Diagnostic Products Division, Corporate R&D Centre, HLL Lifecare Limited, Akkulam, Sreekariyam (P.O), Trivandrum, Kerala India
| | - R. Saikant
- Diagnostic Products Division, Corporate R&D Centre, HLL Lifecare Limited, Akkulam, Sreekariyam (P.O), Trivandrum, Kerala India
| | - S. Lakshmi
- Diagnostic Products Division, Corporate R&D Centre, HLL Lifecare Limited, Akkulam, Sreekariyam (P.O), Trivandrum, Kerala India
| | - R. Kartik
- Diagnostic Products Division, Corporate R&D Centre, HLL Lifecare Limited, Akkulam, Sreekariyam (P.O), Trivandrum, Kerala India
| | - Manoj. G
- Diagnostic Products Division, Corporate R&D Centre, HLL Lifecare Limited, Akkulam, Sreekariyam (P.O), Trivandrum, Kerala India
| |
Collapse
|
109
|
Boldescu V, Behnam MAM, Vasilakis N, Klein CD. Broad-spectrum agents for flaviviral infections: dengue, Zika and beyond. Nat Rev Drug Discov 2017; 16:565-586. [PMID: 28473729 PMCID: PMC5925760 DOI: 10.1038/nrd.2017.33] [Citation(s) in RCA: 217] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Infections with flaviviruses, such as dengue, West Nile virus and the recently re-emerging Zika virus, are an increasing and probably lasting global risk. This Review summarizes and comments on the opportunities for broad-spectrum agents that are active against multiple flaviviruses. Broad-spectrum activity is particularly desirable to prepare for the next flaviviral epidemic, which could emerge from as-yet unknown or neglected viruses. Potential molecular targets for broad-spectrum antiflaviviral compounds include viral proteins, such as the viral protease or polymerase, and host targets that are exploited by these viruses during entry and replication, including α-glucosidase and proteins involved in nucleoside biosynthesis. Numerous compounds with broad-spectrum antiviral activity have already been identified by target-specific or phenotypic assays. For other compounds, broad-spectrum activity can be anticipated because of their mode of action and molecular targets.
Collapse
Affiliation(s)
- Veaceslav Boldescu
- Medicinal Chemistry, Institute of Pharmacy and Molecular Biotechnology IPMB, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
- Laboratory of Organic Synthesis and Biopharmaceuticals, Institute of Chemistry of the Academy of Sciences of Moldova, Academiei 3, 2028 Chisinau, Moldova
| | - Mira A. M. Behnam
- Medicinal Chemistry, Institute of Pharmacy and Molecular Biotechnology IPMB, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| | - Nikos Vasilakis
- Dept. of Pathology and Center for Biodefense and Emerging Infectious Diseases, Center for Tropical Diseases and Institute for Human Infections and Immunity, 2.138D Keiller Bldg, University of Texas Medical Branch, 301 University Blvd, Galveston, TX 77555–0609, USA
| | - Christian D. Klein
- Medicinal Chemistry, Institute of Pharmacy and Molecular Biotechnology IPMB, Heidelberg University, Im Neuenheimer Feld 364, 69120 Heidelberg, Germany
| |
Collapse
|
110
|
Finding a Balance between Protection and Pathology: The Dual Role of Perforin in Human Disease. Int J Mol Sci 2017; 18:ijms18081608. [PMID: 28757574 PMCID: PMC5578000 DOI: 10.3390/ijms18081608] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Revised: 07/20/2017] [Accepted: 07/20/2017] [Indexed: 01/08/2023] Open
Abstract
Perforin is critical for controlling viral infection and tumor surveillance. Clinically, mutations in perforin are viewed as unfavorable, as lack of this pore-forming protein results in lethal, childhood disease, familial hemophagocytic lymphohistiocytosis type 2 (FHL 2). However, many mutations in the coding region of PRF1 are not yet associated with disease. Animal models of viral-associated blood–brain barrier (BBB) disruption and experimental cerebral malaria (ECM) have identified perforin as critical for inducing pathologic central nervous system CNS vascular permeability. This review focuses on the role of perforin in both protecting and promoting human disease. It concludes with a novel hypothesis that diversity observed in the PRF1 gene may be an example of selective advantage that protects an individual from perforin-mediated pathology, such as BBB disruption.
Collapse
|
111
|
Golden JW, Beitzel B, Ladner JT, Mucker EM, Kwilas SA, Palacios G, Hooper JW. An attenuated Machupo virus with a disrupted L-segment intergenic region protects guinea pigs against lethal Guanarito virus infection. Sci Rep 2017; 7:4679. [PMID: 28680057 PMCID: PMC5498534 DOI: 10.1038/s41598-017-04889-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2017] [Accepted: 05/22/2017] [Indexed: 02/07/2023] Open
Abstract
Machupo virus (MACV) is a New World (NW) arenavirus and causative agent of Bolivian hemorrhagic fever (HF). Here, we identified a variant of MACV strain Carvallo termed Car91 that was attenuated in guinea pigs. Infection of guinea pigs with an earlier passage of Carvallo, termed Car68, resulted in a lethal disease with a 63% mortality rate. Sequencing analysis revealed that compared to Car68, Car91 had a 35 nucleotide (nt) deletion and a point mutation within the L-segment intergenic region (IGR), and three silent changes in the polymerase gene that did not impact amino acid coding. No changes were found on the S-segment. Because it was apathogenic, we determined if Car91 could protect guinea pigs against Guanarito virus (GTOV), a distantly related NW arenavirus. While naïve animals succumbed to GTOV infection, 88% of the Car91-exposed guinea pigs were protected. These findings indicate that attenuated MACV vaccines can provide heterologous protection against NW arenaviruses. The disruption in the L-segment IGR, including a single point mutant and 35 nt partial deletion, were the only major variance detected between virulent and avirulent isolates, implicating its role in attenuation. Overall, our data support the development of live-attenuated arenaviruses as broadly protective pan-arenavirus vaccines.
Collapse
Affiliation(s)
- Joseph W Golden
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, 21702, United States.
| | - Brett Beitzel
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, 21702, United States
| | - Jason T Ladner
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, 21702, United States
| | - Eric M Mucker
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, 21702, United States
| | - Steven A Kwilas
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, 21702, United States
| | - Gustavo Palacios
- Center for Genome Sciences, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, 21702, United States
| | - Jay W Hooper
- Department of Molecular Virology, Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, 21702, United States
| |
Collapse
|
112
|
Akhuemokhan OC, Ewah-Odiase RO, Akpede N, Ehimuan J, Adomeh DI, Odia I, Olomu SC, Pahlmann M, Becker-Ziaja B, Happi CT, Asogun DA, Okogbenin SA, Okokhere PO, Dawodu OS, Omoike IU, Sabeti PC, Günther S, Akpede GO. Prevalence of Lassa Virus Disease (LVD) in Nigerian children with fever or fever and convulsions in an endemic area. PLoS Negl Trop Dis 2017; 11:e0005711. [PMID: 28671959 PMCID: PMC5510890 DOI: 10.1371/journal.pntd.0005711] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2016] [Revised: 07/14/2017] [Accepted: 06/13/2017] [Indexed: 12/18/2022] Open
Abstract
Background Convulsions with fever in children are a common neurologic emergency in the tropics, and determining the contribution of endemic viral infections can be challenging. In particular, there is a dearth of data on the prevalence and clinical differentiation of Lassa virus disease (LVD) in febrile children in endemic areas of Nigeria, which has multiple lineages of the virus. The aim of this study was to determine the prevalence and presentation of LVD in febrile children with and without convulsions. Methodology/Principal findings This was a prospective study of consecutive febrile children aged ≥1 month– 15 years admitted to the Children’s Emergency Room of Irrua Specialist Teaching Hospital over a period of 1 year. Febrile children with convulsions (Cases) were compared with those without convulsions (Controls). LVD was defined by the presence of a positive Lassa virus RT-PCR test. Rates were compared between groups using χ2 or Fisher’s exact tests and p <0.05 taken as significant. 373 (40.9%) of 913 admissions had fever. Of these, 108/373 (29%) presented with convulsions. The overall prevalence of LVD was 13/373 (3.5%; 95% CI = 1.9%, 5.7%) in febrile admissions, 3/108 (2.8%) in Cases and 10/265 (3.8%) in Controls [(Odds Ratio (95% Confidence Interval) (OR (95% CI)) of LVD in Cases versus Controls = 0.73 (0.2, 2.7)]. Only vomiting (OR (95% CI) = 0.09 (0.01, 0.70)) and bleeding (OR (95% CI) = 39.56 (8.52, 183.7)) were significantly associated with an increased prevalence of LVD. Conclusions/Significance LVD is an important cause of fever, including undifferentiated fever in children in endemic areas, but it is not significantly associated with convulsions associated with fever. Its prevalence, and lack of clinical differentiation on presentation, underscores the importance of a high index of suspicion in diagnosis. Screening of febrile children with undifferentiated fever in endemic areas for LVD could be an important medical and public health control measure. There has, perhaps, been undue focus on malaria as a cause of childhood fever and convulsions, often with delayed/missed diagnosis of other serious prevalent infections, and correspondingly very little published data on the contribution of Lassa virus disease (LVD) in endemic areas. There is also very little published data on the contribution of LVD to childhood morbidity and mortality in Nigeria, a large LVD-endemic country that has in circulation 3 of the 4 currently known lineages of the Lassa virus. This study was carried out to address these gaps. The results should also be of relevance in the formulation of policies for the treatment and control of viral haemorrhagic fevers. The prevalence of LVD was 5.4% among children with clinically undifferentiated fever (n = 243); 3.9% among those with convulsions associated with fever (n = 77) and 6.0% among those with fever but no convulsions (n = 166). The results underscore the importance of LVD as a cause of acute undifferentiated fever. The results also underscore the need of diagnostic testing for LVD in children with acute undifferentiated fever in endemic areas in order to facilitate control, including the prevention of nosocomial transmission.
Collapse
Affiliation(s)
- Odigie C. Akhuemokhan
- Department of Paediatrics, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | | | - Nosa Akpede
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Jacqueline Ehimuan
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Donatus I. Adomeh
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Ikpomwonsa Odia
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Sylvia C. Olomu
- Department of Paediatrics, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Meike Pahlmann
- Bernhard-Nocht-Institute for Tropical Medicine, WHO Collaborating Centre for Arboviruses and Hemorrhagic Fever Reference and Research, Department of Virology, Bernhard-Nocht-Str. 74, Hamburg, Germany
| | - Beate Becker-Ziaja
- Bernhard-Nocht-Institute for Tropical Medicine, WHO Collaborating Centre for Arboviruses and Hemorrhagic Fever Reference and Research, Department of Virology, Bernhard-Nocht-Str. 74, Hamburg, Germany
| | - Christian T. Happi
- Malaria Research Laboratory, College of Medicine, University of Ibadan, Ibadan, Oyo State, Nigeria
| | - Danny A. Asogun
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Sylvanus A. Okogbenin
- Department of Obstetrics and Gynaecology, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Peter O. Okokhere
- Department of Medicine, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Osagie S. Dawodu
- Department of Paediatrics, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Irekpono U. Omoike
- Department of Paediatrics, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Pardis C. Sabeti
- Department of Organismic Biology, Broad Institute, Harvard University, Cambridge, Massachusetts, United States of America
| | - Stephan Günther
- Bernhard-Nocht-Institute for Tropical Medicine, WHO Collaborating Centre for Arboviruses and Hemorrhagic Fever Reference and Research, Department of Virology, Bernhard-Nocht-Str. 74, Hamburg, Germany
| | - George O. Akpede
- Department of Paediatrics, Faculty of Clinical Sciences, College of Medicine, Ambrose Alli University, Ekpoma, Edo State, Nigeria
- * E-mail:
| |
Collapse
|
113
|
Brody T, Yavatkar AS, Park DS, Kuzin A, Ross J, Odenwald WF. Flavivirus and Filovirus EvoPrinters: New alignment tools for the comparative analysis of viral evolution. PLoS Negl Trop Dis 2017. [PMID: 28622346 PMCID: PMC5489223 DOI: 10.1371/journal.pntd.0005673] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Background Flavivirus and Filovirus infections are serious epidemic threats to human populations. Multi-genome comparative analysis of these evolving pathogens affords a view of their essential, conserved sequence elements as well as progressive evolutionary changes. While phylogenetic analysis has yielded important insights, the growing number of available genomic sequences makes comparisons between hundreds of viral strains challenging. We report here a new approach for the comparative analysis of these hemorrhagic fever viruses that can superimpose an unlimited number of one-on-one alignments to identify important features within genomes of interest. Methodology/Principal finding We have adapted EvoPrinter alignment algorithms for the rapid comparative analysis of Flavivirus or Filovirus sequences including Zika and Ebola strains. The user can input a full genome or partial viral sequence and then view either individual comparisons or generate color-coded readouts that superimpose hundreds of one-on-one alignments to identify unique or shared identity SNPs that reveal ancestral relationships between strains. The user can also opt to select a database genome in order to access a library of pre-aligned genomes of either 1,094 Flaviviruses or 460 Filoviruses for rapid comparative analysis with all database entries or a select subset. Using EvoPrinter search and alignment programs, we show the following: 1) superimposing alignment data from many related strains identifies lineage identity SNPs, which enable the assessment of sublineage complexity within viral outbreaks; 2) whole-genome SNP profile screens uncover novel Dengue2 and Zika recombinant strains and their parental lineages; 3) differential SNP profiling identifies host cell A-to-I hyper-editing within Ebola and Marburg viruses, and 4) hundreds of superimposed one-on-one Ebola genome alignments highlight ultra-conserved regulatory sequences, invariant amino acid codons and evolutionarily variable protein-encoding domains within a single genome. Conclusions/Significance EvoPrinter allows for the assessment of lineage complexity within Flavivirus or Filovirus outbreaks, identification of recombinant strains, highlights sequences that have undergone host cell A-to-I editing, and identifies unique input and database SNPs within highly conserved sequences. EvoPrinter’s ability to superimpose alignment data from hundreds of strains onto a single genome has allowed us to identify unique Zika virus sublineages that are currently spreading in South, Central and North America, the Caribbean, and in China. This new set of integrated alignment programs should serve as a useful addition to existing tools for the comparative analysis of these viruses. Flaviviruses, including Zika and Dengue viruses, and Filoviruses, including Ebola and Marburg viruses, are significant global public health threats. Genetic surveillance of viral isolates provides important insights into the origin of outbreaks, reveals lineage heterogeneity and diversification, and facilitates identification of novel recombinant strains and host cell modified viral genomes. We report the development of EvoPrinter, a web-accessed alignment tool for the rapid comparative analysis of viral genomes. EvoPrinter superimposes alignment data from multiple pairwise comparisons onto a single reference sequence of interest, to reveal both similarities and differences detected in hundreds of selected viral isolates. Evoprinter databases provide easy access to hundreds of non-redundant Flavivirus and Filovirus genomes. allowing the user to distinguish between sublineage identity SNPs and unique strain-specific SNPs, thus facilitating analysis of the history of viral diversification during an epidemic. EvoPrinter also proves useful in identifying recombinant strains and their parental lineages and detecting host-cell genomic editing. EvoPrinter should serve as a useful addition to existing tools for the comparative analysis of these viruses.
Collapse
Affiliation(s)
- Thomas Brody
- Neural Cell-Fate Determinants Section, NINDS, NIH, Bethesda, Maryland, United States of America
- * E-mail: (TB); (WFO)
| | - Amarendra S. Yavatkar
- Division of Intramural Research Information Technology Program, NINDS, NIH, Bethesda, Maryland, United States of America
| | - Dong Sun Park
- Division of Intramural Research Information Technology Program, NINDS, NIH, Bethesda, Maryland, United States of America
| | - Alexander Kuzin
- Neural Cell-Fate Determinants Section, NINDS, NIH, Bethesda, Maryland, United States of America
| | - Jermaine Ross
- Neural Cell-Fate Determinants Section, NINDS, NIH, Bethesda, Maryland, United States of America
| | - Ward F. Odenwald
- Neural Cell-Fate Determinants Section, NINDS, NIH, Bethesda, Maryland, United States of America
- * E-mail: (TB); (WFO)
| |
Collapse
|
114
|
Basler CF. Molecular pathogenesis of viral hemorrhagic fever. Semin Immunopathol 2017; 39:551-561. [PMID: 28555386 DOI: 10.1007/s00281-017-0637-x] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2017] [Accepted: 04/23/2017] [Indexed: 12/11/2022]
Abstract
The clinical syndrome referred to as viral hemorrhagic fever (VHF) can be caused by several different families of RNA viruses, including select members of the arenaviruses, bunyaviruses, filoviruses, and flaviviruses. VHF is characterized by malaise, fever, vascular permeability, decreased plasma volume, coagulation abnormalities, and varying degrees of hemorrhage. Study of the filovirus Ebola virus has demonstrated a critical role for suppression of innate antiviral defenses in viral pathogenesis. Additionally, antigen-presenting cells are targets of productive infection and immune dysregulation. Among these cell populations, monocytes and macrophages are proposed to produce damaging inflammatory cytokines, while infected dendritic cells fail to undergo proper maturation, potentially impairing adaptive immunity. Uncontrolled virus replication and accompanying inflammatory responses are thought to promote vascular leakage and coagulopathy. However, the specific molecular pathways that underlie these features of VHF remain poorly understood. The arenavirus Lassa virus and the flavivirus yellow fever virus exhibit similar molecular pathogenesis suggesting common underlying mechanisms. Because non-human primate models that closely mimic VHF are available for Ebola, Lassa, and yellow fever viruses, we propose that comparative molecular studies using these models will yield new insights into the molecular underpinnings of VHF and suggest new therapeutic approaches.
Collapse
Affiliation(s)
- Christopher F Basler
- Center for Microbial Pathogenesis, Georgia Research Alliance Eminent Scholar in Microbial Pathogenesis, Institute for Biomedical Sciences, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
115
|
Abstract
PURPOSE OF REVIEW The recent 2014-2016 outbreak of Ebola virus disease (EVD) has led to many discoveries regarding Ebola. Although neurological symptoms during EVD had been previously described, many reports since this outbreak have made clear that EVD can lead to neurological issues. This article will review the various neurological manifestations of EVD. RECENT FINDINGS Recently, many neurological symptoms have been described during acute EVD, including altered mental status, seizures, and meningoencephalitis, among others; survivors of EVD also may develop neurological sequelae, such as persistent headache and memory loss and can exhibit abnormalities on neurological exam. Additionally, it is now evident that in rare cases, survivors may experience relapses of EVD months after recovery, including the central nervous system (CNS). EVD can result in many clinical neurological manifestations, both acutely and after recovery. Research is ongoing to further clarify the nature of Ebola in the CNS.
Collapse
Affiliation(s)
- Bridgette Jeanne Billioux
- National Institutes of Neurological Diseases and Stroke National Institutes of Health, 10 Center Drive, Room 5C-103, Bethesda, MD, 20892, USA.
| |
Collapse
|
116
|
Buchko GW, Clifton MC, Wallace EG, Atkins KA, Myler PJ. Backbone chemical shift assignments and secondary structure analysis of the U1 protein from the Bas-Congo virus. BIOMOLECULAR NMR ASSIGNMENTS 2017; 11:51-56. [PMID: 27981424 PMCID: PMC8457264 DOI: 10.1007/s12104-016-9719-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2016] [Accepted: 11/30/2016] [Indexed: 06/06/2023]
Abstract
The Bas-Congo virus (BASV) is the first rhabdovirus associated with a human outbreak of acute hemorrhagic fever. The single-stranded, negative-sense RNA genome of BASV contains the five core genes present in all rhabdoviral genomes plus an additional three genes, annotated U1, U2, and U3, with weak (<21%) sequence similarity only to a handful of genes observed in a few other rhabdoviral genomes. The function of the rhabdoviral U proteins is unknown, but, they are hypothesized to play a role in viral infection or replication. To better understand this unique family of proteins, a construct containing residues 27-203 of the 216-residue U1 protein (BASV-U1*) was prepared. By collecting data in 0.5 M urea it was possible to eliminate transient association enough to enable the assignment of most of the observable 1HN, 1Hα, 15N, 13Cα, 13Cβ, and 13C´ chemical shifts for BASV-U1* that will provide a foundation to study its solution properties. The analyses of these chemical shifts along with 15N-edited NOESY data enabled the identification of the elements of secondary structure present in BASV-U1*.
Collapse
Affiliation(s)
- Garry W Buchko
- Seattle Structural Genomics Center for Infectious Disease, Seattle, USA.
- Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, 99352, USA.
| | - Matthew C Clifton
- Seattle Structural Genomics Center for Infectious Disease, Seattle, USA
- Beryllium, Bainbridge Island, WA, 97331, USA
- Nurix Inc., San Francisco, CA, 94158, USA
| | - Ellen G Wallace
- Seattle Structural Genomics Center for Infectious Disease, Seattle, USA
- Beryllium, Bainbridge Island, WA, 97331, USA
| | - Kateri A Atkins
- Seattle Structural Genomics Center for Infectious Disease, Seattle, USA
- Beryllium, Bainbridge Island, WA, 97331, USA
| | - Peter J Myler
- Seattle Structural Genomics Center for Infectious Disease, Seattle, USA
- Center for Infectious Disease Research, 307 Westlake Ave. N., Seattle, WA, 98109, USA
- Department of Global Health, University of Washington, Seattle, WA, 98165, USA
- Department of Biomedical Informatics and Health Education, University of Washington, Seattle, WA, 98195, USA
| |
Collapse
|
117
|
Wolfe MK, Gallandat K, Daniels K, Desmarais AM, Scheinman P, Lantagne D. Handwashing and Ebola virus disease outbreaks: A randomized comparison of soap, hand sanitizer, and 0.05% chlorine solutions on the inactivation and removal of model organisms Phi6 and E. coli from hands and persistence in rinse water. PLoS One 2017; 12:e0172734. [PMID: 28231311 PMCID: PMC5322913 DOI: 10.1371/journal.pone.0172734] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 02/07/2017] [Indexed: 11/19/2022] Open
Abstract
To prevent Ebola transmission, frequent handwashing is recommended in Ebola Treatment Units and communities. However, little is known about which handwashing protocol is most efficacious. We evaluated six handwashing protocols (soap and water, alcohol-based hand sanitizer (ABHS), and 0.05% sodium dichloroisocyanurate, high-test hypochlorite, and stabilized and non-stabilized sodium hypochlorite solutions) for 1) efficacy of handwashing on the removal and inactivation of non-pathogenic model organisms and, 2) persistence of organisms in rinse water. Model organisms E. coli and bacteriophage Phi6 were used to evaluate handwashing with and without organic load added to simulate bodily fluids. Hands were inoculated with test organisms, washed, and rinsed using a glove juice method to retrieve remaining organisms. Impact was estimated by comparing the log reduction in organisms after handwashing to the log reduction without handwashing. Rinse water was collected to test for persistence of organisms. Handwashing resulted in a 1.94-3.01 log reduction in E. coli concentration without, and 2.18-3.34 with, soil load; and a 2.44-3.06 log reduction in Phi6 without, and 2.71-3.69 with, soil load. HTH performed most consistently well, with significantly greater log reductions than other handwashing protocols in three models. However, the magnitude of handwashing efficacy differences was small, suggesting protocols are similarly efficacious. Rinse water demonstrated a 0.28-4.77 log reduction in remaining E. coli without, and 0.21-4.49 with, soil load and a 1.26-2.02 log reduction in Phi6 without, and 1.30-2.20 with, soil load. Chlorine resulted in significantly less persistence of E. coli in both conditions and Phi6 without soil load in rinse water (p<0.001). Thus, chlorine-based methods may offer a benefit of reducing persistence in rinse water. We recommend responders use the most practical handwashing method to ensure hand hygiene in Ebola contexts, considering the potential benefit of chlorine-based methods in rinse water persistence.
Collapse
Affiliation(s)
- Marlene K. Wolfe
- Department of Civil and Environmental Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Karin Gallandat
- Department of Civil and Environmental Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Kyle Daniels
- Department of Civil and Environmental Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Anne Marie Desmarais
- Department of Civil and Environmental Engineering, Tufts University, Medford, Massachusetts, United States of America
| | - Pamela Scheinman
- Department of Dermatology, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Daniele Lantagne
- Department of Civil and Environmental Engineering, Tufts University, Medford, Massachusetts, United States of America
| |
Collapse
|
118
|
Muñoz-Fontela C, Geisbert TW. The gap between animal and human Ebola virus disease. Future Virol 2017. [DOI: 10.2217/fvl-2016-0128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Affiliation(s)
- César Muñoz-Fontela
- Heinrich Pette Institute, Leibniz Institute for Experimental Virology, Martinistrasse 52, 20251 Hamburg, Germany
- Department of Virology, Bernhard Nocht Institute for Tropical Medicine, Bernhard Nocht Strasse 74, 20359 Hamburg, Germany
| | - Thomas W Geisbert
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
119
|
Abstract
Two of the most important contemporary emerging viruses that affect human health in Africa are Ebola virus (EBOV) and Lassa virus (LASV). The 2013-2016 West African outbreak of EBOV was responsible for more than 11,000 deaths, primarily in Guinea, Sierra Leone and Liberia. LASV is constantly emerging in these and surrounding West African countries, with an estimate of more than 500,000 cases of Lassa fever, and approximately 5,000 deaths, annually. Both EBOV and LASV are zoonotic, and human infection often results in a severe haemorrhagic fever in both cases. However, the contribution of specific immune responses to disease differs between EBOV and LASV. This Review examines innate and adaptive immune responses to these viruses with the goal of delineating responses that are associated with protective versus pathogenic outcomes.
Collapse
|
120
|
Rojek A, Carson G, Kato Y, Horby PW, Leblebicioglu H. Viral Hemorrhagic Fevers. Infect Dis (Lond) 2017. [DOI: 10.1016/b978-0-7020-6285-8.00132-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
121
|
Dengue Fever-Associated Maculopathy and Panuveitis in Australia. Case Rep Ophthalmol Med 2016; 2016:5704695. [PMID: 28078150 PMCID: PMC5204087 DOI: 10.1155/2016/5704695] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2016] [Revised: 10/27/2016] [Accepted: 11/14/2016] [Indexed: 12/27/2022] Open
Abstract
Purpose. To describe a case of dengue fever-associated maculopathy and panuveitis to raise awareness of these ophthalmic complications of dengue in Australia in the light of recent increasing numbers of outbreaks from equatorial through to tropical Australia. Case Report. A 37-year-old Caucasian Australian male returning from Cambodia presented with a bilateral dengue fever-associated maculopathy with left panuveitis diagnosed clinically and haematologically. Automated perimetry revealed bilateral paracentral scotomas while optical coherence tomography demonstrated the maculopathies to be of the diffuse retinal thickening type in the right eye and acute macular neuroretinopathy (AMN) type in the left eye. He was treated conservatively with only topical steroids and cycloplegia and made a full clinical visual recovery. Conclusion. Our case study underscores the importance of the awareness of the ophthalmic complications of dengue fever as despite their rarity they can be potentially sight threatening. The incidence of these complications is likely to rise in Australia with increased global warming and the distribution of Aedes aegypti into subtropical Australia.
Collapse
|
122
|
Marín-López A, Bermúdez R, Calvo-Pinilla E, Moreno S, Brun A, Ortego J. Pathological Characterization Of IFNAR(-/-) Mice Infected With Bluetongue Virus Serotype 4. Int J Biol Sci 2016; 12:1448-1460. [PMID: 27994510 PMCID: PMC5166487 DOI: 10.7150/ijbs.14967] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2016] [Accepted: 06/11/2016] [Indexed: 01/11/2023] Open
Abstract
Bluetongue virus (BTV) replicates in lymphoid tissues where infected mononuclear leukocytes secrete proinflammatory and vasoactive mediators that can contribute to bluetongue (BT) pathogenesis. Using the well-characterized IFNAR(-/-) mice animal model, we have now studied the histopathology and dynamics of leukocyte populations in different target tissues (spleen, thymus, and lung) during BTV-4 infection by histological and immunohistochemical techniques. The spleen and thymus of BTV-4 infected mice showed severe lymphoid depletion on H&E stained sections. This finding was confirmed by IHC, showing moderate decreased immunopositivity against CD3 in the thymus, and scarce immunoreactivity against CD3 and CD79 in the rest of the white pulp in the spleen, together with an increase in MAC387 immunostaining. BTV-4 infection also induced the expression of active caspase-3 in the spleen, where apoptotic debris was observed by H&E. A dramatic increase in iNOS immunoreactivity associated to necrotic areas of the white pulp was observed, being less noticeable in the thymus and the lung. The induction of pro-inflammatory cytokines in tissues where BTV replicates was evaluated by measuring transcript levels by RT-qPCR. BTV-4 infection led to enhance transcription of IFN-γ, TNF, IL-6, IL-12-p40, and IL-1β mRNA in the thymus, spleen and lung, correlating with the level of virus replication in these tissues. Disease progression and pathogenesis in IFNAR(-/-) mice closely mimics hallmarks of bluetongue disease in ruminants. IFNAR(-/-) mice are a good choice to facilitate a faster advance in the field of orbiviruses.
Collapse
Affiliation(s)
| | - Roberto Bermúdez
- Departamento de Ciencias Clínicas Veterinarias, Facultad de Veterinaria, Universidad de Santiago de Compostela, 27002 Lugo, Spain
| | | | - Sandra Moreno
- INIA-CISA, Ctra. Algete-El Casar, Valdeolmos, 28130 Madrid, Spain
| | - Alejandro Brun
- INIA-CISA, Ctra. Algete-El Casar, Valdeolmos, 28130 Madrid, Spain
| | - Javier Ortego
- INIA-CISA, Ctra. Algete-El Casar, Valdeolmos, 28130 Madrid, Spain
| |
Collapse
|
123
|
Koepsell SA, Winkler AM, Roback JD. The Role of the Laboratory and Transfusion Service in the Management of Ebola Virus Disease. Transfus Med Rev 2016; 31:149-153. [PMID: 27894669 PMCID: PMC7126423 DOI: 10.1016/j.tmrv.2016.11.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/09/2016] [Accepted: 11/10/2016] [Indexed: 01/15/2023]
Abstract
The Ebola outbreak that began in 2013 infected and killed record numbers of individuals and created unprecedented challenges, including containment and treatment of the virus in resource-strained West Africa as well as the repatriation and treatment for patients in the United States and Europe. Valuable lessons were learned, especially the important role that the laboratory and transfusion service plays in the treatment for patients with Ebola virus disease (EVD) by providing data for supportive care and fluid resuscitation as well as the generation of investigational therapies such as convalescent plasma (CP). To provide treatment support, laboratories had to evaluate and update procedures to ensure the safety of laboratory personnel. Because there is no licensed EVD-specific treatment, CP was used in more than 99 patients with only 1 possible severe adverse event reported. However, given the biologic variability inherent in CP as well as the small number of patient treated in a nonrandomized fashion, the efficacy of CP in the treatment of EVD remains unknown. Patients with Ebola virus disease were treated in the United States and Europe for the first time. Laboratories played a vital role in supportive care and experimental therapies for Ebola virus disease. Convalescent plasma has unknown efficacy in treating Ebola virus disease.
Collapse
Affiliation(s)
- Scott A Koepsell
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE.
| | | | - John D Roback
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
124
|
Molecular Basis for Antibody-Mediated Neutralization of New World Hemorrhagic Fever Mammarenaviruses. Cell Host Microbe 2016; 18:705-13. [PMID: 26651946 DOI: 10.1016/j.chom.2015.11.005] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 10/18/2015] [Accepted: 11/16/2015] [Indexed: 11/20/2022]
Abstract
In the Western hemisphere, at least five mammarenaviruses cause human viral hemorrhagic fevers with high case fatality rates. Junín virus (JUNV) is the only hemorrhagic fever virus for which transfusion of survivor immune plasma that contains neutralizing antibodies ("passive immunity") is an established treatment. Here, we report the structure of the JUNV surface glycoprotein receptor-binding subunit (GP1) bound to a neutralizing monoclonal antibody. The antibody engages the GP1 site that binds transferrin receptor 1 (TfR1)-the host cell surface receptor for all New World hemorrhagic fever mammarenaviruses-and mimics an important receptor contact. We show that survivor immune plasma contains antibodies that bind the same epitope. We propose that viral receptor-binding site accessibility explains the success of passive immunity against JUNV and that this functionally conserved epitope is a potential target for therapeutics and vaccines to limit infection by all New World hemorrhagic fever mammarenaviruses.
Collapse
|
125
|
Kaner J, Schaack S. Understanding Ebola: the 2014 epidemic. Global Health 2016; 12:53. [PMID: 27624088 PMCID: PMC5022139 DOI: 10.1186/s12992-016-0194-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2015] [Accepted: 08/31/2016] [Indexed: 01/05/2023] Open
Abstract
Near the end of 2013, an outbreak of Zaire ebolavirus (EBOV) began in Guinea, subsequently spreading to neighboring Liberia and Sierra Leone. As this epidemic grew, important public health questions emerged about how and why this outbreak was so different from previous episodes. This review provides a synthetic synopsis of the 2014-15 outbreak, with the aim of understanding its unprecedented spread. We present a summary of the history of previous epidemics, describe the structure and genetics of the ebolavirus, and review our current understanding of viral vectors and the latest treatment practices. We conclude with an analysis of the public health challenges epidemic responders faced and some of the lessons that could be applied to future outbreaks of Ebola or other viruses.
Collapse
Affiliation(s)
- Jolie Kaner
- Department of Epidemiology, Oregon Health and Science University, 3181 S.W. Sam Jackson Park Rd., Portland, OR, 97239, USA.,Department of Biology, Reed College, 3203 SE Woodstock Blvd, Portland, OR, 97202, USA
| | - Sarah Schaack
- Department of Biology, Reed College, 3203 SE Woodstock Blvd, Portland, OR, 97202, USA.
| |
Collapse
|
126
|
Caballero IS, Honko AN, Gire SK, Winnicki SM, Melé M, Gerhardinger C, Lin AE, Rinn JL, Sabeti PC, Hensley LE, Connor JH. In vivo Ebola virus infection leads to a strong innate response in circulating immune cells. BMC Genomics 2016; 17:707. [PMID: 27595844 PMCID: PMC5011782 DOI: 10.1186/s12864-016-3060-0] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Accepted: 09/02/2016] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Ebola virus is the causative agent of a severe syndrome in humans with a fatality rate that can approach 90 %. During infection, the host immune response is thought to become dysregulated, but the mechanisms through which this happens are not entirely understood. In this study, we analyze RNA sequencing data to determine the host response to Ebola virus infection in circulating immune cells. RESULTS Approximately half of the 100 genes with the strongest early increases in expression were interferon-stimulated genes, such as ISG15, OAS1, IFIT2, HERC5, MX1 and DHX58. Other highly upregulated genes included cytokines CXCL11, CCL7, IL2RA, IL2R1, IL15RA, and CSF2RB, which have not been previously reported to change during Ebola virus infection. Comparing this response in two different models of exposure (intramuscular and aerosol) revealed a similar signature of infection. The strong innate response in the aerosol model was seen not only in circulating cells, but also in primary and secondary target tissues. Conversely, the innate immune response of vaccinated macaques was almost non-existent. This suggests that the innate response is a major aspect of the cellular response to Ebola virus infection in multiple tissues. CONCLUSIONS Ebola virus causes a severe infection in humans that is associated with high mortality. The host immune response to virus infection is thought to be an important aspect leading to severe pathology, but the components of this overactive response are not well characterized. Here, we analyzed how circulating immune cells respond to the virus and found that there is a strong innate response dependent on active virus replication. This finding is in stark contrast to in vitro evidence showing a suppression of innate immune signaling, and it suggests that the strong innate response we observe in infected animals may be an important contributor to pathogenesis.
Collapse
Affiliation(s)
| | - Anna N. Honko
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD USA
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, MD USA
| | - Stephen K. Gire
- Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA USA
- Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Sarah M. Winnicki
- Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA USA
- Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Marta Melé
- Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA USA
| | - Chiara Gerhardinger
- Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA USA
| | - Aaron E. Lin
- Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA USA
- Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - John L. Rinn
- Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA USA
- Department of Stem Cell and Regenerative Biology, Harvard University, Cambridge, MA USA
| | - Pardis C. Sabeti
- Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA USA
- Broad Institute of MIT and Harvard, Cambridge, MA USA
| | - Lisa E. Hensley
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD USA
- Integrated Research Facility, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, MD USA
| | - John H. Connor
- Department of Microbiology, Boston University School of Medicine, Boston, MA USA
| |
Collapse
|
127
|
Hsu YL, Wang MY, Ho LJ, Lai JH. Dengue virus infection induces interferon-lambda1 to facilitate cell migration. Sci Rep 2016; 6:24530. [PMID: 27456172 PMCID: PMC4960520 DOI: 10.1038/srep24530] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 03/31/2016] [Indexed: 01/03/2023] Open
Abstract
A marked increase in the rate of dengue virus (DENV) infection has resulted in more than 212 deaths in Taiwan since the beginning of 2015, mostly from fatal outcomes such as dengue hemorrhagic fever and dengue shock syndrome. The pathogenic mechanisms of these fatal manifestations are poorly understood. Cytokines induce an overwhelming immune reaction and thus have crucial roles. Interferon-lambda (IFN-λ), a newly identified IFN subtype, has antiviral effects, but its immunologic effects in DENV infection have not been investigated. In the present study, we show that DENV infection preferentially induced production of IFN-λ1 in human dendritic cells (DCs) and human lung epithelial cells. Virus nonstructural 1 (NS1) glycoprotein was responsible for the effect. DENV-induced production of IFN-λ1 was dependent on signaling pathways involving toll-like receptor (TLR)-3, interferon regulation factor (IRF)-3, and nuclear factor-kappaB (NF-κB). Blocking interaction between IFN-λ1 and its receptor IFN-λR1 through siRNA interference reduced DENV-induced DC migration towards the chemoattractants CCL19 and CCL21, by inhibiting CCR7 expression. Furthermore, IFN-λ1 itself induced CCR7 expression and DC migration. Our study presents the first evidence of the mechanisms and effects of IFN-λ1 induction in DENV-infected DCs and highlights the role of this cytokine in the immunopathogenesis of DENV infection.
Collapse
Affiliation(s)
- Yu-Lin Hsu
- Institute of Preventive Medicine, National Defense Medical Center, Taipei, Taiwan, R.O.C
| | - Mei-Yi Wang
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Tao-Yuan, Taiwan, R.O.C
| | - Ling-Jun Ho
- Institute of Cellular and System Medicine, National Health Research Institute, Zhunan, Taiwan, R.O.C
| | - Jenn-Haung Lai
- Division of Allergy, Immunology, and Rheumatology, Department of Internal Medicine, Chang Gung Memorial Hospital, Chang Gung University, Tao-Yuan, Taiwan, R.O.C.,Graduate Institute of Clinical Research, National Defense Medical Center, Taipei, Taiwan, R.O.C
| |
Collapse
|
128
|
Dietmann A, Putzer D, Beer R, Helbok R, Pfausler B, Nordin AJ, Virgolini I, Grams AE, Schmutzhard E. Cerebral glucose hypometabolism in Tick-Borne Encephalitis, a pilot study in 10 Patients. Int J Infect Dis 2016; 51:73-77. [PMID: 27418580 DOI: 10.1016/j.ijid.2016.06.022] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 06/12/2016] [Accepted: 06/28/2016] [Indexed: 10/21/2022] Open
Abstract
BACKGROUND Tick borne encephalitis (TBE) is an acute meningoencephalitis with or without myelitis caused by an RNA virus from the flavivirus family transmitted by Ixodes spp ticks. The neurotropic TBE virus infects preferentially large neurons in basal ganglia, anterior horns, medulla oblongata, Purkinje cells and thalamus. Brain metabolic changes related to radiologic and clinical findings have not been described so far. METHODS Here we describe the clinical course of 10 consecutive TBE patients with outcome assessment at discharge and after 12 month using a modified Rankin Scale. Patients underwent cerebral MRI after confirmation of diagnosis and before discharge. 18F-FDG PET/CT scans were performed within day 5 to day 14 after TBE diagnosis. Extended analysis of coagulation parameters by thrombelastometry (ROTEM® InTEM, ExTEM, FibTEM) was performed every other day after confirmation of TBE diagnosis up to day 10 after hospital admission or discharge. RESULTS All patients presented with a meningoencephalitic course of disease. Cerebral MRI scans showed unspecific findings at predilection areas in 3 patients. 18F-FDG PET/CT showed increased glucose utilization in one patient and decreased 18F-FDG uptake in seven patients. Changes in coagulation measured by standard parameters and thrombelastometry were not found in any of the patients. DISCUSSION Glucose hypometabolism was present in 7 out of 10 TBE patients reflecting neuronal dysfunction in predilection areas of TBE virus infiltration responsible for development of clinical signs and symptoms.
Collapse
Affiliation(s)
- Anelia Dietmann
- Department of Neurology, Neurological Intensive Care Unit, Innsbruck Medical University, Innsbruck, Austria.
| | - Daniel Putzer
- Department of Nuclear Medicine, Innsbruck Medical University, Innsbruck, Austria; Department of Radiology, Innsbruck Medical University, Innsbruck, Austria
| | - Ronny Beer
- Department of Neurology, Neurological Intensive Care Unit, Innsbruck Medical University, Innsbruck, Austria
| | - Raimund Helbok
- Department of Neurology, Neurological Intensive Care Unit, Innsbruck Medical University, Innsbruck, Austria
| | - Bettina Pfausler
- Department of Neurology, Neurological Intensive Care Unit, Innsbruck Medical University, Innsbruck, Austria
| | - Abdul Jalil Nordin
- Diagnostic Nuklear Imaging Centre, Universiti Putra Malaysia Serdang, Selangor, Malaysia
| | - Irene Virgolini
- Department of Nuclear Medicine, Innsbruck Medical University, Innsbruck, Austria
| | - Astrid E Grams
- Department of Neuroradiology, Innsbruck Medical University, Innsbruck, Austria
| | - Erich Schmutzhard
- Department of Neurology, Neurological Intensive Care Unit, Innsbruck Medical University, Innsbruck, Austria
| |
Collapse
|
129
|
|
130
|
Accuracy, Precision, Ease-Of-Use, and Cost of Methods to Test Ebola-Relevant Chlorine Solutions. PLoS One 2016; 11:e0152442. [PMID: 27243817 PMCID: PMC4887006 DOI: 10.1371/journal.pone.0152442] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Accepted: 03/14/2016] [Indexed: 11/19/2022] Open
Abstract
To prevent transmission in Ebola Virus Disease (EVD) outbreaks, it is recommended to disinfect living things (hands and people) with 0.05% chlorine solution and non-living things (surfaces, personal protective equipment, dead bodies) with 0.5% chlorine solution. In the current West African EVD outbreak, these solutions (manufactured from calcium hypochlorite (HTH), sodium dichloroisocyanurate (NaDCC), and sodium hypochlorite (NaOCl)) have been widely used in both Ebola Treatment Unit and community settings. To ensure solution quality, testing is necessary, however test method appropriateness for these Ebola-relevant concentrations has not previously been evaluated. We identified fourteen commercially-available methods to test Ebola-relevant chlorine solution concentrations, including two titration methods, four DPD dilution methods, and six test strips. We assessed these methods by: 1) determining accuracy and precision by measuring in quintuplicate five different 0.05% and 0.5% chlorine solutions manufactured from NaDCC, HTH, and NaOCl; 2) conducting volunteer testing to assess ease-of-use; and, 3) determining costs. Accuracy was greatest in titration methods (reference-12.4% error compared to reference method), then DPD dilution methods (2.4-19% error), then test strips (5.2-48% error); precision followed this same trend. Two methods had an accuracy of <10% error across all five chlorine solutions with good precision: Hach digital titration for 0.05% and 0.5% solutions (recommended for contexts with trained personnel and financial resources), and Serim test strips for 0.05% solutions (recommended for contexts where rapid, inexpensive, and low-training burden testing is needed). Measurement error from test methods not including pH adjustment varied significantly across the five chlorine solutions, which had pH values 5-11. Volunteers found test strip easiest and titration hardest; costs per 100 tests were $14-37 for test strips and $33-609 for titration. Given the ease-of-use and cost benefits of test strips, we recommend further development of test strips robust to pH variation and appropriate for Ebola-relevant chlorine solution concentrations.
Collapse
|
131
|
Andersen KG, Shapiro BJ, Matranga CB, Sealfon R, Lin AE, Moses LM, Folarin OA, Goba A, Odia I, Ehiane PE, Momoh M, England EM, Winnicki S, Branco LM, Gire SK, Phelan E, Tariyal R, Tewhey R, Omoniwa O, Fullah M, Fonnie R, Fonnie M, Kanneh L, Jalloh S, Gbakie M, Saffa S, Karbo K, Gladden AD, Qu J, Stremlau M, Nekoui M, Finucane HK, Tabrizi S, Vitti JJ, Birren B, Fitzgerald M, McCowan C, Ireland A, Berlin AM, Bochicchio J, Tazon-Vega B, Lennon NJ, Ryan EM, Bjornson Z, Milner DA, Lukens AK, Broodie N, Rowland M, Heinrich M, Akdag M, Schieffelin JS, Levy D, Akpan H, Bausch DG, Rubins K, McCormick JB, Lander ES, Günther S, Hensley L, Okogbenin S, Schaffner SF, Okokhere PO, Khan SH, Grant DS, Akpede GO, Asogun DA, Gnirke A, Levin JZ, Happi CT, Garry RF, Sabeti PC. Clinical Sequencing Uncovers Origins and Evolution of Lassa Virus. Cell 2016; 162:738-50. [PMID: 26276630 DOI: 10.1016/j.cell.2015.07.020] [Citation(s) in RCA: 204] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/26/2015] [Accepted: 06/12/2015] [Indexed: 12/25/2022]
Abstract
The 2013-2015 West African epidemic of Ebola virus disease (EVD) reminds us of how little is known about biosafety level 4 viruses. Like Ebola virus, Lassa virus (LASV) can cause hemorrhagic fever with high case fatality rates. We generated a genomic catalog of almost 200 LASV sequences from clinical and rodent reservoir samples. We show that whereas the 2013-2015 EVD epidemic is fueled by human-to-human transmissions, LASV infections mainly result from reservoir-to-human infections. We elucidated the spread of LASV across West Africa and show that this migration was accompanied by changes in LASV genome abundance, fatality rates, codon adaptation, and translational efficiency. By investigating intrahost evolution, we found that mutations accumulate in epitopes of viral surface proteins, suggesting selection for immune escape. This catalog will serve as a foundation for the development of vaccines and diagnostics. VIDEO ABSTRACT.
Collapse
Affiliation(s)
- Kristian G Andersen
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA; The Scripps Research Institute, Scripps Translational Science Institute, La Jolla, CA 92037, USA.
| | - B Jesse Shapiro
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA; Department of Biological Sciences, University of Montréal, Montréal, QC H2V 2S9, Canada
| | | | - Rachel Sealfon
- Broad Institute, Cambridge, MA 02142, USA; Computer Science and Artificial Intelligence Laboratory, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Aaron E Lin
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Lina M Moses
- Tulane Health Sciences Center, Tulane University, New Orleans, LA 70118, USA
| | - Onikepe A Folarin
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria; Department of Biological Sciences, College of Natural Sciences, Redeemer's University, Redemption City, Osun State, Nigeria
| | - Augustine Goba
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | - Ikponmwonsa Odia
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Philomena E Ehiane
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Mambu Momoh
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone; Eastern Polytechnic College, Kenema, Eastern Province, Sierra Leone
| | | | - Sarah Winnicki
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA
| | | | - Stephen K Gire
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA
| | | | | | - Ryan Tewhey
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Omowunmi Omoniwa
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Mohammed Fullah
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone; Eastern Polytechnic College, Kenema, Eastern Province, Sierra Leone
| | - Richard Fonnie
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | - Mbalu Fonnie
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | - Lansana Kanneh
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | - Simbirie Jalloh
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | - Michael Gbakie
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | - Sidiki Saffa
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | - Kandeh Karbo
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | | | - James Qu
- Broad Institute, Cambridge, MA 02142, USA
| | - Matthew Stremlau
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Mahan Nekoui
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA
| | | | - Shervin Tabrizi
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA
| | - Joseph J Vitti
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA
| | | | | | | | | | | | | | | | | | | | - Zach Bjornson
- Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Danny A Milner
- Department of Immunology and Infectious Disease, Harvard School of Public Health, Boston, MA 02115, USA
| | - Amanda K Lukens
- Department of Immunology and Infectious Disease, Harvard School of Public Health, Boston, MA 02115, USA
| | - Nisha Broodie
- College of Medicine, Columbia University, New York, NY 10032, USA
| | | | | | | | - John S Schieffelin
- Tulane Health Sciences Center, Tulane University, New Orleans, LA 70118, USA
| | - Danielle Levy
- Tulane Health Sciences Center, Tulane University, New Orleans, LA 70118, USA
| | - Henry Akpan
- Nigerian Federal Ministry of Health, Abuja, Federal Capital Territory, Nigeria
| | - Daniel G Bausch
- Tulane Health Sciences Center, Tulane University, New Orleans, LA 70118, USA
| | - Kathleen Rubins
- The National Aeronautics and Space Administration, Johnson Space Center, Houston, TX 77058, USA
| | - Joseph B McCormick
- The University of Texas School of Public Health, Brownsville, TX 77030, USA
| | | | - Stephan Günther
- Department of Virology, Bernhard-Nocht-Institute for Tropical Medicine, 20259 Hamburg, Germany
| | - Lisa Hensley
- NIAID Integrated Research Facility, Frederick, MD 21702, USA
| | - Sylvanus Okogbenin
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | | | | | - Peter O Okokhere
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - S Humarr Khan
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | - Donald S Grant
- Lassa Fever Laboratory, Kenema Government Hospital, Kenema, Eastern Province, Sierra Leone
| | - George O Akpede
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | - Danny A Asogun
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria
| | | | | | - Christian T Happi
- Institute of Lassa Fever Research and Control, Irrua Specialist Teaching Hospital, Irrua, Edo State, Nigeria; Department of Biological Sciences, College of Natural Sciences, Redeemer's University, Redemption City, Osun State, Nigeria.
| | - Robert F Garry
- Tulane Health Sciences Center, Tulane University, New Orleans, LA 70118, USA
| | - Pardis C Sabeti
- FAS Center for Systems Biology, Department of Organismic and Evolutionary Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute, Cambridge, MA 02142, USA; Department of Immunology and Infectious Disease, Harvard School of Public Health, Boston, MA 02115, USA.
| |
Collapse
|
132
|
Abstract
Mammalian arenaviruses are zoonotic viruses that cause asymptomatic, persistent infections in their rodent hosts but can lead to severe and lethal hemorrhagic fever with bleeding and multiorgan failure in human patients. Lassa virus (LASV), for example, is endemic in several West African countries, where it is responsible for an estimated 500,000 infections and 5,000 deaths annually. There are currently no FDA-licensed therapeutics or vaccines available to combat arenavirus infection. A hallmark of arenavirus infection (e.g., LASV) is general immunosuppression that contributes to high viremia. Here, we discuss the early host immune responses to arenavirus infection and the recently discovered molecular mechanisms that enable pathogenic viruses to suppress host immune recognition and to contribute to the high degree of virulence. We also directly compare the innate immune evasion mechanisms between arenaviruses and other hemorrhagic fever-causing viruses, such as Ebola, Marburg, Dengue, and hantaviruses. A better understanding of the immunosuppression and immune evasion strategies of these deadly viruses may guide the development of novel preventative and therapeutic options.
Collapse
Affiliation(s)
- Bjoern Meyer
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Saint Paul, Minnesota, USA
| | - Hinh Ly
- Department of Veterinary and Biomedical Sciences, University of Minnesota, Twin Cities, Saint Paul, Minnesota, USA
| |
Collapse
|
133
|
Bailey AL, Lauck M, Sibley SD, Friedrich TC, Kuhn JH, Freimer NB, Jasinska AJ, Phillips-Conroy JE, Jolly CJ, Marx PA, Apetrei C, Rogers J, Goldberg TL, O'Connor DH. Zoonotic Potential of Simian Arteriviruses. J Virol 2016; 90:630-5. [PMID: 26559828 PMCID: PMC4702702 DOI: 10.1128/jvi.01433-15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Wild nonhuman primates are immediate sources and long-term reservoirs of human pathogens. However, ethical and technical challenges have hampered the identification of novel blood-borne pathogens in these animals. We recently examined RNA viruses in plasma from wild African monkeys and discovered several novel, highly divergent viruses belonging to the family Arteriviridae. Close relatives of these viruses, including simian hemorrhagic fever virus, have caused sporadic outbreaks of viral hemorrhagic fever in captive macaque monkeys since the 1960s. However, arterivirus infection in wild nonhuman primates had not been described prior to 2011. The arteriviruses recently identified in wild monkeys have high sequence and host species diversity, maintain high viremia, and are prevalent in affected populations. Taken together, these features suggest that the simian arteriviruses may be "preemergent" zoonotic pathogens. If not, this would imply that biological characteristics of RNA viruses thought to facilitate zoonotic transmission may not, by themselves, be sufficient for such transmission to occur.
Collapse
Affiliation(s)
- Adam L Bailey
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | - Michael Lauck
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| | - Samuel D Sibley
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Thomas C Friedrich
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Fort Detrick, Frederick, Maryland, USA
| | - Nelson B Freimer
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California, USA
| | - Anna J Jasinska
- Center for Neurobehavioral Genetics, Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, California, USA
| | - Jane E Phillips-Conroy
- Department of Anatomy and Neurobiology, Washington University School of Medicine, and Department of Anthropology, Washington University, Saint Louis, Missouri, USA
| | - Clifford J Jolly
- Department of Anthropology, New York University, New York, New York, USA
| | - Preston A Marx
- Tulane National Primate Research Center, Covington, Louisiana, USA Department of Tropical Medicine, Tulane School of Public Health and Tropical Medicine, New Orleans, Louisiana, USA
| | - Cristian Apetrei
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, USA Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Jeffrey Rogers
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas, USA
| | - Tony L Goldberg
- Wisconsin National Primate Research Center, Madison, Wisconsin, USA Department of Pathobiological Sciences, University of Wisconsin-Madison, Madison, Wisconsin, USA
| | - David H O'Connor
- Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin, USA Wisconsin National Primate Research Center, Madison, Wisconsin, USA
| |
Collapse
|
134
|
Di Minno G, Perno CF, Tiede A, Navarro D, Canaro M, Güertler L, Ironside JW. Current concepts in the prevention of pathogen transmission via blood/plasma-derived products for bleeding disorders. Blood Rev 2016; 30:35-48. [PMID: 26381318 PMCID: PMC7115716 DOI: 10.1016/j.blre.2015.07.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2015] [Revised: 07/11/2015] [Accepted: 07/13/2015] [Indexed: 02/06/2023]
Abstract
The pathogen safety of blood/plasma-derived products has historically been a subject of significant concern to the medical community. Measures such as donor selection and blood screening have contributed to increase the safety of these products, but pathogen transmission does still occur. Reasons for this include lack of sensitivity/specificity of current screening methods, lack of reliable screening tests for some pathogens (e.g. prions) and the fact that many potentially harmful infectious agents are not routinely screened for. Methods for the purification/inactivation of blood/plasma-derived products have been developed in order to further reduce the residual risk, but low concentrations of pathogens do not necessarily imply a low level of risk for the patient and so the overall challenge of minimising risk remains. This review aims to discuss the variable level of pathogenic risk and describes the current screening methods used to prevent/detect the presence of pathogens in blood/plasma-derived products.
Collapse
Affiliation(s)
- Giovanni Di Minno
- Dipartimento di Medicina Clinica e Chirurgia, Regional Reference Centre for Coagulation Disorders, Federico II University, Via S. Pansini 5, 80131 Naples, Italy.
| | - Carlo Federico Perno
- Department of Experimental Medicine and Surgery, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | - Andreas Tiede
- Department of Hematology, Hemostasis, Oncology and Stem Cell Transplantation, Hannover Medical School, Carl-Neuberg-Str. 1, D-30625 Hannover, Germany
| | - David Navarro
- Department of Microbiology, Microbiology Service, Hospital Clínico Universitario, School of Medicine, University of Valencia, Av Blasco Ibáñez 17, 46010 Valencia, Spain
| | - Mariana Canaro
- Department of Hemostasis and Thrombosis, Son Espases University Hospital, Carretera de Valdemossa, 79, 07120 Palma de Mallorca, Spain
| | - Lutz Güertler
- Max von Pettenkofer Institute for Hygiene and Medical Microbiology, University of München, Pettenkofer Str 9A, 80336 Munich, Germany
| | - James W Ironside
- National Creutzfeldt-Jakob Disease Research and Surveillance Unit, School of Clinical Sciences, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| |
Collapse
|
135
|
Abstract
Viral pathogenesis seeks to understand how a virus interacts with its host at multiple levels. Key questions include the source (an infected human, animal, or insect vector), the transmission mechanism, and how the virus is shed and transmitted. Following transmission, pathogenesis is governed by the initial site of replication, whether the virus disseminates within the host, and its tropism for specific tissues and organs. In turn, these steps are dictated by the structure and replication strategy of the virus. In addition to utilizing selected synthetic biochemical pathways in the host cell, viruses frequently reprogram host cells by inducing intracellular signaling pathways that render the cell more permissive. Host–virus interactions also control whether the infection is acute, chronic, latent, or transforming; how the virus interacts with the immune system; and the consequent pathophysiological response of the host. This chapter provides an overview of these basic concepts of viral pathogenesis, with emphasis on the interactions of viruses with their host cells and organisms.
Collapse
|
136
|
Mifsud A, Peelen D, Brincat P, Abela S, Debattista N, Laspina S, Zammit D, Camilleri DJ, Gatt A. A feasibility study on the effects of Triton X-100 for the in vitro inactivation of Ebola virus on haematological assays. J Clin Pathol 2015; 69:637-42. [PMID: 26670745 DOI: 10.1136/jclinpath-2015-203331] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 11/12/2015] [Indexed: 11/04/2022]
Abstract
AIMS The aim of this study was to check the effect of Triton X-100 on various, commonly used haematology test parameters. METHODS Anonymised blood samples were treated with 10 µL of 10% Triton X-100 per 1 mL of blood. Treated and untreated samples were tested in parallel for blood film morphology, complete blood counts (CBCs), flow cytometry, blood grouping and antibody screens. Samples were also taken in 3.2% citrate tubes for coagulation test analyses. RESULTS Statistical differences were noted in all CBC parameters apart from the mean cell volume, eosinophil and basophil counts. Platelet counts were significantly different with an apparent rise after the addition of Triton X-100. Samples were noted to have a high red cell fragmentation index. Immunological platelet counting methods using flow cytometry and fluorescent methods showed no significant differences and gave reliable results. Neither flow cytometry for T-cell subsets nor blood grouping/antibody screens were affected by Triton X-100. However, coagulation samples were severely haemolysed prohibiting analysis. CONCLUSIONS We have demonstrated that the addition of Triton X-100 to haematology blood samples impacts mainly on platelet counts and coagulation studies due to haemolysis. The platelet count is spuriously raised probably due to the presence of red cell fragments. The latter can be circumvented by the use of immunological platelet counting technology.
Collapse
Affiliation(s)
| | - Daphne Peelen
- Department of Haematology, Mater Dei Hospital, Msida, Malta VU University Amsterdam, Amsterdam, The Netherlands
| | | | - Sylvana Abela
- Department of Haematology, Mater Dei Hospital, Msida, Malta
| | | | | | - Daniel Zammit
- Department of Haematology, Mater Dei Hospital, Msida, Malta
| | | | - Alex Gatt
- Department of Haematology, Mater Dei Hospital, Msida, Malta
| |
Collapse
|
137
|
Chang JC. A Thought on Possible Pathogenesis of Ebola Viral Hemorrhagic Disease and Potential Treatments: Could it Be Thrombotic Thrombocytopenic Purpura-Like Syndrome? Ther Apher Dial 2015; 20:93-8. [DOI: 10.1111/1744-9987.12350] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2014] [Revised: 05/06/2015] [Accepted: 06/18/2015] [Indexed: 11/26/2022]
Affiliation(s)
- Jae C. Chang
- Department of Medicine; University of California Irvine School of Medicine; Irvine California
| |
Collapse
|
138
|
Bird BH, Spengler JR, Chakrabarti AK, Khristova ML, Sealy TK, Coleman-McCray JD, Martin BE, Dodd KA, Goldsmith CS, Sanders J, Zaki SR, Nichol ST, Spiropoulou CF. Humanized Mouse Model of Ebola Virus Disease Mimics the Immune Responses in Human Disease. J Infect Dis 2015; 213:703-11. [PMID: 26582961 DOI: 10.1093/infdis/jiv538] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Accepted: 10/01/2015] [Indexed: 11/14/2022] Open
Abstract
Animal models recapitulating human Ebola virus disease (EVD) are critical for insights into virus pathogenesis. Ebola virus (EBOV) isolates derived directly from human specimens do not, without adaptation, cause disease in immunocompetent adult rodents. Here, we describe EVD in mice engrafted with human immune cells (hu-BLT). hu-BLT mice developed EVD following wild-type EBOV infection. Infection with high-dose EBOV resulted in rapid, lethal EVD with high viral loads, alterations in key human antiviral immune cytokines and chemokines, and severe histopathologic findings similar to those shown in the limited human postmortem data available. A dose- and donor-dependent clinical course was observed in hu-BLT mice infected with lower doses of either Mayinga (1976) or Makona (2014) isolates derived from human EBOV cases. Engraftment of the human cellular immune system appeared to be essential for the observed virulence, as nonengrafted mice did not support productive EBOV replication or develop lethal disease. hu-BLT mice offer a unique model for investigating the human immune response in EVD and an alternative animal model for EVD pathogenesis studies and therapeutic screening.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Cynthia S Goldsmith
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jeanine Sanders
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Sherif R Zaki
- Infectious Diseases Pathology Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, Georgia
| | | | | |
Collapse
|
139
|
The Role of Cytokines and Chemokines in Filovirus Infection. Viruses 2015; 7:5489-507. [PMID: 26512687 PMCID: PMC4632400 DOI: 10.3390/v7102892] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 10/09/2015] [Accepted: 10/14/2015] [Indexed: 01/11/2023] Open
Abstract
Ebola- and marburgviruses are highly pathogenic filoviruses and causative agents of viral hemorrhagic fever. Filovirus disease is characterized by a dysregulated immune response, severe organ damage, and coagulation abnormalities. This includes modulation of cytokines, signaling mediators that regulate various components of the immune system as well as other biological processes. Here we examine the role of cytokines in filovirus infection, with an emphasis on understanding how these molecules affect development of the antiviral immune response and influence pathology. These proteins may present targets for immune modulation by therapeutic agents and vaccines in an effort to boost the natural immune response to infection and/or reduce immunopathology.
Collapse
|
140
|
Ocular Manifestations of Ebola Virus Disease: An Ophthalmologist's Guide to Prevent Infection and Panic. BIOMED RESEARCH INTERNATIONAL 2015; 2015:487073. [PMID: 26557674 PMCID: PMC4628748 DOI: 10.1155/2015/487073] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 12/09/2014] [Accepted: 03/11/2015] [Indexed: 11/27/2022]
Abstract
Ebola virus disease (EVD—formerly known as Ebola hemorrhagic fever) is a severe hemorrhagic fever caused by lipid-enveloped, nonsegmented, negative-stranded RNA viruses belonging to the genus Ebolavirus. Case fatality rates may reach up to 76% of infected individuals, making this infection a deadly health problem in the sub-Saharan population. At the moment, there are still no indications on ophthalmological clinical signs and security suggestions for healthcare professionals (doctors and nurses or cooperative persons). This paper provides a short but complete guide to reduce infection risks.
Collapse
|
141
|
Martins K, Cooper C, Warren T, Wells J, Bell T, Raymond J, Stuthman K, Benko J, Garza N, van Tongeren S, Donnelly G, Retterer C, Dong L, Bavari S. Characterization of clinical and immunological parameters during Ebola virus infection of rhesus macaques. Viral Immunol 2015; 28:32-41. [PMID: 25514385 DOI: 10.1089/vim.2014.0085] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The rhesus macaque serves as an animal model for Ebola virus (EBOV) infection. A thorough understanding of EBOV infection in this species would aid in further development of filovirus therapeutics and vaccines. In this study, pathological and immunological data from EBOV-infected rhesus macaques are presented. Changes in blood chemistries, hematology, coagulation, and immune parameters during infection, which were consistently observed in the animals, are presented. In an animal that survived challenge, a delay was observed in the detection of viral RNA and inflammatory cytokines and chemokines which may have contributed to survival. Collectively, these data add to the body of knowledge regarding EBOV pathogenesis in rhesus macaques and emphasize the reproducibility of the rhesus macaque challenge model.
Collapse
Affiliation(s)
- Karen Martins
- 1 Department of Molecular and Translational Sciences, United States Army Medical Research Institute of Infectious Diseases (USAMRIID) , Frederick, Maryland
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Tazawa H, Sato K, Tsutiya A, Tokeshi M, Ohtani-Kaneko R. A microfluidic cell culture system for monitoring of sequential changes in endothelial cells after heat stress. Thromb Res 2015; 136:328-34. [DOI: 10.1016/j.thromres.2015.05.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2015] [Revised: 04/30/2015] [Accepted: 05/11/2015] [Indexed: 12/23/2022]
|
143
|
Addressing Therapeutic Options for Ebola Virus Infection in Current and Future Outbreaks. Antimicrob Agents Chemother 2015; 59:5892-902. [PMID: 26248374 DOI: 10.1128/aac.01105-15] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Ebola virus can cause severe hemorrhagic disease with high fatality rates. Currently, no specific therapeutic agent or vaccine has been approved for treatment and prevention of Ebola virus infection of humans. Although the number of Ebola cases has fallen in the last few weeks, multiple outbreaks of Ebola virus infection and the likelihood of future exposure highlight the need for development and rapid evaluation of pre- and postexposure treatments. Here, we briefly review the existing and future options for anti-Ebola therapy, based on the data coming from rare clinical reports, studies on animals, and results from in vitro models. We also project the mechanistic hypotheses of several potential drugs against Ebola virus, including small-molecule-based drugs, which are under development and being tested in animal models or in vitro using various cell types. Our paper discusses strategies toward identifying and testing anti-Ebola virus properties of known and medically approved drugs, especially those that can limit the pathological inflammatory response in Ebola patients and thereby provide protection from mortality. We underline the importance of developing combinational therapy for better treatment outcomes for Ebola patients.
Collapse
|
144
|
Animal Models for the Study of Rodent-Borne Hemorrhagic Fever Viruses: Arenaviruses and Hantaviruses. BIOMED RESEARCH INTERNATIONAL 2015; 2015:793257. [PMID: 26266264 PMCID: PMC4523679 DOI: 10.1155/2015/793257] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 06/14/2015] [Indexed: 11/20/2022]
Abstract
Human pathogenic hantaviruses and arenaviruses are maintained in nature by persistent infection of rodent carrier populations. Several members of these virus groups can cause significant disease in humans that is generically termed viral hemorrhagic fever (HF) and is characterized as a febrile illness with an increased propensity to cause acute inflammation. Human interaction with rodent carrier populations leads to infection. Arenaviruses are also viewed as potential biological weapons threat agents. There is an increased interest in studying these viruses in animal models to gain a deeper understating not only of viral pathogenesis, but also for the evaluation of medical countermeasures (MCM) to mitigate disease threats. In this review, we examine current knowledge regarding animal models employed in the study of these viruses. We include analysis of infection models in natural reservoirs and also discuss the impact of strain heterogeneity on the susceptibility of animals to infection. This information should provide a comprehensive reference for those interested in the study of arenaviruses and hantaviruses not only for MCM development but also in the study of viral pathogenesis and the biology of these viruses in their natural reservoirs.
Collapse
|
145
|
Matranga CB, Andersen KG, Winnicki S, Busby M, Gladden AD, Tewhey R, Stremlau M, Berlin A, Gire SK, England E, Moses LM, Mikkelsen TS, Odia I, Ehiane PE, Folarin O, Goba A, Kahn SH, Grant DS, Honko A, Hensley L, Happi C, Garry RF, Malboeuf CM, Birren BW, Gnirke A, Levin JZ, Sabeti PC. Enhanced methods for unbiased deep sequencing of Lassa and Ebola RNA viruses from clinical and biological samples. Genome Biol 2015. [PMID: 25403361 PMCID: PMC4262991 DOI: 10.1186/s13059-014-0519-7] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
We have developed a robust RNA sequencing method for generating complete de novo assemblies with intra-host variant calls of Lassa and Ebola virus genomes in clinical and biological samples. Our method uses targeted RNase H-based digestion to remove contaminating poly(rA) carrier and ribosomal RNA. This depletion step improves both the quality of data and quantity of informative reads in unbiased total RNA sequencing libraries. We have also developed a hybrid-selection protocol to further enrich the viral content of sequencing libraries. These protocols have enabled rapid deep sequencing of both Lassa and Ebola virus and are broadly applicable to other viral genomics studies.
Collapse
|
146
|
Matranga CB, Andersen KG, Winnicki S, Busby M, Gladden AD, Tewhey R, Stremlau M, Berlin A, Gire SK, England E, Moses LM, Mikkelsen TS, Odia I, Ehiane PE, Folarin O, Goba A, Kahn SH, Grant DS, Honko A, Hensley L, Happi C, Garry RF, Malboeuf CM, Birren BW, Gnirke A, Levin JZ, Sabeti PC. Enhanced methods for unbiased deep sequencing of Lassa and Ebola RNA viruses from clinical and biological samples. Genome Biol 2015; 15:519. [PMID: 25403361 DOI: 10.1186/preaccept-1698056557139770] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Indexed: 12/20/2022] Open
Abstract
We have developed a robust RNA sequencing method for generating complete de novo assemblies with intra-host variant calls of Lassa and Ebola virus genomes in clinical and biological samples. Our method uses targeted RNase H-based digestion to remove contaminating poly(rA) carrier and ribosomal RNA. This depletion step improves both the quality of data and quantity of informative reads in unbiased total RNA sequencing libraries. We have also developed a hybrid-selection protocol to further enrich the viral content of sequencing libraries. These protocols have enabled rapid deep sequencing of both Lassa and Ebola virus and are broadly applicable to other viral genomics studies.
Collapse
|
147
|
Affiliation(s)
- Baijayantimala Mishra
- Department of Microbiology, All India Institute of Medical Sciences, Bhubaneswar 751 019, Orissa, India
| |
Collapse
|
148
|
Getts DR, Shea LD, Miller SD, King NJC. Harnessing nanoparticles for immune modulation. Trends Immunol 2015; 36:419-27. [PMID: 26088391 PMCID: PMC4603374 DOI: 10.1016/j.it.2015.05.007] [Citation(s) in RCA: 168] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 05/15/2015] [Accepted: 05/17/2015] [Indexed: 01/18/2023]
Abstract
NPs can be generated from numerous biocompatible compounds. Specific physiochemical characteristics can be manipulated to modulate the immune response. Severe inflammation can be treated using NP-based approaches. Antigen delivery via NPs can restore peripheral immune tolerance.
Recent approaches using nanoparticles engineered for immune regulation have yielded promising results in preclinical models of disease. The number of nanoparticle therapies is growing, fueled by innovations in nanotechnology and advances in understanding of the underlying pathogenesis of immune-mediated diseases. In particular, recent mechanistic insight into the ways in which nanoparticles interact with the mononuclear phagocyte system and impact its function during homeostasis and inflammation have highlighted the potential of nanoparticle-based therapies for controlling severe inflammation while concurrently restoring peripheral immune tolerance in autoimmune disease. Here we review recent advances in nanoparticle-based approaches aimed at immune-modulation, and discuss these in the context of concepts in polymeric nanoparticle development, including particle modification, delivery and the factors associated with successful clinical deployment.
Collapse
Affiliation(s)
- Daniel R Getts
- The Discipline of Pathology, School of Medical Sciences, Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia; Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Cour Pharmaceutical Development Company, Elmhurst, IL, USA.
| | - Lonnie D Shea
- Department of Chemical and Biomedical Engineering, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA; Department of Biomedical Engineering, University of Michigan, 2200 Bonisteel Boulevard, Ann Arbor, MI 48109, USA
| | - Stephen D Miller
- Department of Microbiology-Immunology and Interdepartmental Immunobiology Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Nicholas J C King
- The Discipline of Pathology, School of Medical Sciences, Bosch Institute, Sydney Medical School, The University of Sydney, Sydney, NSW 2006, Australia
| |
Collapse
|
149
|
Qureshi AI, Chughtai M, Loua TO, Pe Kolie J, Camara HFS, Ishfaq MF, N'Dour CT, Beavogui K. Study of Ebola Virus Disease Survivors in Guinea. Clin Infect Dis 2015; 61:1035-42. [PMID: 26060289 DOI: 10.1093/cid/civ453] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Accepted: 05/18/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND There is a paucity of data regarding health consequences of Ebola virus disease among survivors. METHODS We surveyed 105 Ebola virus disease survivors postdischarge from an Ebola treatment unit in Guinea using a standard data collection form. Patients rated recovery as the percentage of improvement in functional status, where 0% represents "unable to perform" and 100% represents "able to perform at prior level." RESULTS The mean ± standard deviation time interval between hospital discharge and administration of questionnaire was 103.5 ± 47.9 days in 105 survivors. Anorexia was reported by 103 patients, with varying severity levels: mild (n = 33), moderate (n = 65), or severe (n = 5). Reported pain according to site was chest (30.7%), joint (86.7%), muscle (26.7%), and back (45.7%), among others. Recovery in functional status was graded as mild (10%-30%) (n = 2 [1.9%]), moderate (40%-70%) (n = 52 [50.0%]), and excellent (80%-100%) (n = 50 [48.1%]). Severity of arthralgia (R(2) = 0.09; P = .008) was directly associated with lower recovery in functional status in multivariate analysis. CONCLUSIONS Ebola virus disease survivors frequently reported anorexia and arthralgia. Severity of arthralgia was related to lower functional recovery. There may be a role for focused screening and intervention for symptoms identified in this study of survivors.
Collapse
Affiliation(s)
- Adnan I Qureshi
- Zeenat Qureshi Clinical Neuroscience Institute, Donka National Hospital, Conakry, Guinea Zeenat Qureshi Stroke Institute, St Cloud, Minnesota
| | - Morad Chughtai
- Rubin Institute of Advanced Orthopedics, Sinai Hospital, Baltimore, Maryland
| | | | - Jean Pe Kolie
- Department of Visceral Surgery, China-Guinea Friendship Hospital
| | | | | | - Cheikh Tidane N'Dour
- Department of Infectious Diseases, University of Cheikh Anta Diop of Dakar, Sengal
| | - Kezely Beavogui
- Department of Neurosurgery, Donka National Hospital National Program to Combat Trauma and Violence in Guinea, Ministry of Health and World Health Organization, Conakry
| |
Collapse
|
150
|
Erickson AK, Pfeiffer JK. Spectrum of disease outcomes in mice infected with YFV-17D. J Gen Virol 2015; 96:1328-1339. [PMID: 25646269 PMCID: PMC4635484 DOI: 10.1099/vir.0.000075] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2014] [Accepted: 01/27/2015] [Indexed: 11/18/2022] Open
Abstract
The host and viral factors that influence disease outcome during flavivirus infections are not fully understood. Using the live attenuated yellow fever virus (YFV) vaccine strain 17D as a model system we evaluated how viral dose, inoculation route and immunopathogenesis contributed to disease outcome in mice deficient in the type I IFN response. We found that YFV-17D infection of IFN-α/β receptor knockout mice resulted in three distinct disease outcomes: no clinical signs of disease, fatal viscerotropic disease or fatal neurotropic disease. Interestingly, viral load at disease onset did not correlate with disease outcome. However, we found increased immune infiltrates in the brain tissues of mice that developed neurotropic disease. Additionally, mice that developed viscerotropic disease, as characterized by liver and spleen pathology and/or intestinal haemorrhage, had significantly elevated levels of alanine aminotransferase, monocyte chemotactic protein and IFN-inducible protein (IP)-10 as compared with mice with no clinical signs of disease or neurotropic disease. Furthermore, mice treated with recombinant IP-10 throughout YFV-17D infection showed increased mortality and an increased percentage of mice with viscerotropic disease. Our results demonstrated that viral load did not correlate with pathogenesis, and the host immune response played a pivotal role in disease outcome and contributed to YFV-17D pathogenesis in mice.
Collapse
|