1
|
Wang B, Shen WB, Seif KE, Townsel C, Baracco L, Logue J, Reece EA, Frieman MB, Turan S, Yang P. Placental SARS-CoV-2 Infection and Its Implications for Increased Risk of Adverse Pregnancy Outcomes. Am J Perinatol 2025; 42:52-59. [PMID: 38729183 DOI: 10.1055/a-2323-0854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/12/2024]
Abstract
OBJECTIVE Pregnant women are at increased risk of coronavirus disease 2019 (COVID-19). This could be explained through the prism of physiologic and immunologic changes in pregnancy. In addition, certain immunological reactions originate in the placenta in response to viral infections.This study aimed to investigate whether severe acute respiratory syndrome-coronavirus-2 (SARS-CoV-2) can infect the human placenta and discuss its implications in the pathogenesis of adverse pregnancy outcomes. STUDY DESIGN We conducted a retrospective cohort study in which we collected placental specimens from pregnant women who had a laboratory-confirmed SARS-CoV-2 infection. We performed RNA in situ hybridization assay on formalin-fixed paraffin-embedded tissues to establish the in vivo evidence for placental infectivity by this corona virus. In addition, we infected trophoblast isolated from uninfected term human placenta with SARS-CoV-2 variants to further provide in vitro evidence for such an infectivity. RESULTS There was a total of 21 cases enrolled, which included 5 cases of spontaneous preterm birth (SPTB) and 2 intrauterine fetal demises (IUFDs). Positive staining of positive-sense strand of SARS-CoV-2 virions was detected in 15 placentas including 4 SPTB and both IUFDs. In vitro infection assay demonstrated that SARS-CoV-2 virions were highly capable of infecting both cytotrophoblast and syncytiotrophoblast. CONCLUSION This study implies that placental SARS-CoV-2 infection may be associated with an increased risk of adverse obstetrical outcomes. KEY POINTS · SARS-CoV-2 can effectively infect human placenta.. · Such infectivity is confirmed by in vitro experiments.. · Placental SARS-CoV-2 corelates with adverse obstetrical outcomes..
Collapse
Affiliation(s)
- Bingbing Wang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Wei-Bin Shen
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Karl E Seif
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Courtney Townsel
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Lauren Baracco
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - James Logue
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - E Albert Reece
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Matthew B Frieman
- Department of Microbiology and Immunology, Center for Pathogen Research, University of Maryland School of Medicine, Baltimore, Maryland
| | - Sifa Turan
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| | - Peixin Yang
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, Maryland
| |
Collapse
|
2
|
Kleymann A, Karaaslan E, Scholte FEM, Sorvillo TE, Welch SR, Bergeron É, Elser S, Almanzar-Jordan MR, Velazquez E, Genzer SC, Jean SM, Spiropoulou CF, Spengler JR. Crimean-Congo hemorrhagic fever virus replicon particle vaccine is safe and elicits functional, non-neutralizing anti-nucleoprotein antibodies and T cell activation in rhesus macaques. Antiviral Res 2025; 233:106045. [PMID: 39626793 DOI: 10.1016/j.antiviral.2024.106045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 11/24/2024] [Accepted: 11/30/2024] [Indexed: 12/15/2024]
Abstract
Advancement of vaccine candidates that demonstrate protective efficacy in screening studies necessitates detailed safety and immunogenicity investigations in pre-clinical models. A non-spreading Crimean-Congo hemorrhagic fever virus (CCHFV) viral replicon particle (VRP) vaccine was developed for single-dose administration to protect against disease. To date, several studies have supported safety, immunogenicity, and efficacy of the CCHF VRP in multiple highly sensitive murine models of lethal disease, but the VRP had yet to be evaluated in large animals. Here, we performed studies in non-human primates to further evaluate clinical utility of the VRP vaccine. Twelve adult male and female rhesus macaques were vaccinated intramuscularly and clinical monitoring was performed daily for 28 days. At 3, 7, 14, 21, and 28 days post vaccination, animals were sedated for more detailed clinical assessment; for quantification of vaccine presence in blood and mucosal samples; and for evaluation of hematology, plasma inflammatory markers, and immunogenicity. Consistent with findings in mice, vaccination was well tolerated, with no clinical alterations nor indication of vaccine spread or shedding. In addition, vaccination induced both humoral and cell-mediated responses, with immune profile and kinetics also corroborating data from small animal models. These studies provide key data in non-human primates further supporting development of the VRP for human clinical use.
Collapse
Affiliation(s)
- Alyssa Kleymann
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Elif Karaaslan
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Florine E M Scholte
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Teresa E Sorvillo
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA; Infectious Disease Department, CDC Foundation, Atlanta, GA, USA
| | - Stephen R Welch
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Stephanie Elser
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Melvyn R Almanzar-Jordan
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Eric Velazquez
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Sarah C Genzer
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Sherrie M Jean
- Comparative Medicine Branch, Division of Scientific Resources, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| |
Collapse
|
3
|
Yu KM, Park SJ. Tick-borne viruses: Epidemiology, pathogenesis, and animal models. One Health 2024; 19:100903. [PMID: 39391267 PMCID: PMC11465198 DOI: 10.1016/j.onehlt.2024.100903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/22/2024] [Accepted: 09/22/2024] [Indexed: 10/12/2024] Open
Abstract
Tick-borne viruses, capable of infecting animals and humans, are expanding geographically and increasing in prevalence, posing significant global public health threats. This review explores the current epidemiology of human pathogenic tick-borne viruses, emphasizing their diversity and the spectrum of symptomatic manifestations in humans, which range from mild to severe. We highlight how the infrequent and unpredictable nature of viral outbreaks complicates the precise identification and understanding of these viruses in human infections. Furthermore, we describe the utility of animal models that accurately mimic human clinical symptoms, facilitating the development of effective control strategies. Our comprehensive analysis provides crucial insights into disease progression and emphasizes the urgent need for continued research. This work aims to provide insight into knowledge gaps to mitigate the health burden of tick-borne infections and open an avenue for further study to enhance our understanding of these emerging infectious diseases.
Collapse
Affiliation(s)
- Kwang-Min Yu
- Research Institute of Molecular Alchemy (RIMA), Gyeongsang National University, Jinju 52828, Republic of Korea
| | - Su-Jin Park
- Research Institute of Molecular Alchemy (RIMA), Gyeongsang National University, Jinju 52828, Republic of Korea
- Division of Life Science, Gyeongsang National University, Jinju 52828, Republic of Korea
| |
Collapse
|
4
|
Li H, Pinette M, Smith G, Goolia M, Handel K, Nebroski M, Lung O, Pickering BS. Distinguishing host responses, extensive viral dissemination and long-term viral RNA persistence in domestic sheep experimentally infected with Crimean-Congo haemorrhagic fever virus Kosovo Hoti. Emerg Microbes Infect 2024; 13:2302103. [PMID: 38189080 PMCID: PMC10810640 DOI: 10.1080/22221751.2024.2302103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/31/2023] [Indexed: 01/09/2024]
Abstract
Crimean-Congo haemorrhagic fever orthonairovirus (CCHFV) is a tick-borne, risk group 4 pathogen that often causes a severe haemorrhagic disease in humans (CCHF) with high case fatality rates. The virus is believed to be maintained in a tick-vertebrate-tick ecological cycle involving numerous wild and domestic animal species; however the biology of CCHFV infection in these animals remains poorly understood. Here, we experimentally infect domestic sheep with CCHFV Kosovo Hoti, a clinical isolate representing high pathogenicity to humans and increasingly utilized in current research. In the absence of prominent clinical signs, the infection leads to an acute viremia and coinciding viral shedding, fever and markers for potential impairment in liver and kidney functions. A number of host responses distinguish the subclinical infection in sheep versus fatal infection in humans. These include an early reduction of neutrophil recruitment and its chemoattractant, IL-8, in the blood stream of infected sheep, whereas neutrophil infiltration and elevated IL-8 are features of fatal CCHFV infections reported in immunodeficient mice and humans. Several inflammatory cytokines that correlate with poor disease outcomes in humans and have potential to cause vascular dysfunction, a primary hallmark of severe CCHF, are down-regulated or restricted from increasing in sheep. Of particular interest, the detection of CCHFV RNA (including full-length genome) in a variety of sheep tissues long after the acute phase of infection indicates a widespread viral dissemination in the host and suggests a potentially long-term persisting impact of CCHFV infection. These findings reveal previously unrecognized aspects of CCHFV biology in animals.
Collapse
Affiliation(s)
- Hongzhao Li
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Mathieu Pinette
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Greg Smith
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Melissa Goolia
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Katherine Handel
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Michelle Nebroski
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Oliver Lung
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
| | - Bradley S. Pickering
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, Canada
- Department of Medical Microbiology and Infectious Diseases, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, Canada
| |
Collapse
|
5
|
Wang S, Li W, Wang Z, Yang W, Li E, Xia X, Yan F, Chiu S. Emerging and reemerging infectious diseases: global trends and new strategies for their prevention and control. Signal Transduct Target Ther 2024; 9:223. [PMID: 39256346 PMCID: PMC11412324 DOI: 10.1038/s41392-024-01917-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/13/2024] [Accepted: 07/05/2024] [Indexed: 09/12/2024] Open
Abstract
To adequately prepare for potential hazards caused by emerging and reemerging infectious diseases, the WHO has issued a list of high-priority pathogens that are likely to cause future outbreaks and for which research and development (R&D) efforts are dedicated, known as paramount R&D blueprints. Within R&D efforts, the goal is to obtain effective prophylactic and therapeutic approaches, which depends on a comprehensive knowledge of the etiology, epidemiology, and pathogenesis of these diseases. In this process, the accessibility of animal models is a priority bottleneck because it plays a key role in bridging the gap between in-depth understanding and control efforts for infectious diseases. Here, we reviewed preclinical animal models for high priority disease in terms of their ability to simulate human infections, including both natural susceptibility models, artificially engineered models, and surrogate models. In addition, we have thoroughly reviewed the current landscape of vaccines, antibodies, and small molecule drugs, particularly hopeful candidates in the advanced stages of these infectious diseases. More importantly, focusing on global trends and novel technologies, several aspects of the prevention and control of infectious disease were discussed in detail, including but not limited to gaps in currently available animal models and medical responses, better immune correlates of protection established in animal models and humans, further understanding of disease mechanisms, and the role of artificial intelligence in guiding or supplementing the development of animal models, vaccines, and drugs. Overall, this review described pioneering approaches and sophisticated techniques involved in the study of the epidemiology, pathogenesis, prevention, and clinical theatment of WHO high-priority pathogens and proposed potential directions. Technological advances in these aspects would consolidate the line of defense, thus ensuring a timely response to WHO high priority pathogens.
Collapse
Affiliation(s)
- Shen Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Wujian Li
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin University, Changchun, Jilin, China
| | - Zhenshan Wang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin, China
| | - Wanying Yang
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Entao Li
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China
| | - Xianzhu Xia
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China
| | - Feihu Yan
- Key Laboratory of Jilin Province for Zoonosis Prevention and Control, Changchun Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Changchun, 130000, China.
| | - Sandra Chiu
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, Anhui, China.
- Key Laboratory of Anhui Province for Emerging and Reemerging Infectious Diseases, Hefei, 230027, Anhui, China.
- Department of Laboratory Medicine, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| |
Collapse
|
6
|
Cohen CA, Balinandi S, Kuehne AI, Rock ML, Bonagofski LG, Ricks KM, Davis I, Abelson D, Stonier SW, Odongo M, Bornholdt ZA, Zeitlin L, Moyer C, Cose S, Dye JM, Lutwama JJ, Herbert AS. A Longitudinal Analysis of Memory Immune Responses in Convalescent Crimean-Congo Hemorrhagic Fever Survivors in Uganda. J Infect Dis 2024:jiae395. [PMID: 39248523 DOI: 10.1093/infdis/jiae395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 08/08/2024] [Indexed: 09/10/2024] Open
Abstract
Evaluating the adaptive immune responses to natural infection with Crimean-Congo hemorrhagic fever (CCHF) virus (CCHFV) in human survivors is critical to the development of medical countermeasures. However, the correlates of protection are unknown. As the most prevalent tick-borne human hemorrhagic fever virus with case fatality rates of 5%-30% and worldwide distribution, there is an urgent need to fill these knowledge gaps. Here, we describe adaptive immune responses in a cohort of Ugandan CCHF survivors via serial sampling over 6 years. We demonstrate persistent antibodies after infection and cross-neutralization against various clades of authentic CCHFV, as well as potent effector function. Moreover, we show for the first time persistent, polyfunctional antigen-specific memory T-cell responses to multiple CCHFV proteins up to 9 years after infection. Together, this data provides immunological benchmarks for evaluating CCHFV medical countermeasures and information that can be leveraged toward vaccine immunogen design and viral target identification for monoclonal antibody therapies.
Collapse
Affiliation(s)
- Courtney A Cohen
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, USA
| | - Stephen Balinandi
- Department of Arbovirology, Medical Research Council/Uganda Viral Research Institute, Entebbe, Uganda
| | - Ana I Kuehne
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, USA
| | - Michelle L Rock
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, USA
- The Geneva Foundation, Tacoma, Washington, USA
| | - Luke G Bonagofski
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, USA
| | - Keersten M Ricks
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, USA
| | - Ian Davis
- Diagnostic Systems Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, USA
| | | | - Spencer W Stonier
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, USA
| | - Matthew Odongo
- Department of Arbovirology, Medical Research Council/Uganda Viral Research Institute, Entebbe, Uganda
| | | | | | | | - Stephen Cose
- Department of Arbovirology, Medical Research Council/Uganda Viral Research Institute, Entebbe, Uganda
- London School of Hygiene & Tropical Medicine Uganda Research Unit, Entebbe, Uganda
| | - John M Dye
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, USA
| | - Julius J Lutwama
- Department of Arbovirology, Medical Research Council/Uganda Viral Research Institute, Entebbe, Uganda
| | - Andrew S Herbert
- Virology Division, US Army Medical Research Institute of Infectious Disease, Fort Detrick, Maryland, USA
| |
Collapse
|
7
|
Hawman DW, Leventhal SS, Meade-White K, Khandhar A, Murray J, Lovaglio J, Shaia C, Saturday G, Hinkley T, Erasmus J, Feldmann H. A replicating RNA vaccine confers protection in a rhesus macaque model of Crimean-Congo hemorrhagic fever. NPJ Vaccines 2024; 9:86. [PMID: 38769294 PMCID: PMC11106275 DOI: 10.1038/s41541-024-00887-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/07/2024] [Indexed: 05/22/2024] Open
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is a tick-borne febrile illness with a wide geographic distribution. In recent years the geographic range of Crimean-Congo hemorrhagic fever virus (CCHFV) and its tick vector have increased, placing an increasing number of people at risk of CCHFV infection. Currently, there are no widely available vaccines, and although the World Health Organization recommends ribavirin for treatment, its efficacy is unclear. Here we evaluate a promising replicating RNA vaccine in a rhesus macaque (Macaca mulatta) model of CCHF. This model provides an alternative to the established cynomolgus macaque model and recapitulates mild-to-moderate human disease. Rhesus macaques infected with CCHFV consistently exhibit viremia, detectable viral RNA in a multitude of tissues, and moderate pathology in the liver and spleen. We used this model to evaluate the immunogenicity and protective efficacy of a replicating RNA vaccine. Rhesus macaques vaccinated with RNAs expressing the CCHFV nucleoprotein and glycoprotein precursor developed robust non-neutralizing humoral immunity against the CCHFV nucleoprotein and had significant protection against the CCHFV challenge. Together, our data report a model of CCHF using rhesus macaques and demonstrate that our replicating RNA vaccine is immunogenic and protective in non-human primates after a prime-boost immunization.
Collapse
Affiliation(s)
- David W Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, 59840, USA.
| | - Shanna S Leventhal
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, 59840, USA
| | - Kimberly Meade-White
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, 59840, USA
| | | | - Justin Murray
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, 59840, USA
| | - Jamie Lovaglio
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, 59840, USA
| | - Carl Shaia
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, 59840, USA
| | - Greg Saturday
- Rocky Mountain Veterinary Branch, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, 59840, USA
| | | | | | - Heinz Feldmann
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, 59840, USA.
| |
Collapse
|
8
|
Welch SR, Garrison AR, Bente DA, Burt F, D'Addiego J, Devignot S, Dowall S, Fischer K, Hawman DW, Hewson R, Mirazimi A, Oestereich L, Vatansever Z, Spengler JR, Papa A. Third International Conference on Crimean-Congo Hemorrhagic Fever in Thessaloniki, Greece, September 19-21, 2023. Antiviral Res 2024; 225:105844. [PMID: 38428749 DOI: 10.1016/j.antiviral.2024.105844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 02/19/2024] [Accepted: 02/25/2024] [Indexed: 03/03/2024]
Abstract
The Third International Conference on Crimean-Congo Hemorrhagic Fever (CCHF) was held in Thessaloniki, Greece, September 19-21, 2023, bringing together a diverse group of international partners, including public health professionals, clinicians, ecologists, epidemiologists, immunologists, and virologists. The conference was attended by 118 participants representing 24 countries and the World Health Organization (WHO). Meeting sessions covered the epidemiology of CCHF in humans; Crimean-Congo hemorrhagic fever virus (CCHFV) in ticks; wild and domestic animal hosts; molecular virology; pathogenesis and animal models; immune response related to therapeutics; and CCHF prevention in humans. The concluding session focused on recent WHO recommendations regarding disease prevention, control strategies, and innovations against CCHFV outbreaks. This meeting report summarizes lectures by the invited speakers and highlights advances in the field.
Collapse
Affiliation(s)
- Stephen R Welch
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA.
| | - Aura R Garrison
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, MD, USA
| | - Dennis A Bente
- Galveston National Laboratory, Department of Microbiology and Immunology, Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Felicity Burt
- Division of Virology, National Health Laboratory Service and Division of Virology, University of the Free State, Bloemfontein, South Africa
| | - Jake D'Addiego
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Stephanie Devignot
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Stuart Dowall
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Kerstin Fischer
- Institute of Novel and Emerging Infectious Diseases, Friedrich-Loeffler-Institut, Federal Research Institute of Animal Health, Greifswald-Insel Riems, Germany
| | - David W Hawman
- Laboratory of Virology, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rocky Mountain Laboratories, Hamilton, MT, USA
| | - Roger Hewson
- UK Health Security Agency, Porton Down, Salisbury, Wiltshire, UK
| | - Ali Mirazimi
- Division of Clinical Microbiology, Department of Laboratory Medicine, Karolinska Institutet at Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Lisa Oestereich
- Bernhard Nocht Institute for Tropical Medicine and German Center for Infectious Research, Partner Sites Hamburg-Lübeck-Borstel-Riems, Hamburg, Germany
| | - Zati Vatansever
- Kafkas University, Faculty of Veterinary Medicine, Dept. of Parasitology, Kars, Turkey
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA, USA
| | - Anna Papa
- Department of Microbiology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
| |
Collapse
|
9
|
Cai N, Luo W, Ding L, Chen L, Huang Y. Obesity-related indicators and tuberculosis: A Mendelian randomization study. PLoS One 2024; 19:e0297905. [PMID: 38557966 PMCID: PMC10984409 DOI: 10.1371/journal.pone.0297905] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 01/15/2024] [Indexed: 04/04/2024] Open
Abstract
PURPOSE Obesity is a strong risk factor for many diseases, with controversy regarding the cause(s) of tuberculosis (TB) reflected by contradictory findings. Therefore, a larger sample population is required to determine the relationship between obesity and TB, which may further inform treatment. METHODS Obesity-related indicators and TB mutation data were obtained from a genome-wide association study database, while representative instrumental variables (IVs) were obtained by screening and merging. Causal relationships between exposure factors and outcomes were determined using two-sample Mendelian randomization (MR) analysis. Three tests were used to determine the representativeness and stability of the IVs, supported by sensitivity analysis. RESULTS Initially, 191 single nucleotide polymorphisms were designated as IVs by screening, followed by two-sample MR analysis, which revealed the causal relationship between waist circumference [odds ratio (OR): 2.13 (95% confidence interval (CI): 1.19-3.80); p = 0.011] and TB. Sensitivity analysis verified the credibility of the IVs, none of which were heterogeneous or horizontally pleiotropic. CONCLUSION The present study determined the causal effect between waist circumference and TB by two-sample MR analysis and found both to be likely to be potential risk factors.
Collapse
Affiliation(s)
- Nuannuan Cai
- Pulmonary and Critical Care Medicine, Hainan Provincial People’s Hospital, Haikou, Hainan, China
| | - Weiyan Luo
- Pulmonary and Critical Care Medicine, Hainan Provincial People’s Hospital, Haikou, Hainan, China
| | - Lili Ding
- Pulmonary and Critical Care Medicine, Hainan Provincial People’s Hospital, Haikou, Hainan, China
| | - Lijin Chen
- Pulmonary and Critical Care Medicine, Hainan Provincial People’s Hospital, Haikou, Hainan, China
| | - Yuanjiang Huang
- Infectious and Tropical Disease Dept (Tuberculosis), The Second Affiliated Hospital of Hainan Medical College, Haikou, Hainan, China
| |
Collapse
|
10
|
Ohta K, Saka N, Nishi Y, Nishio M. Nairovirus polymerase mutations associated with the establishment of persistent infection in human cells. J Virol 2024; 98:e0169823. [PMID: 38358288 PMCID: PMC10949423 DOI: 10.1128/jvi.01698-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/23/2024] [Indexed: 02/16/2024] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV), a tick-borne virus of the Orthonairovirus genus, persistently infects tick cells. It has been reported to establish persistent infection in non-human primates, but virological analysis has not yet been performed in human cells. Here, we investigated whether and how nairoviruses persistently infect human cells using Hazara orthonairovirus (HAZV), a surrogate model for CCHFV. We established a human cell line that was persistently infected with HAZV. Surprisingly, virions of persistently infected HAZV (HAZVpi) were not observed in the culture supernatants. There were five mutations (mut1, mut2, mut3, mut4, and mut5) in L protein of HAZVpi. Mutations in L protein of HAZVpi contribute to non-detection of virion in the supernatants. Lmut4 was found to cause low viral growth rate, despite its high polymerase activity. The low growth rate was restored by Lmut2, Lmut3, and Lmut5. The polymerase activity of Lmut1 was extremely low, and recombinant HAZV carrying Lmut1 (rHAZV/Lmut1) was not released into the supernatants. However, genomes of rHAZV/Lmut1 were retained in the infected cells. All mutations (Lmut1-5) found in L protein of HAZVpi were required for experimental reproduction of HAZVpi, and only Lmut1 and Lmut4 were insufficient. We demonstrated that point mutations in viral polymerase contribute to the establishment of persistent HAZV infection. Furthermore, innate immunity was found to be suppressed in HAZVpi-infected cells, which also potentially contributes to viral persistence. This is the first presentation of a possible mechanism behind how nairoviruses establish persistent infection in human cells. IMPORTANCE We investigated whether and how nairoviruses persistently infect human cells, using Hazara orthonairovirus (HAZV), a surrogate model for Crimean-Congo hemorrhagic fever virus. We established a human cell line that was persistently infected with HAZV. Five mutations were found in L protein of persistently infected HAZV (HAZVpi): mut1, mut2, mut3, mut4, and mut5. Among them, Lmut1 and Lmut4 restricted viral growth by low polymerase activity and low growth rate, respectively, leading to inhibition of viral overgrowth. The restriction of viral growth caused by Lmut1 and Lmut4 was compensated by other mutations, including Lmut2, Lmut3, and Lmut5. Each of the mutations found in L protein of HAZVpi was concluded to cooperatively modulate viral growth, which facilitates the establishment of persistent infection. Suppression of innate immunity also potentially contributes to virus persistence. This is the first presentation of a possible mechanism behind how nairoviruses establish persistent infection in human cells.
Collapse
Affiliation(s)
- Keisuke Ohta
- Department of Microbiology, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Naoki Saka
- Department of Microbiology, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Yuzuha Nishi
- Department of Microbiology, School of Medicine, Wakayama Medical University, Wakayama, Japan
| | - Machiko Nishio
- Department of Microbiology, School of Medicine, Wakayama Medical University, Wakayama, Japan
| |
Collapse
|
11
|
Bell TM, Facemire P, Bearss JJ, Raymond JL, Chapman J, Zeng X, Shamblin JD, Williams JA, Grosenbach DW, Hruby DE, Damon IK, Goff AJ, Mucker EM. Smallpox lesion characterization in placebo-treated and tecovirimat-treated macaques using traditional and novel methods. PLoS Pathog 2024; 20:e1012007. [PMID: 38386661 PMCID: PMC10883539 DOI: 10.1371/journal.ppat.1012007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 01/26/2024] [Indexed: 02/24/2024] Open
Abstract
Smallpox was the most rampant infectious disease killer of the 20th century, yet much remains unknown about the pathogenesis of the variola virus. Using archived tissue from a study conducted at the Centers for Disease Control and Prevention we characterized pathology in 18 cynomolgus macaques intravenously infected with the Harper strain of variola virus. Six macaques were placebo-treated controls, six were tecovirimat-treated beginning at 2 days post-infection, and six were tecovirimat-treated beginning at 4 days post-infection. All macaques were treated daily until day 17. Archived tissues were interrogated using immunohistochemistry, in situ hybridization, immunofluorescence, and electron microscopy. Gross lesions in three placebo-treated animals that succumbed to infection primarily consisted of cutaneous vesicles, pustules, or crusts with lymphadenopathy. The only gross lesions noted at the conclusion of the study in the three surviving placebo-treated and the Day 4 treated animals consisted of resolving cutaneous pox lesions. No gross lesions attributable to poxviral infection were present in the Day 2 treated macaques. Histologic lesions in three placebo-treated macaques that succumbed to infection consisted of proliferative and necrotizing dermatitis with intracytoplasmic inclusion bodies and lymphoid depletion. The only notable histologic lesion in the Day 4 treated macaques was resolving dermatitis; no notable lesions were seen in the Day 2 treated macaques. Variola virus was detected in all three placebo-treated animals that succumbed to infection prior to the study's conclusion by all utilized methods (IHC, ISH, IFA, EM). None of the three placebo-treated animals that survived to the end of the study nor the animals in the two tecovirimat treatment groups showed evidence of variola virus by these methods. Our findings further characterize variola lesions in the macaque model and describe new molecular methods for variola detection.
Collapse
Affiliation(s)
- Todd M. Bell
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Paul Facemire
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Jeremy J. Bearss
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Jo Lynne Raymond
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Jennifer Chapman
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Xiankun Zeng
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Joshua D. Shamblin
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Janice A. Williams
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | | | - Dennis E. Hruby
- SIGA Technologies, Inc., Corvallis, Oregon, United States of America
| | - Inger K. Damon
- Poxvirus and Rabies Branch, Centers for Disease Control and Prevention (CDC) Atlanta, Georgia, United States of America
| | - Arthur J. Goff
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| | - Eric M. Mucker
- U.S. Army Medical Research Institute of Infectious Diseases (USAMRIID), Frederick, Maryland, United States of America
| |
Collapse
|
12
|
Ahata B, Akçapınar GB. CCHFV vaccine development, current challenges, limitations, and future directions. Front Immunol 2023; 14:1238882. [PMID: 37753088 PMCID: PMC10518622 DOI: 10.3389/fimmu.2023.1238882] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Accepted: 08/22/2023] [Indexed: 09/28/2023] Open
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is the most prevalent tick-borne viral disease affecting humans. The disease is life-threatening in many regions of the developing world, including Africa, Asia, the Middle East, and Southern Europe. In line with the rapidly increasing disease prevalence, various vaccine strategies are under development. Despite a large number of potential vaccine candidates, there are no approved vaccines as of yet. This paper presents a detailed comparative analysis of current efforts to develop vaccines against CCHFV, limitations associated with current efforts, and future research directions.
Collapse
Affiliation(s)
- Büşra Ahata
- Department of Medical Biotechnology, Institute of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Türkiye
- Health Institutes of Turkey, Istanbul, Türkiye
| | - Günseli Bayram Akçapınar
- Department of Medical Biotechnology, Institute of Health Sciences, Acıbadem Mehmet Ali Aydınlar University, Istanbul, Türkiye
| |
Collapse
|
13
|
Ozdarendeli A. Crimean-Congo Hemorrhagic Fever Virus: Progress in Vaccine Development. Diagnostics (Basel) 2023; 13:2708. [PMID: 37627967 PMCID: PMC10453274 DOI: 10.3390/diagnostics13162708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/27/2023] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV), a member of the Nairoviridae family and Bunyavirales order, is transmitted to humans via tick bites or contact with the blood of infected animals. It can cause severe symptoms, including hemorrhagic fever, with a mortality rate between 5 to 30%. CCHFV is classified as a high-priority pathogen by the World Health Organization (WHO) due to its high fatality rate and the absence of effective medical countermeasures. CCHFV is endemic in several regions across the world, including Africa, Europe, the Middle East, and Asia, and has the potential for global spread. The emergence of the disease in new areas, as well as the presence of the tick vector in countries without reported cases, emphasizes the need for preventive measures to be taken. In the past, the lack of a suitable animal model susceptible to CCHFV infection has been a major obstacle in the development of vaccines and treatments. However, recent advances in biotechnology and the availability of suitable animal models have significantly expedited the development of vaccines against CCHF. These advancements have not only contributed to an enhanced understanding of the pathogenesis of CCHF but have also facilitated the evaluation of potential vaccine candidates. This review outlines the immune response to CCHFV and animal models utilized for the study of CCHFV and highlights the progress made in CCHFV vaccine studies. Despite remarkable advancements in vaccine development for CCHFV, it remains crucial to prioritize continued research, collaboration, and investment in this field.
Collapse
Affiliation(s)
- Aykut Ozdarendeli
- Department of Microbiology, Faculty of Medicine, Erciyes University, 38039 Kayseri, Türkiye;
- Vaccine Research, Development and Application Centre (ERAGEM), Erciyes University, 38039 Kayseri, Türkiye
| |
Collapse
|
14
|
Li H, Smith G, Goolia M, Marszal P, Pickering BS. Comparative characterization of Crimean-Congo hemorrhagic fever virus cell culture systems with application to propagation and titration methods. Virol J 2023; 20:128. [PMID: 37337294 DOI: 10.1186/s12985-023-02089-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Accepted: 06/02/2023] [Indexed: 06/21/2023] Open
Abstract
Crimean-Congo hemorrhagic fever orthonairovirus (CCHFV) is a biosafety level 4 and World Health Organization top priority pathogen. Infection leads to an often fatal hemorrhagic fever disease in humans. The tick-borne virus is endemic in countries across Asia, Europe and Africa, with signs of spreading into new regions. Despite the severity of disease and the potential of CCHFV geographic expansion to cause widespread outbreaks, no approved vaccine or treatment is currently available. Critical for basic research and the development of diagnostics or medical countermeasures, CCHFV viral stocks are commonly produced in Vero E6 and SW-13 cell lines. While a variety of in-house methods are being used across different laboratories, there has been no clear, specific consensus on a standard, optimal system for CCHFV growth and titration. In this study, we perform a systematic, side-by-side characterization of Vero E6 and SW-13 cell lines concerning the replication kinetics of CCHFV under different culture conditions. SW-13 cells are typically cultured in a CO2-free condition (SW-13 CO2-) according to the American Type Culture Collection. However, we identify a CO2-compatible culture condition (SW-13 CO2+) that demonstrates the highest viral load (RNA concentration) and titer (infectious virus concentration) in the culture supernatants, in comparison to SW-13 CO2- and Vero E6 cultures. This optimal viral propagation system also leads to the development of two titration methods: an immunostaining-based plaque assay using a commercial CCHFV antibody and a colorimetric readout, and an antibody staining-free, cytopathic effect-based median tissue culture infectious dose assay using a simple excel calculator. These are anticipated to serve as a basis for a reproducible, standardized and user-friendly platform for CCHFV propagation and titration.
Collapse
Affiliation(s)
- Hongzhao Li
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Greg Smith
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Melissa Goolia
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Peter Marszal
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada
| | - Bradley S Pickering
- National Centre for Foreign Animal Disease, Canadian Food Inspection Agency, Winnipeg, MB, Canada.
- Department of Medical Microbiology and Infectious Diseases, College of Medicine, Faculty of Health Sciences, University of Manitoba, Winnipeg, MB, Canada.
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA, USA.
| |
Collapse
|
15
|
Patel P, Nandi A, Verma SK, Kaushik N, Suar M, Choi EH, Kaushik NK. Zebrafish-based platform for emerging bio-contaminants and virus inactivation research. THE SCIENCE OF THE TOTAL ENVIRONMENT 2023; 872:162197. [PMID: 36781138 PMCID: PMC9922160 DOI: 10.1016/j.scitotenv.2023.162197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/08/2023] [Indexed: 05/27/2023]
Abstract
Emerging bio-contaminants such as viruses have affected health and environment settings of every country. Viruses are the minuscule entities resulting in severe contagious diseases like SARS, MERS, Ebola, and avian influenza. Recent epidemic like the SARS-CoV-2, the virus has undergone mutations strengthen them and allowing to escape from the remedies. Comprehensive knowledge of viruses is essential for the development of targeted therapeutic and vaccination treatments. Animal models mimicking human biology like non-human primates, rats, mice, and rabbits offer competitive advantage to assess risk of viral infections, chemical toxins, nanoparticles, and microbes. However, their economic maintenance has always been an issue. Furthermore, the redundancy of experimental results due to aforementioned aspects is also in examine. Hence, exploration for the alternative animal models is crucial for risk assessments. The current review examines zebrafish traits and explores the possibilities to monitor emerging bio-contaminants. Additionally, a comprehensive picture of the bio contaminant and virus particle invasion and abatement mechanisms in zebrafish and human cells is presented. Moreover, a zebrafish model to investigate the emerging viruses such as coronaviridae and poxviridae has been suggested.
Collapse
Affiliation(s)
- Paritosh Patel
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea
| | - Aditya Nandi
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Suresh K Verma
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India; Condensed Matter Theory Group, Materials Theory Division, Department of Physics and Astronomy, Uppsala University, Box 516, SE-751 20 Uppsala, Sweden
| | - Neha Kaushik
- Department of Biotechnology, College of Engineering, The University of Suwon, 18323 Hwaseong, Republic of Korea
| | - Mrutyunjay Suar
- School of Biotechnology, KIIT University, Bhubaneswar 751024, Odisha, India
| | - Eun Ha Choi
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| | - Nagendra Kumar Kaushik
- Plasma Bioscience Research Center, Department of Electrical and Biological Physics, Kwangwoon University, 01897 Seoul, South Korea.
| |
Collapse
|
16
|
Abstract
Crimean-Congo haemorrhagic fever (CCHF) is a severe tick-borne illness with a wide geographical distribution and case fatality rates of 30% or higher. Caused by infection with the CCHF virus (CCHFV), cases are reported throughout Africa, the Middle East, Asia and southern and eastern Europe. The expanding range of the Hyalomma tick vector is placing new populations at risk for CCHF, and no licensed vaccines or specific antivirals exist to treat CCHF. Furthermore, despite cases of CCHF being reported annually, the host and viral determinants of CCHFV pathogenesis are poorly understood. CCHFV can productively infect a multitude of animal species, yet only humans develop a severe illness. Within human populations, subclinical infections are underappreciated and may represent a substantial proportion of clinical outcomes. Compared with other members of the Bunyavirales order, CCHFV has a more complex genomic organization, with many viral proteins having unclear functions in viral pathogenesis. In recent years, improved animal models have led to increased insights into CCHFV pathogenesis, and several antivirals and vaccines for CCHFV have shown robust efficacy in preclinical models. Translation of these insights and candidate therapeutics to the clinic will hopefully reduce the morbidity and mortality caused by CCHFV.
Collapse
|
17
|
Barboza JJ, León-Figueroa DA, Saldaña-Cumpa HM, Valladares-Garrido MJ, Moreno-Ramos E, Sah R, Bonilla-Aldana DK, Rodriguez-Morales AJ. Virus Identification for Monkeypox in Human Seminal Fluid Samples: A Systematic Review. Trop Med Infect Dis 2023; 8:tropicalmed8030173. [PMID: 36977174 PMCID: PMC10057446 DOI: 10.3390/tropicalmed8030173] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/09/2023] [Accepted: 02/14/2023] [Indexed: 03/17/2023] Open
Abstract
Public health officials around the world are extremely concerned about the global outbreak of monkeypox (MPX), which has been claimed to have originated in Africa. As a result, studies into the origins and reasons behind the outbreak’s rapid spread have been sped up. The goal of the current investigation is to determine whether the monkeypox virus (MPXV) is present in seminal fluid samples from MPX cases that have been verified. Up until 6 January 2023, PubMed, Scopus, Web of Science, Embase, and ScienceDirect databases were used to conduct a thorough evaluation of the literature. The search technique returned a total of 308 items. Fourteen studies reporting the presence of MPXV in the seminal fluid of MPX-confirmed cases were included after the duplicates (n = 158) and searches by title, abstract, and full text were eliminated. In 84 out of the 643 confirmed MPX cases (13.06% or n = 643), MPXV was discovered in seminal fluid. Reverse transcriptase polymerase chain reaction (RT-PCR) was used to identify MPXV, and samples taken from skin lesions (96.27%), pharynx or oropharynx (30.48%), and blood all had higher positivity rates than other samples (12.44%). Additionally, 99.85% of respondents were male with a mean age of 36, 98.45% engaged in MSM (men who have sex with men) sexual conduct, and human immunodeficiency virus (HIV) accounted for 56.9% of all STD cases. This study offers proof that MPXV can be found in the seminal fluid of MPX sufferers. Our data imply that MPXV transmission is a possibility in these samples and that MSM are more vulnerable to it. The creation of hygienic standards is essential for the early identification of MPX cases.
Collapse
Affiliation(s)
- Joshuan J. Barboza
- Escuela de Medicina, Universidad Cesar Vallejo, Trujillo 13007, Peru
- Correspondence: ; Tel.: +51-99-2108-520
| | - Darwin A. León-Figueroa
- Facultad de Medicina Humana, Universidad de San Martín de Porres, Chiclayo 15011, Peru
- Centro de Investigación en Atención Primaria en Salud, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Hortencia M. Saldaña-Cumpa
- Facultad de Medicina Humana, Universidad de San Martín de Porres, Chiclayo 15011, Peru
- Centro de Investigación en Atención Primaria en Salud, Universidad Peruana Cayetano Heredia, Lima 15102, Peru
| | - Mario J. Valladares-Garrido
- South American Center for Education and Research in Public Health, Universidad Norbert Wiener, Lima 15108, Peru
- Oficina de Epidemiología, Hospital Regional Lambayeque, Chiclayo 14012, Peru
| | - Emilly Moreno-Ramos
- División de Revisiones Sistemáticas y Meta-Análisis, Tau-Relaped Group, Trujillo 13007, Peru
| | - Ranjit Sah
- Tribhuvan University Teaching Hospital, Institute of Medicine, Kathmandu 44600, Nepal
- Department of Public Health Dentistry, Dr. D.Y. Patil Dental College and Hospital, Dr. D.Y. Patil Vidyapeeth, Pune 411018, Maharashtra, India
| | | | - Alfonso J. Rodriguez-Morales
- Grupo de Investigación Biomedicina, Faculty of Medicine, Fundación Universitaria Autónoma de las Américas-Institución Universitaria Visión de las Américas, Pereira 660003, Colombia
- Master of Clinical Epidemiology and Biostatistics, Universidad Cientifica del Sur, Lima 15067, Peru
- Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut 1102, Lebanon
| |
Collapse
|
18
|
Hu YL, Zhang LQ, Liu XQ, Ye W, Zhao YX, Zhang L, Qiang ZX, Zhang LX, Lei YF, Jiang DB, Cheng LF, Zhang FL. Construction and evaluation of DNA vaccine encoding Crimean Congo hemorrhagic fever virus nucleocapsid protein, glycoprotein N-terminal and C-terminal fused with LAMP1. Front Cell Infect Microbiol 2023; 13:1121163. [PMID: 37026060 PMCID: PMC10072157 DOI: 10.3389/fcimb.2023.1121163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Accepted: 02/24/2023] [Indexed: 04/08/2023] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) can cause severe hemorrhagic fever in humans and is mainly transmitted by ticks. There is no effective vaccine for Crimean-Congo hemorrhagic fever (CCHF) at present. We developed three DNA vaccines encoding CCHFV nucleocapsid protein (NP), glycoprotein N-terminal (Gn) and C-terminal (Gc) fused with lysosome-associated membrane protein 1 (LAMP1) and assessed their immunogenicity and protective efficacy in a human MHC (HLA-A11/DR1) transgenic mouse model. The mice that were vaccinated three times with pVAX-LAMP1-CCHFV-NP induced balanced Th1 and Th2 responses and could most effectively protect mice from CCHFV transcription and entry-competent virus-like particles (tecVLPs) infection. The mice vaccinated with pVAX-LAMP1-CCHFV-Gc mainly elicited specific anti-Gc and neutralizing antibodies and provided a certain protection from CCHFV tecVLPs infection, but the protective efficacy was less than that of pVAX-LAMP1-CCHFV-NP. The mice vaccinated with pVAX-LAMP1-CCHFV-Gn only elicited specific anti-Gn antibodies and could not provide sufficient protection from CCHFV tecVLPs infection. These results suggest that pVAX-LAMP1-CCHFV-NP would be a potential and powerful candidate vaccine for CCHFV.
Collapse
Affiliation(s)
- Yong-Liang Hu
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
- Department of Dermatology, The Eighth Medical Center of PLA General Hospital, Beijing, China
| | - Lian-Qing Zhang
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
- College of Life Sciences, Northwest University, Xi’an, China
| | - Xiao-Qian Liu
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
- School of Medical Technology, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Wei Ye
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
| | - Yue-Xi Zhao
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
- School of Medical Technology, Shaanxi University of Chinese Medicine, Xianyang, China
| | - Liang Zhang
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
| | - Zun-Xian Qiang
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
| | - Lin-Xuan Zhang
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
| | - Ying-Feng Lei
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
| | - Dong-Bo Jiang
- Department of Immunology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
- *Correspondence: Dong-Bo Jiang, ; Lin-Feng Cheng, ; Fang-Lin Zhang,
| | - Lin-Feng Cheng
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
- *Correspondence: Dong-Bo Jiang, ; Lin-Feng Cheng, ; Fang-Lin Zhang,
| | - Fang-Lin Zhang
- Department of Microbiology, Air Force Medical University (The Fourth Military Medical University), Xi’an, China
- *Correspondence: Dong-Bo Jiang, ; Lin-Feng Cheng, ; Fang-Lin Zhang,
| |
Collapse
|
19
|
Liu J, Mucker EM, Chapman JL, Babka AM, Gordon JM, Bryan AV, Raymond JLW, Bell TM, Facemire PR, Goff AJ, Nalca A, Zeng X. Retrospective detection of monkeypox virus in the testes of nonhuman primate survivors. Nat Microbiol 2022; 7:1980-1986. [PMID: 36253513 DOI: 10.1038/s41564-022-01259-w] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/26/2022] [Indexed: 01/06/2023]
Abstract
Close contact through sexual activity has been associated with the spread of monkeypox virus (MPXV) in the ongoing, global 2022 epidemic. However, it remains unclear whether MPXV replicates in the testes or is transmitted via semen to produce an active infection. We carried out a retrospective analysis of MPXV-infected crab-eating macaque archival tissue samples from acute and convalescent phases of infection of clade I or clade II MPXV using immunostaining and RNA in situ hybridization. We detected MPXV in interstitial cells and seminiferous tubules of testes as well as epididymal lumina, which are the sites of sperm production and maturation. We also detected inflammation and necrosis during the acute phase of the disease by histological analysis. Finally, we found that MPXV was cleared from most organs during convalescence, including healed skin lesions, but could be detected for up to 37 d post-exposure in the testes of convalescent macaques. Our findings highlight the potential for sexual transmission of MPXV in humans.
Collapse
Affiliation(s)
- Jun Liu
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Eric M Mucker
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Jennifer L Chapman
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA.,Labcorp Early Development Laboratories Inc., Chantilly, VA, USA
| | - April M Babka
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Jamal M Gordon
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Ashley V Bryan
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Jo Lynne W Raymond
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Todd M Bell
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Paul R Facemire
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Arthur J Goff
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Aysegul Nalca
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA.
| |
Collapse
|
20
|
Structural characterization of protective non-neutralizing antibodies targeting Crimean-Congo hemorrhagic fever virus. Nat Commun 2022; 13:7298. [PMID: 36435827 PMCID: PMC9701186 DOI: 10.1038/s41467-022-34923-0] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 11/10/2022] [Indexed: 11/28/2022] Open
Abstract
Crimean-Congo Hemorrhagic Fever Virus (CCHFV) causes a life-threatening disease with up to a 40% mortality rate. With no approved medical countermeasures, CCHFV is considered a public health priority agent. The non-neutralizing mouse monoclonal antibody (mAb) 13G8 targets CCHFV glycoprotein GP38 and protects mice from lethal CCHFV challenge when administered prophylactically or therapeutically. Here, we reveal the structures of GP38 bound with a human chimeric 13G8 mAb and a newly isolated CC5-17 mAb from a human survivor. These mAbs bind overlapping epitopes with a shifted angle. The broad-spectrum potential of c13G8 and CC5-17 and the practicality of using them against Aigai virus, a closely related nairovirus were examined. Binding studies demonstrate that the presence of non-conserved amino acids in Aigai virus corresponding region prevent CCHFV mAbs from binding Aigai virus GP38. This information, coupled with in vivo efficacy, paves the way for future mAb therapeutics effective against a wide swath of CCHFV strains.
Collapse
|
21
|
Wang B, Shen WB, Yang P, Turan S. SARS-CoV-2 infection induces activation of ferroptosis in human placenta. Front Cell Dev Biol 2022; 10:1022747. [PMID: 36425527 PMCID: PMC9679405 DOI: 10.3389/fcell.2022.1022747] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 10/27/2022] [Indexed: 09/01/2023] Open
Abstract
Ferroptosis, a regulated non-apoptotic form of cell death, has been implicated in the response to varied types of infectious agents including virus. In this study, we sought to determine whether SARS-CoV-2 infection can induce activation of ferroptosis in the human placenta. We collected placentas from 23 pregnant females with laboratory-confirmed SARS-CoV-2 following delivery and then used RNA in situ hybridization assay for detection of viral positive-sense strand (PSS) to confirm that these placentas have been infected. We also used immunohistochemistry assay to assess expression levels of acyl-CoA synthetase long-chain family member 4 (ACSL4), an essential executioner of ferroptosis in the same specimens. Our results showed that ACSL4 expression was significantly increased in the group with positive positive-sense strand staining compared to their negative counterparts (p = 0.00022). Furthermore, we found that there was a positive trend for increased PSS staining along with increased ACSL4 expression. Our study supports that ferroptosis is activated in the response to SARS-CoV-2 infection in the human placenta, highlighting a molecular mechanism potentially linking this coronavirus infection and pathogenesis of adverse pregnancy outcomes.
Collapse
Affiliation(s)
- Bingbing Wang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | | | - Peixin Yang
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Sifa Turan
- Department of Obstetrics, Gynecology and Reproductive Sciences, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
22
|
Golden JW, Zeng X, Cline CR, Smith JM, Daye SP, Carey BD, Blancett CD, Shoemaker CJ, Liu J, Fitzpatrick CJ, Stefan CP, Garrison AR. The host inflammatory response contributes to disease severity in Crimean-Congo hemorrhagic fever virus infected mice. PLoS Pathog 2022; 18:e1010485. [PMID: 35587473 PMCID: PMC9119488 DOI: 10.1371/journal.ppat.1010485] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/30/2022] [Indexed: 11/23/2022] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is an important human pathogen. In cell culture, CCHFV is sensed by the cytoplasmic RNA sensor retinoic acid-inducible gene I (RIG-I) molecule and its adaptor molecule mitochondrial antiviral signaling (MAVS) protein. MAVS initiates both type I interferon (IFN-I) and proinflammatory responses. Here, we studied the role MAVS plays in CCHFV infection in mice in both the presence and absence of IFN-I activity. MAVS-deficient mice were not susceptible to CCHFV infection when IFN-I signaling was active and showed no signs of disease. When IFN-I signaling was blocked by antibody, MAVS-deficient mice lost significant weight, but were uniformly protected from lethal disease, whereas all control mice succumbed to infection. Cytokine activity in the infected MAVS-deficient mice was markedly blunted. Subsequent investigation revealed that CCHFV infected mice lacking TNF-α receptor signaling (TNFA-R-deficient), but not IL-6 or IL-1 activity, had more limited liver injury and were largely protected from lethal outcomes. Treatment of mice with an anti-TNF-α neutralizing antibody also conferred partial protection in a post-virus exposure setting. Additionally, we found that a disease causing, but non-lethal strain of CCHFV produced more blunted inflammatory cytokine responses compared to a lethal strain in mice. Our work reveals that MAVS activation and cytokine production both contribute to CCHFV pathogenesis, potentially identifying new therapeutic targets to treat this disease.
Collapse
Affiliation(s)
- Joseph W. Golden
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Xiankun Zeng
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Curtis R. Cline
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Jeffrey M. Smith
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Sharon P. Daye
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Brian D. Carey
- Diagnostic Services Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Candace D. Blancett
- Diagnostic Services Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Charles J. Shoemaker
- Diagnostic Services Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Jun Liu
- Pathology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Collin J. Fitzpatrick
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Christopher P. Stefan
- Diagnostic Services Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| | - Aura R. Garrison
- Virology Division, United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Maryland, United States of America
| |
Collapse
|
23
|
Rodriguez SE, Hawman DW, Sorvillo TE, O'Neal TJ, Bird BH, Rodriguez LL, Bergeron É, Nichol ST, Montgomery JM, Spiropoulou CF, Spengler JR. Immunobiology of Crimean-Congo hemorrhagic fever. Antiviral Res 2022; 199:105244. [PMID: 35026307 PMCID: PMC9245446 DOI: 10.1016/j.antiviral.2022.105244] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 01/05/2022] [Accepted: 01/06/2022] [Indexed: 12/29/2022]
Abstract
Human infection with Crimean-Congo hemorrhagic fever virus (CCHFV), a tick-borne pathogen in the family Nairoviridae, can result in a spectrum of outcomes, ranging from asymptomatic infection through mild clinical signs to severe or fatal disease. Studies of CCHFV immunobiology have investigated the relationship between innate and adaptive immune responses with disease severity, attempting to elucidate factors associated with differential outcomes. In this article, we begin by highlighting unanswered questions, then review current efforts to answer them. We discuss in detail current clinical studies and research in laboratory animals on CCHF, including immune targets of infection and adaptive and innate immune responses. We summarize data about the role of the immune response in natural infections of animals and humans and experimental studies in vitro and in vivo and from evaluating immune-based therapies and vaccines, and present recommendations for future research.
Collapse
Affiliation(s)
- Sergio E Rodriguez
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia; Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA
| | - David W Hawman
- Laboratory of Virology, Rocky Mountain Laboratories, Division of Intramural Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Hamilton, MT, USA
| | - Teresa E Sorvillo
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia; Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX, USA; Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX, USA; One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - T Justin O'Neal
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Brian H Bird
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia; One Health Institute, School of Veterinary Medicine, University of California Davis, Davis, CA, USA
| | - Luis L Rodriguez
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient Point, New York, USA
| | - Éric Bergeron
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia; Department of Pharmaceutical and Biomedical Sciences, University of Georgia, Athens, Georgia
| | - Stuart T Nichol
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Joel M Montgomery
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Christina F Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jessica R Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, National Center for Emerging and Zoonotic Infectious Diseases, Centers for Disease Control and Prevention, Atlanta, Georgia.
| |
Collapse
|
24
|
Host response transcriptomic analysis of Crimean-Congo hemorrhagic fever pathogenesis in the cynomolgus macaque model. Sci Rep 2021; 11:19807. [PMID: 34615921 PMCID: PMC8494817 DOI: 10.1038/s41598-021-99130-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Accepted: 09/14/2021] [Indexed: 11/09/2022] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is a highly pathogenic tick-borne RNA virus prevalent in Asia, Europe, and Africa, and can cause a hemorrhagic disease (CCHF) in humans with mortality rates as high as 60%. A general lack of both effective medical countermeasures and a comprehensive understanding of disease pathogenesis is partly driven by an historical lack of viable CCHF animal models. Recently, a cynomolgous macaque model of CCHF disease was developed. Here, we document the targeted transcriptomic response of non-human primates (NHP) to two different CCHFV strains; Afghan09-2990 and Kosova Hoti that both yielded a mild CCHF disease state. We utilized a targeted gene panel to elucidate the transcriptomic changes occurring in NHP whole blood during CCHFV infection; a first for any primate species. We show numerous upregulated genes starting at 1 day post-challenge through 14 days post-challenge. Early gene changes fell predominantly in the interferon stimulated gene family with later gene changes coinciding with an adaptive immune response to the virus. There are subtle differences between viral strains, namely duration of the differentially expressed gene response and biological pathways enriched. After recovery, NHPs showed no lasting transcriptomic changes at the end of sample collection.
Collapse
|
25
|
Evans TS, Myat TW, Hom NS, Ricks KM, Maw MT, Oo ZM, Toe AT, Aung NT, Aung P, Aung TH, Kuehnert P, Thant KZ, Win YT, Thein WZ, Gardner NR, Schoepp RJ, Johnson CK, Thu HM. Seroepidemiologic Survey of Crimean-Congo Hemorrhagic Fever Virus in Logging Communities, Myanmar. Emerg Infect Dis 2021; 27:1709-1713. [PMID: 34013868 PMCID: PMC8153884 DOI: 10.3201/eid2706.203223] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Crimean-Congo hemorrhagic fever virus (CCHFV) is endemic in Asia, infecting many animal hosts, but CCHFV has not been reported in Myanmar. We conducted a seroepidemiologic survey of logging communities in Myanmar and found CCHFV exposure was common (9.8%) and exposure to wild animal blood and body fluids was associated with seropositivity.
Collapse
|
26
|
Washburn RL, Hibler T, Thompson LA, Kaur G, Dufour JM. Therapeutic application of Sertoli cells for treatment of various diseases. Semin Cell Dev Biol 2021; 121:10-23. [PMID: 33910764 DOI: 10.1016/j.semcdb.2021.04.007] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 04/07/2021] [Indexed: 12/11/2022]
Abstract
Sertoli cells (SCs) are immune privileged cells found in the testis that function to immunologically protect maturing germ cells from immune destruction. This immune protection is due to the blood-testis-barrier, which prevents infiltration of cytotoxic immune cells and antibodies, and SC production of immunomodulatory factors, that favor a tolerogenic environment. The ability of SCs to create an immune privileged environment has led to the exploration of their potential use in the treatment of various diseases. SCs have been utilized to create a tolerogenic ectopic microenvironment, to protect co-grafted cells, and to deliver therapeutic proteins through gene therapy. To date, numerous studies have reported the potential use of SCs for the treatment of diabetes, neurodegenerative disorders, and restoration of spermatogenesis. Additionally, SCs have been investigated as a delivery vehicle for therapeutic products to treat other diseases like Laron syndrome, muscular dystrophy, and infections. This review will provide an overview of these therapeutic applications.
Collapse
Affiliation(s)
- Rachel L Washburn
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Taylor Hibler
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Lea Ann Thompson
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA
| | - Gurvinder Kaur
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| | - Jannette M Dufour
- Department of Cell Biology and Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA; Department of Medical Education, Texas Tech University Health Sciences Center, Lubbock, TX 79430, USA.
| |
Collapse
|
27
|
Pathogen Dose in Animal Models of Hemorrhagic Fever Virus Infections and the Potential Impact on Studies of the Immune Response. Pathogens 2021; 10:pathogens10030275. [PMID: 33804381 PMCID: PMC7999429 DOI: 10.3390/pathogens10030275] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/22/2021] [Accepted: 02/25/2021] [Indexed: 12/24/2022] Open
Abstract
Viral hemorrhagic fever viruses come from a wide range of virus families and are a significant cause of morbidity and mortality worldwide each year. Animal models of infection with a number of these viruses have contributed to our knowledge of their pathogenesis and have been crucial for the development of therapeutics and vaccines that have been approved for human use. Most of these models use artificially high doses of virus, ensuring lethality in pre-clinical drug development studies. However, this can have a significant effect on the immune response generated. Here I discuss how the dose of antigen or pathogen is a critical determinant of immune responses and suggest that the current study of viruses in animal models should take this into account when developing and studying animal models of disease. This can have implications for determination of immune correlates of protection against disease as well as informing relevant vaccination and therapeutic strategies.
Collapse
|
28
|
Serretiello E, Astorri R, Chianese A, Stelitano D, Zannella C, Folliero V, Santella B, Galdiero M, Franci G, Galdiero M. The emerging tick-borne Crimean-Congo haemorrhagic fever virus: A narrative review. Travel Med Infect Dis 2020; 37:101871. [PMID: 32891725 DOI: 10.1016/j.tmaid.2020.101871] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 08/25/2020] [Accepted: 08/26/2020] [Indexed: 01/31/2023]
Abstract
Crimean-Congo Haemorrhagic Fever (CCHF) is an increasingly relevant viral zoonosis caused by the negative-sense single-stranded (ss) RNA Crimean-Congo Haemorrhagic Fever Orthonairovirus (CCHFV) (Nairoviridae family, Bunyavirales order). The viral genome is divided into three segments (L-M-S) of distinct size and functions. The infection is generally mediated by a tick vector, in particular belonging to the Hyalomma genus, and the transmission follows a tick-vertebrate-tick ecologic cycle, with asymptomatic infected animals functioning as reservoirs and amplifiers for CCHFV. Human hosts could be infected primarily through infected ticks or by contact with infected hosts or their body fluids and tissues, also in a nosocomial way and in occupational contexts. Infected symptomatic patients generally manifest a nonspecific illness, which progresses across four stages, with possibly lethal outcomes. Disease outbreaks show a widespread geographic diffusion and a highly variable mortality rate, dramatically peaking in untreated patients. The lack of an adequate animal model and the elevated virus biological risk (only manageable under biosafety level 4 conditions) represent strongly limiting factors for a better characterization of the disease and for the development of specific therapies and vaccines. The present review discusses updated information on CCHFV-related disease, including details about the virus (taxonomy, structure, life cycle, transmission modalities) and considering CCHF pathogenesis, epidemiology and current strategies (diagnostic, therapeutic and preventive).
Collapse
Affiliation(s)
- Enrica Serretiello
- Section of Microbiology and Virology, University Hospital Luigi Vanvitelli of Naples, Naples, Italy
| | - Roberta Astorri
- Department of Mental Health and Public Medicine, Infectious Diseases Unit, University of Campania "Luigi Vanvitelli", Naples, Italy; Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Annalisa Chianese
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Debora Stelitano
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Carla Zannella
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Veronica Folliero
- Section of Microbiology and Virology, University Hospital Luigi Vanvitelli of Naples, Naples, Italy
| | - Biagio Santella
- Section of Microbiology and Virology, University Hospital Luigi Vanvitelli of Naples, Naples, Italy
| | - Marilena Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Gianluigi Franci
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy; Department of Medicine, Surgery and Dentistry "Scuola Medica Salernitana", University of Salerno, Baronissi (SA), Italy.
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy.
| |
Collapse
|
29
|
Blitvich BJ, Magalhaes T, Laredo-Tiscareño SV, Foy BD. Sexual Transmission of Arboviruses: A Systematic Review. Viruses 2020; 12:v12090933. [PMID: 32854298 PMCID: PMC7552039 DOI: 10.3390/v12090933] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 08/20/2020] [Accepted: 08/22/2020] [Indexed: 12/15/2022] Open
Abstract
Arthropod-borne viruses (arboviruses) are primarily maintained in nature in transmission cycles between hematophagous arthropods and vertebrate hosts, but an increasing number of arboviruses have been isolated from or indirectly detected in the urogenital tract and sexual secretions of their vertebrate hosts, indicating that further investigation on the possibility of sexual transmission of these viruses is warranted. The most widely recognized sexually-transmitted arbovirus is Zika virus but other arboviruses, including Crimean-Congo hemorrhagic fever virus and dengue virus, might also be transmitted, albeit occasionally, by this route. This review summarizes our current understanding on the ability of arboviruses to be sexually transmitted. We discuss the sexual transmission of arboviruses between humans and between vertebrate animals, but not arthropod vectors. Every taxonomic group known to contain arboviruses (Asfarviridae, Bunyavirales, Flaviviridae, Orthomyxoviridae, Reoviridae, Rhabdoviridae and Togaviridae) is covered.
Collapse
Affiliation(s)
- Bradley J. Blitvich
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
- Correspondence: ; Tel.: +1-515-294-9861; Fax: +1-515-294-8500
| | - Tereza Magalhaes
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (T.M.); (B.D.F.)
| | - S. Viridiana Laredo-Tiscareño
- Department of Veterinary Microbiology and Preventive Medicine, College of Veterinary Medicine, Iowa State University, Ames, IA 50011, USA;
| | - Brian D. Foy
- Arthropod-Borne and Infectious Diseases Laboratory, Department of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO 80523, USA; (T.M.); (B.D.F.)
| |
Collapse
|
30
|
Smith DR, Schmaljohn CS, Badger C, Ostrowski K, Zeng X, Grimes SD, Rayner JO. Comparative pathology study of Venezuelan, eastern, and western equine encephalitis viruses in non-human primates. Antiviral Res 2020; 182:104875. [PMID: 32755661 DOI: 10.1016/j.antiviral.2020.104875] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 07/02/2020] [Accepted: 07/06/2020] [Indexed: 11/16/2022]
Abstract
Venezuelan, eastern, and western equine encephalitis viruses (VEEV, EEEV, and WEEV) are mosquito-borne viruses in the Americas that cause central nervous system (CNS) disease in humans and equids. In this study, we directly characterized the pathogenesis of VEEV, EEEV, and WEEV in cynomolgus macaques following subcutaneous exposure because this route more closely mimics natural infection via mosquito transmission or by an accidental needle stick. Our results highlight how EEEV is significantly more pathogenic compared to VEEV similarly to what is observed in humans. Interestingly, EEEV appears to be just as neuropathogenic by subcutaneous exposure as it was in previously completed aerosol exposure studies. In contrast, subcutaneous exposure of cynomolgus macaques with WEEV caused limited disease and is contradictory to what has been reported for aerosol exposure. Several differences in viremia, hematology, or tissue tropism were noted when animals were exposed subcutaneously compared to prior aerosol exposure studies. This study provides a more complete picture of the pathogenesis of the encephalitic alphaviruses and highlights how further defining the neuropathology of these viruses could have important implications for the development of medical countermeasures for the neurovirulent alphaviruses.
Collapse
Affiliation(s)
- Darci R Smith
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, 21702, MD, USA.
| | - Connie S Schmaljohn
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, 21702, MD, USA
| | - Catherine Badger
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, 21702, MD, USA
| | - Kristen Ostrowski
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, 21702, MD, USA
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, 21702, MD, USA
| | | | | |
Collapse
|
31
|
Cross RW, Prasad AN, Borisevich V, Geisbert JB, Agans KN, Deer DJ, Fenton KA, Geisbert TW. Crimean-Congo hemorrhagic fever virus strains Hoti and Afghanistan cause viremia and mild clinical disease in cynomolgus monkeys. PLoS Negl Trop Dis 2020; 14:e0008637. [PMID: 32790668 PMCID: PMC7447009 DOI: 10.1371/journal.pntd.0008637] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2019] [Revised: 08/25/2020] [Accepted: 07/24/2020] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Development of vaccines and therapies against Crimean-Congo hemorrhagic fever virus (CCHFV) have been hindered by the lack of immunocompetent animal models. Recently, a lethal nonhuman primate model based on the CCHFV Hoti strain was reported. CCHFV Hoti caused severe disease in cynomolgus monkeys with 75% lethality when given by the intravenous (i.v.) route. METHODOLOGY/PRINCIPAL FINDINGS In a series of experiments, eleven cynomologus monkeys were exposed i.v. to CCHFV Hoti and four macaques were exposed i.v. to CCHFV Afghanistan. Despite transient viremia and changes in clinical pathology such as leukopenia and thrombocytopenia developing in all 15 animals, all macaques survived to the study endpoint without developing severe disease. CONCLUSIONS/SIGNIFICANCE We were unable to attribute differences in the results of our study versus the previous report to differences in the CCHFV Hoti stock, challenge dose, origin, or age of the macaques. The observed differences are most likely the result of the outbred nature of macaques and low animal numbers often used by necessity and for ethical considerations in BSL-4 studies. Nonetheless, while we were unable to achieve severe disease or lethality, the CCHFV Hoti and Afghanistan macaque models are useful for screening medical countermeasures using biomarkers including viremia and clinical pathology to assess efficacy.
Collapse
Affiliation(s)
- Robert W. Cross
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Abhishek N. Prasad
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Viktoriya Borisevich
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Joan B. Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Krystle N. Agans
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Daniel J. Deer
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Karla A. Fenton
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Thomas W. Geisbert
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, Texas, United States of America
| |
Collapse
|
32
|
Sorvillo TE, Rodriguez SE, Hudson P, Carey M, Rodriguez LL, Spiropoulou CF, Bird BH, Spengler JR, Bente DA. Towards a Sustainable One Health Approach to Crimean-Congo Hemorrhagic Fever Prevention: Focus Areas and Gaps in Knowledge. Trop Med Infect Dis 2020; 5:tropicalmed5030113. [PMID: 32645889 PMCID: PMC7558268 DOI: 10.3390/tropicalmed5030113] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/13/2022] Open
Abstract
Crimean–Congo hemorrhagic fever virus (CCHFV) infection is identified in the 2018 World Health Organization Research and Development Blueprint and the National Institute of Allergy and Infectious Diseases (NIH/NIAID) priority A list due to its high risk to public health and national security. Tick-borne CCHFV is widespread, found in Europe, Asia, Africa, the Middle East, and the Indian subcontinent. It circulates between ticks and several vertebrate hosts without causing overt disease, and thus can be present in areas without being noticed by the public. As a result, the potential for zoonotic spillover from ticks and animals to humans is high. In contrast to other emerging viruses, human-to-human transmission of CCHFV is typically limited; therefore, prevention of spillover events should be prioritized when considering countermeasures. Several factors in the transmission dynamics of CCHFV, including a complex transmission cycle that involves both ticks and vertebrate hosts, lend themselves to a One Health approach for the prevention and control of the disease that are often overlooked by current strategies. Here, we examine critical focus areas to help mitigate CCHFV spillover, including surveillance, risk assessment, and risk reduction strategies concentrated on humans, animals, and ticks; highlight gaps in knowledge; and discuss considerations for a more sustainable One Health approach to disease control.
Collapse
Affiliation(s)
- Teresa E. Sorvillo
- One Health Institute, School of Veterinary Medicine, University of California Davis, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA;
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; (S.E.R.); (C.F.S.); (J.R.S.)
- Correspondence: ; Tel.: +1-530-752-7526
| | - Sergio E. Rodriguez
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; (S.E.R.); (C.F.S.); (J.R.S.)
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (M.C.); (D.A.B.)
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Peter Hudson
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA;
| | - Megan Carey
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (M.C.); (D.A.B.)
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| | - Luis L. Rodriguez
- Foreign Animal Disease Research Unit, Plum Island Animal Disease Center, Agricultural Research Service, United States Department of Agriculture, Orient Point, NY 11957, USA;
| | - Christina F. Spiropoulou
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; (S.E.R.); (C.F.S.); (J.R.S.)
| | - Brian H. Bird
- One Health Institute, School of Veterinary Medicine, University of California Davis, 1089 Veterinary Medicine Drive, Davis, CA 95616, USA;
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; (S.E.R.); (C.F.S.); (J.R.S.)
| | - Jessica R. Spengler
- Viral Special Pathogens Branch, Division of High-Consequence Pathogens and Pathology, Centers for Disease Control and Prevention, Atlanta, GA 30333, USA; (S.E.R.); (C.F.S.); (J.R.S.)
| | - Dennis A. Bente
- Department of Microbiology & Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA; (M.C.); (D.A.B.)
- Galveston National Laboratory, University of Texas Medical Branch, Galveston, TX 77555, USA
| |
Collapse
|
33
|
Di Paola N, Sanchez-Lockhart M, Zeng X, Kuhn JH, Palacios G. Viral genomics in Ebola virus research. Nat Rev Microbiol 2020; 18:365-378. [PMID: 32367066 PMCID: PMC7223634 DOI: 10.1038/s41579-020-0354-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/09/2020] [Indexed: 12/20/2022]
Abstract
Filoviruses such as Ebola virus continue to pose a substantial health risk to humans. Advances in the sequencing and functional characterization of both pathogen and host genomes have provided a wealth of knowledge to clinicians, epidemiologists and public health responders during outbreaks of high-consequence viral disease. Here, we describe how genomics has been historically used to investigate Ebola virus disease outbreaks and how new technologies allow for rapid, large-scale data generation at the point of care. We highlight how genomics extends beyond consensus-level sequencing of the virus to include intra-host viral transcriptomics and the characterization of host responses in acute and persistently infected patients. Similar genomics techniques can also be applied to the characterization of non-human primate animal models and to known natural reservoirs of filoviruses, and metagenomic sequencing can be the key to the discovery of novel filoviruses. Finally, we outline the importance of reverse genetics systems that can swiftly characterize filoviruses as soon as their genome sequences are available.
Collapse
Affiliation(s)
- Nicholas Di Paola
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Mariano Sanchez-Lockhart
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA
| | - Jens H Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Frederick, MD, USA
| | - Gustavo Palacios
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, MD, USA.
| |
Collapse
|
34
|
Liu J, Babka AM, Kearney BJ, Radoshitzky SR, Kuhn JH, Zeng X. Molecular detection of SARS-CoV-2 in formalin-fixed, paraffin-embedded specimens. JCI Insight 2020; 5:139042. [PMID: 32379723 PMCID: PMC7406253 DOI: 10.1172/jci.insight.139042] [Citation(s) in RCA: 70] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 05/06/2020] [Indexed: 01/08/2023] Open
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of human coronavirus disease 2019 (COVID-19), emerged in Wuhan, China, in December 2019. The virus rapidly spread globally, resulting in a public health crisis including almost 5 million cases and 323,256 deaths as of May 21, 2020. Here, we describe the identification and evaluation of commercially available reagents and assays for the molecular detection of SARS-CoV-2 in infected FFPE cell pellets. We identified a suitable rabbit polyclonal anti-SARS-CoV spike protein antibody and a mouse monoclonal anti-SARS-CoV nucleocapsid protein (NP) antibody for cross-detection of the respective SARS-CoV-2 proteins by IHC and immunofluorescence assay (IFA). Next, we established RNAscope in situ hybridization (ISH) to detect SARS-CoV-2 RNA. Furthermore, we established a multiplex FISH (mFISH) to detect positive-sense SARS-CoV-2 RNA and negative-sense SARS-CoV-2 RNA (a replicative intermediate indicating viral replication). Finally, we developed a dual staining assay using IHC and ISH to detect SARS-CoV-2 antigen and RNA in the same FFPE section. It is hoped that these reagents and assays will accelerate COVID-19 pathogenesis studies in humans and in COVID-19 animal models.
Collapse
Affiliation(s)
- Jun Liu
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, Maryland, USA
| | - April M. Babka
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, Maryland, USA
| | - Brian J. Kearney
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, Maryland, USA
| | - Sheli R. Radoshitzky
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, Maryland, USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases (NIAID), NIH, Fort Detrick, Frederick, Maryland, USA
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases (USAMRIID), Fort Detrick, Frederick, Maryland, USA
| |
Collapse
|
35
|
Tipih T, Burt FJ. Crimean-Congo Hemorrhagic Fever Virus: Advances in Vaccine Development. Biores Open Access 2020; 9:137-150. [PMID: 32461819 PMCID: PMC7247048 DOI: 10.1089/biores.2019.0057] [Citation(s) in RCA: 41] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/27/2020] [Indexed: 01/12/2023] Open
Abstract
Crimean-Congo hemorrhagic fever (CCHF) is a severe human disease with mortality rates of up to 30%. The disease is widespread in Africa, Asia, the Middle East and Eastern Europe. The last few years have seen disease emergence in Spain for the first time and disease re-emergence in other regions of the world after periods of inactivity. Factors, such as climate change, movement of infected ticks, animals, and changes in human activity, are likely to broaden endemic foci. There are therefore concerns that CCHF might emerge in currently nonendemic regions. The absence of approved vaccines or therapies heightens these concerns; thus Crimean-Congo hemorrhagic fever virus (CCHFV) is listed by the World Health Organization as a priority organism. However, the current sporadic nature of CCHF cases may call for targeted vaccination of risk groups as opposed to mass vaccinations. CCHF vaccine development has accelerated in recent years, partly because of the discovery of CCHF animal models. In this review, we discuss CCHF risk groups who are most likely to benefit from vaccine development, the merits and demerits of available CCHF animal models, and the various approaches which have been explored for CCHF vaccine development. Lastly, we present concluding remarks and research areas which can be further explored to enhance the available CCHFV vaccine data.
Collapse
Affiliation(s)
- Thomas Tipih
- Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
| | - Felicity Jane Burt
- Division of Virology, Faculty of Health Sciences, University of the Free State, Bloemfontein, South Africa
- National Health Laboratory Service, Bloemfontein, South Africa
| |
Collapse
|
36
|
Liu J, Babka AM, Kearney BJ, Radoshitzky SR, Kuhn JH, Zeng X. Molecular Detection of SARS-CoV-2 in Formalin Fixed Paraffin Embedded Specimens. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2020:2020.04.21.042911. [PMID: 32511350 PMCID: PMC7255791 DOI: 10.1101/2020.04.21.042911] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the cause of human coronavirus disease 2019 (COVID-19), emerged in Wuhan, China in December 2019. The virus rapidly spread globally, resulting in a public-health crisis including more than one million cases and tens of thousands of deaths. Here, we describe the identification and evaluation of commercially available reagents and assays for the molecular detection of SARS-CoV-2 in infected formalin fixed paraffin embedded (FFPE) cell pellets. We identified a suitable rabbit polyclonal anti-SARS-CoV spike protein antibody and a mouse monoclonal anti-SARS-CoV nucleocapsid protein (NP) antibody for cross detection of the respective SARS-CoV-2 proteins by immunohistochemistry (IHC) and immunofluorescence assay (IFA). Next, we established RNAscope in situ hybridization (ISH) to detect SARS-CoV-2 RNA. Furthermore, we established a multiplex fluorescence ISH (mFISH) to detect positive-sense SARS-CoV-2 RNA and negative-sense SARS-CoV-2 RNA (a replicative intermediate indicating viral replication). Finally, we developed a dual staining assay using IHC and ISH to detect SARS-CoV-2 antigen and RNA in the same FFPE section. These reagents and assays will accelerate COVID-19 pathogenesis studies in humans and in COVID-19 animal models.
Collapse
Affiliation(s)
- Jun Liu
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702, USA
| | - April M. Babka
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702, USA
| | - Brian J. Kearney
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702, USA
| | - Sheli R. Radoshitzky
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702, USA
| | - Jens H. Kuhn
- Integrated Research Facility at Fort Detrick, National Institute of Allergy and Infectious Diseases, National, Institutes of Health, Fort Detrick, Frederick, MD 21702, USA
| | - Xiankun Zeng
- United States Army Medical Research Institute of Infectious Diseases, Fort Detrick, Frederick, Maryland 21702, USA
| |
Collapse
|