1
|
Kimble JB, Noronha L, Trujillo JD, Mitzel D, Richt JA, Wilson WC. Rift Valley Fever. Vet Clin North Am Food Anim Pract 2024; 40:293-304. [PMID: 38453549 DOI: 10.1016/j.cvfa.2024.01.004] [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] [Indexed: 03/09/2024] Open
Abstract
Rift Valley fever (RVF) is a zoonotic viral disease that affects domestic and wild ruminants such as cattle, sheep, goats, camels, and buffaloes. Rift valley fever virus (RVFV), the causative agent of RVF, can also infect humans. RVFV is an arthropod-borne virus (arbovirus) that is primarily spread through the bites of infected mosquitoes or exposure to infected blood. RVFV was first isolated and characterized in the Rift Valley of Kenya in 1931 and is endemic throughout sub-Saharan Africa, including Comoros and Madagascar, the Arabian Peninsula (Saudi Arabia and Yemen), and Mayotte.
Collapse
Affiliation(s)
- J Brian Kimble
- Foreign Arthropod-Borne Animal Diseases Research Unit, National Bio and Agro-Defense Facility, USDA, ARS, Manhattan, KS, USA
| | - Leela Noronha
- Foreign Arthropod-Borne Animal Diseases Research Unit, National Bio and Agro-Defense Facility, USDA, ARS, Manhattan, KS, USA
| | - Jessie D Trujillo
- Diganostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA
| | - Dana Mitzel
- Foreign Arthropod-Borne Animal Diseases Research Unit, National Bio and Agro-Defense Facility, USDA, ARS, Manhattan, KS, USA
| | - Juergen A Richt
- Diganostic Medicine/Pathobiology, Kansas State University, Manhattan, KS, USA; Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, South Africa
| | - William C Wilson
- Foreign Arthropod-Borne Animal Diseases Research Unit, National Bio and Agro-Defense Facility, USDA, ARS, Manhattan, KS, USA.
| |
Collapse
|
2
|
Balaraman V, Indran SV, Kim IJ, Trujillo JD, Meekins DA, Shivanna V, Zajac MD, Urbaniak K, Morozov I, Sunwoo SY, Faburay B, Osterrieder K, Gaudreault NN, Wilson WC, Richt JA. Rift Valley Fever Phlebovirus Reassortment Study in Sheep. Viruses 2024; 16:880. [PMID: 38932172 PMCID: PMC11209395 DOI: 10.3390/v16060880] [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: 03/25/2024] [Revised: 05/21/2024] [Accepted: 05/26/2024] [Indexed: 06/28/2024] Open
Abstract
Rift Valley fever (RVF) in ungulates and humans is caused by a mosquito-borne RVF phlebovirus (RVFV). Live attenuated vaccines are used in livestock (sheep and cattle) to control RVF in endemic regions during outbreaks. The ability of two or more different RVFV strains to reassort when co-infecting a host cell is a significant veterinary and public health concern due to the potential emergence of newly reassorted viruses, since reassortment of RVFVs has been documented in nature and in experimental infection studies. Due to the very limited information regarding the frequency and dynamics of RVFV reassortment, we evaluated the efficiency of RVFV reassortment in sheep, a natural host for this zoonotic pathogen. Co-infection experiments were performed, first in vitro in sheep-derived cells, and subsequently in vivo in sheep. Two RVFV co-infection groups were evaluated: group I consisted of co-infection with two wild-type (WT) RVFV strains, Kenya 128B-15 (Ken06) and Saudi Arabia SA01-1322 (SA01), while group II consisted of co-infection with the live attenuated virus (LAV) vaccine strain MP-12 and a WT strain, Ken06. In the in vitro experiments, the virus supernatants were collected 24 h post-infection. In the in vivo experiments, clinical signs were monitored, and blood and tissues were collected at various time points up to nine days post-challenge for analyses. Cell culture supernatants and samples from sheep were processed, and plaque-isolated viruses were genotyped to determine reassortment frequency. Our results show that RVFV reassortment is more efficient in co-infected sheep-derived cells compared to co-infected sheep. In vitro, the reassortment frequencies reached 37.9% for the group I co-infected cells and 25.4% for the group II co-infected cells. In contrast, we detected just 1.7% reassortant viruses from group I sheep co-infected with the two WT strains, while no reassortants were detected from group II sheep co-infected with the WT and LAV strains. The results indicate that RVFV reassortment occurs at a lower frequency in vivo in sheep when compared to in vitro conditions in sheep-derived cells. Further studies are needed to better understand the implications of RVFV reassortment in relation to virulence and transmission dynamics in the host and the vector. The knowledge learned from these studies on reassortment is important for understanding the dynamics of RVFV evolution.
Collapse
Affiliation(s)
- Velmurugan Balaraman
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - Sabarish V. Indran
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - In Joong Kim
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - Jessie D. Trujillo
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - David A. Meekins
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - Vinay Shivanna
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - Michelle D. Zajac
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
- Foreign Arthropod-Borne Animal Diseases Research Unit, United States Department of Agriculture, Agricultural Research Service, National Bio and Agro-Defense Facility, Manhattan, KS 66505, USA
| | - Kinga Urbaniak
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - Igor Morozov
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - Sun-Young Sunwoo
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - Bonto Faburay
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
- Foreign Arthropod-Borne Animal Diseases Research Unit, United States Department of Agriculture, Agricultural Research Service, National Bio and Agro-Defense Facility, Manhattan, KS 66505, USA
| | - Klaus Osterrieder
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - Natasha N. Gaudreault
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| | - William C. Wilson
- Foreign Arthropod-Borne Animal Diseases Research Unit, United States Department of Agriculture, Agricultural Research Service, National Bio and Agro-Defense Facility, Manhattan, KS 66505, USA
| | - Juergen A. Richt
- Center of Excellence for Emerging and Zoonotic Animal Diseases, Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA; (V.B.); (S.V.I.); (J.D.T.); (S.-Y.S.)
| |
Collapse
|
3
|
Tinto B, Quellec J, Cêtre-Sossah C, Dicko A, Salinas S, Simonin Y. Rift Valley fever in West Africa: A zoonotic disease with multiple socio-economic consequences. One Health 2023; 17:100583. [PMID: 37664171 PMCID: PMC10474305 DOI: 10.1016/j.onehlt.2023.100583] [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: 02/20/2023] [Revised: 06/12/2023] [Accepted: 06/12/2023] [Indexed: 09/05/2023] Open
Abstract
Rift Valley fever virus (RVFV) is an arbovirus that causes Rift Valley fever (RVF), a zoonotic disease that mainly affects domestic and wildlife ruminants and humans. The first epidemic in North-Western and West Africa occurred in Senegal and Mauritania in 1987, two countries where RVF is now endemic. Slaughterhouse workers, farmers, herders and veterinarians are at high risk of exposure to RVF. Beyond the health threat, RVF is considered to cause major socio-economic problems, specifically in developing countries where livestock farming and trade are important economic activities. Indeed, the mortality rate linked to RVF infection can reach 95-100% in newborns and young animals. In West Africa, livestock production is a key factor for food production and for national economics. Epizootics caused by RVF can therefore have serious socio-economic consequences by impacting multisectoral economics, the psycho-social health of pastoral communities, and food security. Improving prevention strategies against RVF, including vaccination, enhancing knowledge of RVF and correcting any inappropriate behaviors by populations of endemics areas, as well as better monitoring of RVF ecological factors are effective ways to better foresee and control outbreaks of RVF and its socio-economical side-effects in countries at high risk of occurrence of the disease.
Collapse
Affiliation(s)
- Bachirou Tinto
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Montpellier, France
- Laboratoire National de Référence des Fièvres Hémorragiques Virale, Centre MURAZ, Institut National de Santé Publique (INSP), Bobo-Dioulasso, Burkina Faso
| | - Jordan Quellec
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Montpellier, France
- ASTRE, University of Montpellier, CIRAD, INRAe, Montpellier, France
| | | | - Amadou Dicko
- Laboratoire central de référence, Institut National de Santé Publique (INSP), Ouagadougou, Burkina Faso
- Ministère de l'Agriculture, des ressources animales et halieutiques du Burkina Faso, Ouagadougou, Burkina Faso
| | - Sara Salinas
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Montpellier, France
| | - Yannick Simonin
- Pathogenesis and Control of Chronic and Emerging Infections, University of Montpellier, INSERM, Etablissement Français du Sang, Montpellier, France
- ASTRE, University of Montpellier, CIRAD, INRAe, Montpellier, France
| |
Collapse
|
4
|
Abstract
Rift Valley Fever Virus (RVFV) is a negative sense segmented RNA virus that can cause severe hemorrhagic fever. The tri-segmented virus genome encodes for six (6) multifunctional proteins that engage host factors at a variety of different stages in the replication cycle. The S segment encodes nucleoprotein (N) and nonstructural protein S (NSs), the M segment encodes viral glycoproteins Gn and Gc as well as nonstructural protein M (NSm) and the L segment encodes the viral polymerase (L). Viral glycoproteins Gn and Gc are responsible for entry by binding to a number of host factors. Our recent studies identified a scavenger receptor, LDL receptor related protein 1 (Lrp1), as a potential pro-viral host factor for RVFV and related viruses, including Oropouche virus (OROV) infection. Coincidentally, several recent studies identified other LDL family proteins as viral entry factors and receptors for other viral families. Collectively, these observations suggest that highly conserved LDL family proteins may play a significant role in facilitating entry of viruses from several distinct families. Given the significant roles of viral and host factors during infection, characterization of these interactions is critical for therapeutic targeting with neutralizing antibodies and vaccines.
Collapse
Affiliation(s)
- Safder S Ganaie
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States
| | - Daisy W Leung
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States; Department of Medicine, Washington University School of Medicine, St Louis, MO, United States
| | - Amy L Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, United States; Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, United States
| | - Gaya K Amarasinghe
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO, United States.
| |
Collapse
|
5
|
Nair N, Osterhaus ADME, Rimmelzwaan GF, Prajeeth CK. Rift Valley Fever Virus-Infection, Pathogenesis and Host Immune Responses. Pathogens 2023; 12:1174. [PMID: 37764982 PMCID: PMC10535968 DOI: 10.3390/pathogens12091174] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/09/2023] [Accepted: 09/14/2023] [Indexed: 09/29/2023] Open
Abstract
Rift Valley Fever Virus is a mosquito-borne phlebovirus causing febrile or haemorrhagic illness in ruminants and humans. The virus can prevent the induction of the antiviral interferon response through its NSs proteins. Mutations in the NSs gene may allow the induction of innate proinflammatory immune responses and lead to attenuation of the virus. Upon infection, virus-specific antibodies and T cells are induced that may afford protection against subsequent infections. Thus, all arms of the adaptive immune system contribute to prevention of disease progression. These findings will aid the design of vaccines using the currently available platforms. Vaccine candidates have shown promise in safety and efficacy trials in susceptible animal species and these may contribute to the control of RVFV infections and prevention of disease progression in humans and ruminants.
Collapse
|
6
|
Schwarz MM, Ganaie SS, Feng A, Brown G, Yangdon T, White JM, Hoehl RM, McMillen CM, Rush RE, Connors KA, Cui X, Leung DW, Egawa T, Amarasinghe GK, Hartman AL. Lrp1 is essential for lethal Rift Valley fever hepatic disease in mice. SCIENCE ADVANCES 2023; 9:eadh2264. [PMID: 37450601 PMCID: PMC10348670 DOI: 10.1126/sciadv.adh2264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Accepted: 06/12/2023] [Indexed: 07/18/2023]
Abstract
Rift Valley fever virus (RVFV) is an emerging arbovirus found in Africa. While RVFV is pantropic and infects many cells and tissues, viral replication and necrosis within the liver play a critical role in mediating severe disease. The low-density lipoprotein receptor-related protein 1 (Lrp1) is a recently identified host factor for cellular entry and infection by RVFV. The biological significance of Lrp1, including its role in hepatic disease in vivo, however, remains to be determined. Because Lrp1 has a high expression level in hepatocytes, we developed a mouse model in which Lrp1 is specifically deleted in hepatocytes to test how the absence of liver Lrp1 expression affects RVF pathogenesis. Mice lacking Lrp1 expression in hepatocytes showed minimal RVFV replication in the liver, longer time to death, and altered clinical signs toward neurological disease. In contrast, RVFV infection levels in other tissues showed no difference between the two genotypes. Therefore, Lrp1 is essential for RVF hepatic disease in mice.
Collapse
Affiliation(s)
- Madeline M. Schwarz
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Safder S. Ganaie
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Annie Feng
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Griffin Brown
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Tenzin Yangdon
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - J. Michael White
- Transgenic, Knockout and Micro-Injection Core, Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
| | - Cynthia M. McMillen
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Rachael E. Rush
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Kaleigh A. Connors
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| | - Xiaoxia Cui
- Genome Engineering & Stem Cell Center, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Daisy W. Leung
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
- Department of Medicine, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Takeshi Egawa
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Gaya K. Amarasinghe
- Department of Pathology & Immunology, Washington University School of Medicine in St. Louis, St. Louis, MO, USA
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Infectious Diseases and Microbiology, School of Public Health, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
7
|
Bermúdez-Méndez E, Angelino P, van Keulen L, van de Water S, Rockx B, Pijlman GP, Ciuffi A, Kortekaas J, Wichgers Schreur PJ. Transcriptomic Profiling Reveals Intense Host-Pathogen Dispute Compromising Homeostasis during Acute Rift Valley Fever Virus Infection. J Virol 2023; 97:e0041523. [PMID: 37306574 PMCID: PMC10308945 DOI: 10.1128/jvi.00415-23] [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: 03/17/2023] [Accepted: 05/13/2023] [Indexed: 06/13/2023] Open
Abstract
Rift Valley fever virus (RVFV) (family Phenuiviridae) can cause severe disease, and outbreaks of this mosquito-borne pathogen pose a significant threat to public and animal health. Yet many molecular aspects of RVFV pathogenesis remain incompletely understood. Natural RVFV infections are acute, characterized by a rapid onset of peak viremia during the first days post-infection, followed by a rapid decline. Although in vitro studies identified a major role of interferon (IFN) responses in counteracting the infection, a comprehensive overview of the specific host factors that play a role in RVFV pathogenesis in vivo is still lacking. Here, the host in vivo transcriptional profiles in the liver and spleen tissues of lambs exposed to RVFV are studied using RNA sequencing (RNA-seq) technology. We validate that IFN-mediated pathways are robustly activated in response to infection. We also link the observed hepatocellular necrosis with severely compromised organ function, which is reflected as a marked downregulation of multiple metabolic enzymes essential for homeostasis. Furthermore, we associate the elevated basal expression of LRP1 in the liver with RVFV tissue tropism. Collectively, the results of this study deepen the knowledge of the in vivo host response during RVFV infection and reveal new insights into the gene regulation networks underlying pathogenesis in a natural host. IMPORTANCE Rift Valley fever virus (RVFV) is a mosquito-transmitted pathogen capable of causing severe disease in animals and humans. Outbreaks of RVFV pose a significant threat to public health and can result in substantial economic losses. Little is known about the molecular basis of RVFV pathogenesis in vivo, particularly in its natural hosts. We employed RNA-seq technology to investigate genome-wide host responses in the liver and spleen of lambs during acute RVFV infection. We show that RVFV infection drastically decreases the expression of metabolic enzymes, which impairs normal liver function. Moreover, we highlight that basal expression levels of the host factor LRP1 may be a determinant of RVFV tissue tropism. This study links the typical pathological phenotype induced by RVFV infection with tissue-specific gene expression profiles, thereby improving our understanding of RVFV pathogenesis.
Collapse
Affiliation(s)
- Erick Bermúdez-Méndez
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
- Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
- Institute of Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Paolo Angelino
- Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
- Institute of Microbiology, University of Lausanne, Lausanne, Switzerland
- Bioinformatics Core Facility, Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Lucien van Keulen
- Department of Bacteriology, Host-Pathogen Interaction and Diagnostics Development, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Sandra van de Water
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Barry Rockx
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| | - Gorben P. Pijlman
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| | - Angela Ciuffi
- Institute of Microbiology, Lausanne University Hospital, Lausanne, Switzerland
- Institute of Microbiology, University of Lausanne, Lausanne, Switzerland
| | - Jeroen Kortekaas
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
- Laboratory of Virology, Wageningen University & Research, Wageningen, The Netherlands
| | - Paul J. Wichgers Schreur
- Department of Virology and Molecular Biology, Wageningen Bioveterinary Research, Lelystad, The Netherlands
| |
Collapse
|
8
|
Experimental Infection of Domestic Pigs ( Sus scrofa) with Rift Valley Fever Virus. Viruses 2023; 15:v15020545. [PMID: 36851759 PMCID: PMC9964260 DOI: 10.3390/v15020545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Revised: 02/09/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
Rift valley fever (RVF), caused by the RVF virus (RVFV), is a vector-borne zoonotic disease that primarily affects domestic ruminants. Abortion storms and neonatal deaths characterise the disease in animals. Humans develop flu-like symptoms, which can progress to severe disease. The susceptibility of domestic pigs (Sus scrofa domesticus) to RVFV remains unresolved due to conflicting experimental infection results. To address this, we infected two groups of pregnant sows, neonates and weaners, each with a different RVFV isolate, and a third group of weaners with a mixture of the two viruses. Serum, blood and oral, nasal and rectal swabs were collected periodically, and two neonates and a weaner from group 1 and 2 euthanised from 2 days post infection (DPI), with necropsy and histopathology specimens collected. Sera and organ pools, blood and oronasorectal swabs were tested for RVFV antibodies and RNA. Results confirmed that pigs can be experimentally infected with RVFV, although subclinically, and that pregnant sows can abort following infection. Presence of viral RNA in oronasorectal swab pools on 28 DPI suggest that pigs may shed RVFV for at least one month. It is concluded that precautions should be applied when handling pig body fluids and carcasses during RVF outbreaks.
Collapse
|
9
|
Rift Valley Fever Virus Non-Structural Protein S Is Associated with Nuclear Translocation of Active Caspase-3 and Inclusion Body Formation. Viruses 2022; 14:v14112487. [PMID: 36366585 PMCID: PMC9698985 DOI: 10.3390/v14112487] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/06/2022] [Accepted: 11/06/2022] [Indexed: 11/12/2022] Open
Abstract
Rift Valley fever phlebovirus (RVFV) causes Rift Valley fever (RVF), an emerging zoonotic disease that causes abortion storms and high mortality rates in young ruminants as well as severe or even lethal complications in a subset of human patients. This study investigates the pathomechanism of intranuclear inclusion body formation in severe RVF in a mouse model. Liver samples from immunocompetent mice infected with virulent RVFV 35/74, and immunodeficient knockout mice that lack interferon type I receptor expression and were infected with attenuated RVFV MP12 were compared to livers from uninfected controls using histopathology and immunohistochemistry for RVFV nucleoprotein, non-structural protein S (NSs) and pro-apoptotic active caspase-3. Histopathology of the livers showed virus-induced, severe hepatic necrosis in both mouse strains. However, immunohistochemistry and immunofluorescence revealed eosinophilic, comma-shaped, intranuclear inclusions and an intranuclear (co-)localization of RVFV NSs and active caspase-3 only in 35/74-infected immunocompetent mice, but not in MP12-infected immunodeficient mice. These results suggest that intranuclear accumulation of RVFV 35/74 NSs is involved in nuclear translocation of active caspase-3, and that nuclear NSs and active caspase-3 are involved in the formation of the light microscopically visible inclusion bodies.
Collapse
|
10
|
McMillen CM, Boyles DA, Kostadinov SG, Hoehl RM, Schwarz MM, Albe JR, Demers MJ, Hartman AL. Congenital Rift Valley fever in Sprague Dawley rats is associated with diffuse infection and pathology of the placenta. PLoS Negl Trop Dis 2022; 16:e0010898. [PMID: 36315601 PMCID: PMC9648853 DOI: 10.1371/journal.pntd.0010898] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 11/10/2022] [Accepted: 10/17/2022] [Indexed: 11/12/2022] Open
Abstract
Rift Valley fever (RVF) is a disease of animals and humans associated with abortions in ruminants and late-gestation miscarriages in women. Here, we use a rat model of congenital RVF to identify tropisms, pathologies, and immune responses in the placenta during vertical transmission. Infection of late-gestation pregnant rats resulted in vertical transmission to the placenta and widespread infection throughout the decidua, basal zone, and labyrinth zone. Some pups from infected dams appeared normal while others had gross signs of teratogenicity including death. Histopathological lesions were detected in placenta from pups regardless of teratogenicity, while teratogenic pups had widespread hemorrhage throughout multiple placenta layers. Teratogenic events were associated with significant increases in placental pro-inflammatory cytokines, type I interferons, and chemokines. RVFV displays a high degree of tropism for all placental tissue layers and the degree of hemorrhage and inflammatory mediator production is highest in placenta from pups with adverse outcomes. Given the potential for RVFV to emerge in new locations and the recent evidence of emerging viruses, like Zika and SARS-CoV-2, to undergo vertical transmission, this study provides essential understanding regarding the mechanisms by which RVFV crosses the placenta barrier. Rift Valley fever virus (RVFV) infections cause human health and economical burdens given its ability to induce high rates of abortions in ruminants and possible contributions towards late-term miscarriages in women. In this study, we have identified important structures in the placenta targeted by this emerging bunyavirus. Inflammation was associated with more severe fetal outcomes such as death and fetal deformities. The striking similarities between the pathologies of the placenta in the rat model of congenital RVF and those observed in naturally infected ruminants highlight the utility of this rodent model. These findings may be further translated towards understanding the mechanisms involved in vertical transmission of RVFV in humans.
Collapse
Affiliation(s)
- Cynthia M. McMillen
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Devin A. Boyles
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Stefan G. Kostadinov
- Department of Pathology, Magee Women’s Hospital of the University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, United States of America
| | - Ryan M. Hoehl
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Madeline M. Schwarz
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Joseph R. Albe
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
| | - Matthew J. Demers
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
| | - Amy L. Hartman
- Center for Vaccine Research, University of Pittsburgh, Pittsburgh, Pennsylvania, United States of America
- Department of Infectious Diseases and Microbiology, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania, United States of America
- * E-mail:
| |
Collapse
|
11
|
Yao X, Yin Q, Hu D, Fu S, Zhang W, Nie K, Li F, Xu S, He Y, Liang G, Li X, Wang H. In Vitro Infection Dynamics of Wuxiang Virus in Different Cell Lines. Viruses 2022; 14:2383. [PMID: 36366481 PMCID: PMC9699334 DOI: 10.3390/v14112383] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Revised: 10/21/2022] [Accepted: 10/25/2022] [Indexed: 01/31/2023] Open
Abstract
Wuxiang virus (WUXV) is a newly discovered Bunyavirales transmitted by sandflies. It is found to infect humans and chickens and can cause neurological symptoms and even death in mice. However, the susceptibility of different hosts and tissue-derived cells to this virus is unclear. In this study, we examined cells derived from murine (BHK-21, N2A), human (HEK-293T, SH-SY5Y), dog (MDCK), pig (PK-15), monkey (Vero), and chicken (DF1), which were inoculated with WUXV at 0.05 MOI, and monitored for monolayer cytopathic effect (CPE). Culture supernatants and cells were collected from 0 to 96 h post-infection, cell viability was determined by trypan blue staining, numbers of infectious virus particles were quantified using plaque tests, and viral nucleic acid contents were determined by RT-qPCR. The presence of WUXV N antigen in infected cells was detected by Western blotting (WB). In response to virus infection, BHK-21, MDCK, and PK-15 cells were characterized by a clear CPE, and we observed reductions in the proportion of viable cells after 96 h. By contrast, no significant CPEs were observed in the other cell lines. We detected increases in viral titers, viral nucleic acid content, and N antigen expression in BHK-21, MDCK, PK-15, HEK-293T, N2A, SH-SY5Y, and DF1 cells post-infection. Vero cells showed no CPE, and the findings for other tests were negative. In conclusion, we tested the susceptibility of different cell lines to WUXV, enhanced our current understanding of WUXV biology at the cellular level, and laid the foundations for further investigation of the underlying virus infection mechanisms.
Collapse
Affiliation(s)
- Xiaohui Yao
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Qikai Yin
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Danhe Hu
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Shihong Fu
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Weijia Zhang
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Kai Nie
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Fan Li
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Songtao Xu
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Ying He
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Guodong Liang
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| | - Xiangdong Li
- Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, College of Veterinary Medicine, Yangzhou University, Yangzhou 225009, China
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
- Joint International Research Laboratory of Agriculture and Agri-Product Safety, The Ministry of Education of China, Yangzhou University, Yangzhou 225009, China
| | - Huanyu Wang
- Department of Arboviruses, NHC Key Laboratory of Biosafety, National Institute for Viral Disease Control and Prevention, State Key Laboratory for Infectious Disease Prevention and Control, Chinese Center for Disease Control and Prevention, Beijing 102206, China
| |
Collapse
|
12
|
Campbell CL, Snell TK, Bennett S, Wyckoff JH, Heaslip D, Flatt J, Harris EK, Hartman DA, Lian E, Bird BH, Stenglein MD, Bowen RA, Kading RC. Safety study of Rift Valley Fever human vaccine candidate (DDVax) in mosquitoes. Transbound Emerg Dis 2022; 69:2621-2633. [PMID: 34890118 PMCID: PMC9788258 DOI: 10.1111/tbed.14415] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 11/20/2021] [Accepted: 11/30/2021] [Indexed: 12/30/2022]
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne pathogen with significant human and veterinary health consequences that periodically emerges in epizootics. RVFV causes fetal loss and death in ruminants and in humans can lead to liver and renal disease, delayed-onset encephalitis, retinitis, and in some cases severe haemorrhagic fever. A live attenuated vaccine candidate (DDVax), was developed by the deletion of the virulence factors NSs and NSm from a clinical isolate, ZH501, and has proven safe and immunogenic in rodents, pregnant sheep and non-human primates. Deletion of NSm also severely restricted mosquito midgut infection and inhibited vector-borne transmission. To demonstrate environmental safety, this study investigated the replication, dissemination and transmission efficiency of DDVax in mosquitoes following oral exposure compared to RVFV strains MP-12 and ZH501. Infection and dissemination profiles were also measured in mosquitoes 7 days after they fed on goats inoculated with DDvax or MP-12. We hypothesized that DDVax would infect mosquitoes at significantly lower rates than other RVFV strains and, due to lack of NSm, be transmission incompetent. Exposure of Ae. aegypti and Cx. tarsalis to 8 log10 plaque forming units (PFU)/ml DDVax by artificial bloodmeal resulted in significantly reduced DDVax infection rates in mosquito bodies compared to controls. Plaque assays indicated negligible transmission of infectious DDVax in Cx. tarsalis saliva (1/140 sampled) and none in Ae. aegypti saliva (0/120). Serum from goats inoculated with DDVax or MP-12 did not harbour detectable infectious virus by plaque assay at 1, 2 or 3 days post-inoculation. Infectious virus was, however, recovered from Aedes and Culex bodies that fed on goats vaccinated with MP-12 (13.8% and 4.6%, respectively), but strikingly, DDvax-positive mosquito bodies were greatly reduced (4%, and 0%, respectively). Furthermore, DDVax did not disseminate to legs/wings in any of the goat-fed mosquitoes. Collectively, these results are consistent with a beneficial environmental safety profile.
Collapse
Affiliation(s)
- Corey L. Campbell
- Department of MicrobiologyImmunology, and PathologyCenter for Vector‐Borne Infectious DiseasesColorado State UniversityFort CollinsColorado
| | - Trey K. Snell
- Department of MicrobiologyImmunology, and PathologyCenter for Vector‐Borne Infectious DiseasesColorado State UniversityFort CollinsColorado
| | - Susi Bennett
- Department of MicrobiologyImmunology, and PathologyCenter for Vector‐Borne Infectious DiseasesColorado State UniversityFort CollinsColorado
| | - John H. Wyckoff
- BioMARC, Infectious Diseases Research Center, Colorado State UniversityFort CollinsColorado
| | - Darragh Heaslip
- BioMARC, Infectious Diseases Research Center, Colorado State UniversityFort CollinsColorado
| | - Jordan Flatt
- BioMARC, Infectious Diseases Research Center, Colorado State UniversityFort CollinsColorado
| | - Emma K. Harris
- Department of MicrobiologyImmunology, and PathologyCenter for Vector‐Borne Infectious DiseasesColorado State UniversityFort CollinsColorado
| | - Daniel A. Hartman
- Department of MicrobiologyImmunology, and PathologyCenter for Vector‐Borne Infectious DiseasesColorado State UniversityFort CollinsColorado
| | - Elena Lian
- Department of MicrobiologyImmunology, and PathologyCenter for Vector‐Borne Infectious DiseasesColorado State UniversityFort CollinsColorado
| | - Brian H. Bird
- School of Veterinary MedicineOne Health InstituteUniversity of CaliforniaDavisCalifornia
| | - Mark D. Stenglein
- Department of MicrobiologyImmunology, and PathologyCenter for Vector‐Borne Infectious DiseasesColorado State UniversityFort CollinsColorado
| | - Richard A. Bowen
- Department of MicrobiologyImmunology, and PathologyCenter for Vector‐Borne Infectious DiseasesColorado State UniversityFort CollinsColorado
| | - Rebekah C. Kading
- Department of MicrobiologyImmunology, and PathologyCenter for Vector‐Borne Infectious DiseasesColorado State UniversityFort CollinsColorado
| |
Collapse
|
13
|
Alzuheir I, Helal BA, Helal MA, Fayyad A, Jalboush N. No evidence of Rift Valley fever antibodies in veterinarians and sheep in Northern Palestine. Vet World 2022; 15:1990-1995. [PMID: 36313834 PMCID: PMC9615502 DOI: 10.14202/vetworld.2022.1990-1995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/05/2022] [Indexed: 11/16/2022] Open
Abstract
Background and Aim: Rift Valley fever virus (RVFV) is a vector-borne virus that causes RVF in humans and ruminants. The clinical symptoms in humans and animals are non-specific and often misdiagnosed, but abortions in ruminants and high mortality in young animals are characteristic. Since the initial outbreak in the Rift Valley area in Kenya, the disease has spread to most African countries and the Middle East. The presence and epidemiological status of RVFV in humans and animals in Palestine are unknown. This study aimed to investigate the presence and risk factors for RVF seroprevalence in veterinarians, as occupational hazard professionals, and sheep, as highly susceptible animals, in Northern Palestine. Materials and Methods: A cross-sectional study was conducted. Data and blood samples of 280 Assaf sheep and 100 veterinarians in close occupational contact with sheep were collected between August and September 2020 using an indirect enzyme-linked immunosorbent assay. Results: No evidence of RVF antibodies was found in any human or animal sample. Conclusion: Our results suggest that RVFV has not circulated in livestock in Northern Palestine, yet. Surveillance and response capabilities and cooperation with the nearby endemic regions are recommended. The distribution of competent vectors in Palestine, associated with global climate change and the role of wild animals, might be a possible route for RVF spreading to Palestine from neighboring countries.
Collapse
Affiliation(s)
- Ibrahim Alzuheir
- Department of Veterinary Medicine, An-Najah National University, P.O. Box 7 Nablus, Palestine
| | - Belal Abu Helal
- Department of Veterinary Medicine, An-Najah National University, P.O. Box 7 Nablus, Palestine
| | - Mohammad Abu Helal
- Department of Public Health Sciences, Faculty of Graduate Studies, An-Najah National University, P.O. Box 7 Nablus, Palestine
| | - Adnan Fayyad
- Department of Veterinary Medicine, An-Najah National University, P.O. Box 7 Nablus, Palestine
| | - Nasr Jalboush
- Department of Veterinary Medicine, An-Najah National University, P.O. Box 7 Nablus, Palestine
| |
Collapse
|
14
|
Terasaki K, Kalveram B, Johnson KN, Juelich T, Smith JK, Zhang L, Freiberg AN, Makino S. Rift Valley fever virus 78kDa envelope protein attenuates virus replication in macrophage-derived cell lines and viral virulence in mice. PLoS Negl Trop Dis 2021; 15:e0009785. [PMID: 34516560 PMCID: PMC8460012 DOI: 10.1371/journal.pntd.0009785] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2021] [Revised: 09/23/2021] [Accepted: 09/02/2021] [Indexed: 12/27/2022] Open
Abstract
Rift Valley fever virus (RVFV) is a mosquito-borne bunyavirus with a wide host range including ruminants and humans. RVFV outbreaks have had devastating effects on public health and the livestock industry in African countries. However, there is no approved RVFV vaccine for human use in non-endemic countries and no FDA-approved antiviral drug for RVFV treatment. The RVFV 78kDa protein (P78), which is a membrane glycoprotein, plays a role in virus dissemination in the mosquito host, but its biological role in mammalian hosts remains unknown. We generated an attenuated RVFV MP-12 strain-derived P78-High virus and a virulent ZH501 strain-derived ZH501-P78-High virus, both of which expressed a higher level of P78 and carried higher levels of P78 in the virion compared to their parental viruses. We also generated another MP-12-derived mutant virus (P78-KO virus) that does not express P78. MP-12 and P78-KO virus replicated to similar levels in fibroblast cell lines and Huh7 cells, while P78-High virus replicated better than MP-12 in Vero E6 cells, fibroblast cell lines, and Huh7 cells. Notably, P78-High virus and P78-KO virus replicated less efficiently and more efficiently, respectively, than MP-12 in macrophage cell lines. ZH501-P78-High virus also replicated poorly in macrophage cell lines. Our data further suggest that inefficient binding of P78-High virus to the cells led to inefficient virus internalization, low virus infectivity and reduced virus replication in a macrophage cell line. P78-High virus and P78-KO virus showed lower and higher virulence than MP-12, respectively, in young mice. ZH501-P78-High virus also exhibited lower virulence than ZH501 in mice. These data suggest that high levels of P78 expression attenuate RVFV virulence by preventing efficient virus replication in macrophages. Genetic alteration leading to increased P78 expression may serve as a novel strategy for the attenuation of RVFV virulence and generation of safe RVFV vaccines.
Collapse
Affiliation(s)
- Kaori Terasaki
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute of Human Infection and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail: (KT); (SM)
| | - Birte Kalveram
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Kendra N. Johnson
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Terry Juelich
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Jennifer K. Smith
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Lihong Zhang
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Alexander N. Freiberg
- Institute of Human Infection and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Department of Pathology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Biodefense and Emerging Infectious Diseases, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Institute for Vaccine Sciences, The University of Texas Medical Branch, Galveston, Texas, United States of America
| | - Shinji Makino
- Department of Microbiology and Immunology, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Institute of Human Infection and Immunity, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Biodefense and Emerging Infectious Diseases, the University of Texas Medical Branch, Galveston, Texas, United States of America
- Center for Tropical Diseases, The University of Texas Medical Branch, Galveston, Texas, United States of America
- Sealy Institute for Vaccine Sciences, The University of Texas Medical Branch, Galveston, Texas, United States of America
- * E-mail: (KT); (SM)
| |
Collapse
|
15
|
Anthony T, van Schalkwyk A, Romito M, Odendaal L, Clift SJ, Davis AS. Vaccination with Rift Valley fever virus live attenuated vaccine strain Smithburn caused meningoencephalitis in alpacas. J Vet Diagn Invest 2021; 33:777-781. [PMID: 34041966 DOI: 10.1177/10406387211015294] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Rift Valley fever (RVF) is a zoonotic, viral, mosquito-borne disease that causes considerable morbidity and mortality in humans and livestock in Africa and the Arabian Peninsula. In June 2018, 4 alpaca inoculated subcutaneously with live attenuated RVF virus (RVFV) Smithburn strain exhibited pyrexia, aberrant vocalization, anorexia, neurologic signs, and respiratory distress. One animal died the evening of inoculation, and 2 at ~20 d post-inoculation. Concern regarding potential vaccine strain reversion to wild-type RVFV or vaccine-induced disease prompted autopsy of the latter two. Macroscopically, both alpacas had severe pulmonary edema and congestion, myocardial hemorrhages, and cyanotic mucous membranes. Histologically, they had cerebral nonsuppurative encephalomyelitis with perivascular cuffing, multifocal neuronal necrosis, gliosis, and meningitis. Lesions were more severe in the 4-mo-old cria. RVFV antigen and RNA were present in neuronal cytoplasm, by immunohistochemistry and in situ hybridization (ISH) respectively, and cerebrum was also RVFV positive by RT-rtPCR. The virus clustered in lineage K (100% sequence identity), with close association to Smithburn sequences published previously (identity: 99.1-100%). There was neither evidence of an aberrant immune-mediated reaction nor reassortment with wild-type virus. The evidence points to a pure infection with Smithburn vaccine strain as the cause of the animals' disease.
Collapse
Affiliation(s)
- Tasneem Anthony
- Provincial Veterinary Laboratory, Department of Agriculture, Western Cape Government, Capetown, South Africa
| | - Antoinette van Schalkwyk
- South Africa Agricultural Research Council-Onderstepoort Veterinary Institute, Onderstepoort, South Africa
| | - Marco Romito
- South Africa Agricultural Research Council-Onderstepoort Veterinary Institute, Onderstepoort, South Africa
| | - Lieza Odendaal
- Department of Paraclinical Sciences, University of Pretoria, Onderstepoort, South Africa
| | - Sarah J Clift
- Department of Paraclinical Sciences, University of Pretoria, Onderstepoort, South Africa
| | - A Sally Davis
- Department of Paraclinical Sciences, University of Pretoria, Onderstepoort, South Africa.,Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| |
Collapse
|
16
|
Rift Valley fever virus detection in susceptible hosts with special emphasis in insects. Sci Rep 2021; 11:9822. [PMID: 33972596 PMCID: PMC8110843 DOI: 10.1038/s41598-021-89226-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 04/15/2021] [Indexed: 11/09/2022] Open
Abstract
Rift Valley fever phlebovirus (RVFV, Phenuiviridae) is an emerging arbovirus that can cause potentially fatal disease in many host species including ruminants and humans. Thus, tools to detect this pathogen within tissue samples from routine diagnostic investigations or for research purposes are of major interest. This study compares the immunohistological usefulness of several mono- and polyclonal antibodies against RVFV epitopes in tissue samples derived from natural hosts of epidemiologic importance (sheep), potentially virus transmitting insect species (Culex quinquefasciatus, Aedes aegypti) as well as scientific infection models (mouse, Drosophila melanogaster, C6/36 cell pellet). While the nucleoprotein was the epitope most prominently detected in mammal and mosquito tissue samples, fruit fly tissues showed expression of glycoproteins only. Antibodies against non-structural proteins exhibited single cell reactions in salivary glands of mosquitoes and the C6/36 cell pellet. However, as single antibodies exhibited a cross reactivity of varying degree in non-infected specimens, a careful interpretation of positive reactions and consideration of adequate controls remains of critical importance. The results suggest that primary antibodies directed against viral nucleoproteins and glycoproteins can facilitate RVFV detection in mammals and insects, respectively, and therefore will allow RVFV detection for diagnostic and research purposes.
Collapse
|
17
|
Odendaal L, Davis AS, Venter EH. Insights into the Pathogenesis of Viral Haemorrhagic Fever Based on Virus Tropism and Tissue Lesions of Natural Rift Valley Fever. Viruses 2021; 13:v13040709. [PMID: 33923863 PMCID: PMC8073615 DOI: 10.3390/v13040709] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2021] [Revised: 03/26/2021] [Accepted: 04/02/2021] [Indexed: 12/20/2022] Open
Abstract
Rift Valley fever phlebovirus (RVFV) infects humans and a wide range of ungulates and historically has caused devastating epidemics in Africa and the Arabian Peninsula. Lesions of naturally infected cases of Rift Valley fever (RVF) have only been described in detail in sheep with a few reports concerning cattle and humans. The most frequently observed lesion in both ruminants and humans is randomly distributed necrosis, particularly in the liver. Lesions supportive of vascular endothelial injury are also present and include mild hydropericardium, hydrothorax and ascites; marked pulmonary congestion and oedema; lymph node congestion and oedema; and haemorrhages in many tissues. Although a complete understanding of RVF pathogenesis is still lacking, antigen-presenting cells in the skin are likely the early targets of the virus. Following suppression of type I IFN production and necrosis of dermal cells, RVFV spreads systemically, resulting in infection and necrosis of other cells in a variety of organs. Failure of both the innate and adaptive immune responses to control infection is exacerbated by apoptosis of lymphocytes. An excessive pro-inflammatory cytokine and chemokine response leads to microcirculatory dysfunction. Additionally, impairment of the coagulation system results in widespread haemorrhages. Fatal outcomes result from multiorgan failure, oedema in many organs (including the lungs and brain), hypotension, and circulatory shock. Here, we summarize current understanding of RVF cellular tropism as informed by lesions caused by natural infections. We specifically examine how extant knowledge informs current understanding regarding pathogenesis of the haemorrhagic fever form of RVF, identifying opportunities for future research.
Collapse
Affiliation(s)
- Lieza Odendaal
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria 0002, South Africa
- Correspondence: (L.O.); (A.S.D.)
| | - A Sally Davis
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria 0002, South Africa
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS 66506, USA
- Correspondence: (L.O.); (A.S.D.)
| | - Estelle H Venter
- Department of Veterinary Tropical Diseases, Faculty of Veterinary Science, University of Pretoria, Pretoria 0002, South Africa;
- College of Public Health Medical and Veterinary Sciences, Discipline Veterinary Science, James Cook University, Townsville, QLD 4811, Australia
| |
Collapse
|
18
|
van Schalkwyk A, Gwala S, Schuck KN, Quan M, Davis AS, Romito M, Odendaal L. Retrospective phylogenetic analyses of formalin-fixed paraffin-embedded samples from the 2011 Rift Valley fever outbreak in South Africa, through sequencing of targeted regions. J Virol Methods 2020; 287:114003. [PMID: 33164863 DOI: 10.1016/j.jviromet.2020.114003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2020] [Revised: 10/13/2020] [Accepted: 10/18/2020] [Indexed: 11/25/2022]
Abstract
The last major Rift Valley fever outbreak in South Africa was between 2008 and 2011. Viruses isolated between 2008 and 2010 were phylogenetically assigned to Lineage C, Lineage K and the novel lineage H. The 2011 outbreaks occurred primarily in the Eastern, Western and Northern Cape provinces, with no sequence data or phylogenetic relationship published. Samples from these outbreaks were submitted to the Faculty of Veterinary Sciences, University of Pretoria, for immunohistochemical confirmation of Rift Valley fever phlebovirus presence. These samples were formalin-fixed and paraffin-embedded (FFPE) and stored at the Pathology section for several years. This study describes a modified extraction method used to obtain RNA from the FFPE samples, as well as the primer combinations used to phylogenetically classify them as belonging to the novel lineage H.
Collapse
Affiliation(s)
- Antoinette van Schalkwyk
- Agricultural Research Council - Onderstepoort Veterinary Institute. Onderstepoort, Pretoria, South Africa.
| | - Sipho Gwala
- Faculty of Veterinary Sciences, University of Pretoria, Onderstepoort, Pretoria, South Africa
| | - Kaitlyn N Schuck
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine. Kansas State University, Manhattan, KS, USA
| | - Melvyn Quan
- Faculty of Veterinary Sciences, University of Pretoria, Onderstepoort, Pretoria, South Africa
| | - Anne Sally Davis
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine. Kansas State University, Manhattan, KS, USA
| | - Marco Romito
- Agricultural Research Council - Onderstepoort Veterinary Institute. Onderstepoort, Pretoria, South Africa
| | - Lieza Odendaal
- Faculty of Veterinary Sciences, University of Pretoria, Onderstepoort, Pretoria, South Africa
| |
Collapse
|
19
|
Odendaal L, Clift SJ, Fosgate GT, Davis AS. Ovine Fetal and Placental Lesions and Cellular Tropism in Natural Rift Valley Fever Virus Infections. Vet Pathol 2020; 57:791-806. [PMID: 32885745 DOI: 10.1177/0300985820954549] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Infection with Rift Valley fever phlebovirus (RVFV) causes abortion storms and a wide variety of outcomes for both ewes and fetuses. Sheep fetuses and placenta specimens were examined during the 2010-2011 River Valley fever (RVF) outbreak in South Africa. A total of 72 fetuses were studied of which 58 were confirmed positive for RVF. Placenta specimens were available for 35 cases. Macroscopic lesions in fetuses were nonspecific and included marked edema and occasional hemorrhages in visceral organs. Microscopically, multifocal hepatic necrosis was present in 48 of 58 cases, and apoptotic bodies, foci of liquefactive hepatic necrosis (primary foci), and eosinophilic intranuclear inclusions in hepatocytes were useful diagnostic features. Lymphocytolysis was present in all lymphoid organs examined with the exception of thymus and Peyer's patches, and pyknosis or karyorrhexis was often present in renal glomeruli. The most significant histologic lesion in the placenta was necrosis of trophoblasts and endothelial cells in the cotyledonary and intercotyledonary chorioallantois. Immunolabeling for RVFV was most consistent in trophoblasts of the cotyledon or caruncle. Other antigen-positive cells included hepatocytes, renal tubular epithelial, juxtaglomerular and extraglomerular mesangial cells, vascular smooth muscle, endothelial and adrenocortical cells, cardiomyocytes, Purkinje fibers, and macrophages. Fetal organ samples for diagnosis must minimally include liver, kidney, and spleen. From the placenta, the minimum recommended specimens for histopathology include the cotyledonary units and caruncles from the endometrium, if available. The diagnostic investigation of abortion in endemic areas should always include routine testing for RVFV, and a diagnosis during interepidemic periods might be missed if only limited specimens are available for examination.
Collapse
Affiliation(s)
- Lieza Odendaal
- 56410University of Pretoria, Onderstepoort, Pretoria, South Africa
| | - Sarah J Clift
- 56410University of Pretoria, Onderstepoort, Pretoria, South Africa
| | | | - A Sally Davis
- 56410University of Pretoria, Onderstepoort, Pretoria, South Africa.,5308Kansas State University, Manhattan, KS, USA
| |
Collapse
|
20
|
Calvo-Pinilla E, Marín-López A, Moreno S, Lorenzo G, Utrilla-Trigo S, Jiménez-Cabello L, Benavides J, Nogales A, Blasco R, Brun A, Ortego J. A protective bivalent vaccine against Rift Valley fever and bluetongue. NPJ Vaccines 2020; 5:70. [PMID: 32793399 PMCID: PMC7393076 DOI: 10.1038/s41541-020-00218-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 07/06/2020] [Indexed: 11/09/2022] Open
Abstract
Rift Valley fever (RVF) and bluetongue (BT) are two important ruminant diseases transmitted by arthropods. Both viruses have shown important geographic spread leading to endemicity of BT virus (BTV) in Africa and Europe. In this work, we report a dual vaccine that simultaneously induces protective immune responses against BTV and RVFV based on modified vaccinia Ankara virus (MVA) expressing BTV proteins VP2, NS1, or a truncated form of NS1 (NS1-Nt), and RVFV Gn and Gc glycoproteins. IFNAR(-/-) mice immunized with two doses of MVA-GnGc-VP2 developed a significant neutralizing antibody response against BTV-4 and RVFV. Furthermore, the homologous prime-boost immunization with MVA-GnGc-NS1 or MVA-GnGc-NS1-Nt triggered neutralizing antibodies against RVFV and NS1-specific cytotoxic CD8+ T cells in mice. Moreover, all mice immunized with MVA-GnGc-NS1 or MVA-GnGc-NS1-Nt remained healthy after lethal challenge with RVFV or BTV-4. The homologous prime-boost vaccination with MVA-GnGc-NS1, which was the best immunization strategy observed in mice, was assayed in sheep. Clinical signs and viremia were absent or highly reduced in vaccinated sheep after challenge with BTV-4 or RVFV. These results indicate that MVA-GnGc-NS1 vaccination elicits immune protection against RVFV and BTV in sheep.
Collapse
Affiliation(s)
- Eva Calvo-Pinilla
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Centro de Investigación en Sanidad Animal (INIA-CISA), Madrid, Spain
| | - Alejandro Marín-López
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Centro de Investigación en Sanidad Animal (INIA-CISA), Madrid, Spain.,Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, CT USA
| | - Sandra Moreno
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Centro de Investigación en Sanidad Animal (INIA-CISA), Madrid, Spain
| | - Gema Lorenzo
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Centro de Investigación en Sanidad Animal (INIA-CISA), Madrid, Spain
| | - Sergio Utrilla-Trigo
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Centro de Investigación en Sanidad Animal (INIA-CISA), Madrid, Spain
| | - Luis Jiménez-Cabello
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Centro de Investigación en Sanidad Animal (INIA-CISA), Madrid, Spain
| | - Julio Benavides
- Instituto de Ganadería de Montaña (CSIC-Universidad de León), León, Spain
| | - Aitor Nogales
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Centro de Investigación en Sanidad Animal (INIA-CISA), Madrid, Spain
| | - Rafael Blasco
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Departamento de Biotecnología, Madrid, Spain
| | - Alejandro Brun
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Centro de Investigación en Sanidad Animal (INIA-CISA), Madrid, Spain
| | - Javier Ortego
- Instituto Nacional de Investigación y Tecnología Agraria y Alimentaria, Centro de Investigación en Sanidad Animal (INIA-CISA), Madrid, Spain
| |
Collapse
|
21
|
Smith MR, Schirtzinger EE, Wilson WC, Davis AS. Rift Valley Fever Virus: Propagation, Quantification, and Storage. ACTA ACUST UNITED AC 2020; 55:e92. [PMID: 31763765 DOI: 10.1002/cpmc.92] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Rift Valley fever virus (RVFV) is an arthropod-borne, zoonotic disease endemic to sub-Saharan Africa and the Arabian Peninsula. Outbreaks of Rift Valley fever have had up to 100% mortality rates in fetal and neonatal sheep. Upon infection of ruminant and human hosts alike, RVFV infection causes an at times severe hepatitis and pathology in many other organs. The enveloped virion contains a tripartite, predominantly negative-sense, single-stranded RNA genome, which codes for the proteins the virus needs to replicate both in mammalian hosts and insect vectors. Endemic countries often use attenuated RVFV strains for vaccination of livestock but there are no commercially licensed vaccines for humans or livestock in non-endemic areas. In the laboratory, RVFV can be readily propagated and manipulated in vitro using cell culture systems. Presented in this article are techniques routinely used in RVFV research that have proven successful in our laboratories. © 2019 by John Wiley & Sons, Inc. Basic Protocol 1: Propagation of Rift Valley fever virus in mammalian cells Basic Protocol 2: Quantification of Rift Valley fever virus by plaque assay Basic Protocol 3: Quantification of Rift Valley fever virus by 50% tissue culture infectious dose (TCID50 ) assay Basic Protocol 4: Quantification of Rift Valley fever virus by focus-forming assay Basic Protocol 5: Storage and disinfection Alternate Protocol 1: Propagation of Rift Valley fever virus in MRC-5 cells Alternate Protocol 2: Propagation of RVFV in mosquito-derived cells Alternate Protocol 3: TCID50 detection using fluorescence visualization Support Protocol 1: Calculation of the amount of virus needed to infect a flask at a chosen multiplicity of infection Support Protocol 2: Calculation of the virus titer by plaque assay or focus-forming assay Support Protocol 3: Calculation of the TCID50 titer by the method of Reed and Muench Support Protocol 4: Calculation of the antibody volume for the focus-forming assay.
Collapse
Affiliation(s)
- MaRyka R Smith
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Erin E Schirtzinger
- Arthropod-Borne Animal Diseases Research Unit, Agricultural Research Service, United States Department of Agriculture, Manhattan, Kansas
| | - William C Wilson
- Arthropod-Borne Animal Diseases Research Unit, Agricultural Research Service, United States Department of Agriculture, Manhattan, Kansas
| | - A Sally Davis
- Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| |
Collapse
|
22
|
Batista L, Jouvion G, Simon-Chazottes D, Houzelstein D, Burlen-Defranoux O, Boissière M, Tokuda S, do Valle TZ, Cumano A, Flamand M, Montagutelli X, Panthier JJ. Genetic dissection of Rift Valley fever pathogenesis: Rvfs2 locus on mouse chromosome 11 enables survival to early-onset hepatitis. Sci Rep 2020; 10:8734. [PMID: 32457349 PMCID: PMC7250886 DOI: 10.1038/s41598-020-65683-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Accepted: 05/04/2020] [Indexed: 11/19/2022] Open
Abstract
Infection of mice with Rift Valley fever virus (RVFV) reproduces major pathological features of severe human disease, notably the early-onset hepatitis and delayed-onset encephalitis. We previously reported that the Rvfs2 locus from the susceptible MBT/Pas strain reduces survival time after RVFV infection. Here, we used BALB/cByJ (BALB) mice congenic for Rvfs2 (C.MBT-Rvfs2) to investigate the pathophysiological mechanisms impacted by Rvfs2. Clinical, biochemical and histopathological features indicated similar liver damage in BALB and C.MBT-Rvfs2 mice until day 5 after infection. However, while C.MBT-Rvfs2 mice succumbed from acute liver injury, most BALB mice recovered and died later of encephalitis. Hepatocytes of BALB infected liver proliferated actively on day 6, promoting organ regeneration and recovery from liver damage. By comparison with C.MBT-Rvfs2, BALB mice had up to 100-fold lower production of infectious virions in the peripheral blood and liver, strongly decreased RVFV protein in liver and reduced viral replication in primary cultured hepatocytes, suggesting that the BALB Rvfs2 haplotype limits RVFV pathogenicity through decreased virus replication. Moreover, bone marrow chimera experiments showed that both hematopoietic and non-hematopoietic cells are required for the protective effect of the BALB Rvfs2 haplotype. Altogether, these results indicate that Rvfs2 controls critical events which allow survival to RVFV-induced hepatitis.
Collapse
Affiliation(s)
- Leandro Batista
- Mouse Functional Genetics, Institut Pasteur, UMR3738, CNRS, Paris, 75015, France.,Sorbonne Université, IFD, Paris, 75005, France
| | - Gregory Jouvion
- Experimental Neuropathology, Institut Pasteur, Paris, 75015, France.,Sorbonne Université, INSERM, Physiopathologie des Maladies Génétiques d'Expression Pédiatrique, APHP, Hôpital Armand Trousseau, UF de Génétique Moléculaire, Paris, 75012, France
| | - Dominique Simon-Chazottes
- Mouse Functional Genetics, Institut Pasteur, UMR3738, CNRS, Paris, 75015, France.,Mouse Genetics, Institut Pasteur, Paris, 75015, France
| | - Denis Houzelstein
- Mouse Functional Genetics, Institut Pasteur, UMR3738, CNRS, Paris, 75015, France
| | | | | | - Satoko Tokuda
- Mouse Functional Genetics, Institut Pasteur, UMR3738, CNRS, Paris, 75015, France
| | - Tania Zaverucha do Valle
- Mouse Functional Genetics, Institut Pasteur, UMR3738, CNRS, Paris, 75015, France.,Laboratório de Imunomodulação e Protozoologia, Instituto Oswaldo Cruz, Fiocruz, Rio de Janeiro, Brasil
| | - Ana Cumano
- Lymphopoiesis, Institut Pasteur, U668, INSERM, Paris, 75015, France
| | - Marie Flamand
- Structural Virology, Institut Pasteur, Paris, 75015, France
| | - Xavier Montagutelli
- Mouse Functional Genetics, Institut Pasteur, UMR3738, CNRS, Paris, 75015, France. .,Mouse Genetics, Institut Pasteur, Paris, 75015, France.
| | | |
Collapse
|
23
|
Shiell BJ, Ye S, Harper JA, van der Heide B, Beddome G, Foord AJ, Michalski WP, Bingham J, Peck GR. Reagents for detection of Rift Valley fever virus infection in sheep. J Vet Diagn Invest 2020; 32:577-580. [PMID: 32450762 DOI: 10.1177/1040638720926476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Rift Valley fever virus (RVFV) causes Rift Valley fever (RVF), resulting in morbidity and mortality in humans and ruminants. Evidence of transboundary outbreaks means that RVFV remains a threat to human health and livestock industries in countries that are free from the disease. To enhance surveillance capability, methods for detection of RVFV are required. The generation of reagents suitable for the detection of RVFV antigen in formalin-fixed, paraffin-embedded tissues from infected animals have been developed and are described herein. Recombinant nucleoprotein (rNP) was expressed in Escherichia coli and purified using immobilized metal ion affinity chromatography. Purified rNP was used as an immunogen to produce anti-NP polyclonal antisera in rabbits for use in detection of RVFV NP in experimentally infected animals by immunohistochemistry. Antisera raised in rabbits against rNP were able to recognize viral NP antigen in fixed infected Vero cell pellets and sheep liver. Therefore, the methods and reagents described herein are useful in assays for detection of RVFV infections in animals, for research and surveillance purposes.
Collapse
Affiliation(s)
- Brian J Shiell
- CSIRO-Australian Animal Health Laboratory, East Geelong, Victoria, Australia
| | - Siying Ye
- CSIRO-Australian Animal Health Laboratory, East Geelong, Victoria, Australia
| | - Jennifer A Harper
- CSIRO-Australian Animal Health Laboratory, East Geelong, Victoria, Australia
| | | | - Gary Beddome
- CSIRO-Australian Animal Health Laboratory, East Geelong, Victoria, Australia
| | - Adam J Foord
- CSIRO-Australian Animal Health Laboratory, East Geelong, Victoria, Australia
| | - Wojtek P Michalski
- CSIRO-Australian Animal Health Laboratory, East Geelong, Victoria, Australia
| | - John Bingham
- CSIRO-Australian Animal Health Laboratory, East Geelong, Victoria, Australia
| | - Grantley R Peck
- CSIRO-Australian Animal Health Laboratory, East Geelong, Victoria, Australia
| |
Collapse
|
24
|
Le Tortorec A, Matusali G, Mahé D, Aubry F, Mazaud-Guittot S, Houzet L, Dejucq-Rainsford N. From Ancient to Emerging Infections: The Odyssey of Viruses in the Male Genital Tract. Physiol Rev 2020; 100:1349-1414. [PMID: 32031468 DOI: 10.1152/physrev.00021.2019] [Citation(s) in RCA: 56] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The male genital tract (MGT) is the target of a number of viral infections that can have deleterious consequences at the individual, offspring, and population levels. These consequences include infertility, cancers of male organs, transmission to the embryo/fetal development abnormalities, and sexual dissemination of major viral pathogens such as human immunodeficiency virus (HIV) and hepatitis B virus. Lately, two emerging viruses, Zika and Ebola, have additionally revealed that the human MGT can constitute a reservoir for viruses cleared from peripheral circulation by the immune system, leading to their sexual transmission by cured men. This represents a concern for future epidemics and further underlines the need for a better understanding of the interplay between viruses and the MGT. We review here how viruses, from ancient viruses that integrated the germline during evolution through old viruses (e.g., papillomaviruses originating from Neanderthals) and more modern sexually transmitted infections (e.g., simian zoonotic HIV) to emerging viruses (e.g., Ebola and Zika) take advantage of genital tract colonization for horizontal dissemination, viral persistence, vertical transmission, and endogenization. The MGT immune responses to viruses and the impact of these infections are discussed. We summarize the latest data regarding the sources of viruses in semen and the complex role of this body fluid in sexual transmission. Finally, we introduce key animal findings that are relevant for our understanding of viral infection and persistence in the human MGT and suggest future research directions.
Collapse
Affiliation(s)
- Anna Le Tortorec
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Giulia Matusali
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Dominique Mahé
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Florence Aubry
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Séverine Mazaud-Guittot
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Laurent Houzet
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| | - Nathalie Dejucq-Rainsford
- University of Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail)-UMR_S1085, Rennes, France
| |
Collapse
|
25
|
Abstract
Rift Valley fever (RVF) is a mosquito-borne viral disease, principally of ruminants, that is endemic to Africa. The causative Phlebovirus, Rift Valley fever virus (RVFV), has a broad host range and, as such, also infects humans to cause primarily a self-limiting febrile illness. A small number of human cases will also develop severe complications, including haemorrhagic fever, encephalitis and visual impairment. In parts of Africa, it is a major disease of domestic ruminants, causing epidemics of abortion and mortality. It infects and can be transmitted by a broad range of mosquitos, with those of the genus Aedes and Culex thought to be the major vectors. Therefore, the virus has the potential to become established beyond Africa, including in Australia, where competent vector hosts are endemic. Vaccines for humans have not yet been developed to the commercial stage. This review examines the threat of this virus, with particular reference to Australia, and assesses gaps in our knowledge that may benefit from research focus.
Collapse
|
26
|
Teffera M, Babiuk S. Potential of Using Capripoxvirus Vectored Vaccines Against Arboviruses in Sheep, Goats, and Cattle. Front Vet Sci 2019; 6:450. [PMID: 31921911 PMCID: PMC6932975 DOI: 10.3389/fvets.2019.00450] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Accepted: 11/27/2019] [Indexed: 11/26/2022] Open
Abstract
The genus capripoxvirus consists of sheeppox virus, goatpox virus, and lumpy skin disease virus, which affect sheep, goats, and cattle, respectively. Together capripoxviruses cause significant economic losses to the sheep, goat, and cattle industry where these diseases are present. These diseases have spread into previously free bordering regions most recently demonstrated with the spread of lumpy skin disease virus into the Middle East, some Eastern European countries, and Russia. This recent spread has highlighted the transboundary nature of these diseases. To control lumpy skin disease virus, live attenuated viral vaccines are used in endemic countries as well as in response to an outbreak. For sheeppox and goatpox, live attenuated viral vaccines are used in endemic countries; these diseases can also be contained through slaughter of infected animals to stamp out the disease. The thermostability, narrow host range, and ability of capripoxviruses to express a wide variety of antigens make capripoxviruses ideal vectors. The ability to immunize animals against multiple diseases simultaneously increases vaccination efficiency by decreasing the number of vaccinations required. Additionally, the use of capripoxvirus vectored vaccines allows the possibility of differentiating infected from vaccinated animals. Arboviruses such as bluetongue virus and Rift Valley fever viruses are also responsible for significant economic losses in endemic countries. In the case of Rift Valley fever virus, vaccination is not routinely practiced unless there is an outbreak making vaccination not as effective, therefore, incorporating Rift Valley fever vaccination into routine capripoxvirus vaccination would be highly beneficial. This review will discuss the potential of using capripoxvirus as a vector expressing protective arboviral antigens.
Collapse
Affiliation(s)
- Mahder Teffera
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| | - Shawn Babiuk
- Canadian Food Inspection Agency, National Centre for Foreign Animal Disease, Winnipeg, MB, Canada
- Department of Immunology, University of Manitoba, Winnipeg, MB, Canada
| |
Collapse
|
27
|
Odendaal L, Davis AS, Fosgate GT, Clift SJ. Lesions and Cellular Tropism of Natural Rift Valley Fever Virus Infection in Young Lambs. Vet Pathol 2019; 57:66-81. [PMID: 31842723 DOI: 10.1177/0300985819882633] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
A clear distinction can be made regarding the susceptibility to and the severity of lesions in young lambs when compared to adult sheep. In particular, there are important differences in the lesions and tropism of Rift Valley fever virus (RVFV) in the liver, kidneys, and lymphoid tissues of young lambs. A total of 84 lambs (<6 weeks old), necropsied during the 2010 to 2011 Rift Valley fever (RVF) outbreak in South Africa, were examined by histopathology and immunohistochemistry (IHC). Of the 84 lambs, 71 were positive for RVFV. The most striking diagnostic feature in infected lambs was diffuse necrotizing hepatitis with multifocal liquefactive hepatic necrosis (primary foci) against a background of diffuse hepatocellular death. Lymphocytolysis was present in all lymphoid organs except for the thymus. Lesions in the kidney rarely progressed beyond hydropic change and occasional pyknosis or karyolysis in renal tubular epithelial cells. Viral antigen was diffusely present in the cytoplasm of hepatocytes, but this labeling was noticeably sparse in primary foci. Immunolabeling for RVFV in young lambs was also detected in macrophages, vascular smooth muscle cells, adrenocortical epithelial cells, renal tubular epithelial cells, renal perimacular cells, and cardiomyocytes. RVFV immunolabeling was also often present in capillaries and small blood vessels either as non-cell-associated viral antigen, as antigen in endothelial cells, or intravascular cellular debris. Specimens from the liver, spleen, kidney, and lungs were adequate to confirm a diagnosis of RVF. Characteristic lesions were present in these organs with the liver and spleen being the most consistently positive for RVFV by IHC.
Collapse
Affiliation(s)
- Lieza Odendaal
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa
| | - A Sally Davis
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa.,Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, KS, USA
| | - Geoffrey T Fosgate
- Department of Production Animal Studies, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa
| | - Sarah J Clift
- Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa
| |
Collapse
|
28
|
Ragan IK, Schuck KN, Upreti D, Odendaal L, Richt JA, Trujillo JD, Wilson WC, Davis AS. Rift Valley Fever Viral RNA Detection by In Situ Hybridization in Formalin-Fixed, Paraffin-Embedded Tissues. Vector Borne Zoonotic Dis 2019; 19:553-556. [PMID: 30720389 DOI: 10.1089/vbz.2018.2383] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Sporadic outbreaks of Rift Valley fever virus (RVFV), a zoonotic, mosquito-borne Phlebovirus, cause abortion storms and death in sheep and cattle resulting in catastrophic economic impacts in endemic regions of Africa. More recently, with changes in competent vector distribution, growing international trade, and its potential use for bioterrorism, RVFV has become a transboundary animal disease of significant concern. New and sensitive techniques that determine RVFV presence, while lessening the potential for environmental contamination and human risk, through the use of inactivated, noninfectious samples such as formalin-fixed, paraffin-embedded (FFPE) tissues are needed. FFPE tissue in situ hybridization (ISH) enables the detection of nucleic acid sequences within the visual context of cellular and tissue morphology. Here, we present a chromogenic pan-RVFV ISH assay based on RNAscope® technology, which is able to detect multiple RVFV strains in FFPE tissues, enabling visual correlation of RVFV RNA presence with histopathologic lesions.
Collapse
Affiliation(s)
- Izabela K Ragan
- 1 Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Kaitlynn N Schuck
- 1 Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Deepa Upreti
- 1 Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Lieza Odendaal
- 2 Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa
| | - Juergen A Richt
- 1 Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - Jessie D Trujillo
- 1 Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas
| | - William C Wilson
- 3 USDA-ARS Arthropod-Borne Animal Diseases Research Unit, Center for Grain and Animal Health Research, Manhattan, Kansas
| | - A Sally Davis
- 1 Department of Diagnostic Medicine/Pathobiology, College of Veterinary Medicine, Kansas State University, Manhattan, Kansas.,2 Department of Paraclinical Sciences, Faculty of Veterinary Science, University of Pretoria, Onderstepoort, Pretoria, South Africa
| |
Collapse
|
29
|
Affiliation(s)
- Reiner Ulrich
- 1 Department of Experimental Animal Facilities and Biorisk Management, Friedrich-Loeffler-Institute, Greifswald-Island of Riems, Germany
| |
Collapse
|