Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 188
Filter
1.
Antiviral Res ; 226: 105895, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38679165

ABSTRACT

Rift Valley fever virus (RVFV) is an arbovirus in the Phenuiviridae family identified initially by the large 'abortion storms' observed among ruminants; RVFV can also infect humans. In humans, there is a wide variation of clinical symptoms ranging from subclinical to mild febrile illness to hepatitis, retinitis, delayed-onset encephalitis, or even hemorrhagic fever. The RVFV is a tri-segmented negative-sense RNA virus consisting of S, M, and L segments. The L segment encodes the RNA-dependent RNA polymerase (RdRp), termed the L protein, which is responsible for both viral mRNA synthesis and genome replication. Phosphorylation of viral RdRps is known to regulate viral replication. This study shows that RVFV L protein is serine phosphorylated and identified Casein Kinase 1 alpha (CK1α) and protein phosphatase 1 alpha (PP1α) as L protein binding partners. Inhibition of CK1 and PP1 through small molecule inhibitor treatment, D4476 and 1E7-03, respectively, caused a change in the phosphorylated status of the L protein. Inhibition of PP1α resulted in increased L protein phosphorylation whereas inhibition of CK1α decreased L protein phosphorylation. It was also found that in RVFV infected cells, PP1α localized to the cytoplasmic compartment. Treatment of RVFV infected cells with CK1 inhibitors reduced virus production in both mammalian and mosquito cells. Lastly, inhibition of either CK1 or PP1 reduced viral genomic RNA levels. These data indicate that L protein is phosphorylated and that CK1 and PP1 play a crucial role in regulating the L protein phosphorylation cycle, which is critical to viral RNA production and viral replication.


Subject(s)
Protein Phosphatase 1 , Rift Valley fever virus , Virus Replication , Rift Valley fever virus/physiology , Rift Valley fever virus/genetics , Phosphorylation , Humans , Animals , Protein Phosphatase 1/metabolism , Protein Phosphatase 1/genetics , Genome, Viral , Viral Proteins/metabolism , Viral Proteins/genetics , Casein Kinase Ialpha/metabolism , Casein Kinase Ialpha/genetics , Chlorocebus aethiops , Cell Line , RNA-Dependent RNA Polymerase/metabolism , RNA-Dependent RNA Polymerase/genetics , Vero Cells , RNA, Viral/genetics , RNA, Viral/metabolism , Rift Valley Fever/virology
2.
J Gen Virol ; 105(3)2024 03.
Article in English | MEDLINE | ID: mdl-38546100

ABSTRACT

Rift Valley fever virus (RVFV) is an emerging arboviral disease with pandemic potential. While infection is often self-limiting, a subset of individuals may develop late-onset encephalitis, accounting for up to 20 % of severe cases. Importantly, individuals displaying neurologic disease have up to a 53 % case fatality rate, yet the neuropathogenesis of RVFV infection remains understudied. In this study, we evaluated whether ex vivo postnatal rat brain slice cultures (BSCs) could be used to evaluate RVFV infection in the central nervous system. BSCs mounted an inflammatory response after slicing, which resolved over time, and they were viable in culture for at least 12 days. Infection of rat BSCs with pathogenic RVFV strain ZH501 induced tissue damage and apoptosis over 48 h. Viral replication in BSCs reached up to 1×107 p.f.u. equivalents/ml, depending on inoculation dose. Confocal immunofluorescent microscopy of cleared slices confirmed direct infection of neurons as well as activation of microglia and astrocytes. Further, RVFV-infected rat BSCs produced antiviral cytokines and chemokines, including MCP-1 and GRO/KC. This study demonstrates that rat BSCs support replication of RVFV for ex vivo studies of neuropathogenesis. This allows for continued and complementary investigation into RVFV infection in an ex vivo postnatal brain slice culture format.


Subject(s)
Rift Valley Fever , Rift Valley fever virus , Rats , Animals , Rift Valley fever virus/physiology , Cytokines , Brain , Cell Death
3.
J Virol ; 97(9): e0085323, 2023 09 28.
Article in English | MEDLINE | ID: mdl-37695055

ABSTRACT

Rift Valley fever virus (RVFV) causes mild to severe disease in humans and livestock. Outbreaks of RVFV have been reported throughout Africa and have spread outside Africa since 2000, calling for urgent worldwide attention to this emerging virus. RVFV directly infects the liver, and elevated transaminases are a hallmark of severe RVFV infection. However, the specific contribution of viral replication in hepatocytes to pathogenesis of RVFV remains undefined. To address this, we generated a recombinant miRNA-targeted virus, RVFVmiR-122, to limit hepatocellular replication. MicroRNAs are evolutionarily conserved non-coding RNAs that regulate mRNA expression by targeting them for degradation. RVFVmiR-122 includes an insertion of four target sequences of the liver-specific miR-122. In contrast to control RVFVmiR-184, which contains four target sequences of mosquito-specific miR-184, RVFVmiR-122 has restricted replication in vitro in primary mouse hepatocytes. RVFVmiR-122-infected C57BL/6 mice survived acute hepatitis and instead developed late-onset encephalitis. This difference in clinical outcome was eliminated in Mir-122 KO mice, confirming the specificity of the finding. Interestingly, C57BL/6 mice infected with higher doses of RVFVmiR-122 had a higher survival rate which was correlated with faster clearance of virus from the liver, suggesting a role for activation of host immunity in the phenotype. Together, our data demonstrate that miR-122 can specifically restrict the replication of RVFVmiR-122 in liver tissue both in vitro and in vivo, and this restriction alters the clinical course of disease following RVFVmiR-122 infection. IMPORTANCE Rift Valley fever virus (RVFV) is a hemorrhagic fever virus that causes outbreaks in humans and livestock throughout Africa and has spread to continents outside Africa since 2000. However, no commercial vaccine or treatment is currently available for human use against RVFV. Although the liver has been demonstrated as a key target of RVFV, the contribution of viral replication in hepatocytes to overall RVFV pathogenesis is less well defined. In this study we addressed this question by using a recombinant miRNA-targeted virus with restricted replication in hepatocytes. We gained a better understanding of how this individual cell type contributes to the development of disease caused by RVFV. Techniques used in this study provide an innovative tool to the RVFV field that could be applied to study the consequences of limited RVFV replication in other target cells.


Subject(s)
Hepatocytes , Rift Valley Fever , Rift Valley fever virus , Virus Replication , Animals , Humans , Mice , Hepatocytes/pathology , Hepatocytes/virology , Mice, Inbred C57BL , MicroRNAs/genetics , Rift Valley Fever/virology , Rift Valley fever virus/physiology
4.
J Virol ; 96(20): e0111222, 2022 10 26.
Article in English | MEDLINE | ID: mdl-36194021

ABSTRACT

People infected with the mosquito-borne Rift Valley fever virus (RVFV) can suffer from eye-related problems resulting in ongoing vision issues or even permanent blindness. Despite ocular disease being the most frequently reported severe outcome, it is vastly understudied compared to other disease outcomes caused by RVFV. Ocular manifestations of RVFV include blurred vision, uveitis, and retinitis. When an infected individual develops macular or paramacular lesions, there is a 50% chance of permanent vision loss in one or both eyes. The cause of blinding ocular pathology remains unknown in part due to the lack of a tractable animal model. Using 3 relevant exposure routes, both subcutaneous (SC) and aerosol inoculation of Sprague Dawley rats led to RVFV infection of the eye. Surprisingly, direct inoculation of the conjunctiva did not result in successful ocular infection. The posterior segment of the eye, including the optic nerve, choroid, ciliary body, and retina, were all positive for RVFV antigen in SC-infected rats, and live virus was isolated from the eyes. Proinflammatory cytokines and increased leukocyte counts were also found in the eyes of infected rats. Additionally, human ocular cell lines were permissive for Lrp1-dependent RVFV infection. This study experimentally defines viral tropism of RVFV in the posterior segment of the rat eye and characterizes virally-mediated ocular inflammation, providing a foundation for evaluation of vaccines and therapeutics to protect against adverse ocular outcomes. IMPORTANCE Rift Valley fever virus (RVFV) infection leads to eye damage in humans in up to 10% of reported cases. Permanent blindness occurs in 50% of individuals with significant retinal scarring. Despite the prevalence and severity of this outcome, very little is known about the mechanisms of pathogenesis. We addressed this gap by developing a rodent model of ocular disease. Subcutaneous infection of Sprague Dawley rats resulted in infection of the uvea, retina, and optic nerve along with the induction of inflammation within the posterior eye. Infection of human ocular cells induced inflammatory responses and required host entry factors for RVFV infection similar to rodents. This work provides evidence of how RVFV infects the eye, and this information can be applied to help mitigate the devastating outcomes of RVF ocular disease through vaccines or treatments.


Subject(s)
Eye Diseases , Rift Valley Fever , Rift Valley fever virus , Rats , Humans , Animals , Rift Valley fever virus/physiology , Rats, Sprague-Dawley , Inflammation , Cytokines , Aerosols , Blindness
5.
J Virol ; 96(16): e0067222, 2022 08 24.
Article in English | MEDLINE | ID: mdl-35894603

ABSTRACT

Rift Valley fever virus (RVFV) is endemic in sub-Saharan Africa (SSA), with outbreaks reported in the Arabian Peninsula and throughout SSA. The natural reservoir for RVFV are ruminants, with livestock populations exceeding 50% exposure rates in some areas of SSA. Transmission to humans can occur through exposure to infected livestock products or multiple species of mosquito vectors. In 2013 and 2014, cross-sectional surveys occurred in two districts of Nacala-a-Velha and Mecubúri in northern Mozambique, and participants provided blood samples for later serological assays. IgG against the N protein of RVFV was detected through multiplex bead assay (MBA). Of the 2,278 persons enrolled between the two surveys and study sites, 181 (7.9%, 95% confidence interval (CI): 6.9%-9.1%) were found to be IgG seropositive with increasing seroprevalence with older age and significantly higher seroprevalence in Nacala-a-Velha (10.5%, 8.8%-12.5%) versus Mecubúri (5.7%, 4.5%-7.1%). Seroprevalence estimates were not significantly different between the 2013 and 2014 surveys. Significant spatial clustering of IgG positive persons were consistent among surveys and within the two districts, pointing toward the consistency of serology data for making population-level assumptions regarding RVFV seroprevalence. A subset of persons (n = 539) provided samples for both the 2013 and 2014 surveys, and a low percentage (0.81%) of these were found to seroconvert between these two surveys. Including the RVFV N protein in an MBA antigen panel could assist elucidate RVFV exposure in SSA. IMPORTANCE Due to sporadic transmission, human contact with Rift Valley Fever Virus (RVFV) is difficult to ascertain at a population level. Detection of antibodies against RVFV antigens assist in estimating exposure as antibodies remain in the host long after the virus has been cleared. In this study, we show that antibodies against RVFV N protein can be detected from dried blood spot (DBS) samples being assayed by multiplex bead assay. DBS from two districts in northern Mozambique were tested for IgG against the N protein, and 7.9% of all enrolled persons were seropositive. Older persons, males, and persons residing closer to the coast had higher RVFV N protein seroprevalence. Spatial clustering of IgG positive persons was noted in both districts. These results show low exposure rates to RVFV in these two northern districts in Mozambique, and the ability to perform serology for the RVFV N protein from dried blood samples.


Subject(s)
Microbiological Techniques/methods , Nucleocapsid Proteins/analysis , Rift Valley Fever , Rift Valley fever virus , Aged , Aged, 80 and over , Animals , Antibodies, Viral , Cross-Sectional Studies , Female , Humans , Immunoglobulin G , Livestock , Male , Mozambique/epidemiology , Rift Valley Fever/epidemiology , Rift Valley fever virus/physiology , Seroepidemiologic Studies
6.
Viruses ; 14(2)2022 02 08.
Article in English | MEDLINE | ID: mdl-35215938

ABSTRACT

Rift Valley fever (RVF) is a zoonotic disease caused by RVF Phlebovirus (RVFV). The RVFV MP-12 vaccine strain is known to exhibit residual virulence in the case of a deficient interferon type 1 response. The hypothesis of this study is that virus replication and severity of lesions induced by the MP-12 strain in immunocompromised mice depend on the specific function of the disturbed pathway. Therefore, 10 strains of mice with deficient innate immunity (B6-IFNARtmAgt, C.129S7(B6)-Ifngtm1Ts/J, B6-TLR3tm1Flv, B6-TLR7tm1Aki, NOD/ShiLtJ), helper T-cell- (CD4tm1Mak), cytotoxic T-cell- (CD8atm1Mak), B-cell- (Igh-Jtm1DhuN?+N2), combined T- and B-cell- (NU/J) and combined T-, B-, natural killer (NK) cell- and macrophage-mediated immunity (NOD.Cg-PrkdcscidIl2rgtm1WjI/SzJ (NSG) mice) were subcutaneously infected with RVFV MP-12. B6-IFNARtmAgt mice were the only strain to develop fatal disease due to RVFV-induced severe hepatocellular necrosis and apoptosis. Notably, no clinical disease and only mild multifocal hepatocellular necrosis and apoptosis were observed in NSG mice, while immunohistochemistry detected the RVFV antigen in the liver and the brain. No or low virus expression and no lesions were observed in the other mouse strains. Conclusively, the interferon type 1 response is essential for early control of RVFV replication and disease, whereas functional NK cells, macrophages and lymphocytes are essential for virus clearance.


Subject(s)
Adaptive Immunity , Immunity, Innate , Rift Valley Fever/immunology , Rift Valley fever virus/physiology , Animals , Apoptosis , Female , Humans , Killer Cells, Natural/immunology , Killer Cells, Natural/virology , Liver/immunology , Liver/virology , Macrophages/immunology , Macrophages/virology , Male , Mice , Mice, Inbred NOD , Rift Valley Fever/genetics , Rift Valley Fever/physiopathology , Rift Valley Fever/virology , Rift Valley fever virus/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/virology , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/virology
7.
Viruses ; 13(12)2021 12 02.
Article in English | MEDLINE | ID: mdl-34960686

ABSTRACT

Rift Valley fever virus (RVFV) is a negative-sense, tripartite RNA virus that is endemic to Africa and the Arabian Peninsula. It can cause severe disease and mortality in humans and domestic livestock and is a concern for its potential to spread more globally. RVFV's nucleocapsid protein (N) is an RNA-binding protein that is necessary for viral transcription, replication, and the production of nascent viral particles. We have conducted crosslinking, immunoprecipitation, and sequencing (CLIP-seq) to characterize N interactions with host and viral RNAs during infection. In parallel, to precisely measure intracellular N levels, we employed multiple reaction monitoring mass spectrometry (MRM-MS). Our results show that N binds mostly to host RNAs at early stages of infection, yielding nascent virus particles of reduced infectivity. The expression of N plateaus 10 h post-infection, whereas the intracellular viral RNA concentration continues to increase. Moreover, the virions produced later in infection have higher infectivity. Taken together, the detailed examination of these N-RNA interactions provides insight into how the regulated expression of N and viral RNA produces both infectious and incomplete, noninfectious particles.


Subject(s)
Capsid/metabolism , Nucleocapsid Proteins/metabolism , RNA, Viral/metabolism , Rift Valley fever virus/physiology , Viral Genome Packaging , Animals , Chlorocebus aethiops , HEK293 Cells , Humans , Nucleocapsid Proteins/genetics , Protein Binding , RNA/metabolism , RNA, Antisense/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA-Binding Proteins/metabolism , Vero Cells , Virion/metabolism
8.
Virologie (Montrouge) ; 25(5): 263-279, 2021 10 01.
Article in French | MEDLINE | ID: mdl-34762049

ABSTRACT

Rift Valley fever (RVF) is a major emerging arboviral disease with a complex epidemiological cycle. RVF virus (RVFV) is transmitted by mosquito vectors to ruminants, causing epizootics, and then from animals to humans, triggering epidemics. During its cycle, RVFV infects a wide range of hosts, but the associated pathogenesis has yet to be elucidated. RVFV displays a predominant hepatic tropism, but also has a multicellular tropism inducing physiopathological effects in several tissues. However, there is variability between species in terms of physiopathology : a common clinical picture is found (severe hepatitis, hemorrhages, leukopenia), but certain forms are mainly found in humans (neurological and ocular damage) or in ruminant herds (waves of abortions). Although the molecular mechanisms involved are still poorly understood, it seems that early inflammatory response is related to the severity of the pathology. A better understanding of the pathogenesis of RVFV seems essential, especially since no specific treatment exists to date.


Subject(s)
Epidemics , Rift Valley Fever , Rift Valley fever virus , Animals , Humans , Mosquito Vectors , Rift Valley Fever/epidemiology , Rift Valley fever virus/physiology , Ruminants
9.
PLoS Negl Trop Dis ; 15(10): e0009837, 2021 10.
Article in English | MEDLINE | ID: mdl-34695125

ABSTRACT

Rift Valley fever virus (RVFV) causes morbidity and mortality in humans and domestic ungulates in sub-Saharan Africa, Egypt, and the Arabian Peninsula. Mosquito vectors transmit RVFV between vertebrates by bite, and also vertically to produce infectious progeny. Arrival of RVFV into the United States by infected mosquitoes or humans could result in significant impacts on food security, human health, and wildlife health. Elucidation of the vectors involved in the post-introduction RVFV ecology is paramount to rapid implementation of vector control. We performed vector competence experiments in which field-collected mosquitoes were orally exposed to an epidemic strain of RVFV via infectious blood meals. We targeted floodwater Aedes species known to feed on cattle, and/or deer species (Aedes melanimon Dyar, Aedes increpitus Dyar, Aedes vexans [Meigen]). Two permanent-water-breeding species were targeted as well: Culiseta inornata (Williston) of unknown competence considering United States populations, and Culex tarsalis Coquillett as a control species for which transmission efficiency is known. We tested the potential for midgut infection, midgut escape (dissemination), ovarian infection (vertical transmission), and transmission by bite (infectious saliva). Tissues were assayed by plaque assay and RT-qPCR, to quantify infectious virus and confirm virus identity. Tissue infection data were analyzed using a within-host model under a Bayesian framework to determine the probabilities of infection outcomes (midgut-limited infection, disseminated infection, etc.) while estimating barriers to infection between tissues. Permanent-water-breeding mosquitoes (Cx. tarsalis and Cs. inornata) exhibited more efficient horizontal transmission, as well as potential for vertical transmission, which is contrary to the current assumptions of RVFV ecology. Barrier estimates trended higher for Aedes spp., suggesting systemic factors in the differences between these species and Cx. tarsalis and Cs. inornata. These data indicate higher potential for vertical transmission than previously appreciated, and support the consensus of RVFV transmission including a broad range of potential vectors.


Subject(s)
Aedes/virology , Culex/virology , Mosquito Vectors/virology , Rift Valley Fever/transmission , Rift Valley fever virus/physiology , Aedes/genetics , Aedes/physiology , Animals , Cattle/virology , Colorado , Culex/physiology , Deer/virology , Mosquito Vectors/classification , Mosquito Vectors/physiology , Rift Valley Fever/virology , Rift Valley fever virus/genetics , Rift Valley fever virus/isolation & purification , Saliva/virology
10.
Cell ; 184(20): 5163-5178.e24, 2021 09 30.
Article in English | MEDLINE | ID: mdl-34559985

ABSTRACT

Rift Valley fever virus (RVFV) is a zoonotic pathogen with pandemic potential. RVFV entry is mediated by the viral glycoprotein (Gn), but host entry factors remain poorly defined. Our genome-wide CRISPR screen identified low-density lipoprotein receptor-related protein 1 (mouse Lrp1/human LRP1), heat shock protein (Grp94), and receptor-associated protein (RAP) as critical host factors for RVFV infection. RVFV Gn directly binds to specific Lrp1 clusters and is glycosylation independent. Exogenous addition of murine RAP domain 3 (mRAPD3) and anti-Lrp1 antibodies neutralizes RVFV infection in taxonomically diverse cell lines. Mice treated with mRAPD3 and infected with pathogenic RVFV are protected from disease and death. A mutant mRAPD3 that binds Lrp1 weakly failed to protect from RVFV infection. Together, these data support Lrp1 as a host entry factor for RVFV infection and define a new target to limit RVFV infections.


Subject(s)
Host-Pathogen Interactions , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Rift Valley fever virus/physiology , Virus Internalization , Animals , Antibody Specificity/immunology , Base Sequence , Brain/pathology , Brain/virology , CRISPR-Cas Systems/genetics , Cell Membrane/metabolism , Cells, Cultured , Glycoproteins/metabolism , Glycosaminoglycans/metabolism , Glycosylation , Humans , LDL-Receptor Related Protein-Associated Protein/metabolism , Ligands , Low Density Lipoprotein Receptor-Related Protein-1/deficiency , Membrane Glycoproteins/metabolism , Mice , Protein Binding , Protein Denaturation , Rift Valley Fever/pathology , Rift Valley Fever/prevention & control , Rift Valley Fever/virology , Rift Valley fever virus/immunology
11.
J Virol ; 95(23): e0150621, 2021 11 09.
Article in English | MEDLINE | ID: mdl-34495703

ABSTRACT

Rift Valley fever virus (RVFV) is an arbovirus found throughout Africa. It causes disease that is typically mild and self-limiting; however, some infected individuals experience severe manifestations, including hepatitis, encephalitis, or even death. Reports of RVFV encephalitis are notable among immunosuppressed individuals, suggesting a role for adaptive immunity in preventing this severe complication. This phenomenon has been modeled in C57BL/6 mice depleted of CD4 T cells prior to infection with DelNSs RVFV (RVFV containing a deletion of nonstructural protein NSs), resulting in late-onset encephalitis accompanied by high levels of viral RNA in the brain in 30% of animals. In this study, we sought to define the specific type(s) of CD4 T cells that mediate protection from RVFV encephalitis. The viral epitopes targeted by CD4 and CD8 T cells were defined in C57BL/6 mice, and tetramers for both CD4 and CD8 T cells were generated. RVFV-specific CD8 T cells were expanded and of a cytotoxic and proliferating phenotype in the liver following infection. RVFV-specific CD4 T cells were identified in the liver and spleen following infection and phenotyped as largely Th1 or Tfh subtypes. Knockout mice lacking various aspects of pathways important in Th1 and Tfh development and function were used to demonstrate that T-bet, CD40, CD40L, and major histocompatibility complex class II (MHC-II) mediated protection from RVFV encephalitis, while gamma interferon (IFN-γ) and interleukin-12 (IL-12) were dispensable. Virus-specific antibody responses correlated with protection from encephalitis in all mouse strains, suggesting that Tfh/B cell interactions modulate clinical outcome in this model. IMPORTANCE The prevention of RVFV encephalitis requires intact adaptive immunity. In this study, we developed reagents to detect RVFV-specific T cells and provide evidence for Tfh cells and CD40/CD40L interactions as critical mediators of this protection.


Subject(s)
CD40 Antigens , CD40 Ligand , Encephalitis, Viral/prevention & control , Rift Valley Fever/immunology , Rift Valley fever virus/immunology , Rift Valley fever virus/physiology , T-Lymphocytes/immunology , Africa , Animals , Antibody Formation , B-Lymphocytes/immunology , Brain/virology , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Encephalitis, Viral/immunology , Encephalitis, Viral/virology , Epitopes , Female , Liver/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
12.
PLoS Negl Trop Dis ; 15(9): e0009785, 2021 09.
Article in English | MEDLINE | ID: mdl-34516560

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.


Subject(s)
Macrophages/virology , Rift Valley Fever/virology , Rift Valley fever virus/physiology , Viral Envelope Proteins/metabolism , Virus Replication/physiology , Animals , Mice , Rift Valley fever virus/pathogenicity , Viral Envelope Proteins/genetics , Virulence
13.
Nat Commun ; 12(1): 5593, 2021 09 22.
Article in English | MEDLINE | ID: mdl-34552082

ABSTRACT

The persistence mechanisms of Rift Valley fever (RVF), a zoonotic arboviral haemorrhagic fever, at both local and broader geographical scales have yet to be fully understood and rigorously quantified. We developed a mathematical metapopulation model describing RVF virus transmission in livestock across the four islands of the Comoros archipelago, accounting for island-specific environments and inter-island animal movements. By fitting our model in a Bayesian framework to 2004-2015 surveillance data, we estimated the importance of environmental drivers and animal movements on disease persistence, and tested the impact of different control scenarios on reducing disease burden throughout the archipelago. Here we report that (i) the archipelago network was able to sustain viral transmission in the absence of explicit disease introduction events after early 2007, (ii) repeated outbreaks during 2004-2020 may have gone under-detected by local surveillance, and (iii) co-ordinated within-island control measures are more effective than between-island animal movement restrictions.


Subject(s)
Models, Theoretical , Rift Valley Fever/prevention & control , Rift Valley Fever/transmission , Rift Valley fever virus/physiology , Animals , Comoros/epidemiology , Livestock/virology , Rift Valley Fever/epidemiology , Seroepidemiologic Studies , Zoonoses/epidemiology , Zoonoses/prevention & control , Zoonoses/transmission
14.
PLoS Negl Trop Dis ; 15(8): e0009705, 2021 08.
Article in English | MEDLINE | ID: mdl-34424895

ABSTRACT

The Government of Rwanda reported an outbreak of Rift Valley fever (RVF) in the Eastern Province in 2018. To respond to the outbreak, vaccination and education campaigns about the disease were carried out. Because RVF cases continue to be detected in Rwanda and the disease impacts livelihoods and health, accurate knowledge and communication are imperative. The objectives of this study were to evaluate knowledge and risk perceptions of RVF transmission among livestock farmers in Nyagatare District, Eastern Province, Rwanda, and to compare RVF knowledge, risk perceptions, and farming practices between male and female livestock farmers. This cross-sectional, quantitative study was conducted in selected sectors of Nyagatare District in the Eastern Province of Rwanda in June 2019. A 34-question survey was used to ask about demographics, livestock ownership, risk perceptions about zoonotic diseases and livestock management, RVF knowledge, preferred communication sources and information sharing strategies, and protective strategies for RVF mitigation while working with livestock. Livestock farmers were interviewed at three milk collection centers, two village meeting points, a farm cooperative meeting, and during door-to-door visits in villages. In total, 123 livestock farmers were interviewed. The survey found that most livestock farmers lacked knowledge about epizootic and zoonotic transmission of RVF, more male livestock farmers were familiar with RVF and risk mitigation strategies, and female livestock farmers are not viewed as reliable sources of information. Additionally, most livestock farmers had not vaccinated their animals against RVF despite past vaccination campaigns. Radio was the most popular communication channel. These findings show that RVF knowledge and information sharing are inadequate among livestock farmers in Eastern Province. Therefore, vaccination and education campaigns may need to be reevaluated within the context of these trends in order to prepare for future RVF outbreaks.


Subject(s)
Farmers/psychology , Health Knowledge, Attitudes, Practice , Rift Valley Fever/psychology , Viral Zoonoses/psychology , Adult , Aged , Aged, 80 and over , Animals , Communication , Cross-Sectional Studies , Female , Humans , Information Dissemination , Livestock/growth & development , Male , Middle Aged , Perception , Rift Valley Fever/virology , Rift Valley fever virus/physiology , Rwanda , Viral Zoonoses/virology , Young Adult
15.
Sci Rep ; 11(1): 12395, 2021 06 11.
Article in English | MEDLINE | ID: mdl-34117312

ABSTRACT

Viral interference is a common occurrence that has been reported in cell culture in many cases. In the present study, viral interference between two capripox viruses (sheeppox SPPV and lumpy skin disease virus LSDV in cattle) with Rift Valley fever virus (RVFV) was investigated in vitro and in their natural hosts, sheep and cattle. A combination of SPPV/RVFV and LSDV/RVFV was used to co-infect susceptible cells and animals to detect potential competition. In-vitro interference was evaluated by estimating viral infectivity and copies of viral RNA by a qPCR during three serial passages in cell cultures, whereas in-vivo interference was assessed through antibody responses to vaccination. When lamb testis primary cells were infected with the mixture of capripox and RVFV, the replication of both SPPV and LSDV was inhibited by RVFV. In animals, SPPV/RVFV or LSDV/RVFV combinations inhibited the replication SPPV and LSDV and the antibody response following vaccination. The combined SPPV/RVFV did not protect sheep after challenging with the virulent strain of SPPV and the LSDV/RVFV did not induce interferon Gamma to LSDV, while immunological response to RVFV remain unaffected. Our goal was to assess this interference response to RVFV/capripoxviruses' coinfection in order to develop effective combined live-attenuated vaccines as a control strategy for RVF and SPP/LSD diseases. Our findings indicated that this approach was not suitable for developing a combined SPPV/LSDV/RVFV vaccine candidate because of interference of replication and the immune response among these viruses.


Subject(s)
Capripoxvirus/genetics , Rift Valley fever virus/genetics , Animals , Antibodies, Viral/biosynthesis , Capripoxvirus/immunology , Capripoxvirus/physiology , Cattle , Cells, Cultured , Chlorocebus aethiops , Genes, Viral , In Vitro Techniques , RNA, Viral/genetics , Real-Time Polymerase Chain Reaction , Rift Valley fever virus/immunology , Rift Valley fever virus/physiology , Sheep , Vero Cells , Viral Vaccines/immunology , Virus Replication
16.
Mamm Genome ; 32(1): 30-37, 2021 02.
Article in English | MEDLINE | ID: mdl-33420513

ABSTRACT

Rift Valley fever (RVF) is an emerging viral zoonosis that primarily affects ruminants and humans. We have previously shown that wild-derived MBT/Pas mice are highly susceptible to RVF virus and that part of this phenotype is controlled by a locus located on distal Chromosome 11. Using congenic strains, we narrowed down the critical interval to a 530 kb region containing five protein-coding genes among which Rnf213 emerged as a potential candidate. We generated Rnf213-deficient mice by CRISPR/CAS9 on the C57BL/6 J background and showed that they were significantly more susceptible to RVF than control mice, with an average survival time post-infection reduced from 7 to 4 days. The human RNF213 gene had been associated with the cerebrovascular Moyamoya disease (MMD or MYMY) but the inactivation of this gene in the mouse resulted only in mild anomalies of the neovascularization. This study provides the first evidence that the Rnf213 gene may also impact the resistance to infectious diseases such as RVF.


Subject(s)
Adenosine Triphosphatases/genetics , Disease Resistance/genetics , Host-Pathogen Interactions/genetics , Rift Valley Fever/genetics , Rift Valley Fever/virology , Rift Valley fever virus/physiology , Ubiquitin-Protein Ligases/genetics , Animals , CRISPR-Cas Systems , Chromosome Mapping , Disease Models, Animal , Mice , Mice, Inbred C57BL , Mice, Knockout
17.
Trop Anim Health Prod ; 53(1): 92, 2021 Jan 07.
Article in English | MEDLINE | ID: mdl-33415465

ABSTRACT

West Nile fever (WNF) and Rift Valley fever (RVF) are emerging and re-emerging zoonotic diseases of veterinary and public health importance in Africa. Despite the existence of potential vectors and a wide range of hosts, the transmission of these diseases in domestic animals has not been well documented in the South Omo area of Ethiopia. This study aimed to estimate the sero-prevalence of IgG antibodies produced against West Nile virus (WNV) and Rift Valley fever virus (RVFV) infections among cattle in the South Omo area. Between May and June 2019, blood samples were collected from 397 cattle and screened for IgG antibodies against WNV and RVFV infections using enzyme-linked immunosorbent assay (ELISA). The overall sero-prevalence of IgG antibody to WNV infection was 4.8% (95% CI: 2.67-6.88%), while it was 5.0% to RVFV infection (95% CI: 2.87-7.18). Compared to 1-3 years old cattle, those in the age group ≥ 7 years had significantly higher odds of being positive for WNV (AOR = 6.82; 95% CI: 1.72-26.99) and RVFV (AOR = 4.38; 95% CI: 1.08-17.88) infections. The occurrence of WNV and RVFV infections in cattle population in the present study area indicates the risk of transmission to humans. Strengthening the surveillance system and conducting further studies to identify active cases in domestic and wild animals as well as in humans is crucial to reduce the risk of possible outbreaks.


Subject(s)
Cattle Diseases/epidemiology , Rift Valley Fever/epidemiology , Rift Valley fever virus/physiology , West Nile Fever/veterinary , West Nile virus/physiology , Animal Husbandry , Animals , Cattle , Cattle Diseases/virology , Ethiopia/epidemiology , Prevalence , Rift Valley Fever/virology , Seroepidemiologic Studies , West Nile Fever/epidemiology , West Nile Fever/virology
18.
J Gen Virol ; 102(2)2021 02.
Article in English | MEDLINE | ID: mdl-33231535

ABSTRACT

The zoonotic emerging Rift Valley fever virus (RVFV) causes sporadic disease in livestock and humans throughout Africa and the Saudi Arabian peninsula. Infection of people with RVFV can occur through mosquito bite or mucosal exposure during butchering or milking of infected livestock. Disease typically presents as a self-limiting fever; however, in rare cases, hepatitis, encephalitis and ocular disease may occur. Recent studies have illuminated the neuropathogenic mechanisms of RVFV in a rat aerosol infection model. Neurological disease in rats is characterized by breakdown of the blood-brain barrier late in infection, infiltration of leukocytes to the central nervous system (CNS) and massive viral replication in the brain. However, the route of RVFV entry into the CNS after inhalational exposure remains unknown. Here, we visualized the entire nasal olfactory route from snout to brain after RVFV infection using RNA in situ hybridization and immunofluorescence microscopy. We found widespread RVFV-infected cells within the olfactory epithelium, across the cribriform plate, and in the glomerular region of the olfactory bulb within 2 days of infection. These results indicate that the olfactory tract is a major route of infection of the brain after inhalational exposure. A better understanding of potential neuroinvasion pathways can support the design of more effective therapeutic regiments for the treatment of neurological disease caused by RVFV.


Subject(s)
Encephalitis, Viral/virology , Ethmoid Bone/virology , Olfactory Mucosa/virology , Rift Valley Fever/pathology , Rift Valley fever virus/physiology , Animals , Disease Models, Animal , Encephalitis, Viral/pathology , Ethmoid Bone/pathology , Female , Inhalation Exposure , Olfactory Mucosa/pathology , Rats , Rats, Inbred Lew , Rift Valley Fever/virology
19.
PLoS Negl Trop Dis ; 14(12): e0008870, 2020 12.
Article in English | MEDLINE | ID: mdl-33301456

ABSTRACT

Rift Valley fever phlebovirus (RVFV) causes an emerging zoonotic disease and is mainly transmitted by Culex and Aedes mosquitoes. While Aedes aegypti-dengue virus (DENV) is the most studied model, less is known about the genes involved in infection-responses in other mosquito-arboviruses pairing. The main objective was to investigate the molecular responses of Cx. pipiens to RVFV exposure focusing mainly on genes implicated in innate immune responses. Mosquitoes were fed with blood spiked with RVFV. The fully-engorged females were pooled at 3 different time points: 2 hours post-exposure (hpe), 3- and 14-days post-exposure (dpe). Pools of mosquitoes fed with non-infected blood were also collected for comparisons. Total RNA from each mosquito pool was subjected to RNA-seq analysis and a de novo transcriptome was constructed. A total of 451 differentially expressed genes (DEG) were identified. Most of the transcriptomic alterations were found at an early infection stage after RVFV exposure. Forty-eight DEG related to immune infection-response were characterized. Most of them were related with the RNAi system, Toll and IMD pathways, ubiquitination pathway and apoptosis. Our findings provide for the first time a comprehensive view on Cx. pipiens-RVFV interactions at the molecular level. The early depletion of RNAi pathway genes at the onset of the RVFV infection would allow viral replication in mosquitoes. While genes from the Toll and IMD immune pathways were altered in response to RVFV none of the DEG were related to the JAK/STAT pathway. The fact that most of the DEG involved in the Ubiquitin-proteasome pathway (UPP) or apoptosis were found at an early stage of infection would suggest that apoptosis plays a regulatory role in infected Cx. pipiens midguts. This study provides a number of target genes that could be used to identify new molecular targets for vector control.


Subject(s)
Culex/virology , Host-Pathogen Interactions , Rift Valley fever virus/physiology , Animals , Biological Evolution , Culex/immunology , Gene Expression Regulation/immunology , RNA, Viral , Transcriptome
20.
Proc Natl Acad Sci U S A ; 117(39): 24567-24574, 2020 09 29.
Article in English | MEDLINE | ID: mdl-32929025

ABSTRACT

Rift Valley fever (RVF) is an emerging, zoonotic, arboviral hemorrhagic fever threatening livestock and humans mainly in Africa. RVF is of global concern, having expanded its geographical range over the last decades. The impact of control measures on epidemic dynamics using empirical data has not been assessed. Here, we fitted a mathematical model to seroprevalence livestock and human RVF case data from the 2018-2019 epidemic in Mayotte to estimate viral transmission among livestock, and spillover from livestock to humans through both direct contact and vector-mediated routes. Model simulations were used to assess the impact of vaccination on reducing the epidemic size. The rate of spillover by direct contact was about twice as high as vector transmission. Assuming 30% of the population were farmers, each transmission route contributed to 45% and 55% of the number of human infections, respectively. Reactive vaccination immunizing 20% of the livestock population reduced the number of human cases by 30%. Vaccinating 1 mo later required using 50% more vaccine doses for a similar reduction. Vaccinating only farmers required 10 times as more vaccine doses for a similar reduction in human cases. Finally, with 52.0% (95% credible interval [CrI] [42.9-59.4]) of livestock immune at the end of the epidemic wave, viral reemergence in the next rainy season (2019-2020) is unlikely. Coordinated human and animal health surveillance, and timely livestock vaccination appear to be key to controlling RVF in this setting. We furthermore demonstrate the value of a One Health quantitative approach to surveillance and control of zoonotic infectious diseases.


Subject(s)
Rift Valley Fever/epidemiology , Zoonoses/epidemiology , Animals , Comoros/epidemiology , Epidemics , Humans , Livestock/virology , Rift Valley Fever/prevention & control , Rift Valley Fever/transmission , Rift Valley Fever/virology , Rift Valley fever virus/genetics , Rift Valley fever virus/isolation & purification , Rift Valley fever virus/physiology , Seasons , Seroepidemiologic Studies , Vaccination , Viral Vaccines/administration & dosage , Zoonoses/transmission , Zoonoses/virology
SELECTION OF CITATIONS
SEARCH DETAIL
...