Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 294
Filtrar
1.
Viruses ; 16(6)2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38932175
2.
Nat Commun ; 15(1): 4906, 2024 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-38851803

RESUMEN

Various low-density lipoprotein receptors (LPRs) have been identified as entry factors for alphaviruses, and structures of the corresponding virion-receptor complexes have been determined. Here, we analyze the similarities and differences in the receptor binding modes of multiple alphaviruses to understand their ability to infect a wide range of hosts. We further discuss the challenges associated with the development of broad-spectrum treatment strategies against a diverse range of alphaviruses.


Asunto(s)
Alphavirus , Antivirales , Receptores de LDL , Internalización del Virus , Animales , Humanos , Alphavirus/efectos de los fármacos , Alphavirus/fisiología , Alphavirus/genética , Infecciones por Alphavirus/tratamiento farmacológico , Infecciones por Alphavirus/virología , Antivirales/uso terapéutico , Antivirales/farmacología , Unión Proteica , Receptores de LDL/metabolismo , Receptores de LDL/genética , Receptores Virales/metabolismo , Receptores Virales/química , Virión/metabolismo , Internalización del Virus/efectos de los fármacos
3.
Front Immunol ; 15: 1401086, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38903507

RESUMEN

The mitochondrial anti-viral signaling (MAVS) protein is an intermediary adaptor protein of retinoic acid-inducible gene-1 (RIG-I) like receptor (RLR) signaling, which activates the transcription factor interferon (IFN) regulatory factor 3 (IRF3) and NF-kB to produce type I IFNs. MAVS expression has been reported in different fish species, but few studies have shown its functional role in anti-viral responses to fish viruses. In this study, we used the transcription activator-like effector nuclease (TALEN) as a gene editing tool to disrupt the function of MAVS in Chinook salmon (Oncorhynchus tshawytscha) embryonic cells (CHSE) to understand its role in induction of interferon I responses to infections with the (+) RNA virus salmonid alphavirus subtype 3 (SAV-3), and the dsRNA virus infectious pancreatic necrosis virus (IPNV) infection. A MAVS-disrupted CHSE clone with a 7-aa polypeptide (GVFVSRV) deletion mutation at the N-terminal of the CARD domain infected with SAV-3 resulted in significantly lower expression of IRF3, IFNa, and ISGs and increased viral titer (1.5 log10) compared to wild-type. In contrast, the IPNV titer in MAVS-disrupted cells was not different from the wild-type. Furthermore, overexpression of salmon MAVS in MAVS-disrupted CHSE cells rescued the impaired type I IFN-mediated anti-viral effect against SAV-3.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales , Infecciones por Alphavirus , Alphavirus , Enfermedades de los Peces , Virus de la Necrosis Pancreática Infecciosa , Transducción de Señal , Replicación Viral , Animales , Virus de la Necrosis Pancreática Infecciosa/fisiología , Virus de la Necrosis Pancreática Infecciosa/inmunología , Alphavirus/inmunología , Alphavirus/fisiología , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Enfermedades de los Peces/inmunología , Enfermedades de los Peces/virología , Infecciones por Alphavirus/inmunología , Infecciones por Alphavirus/virología , Salmón/virología , Salmón/inmunología , Proteínas de Peces/genética , Proteínas de Peces/inmunología , Proteínas de Peces/metabolismo , Factor 3 Regulador del Interferón/metabolismo , Factor 3 Regulador del Interferón/genética , Infecciones por Birnaviridae/inmunología , Infecciones por Birnaviridae/veterinaria , Infecciones por Birnaviridae/virología
4.
Parasit Vectors ; 17(1): 200, 2024 May 04.
Artículo en Inglés | MEDLINE | ID: mdl-38704595

RESUMEN

BACKGROUND: Mayaro virus (MAYV) is an emerging alphavirus, primarily transmitted by the mosquito Haemagogus janthinomys in Central and South America. However, recent studies have shown that Aedes aegypti, Aedes albopictus and various Anopheles mosquitoes can also transmit the virus under laboratory conditions. MAYV causes sporadic outbreaks across the South American region, particularly in areas near forests. Recently, cases have been reported in European and North American travelers returning from endemic areas, raising concerns about potential introductions into new regions. This study aims to assess the vector competence of three potential vectors for MAYV present in Europe. METHODS: Aedes albopictus from Italy, Anopheles atroparvus from Spain and Culex pipiens biotype molestus from Belgium were exposed to MAYV and maintained under controlled environmental conditions. Saliva was collected through a salivation assay at 7 and 14 days post-infection (dpi), followed by vector dissection. Viral titers were determined using focus forming assays, and infection rates, dissemination rates, and transmission efficiency were calculated. RESULTS: Results indicate that Ae. albopictus and An. atroparvus from Italy and Spain, respectively, are competent vectors for MAYV, with transmission possible starting from 7 dpi under laboratory conditions. In contrast, Cx. pipiens bioform molestus was unable to support MAYV infection, indicating its inability to contribute to the transmission cycle. CONCLUSIONS: In the event of accidental MAYV introduction in European territories, autochthonous outbreaks could potentially be sustained by two European species: Ae. albopictus and An. atroparvus. Entomological surveillance should also consider certain Anopheles species when monitoring MAYV transmission.


Asunto(s)
Aedes , Infecciones por Alphavirus , Alphavirus , Culex , Mosquitos Vectores , Animales , Aedes/virología , Mosquitos Vectores/virología , Alphavirus/fisiología , Alphavirus/aislamiento & purificación , Culex/virología , Europa (Continente) , Infecciones por Alphavirus/transmisión , Infecciones por Alphavirus/virología , Saliva/virología , Anopheles/virología , España , Italia , Femenino , Bélgica
5.
mBio ; 15(6): e0042024, 2024 Jun 12.
Artículo en Inglés | MEDLINE | ID: mdl-38700353

RESUMEN

Chikungunya virus (CHIKV) is an enveloped, positive-sense RNA virus that has re-emerged to cause millions of human infections worldwide. In humans, acute CHIKV infection causes fever and severe muscle and joint pain. Chronic and debilitating arthritis and joint pain can persist for months to years. To date, there are no approved antivirals against CHIKV. Recently, the ribonucleoside analog 4'-fluorouridine (4'-FlU) was reported as a highly potent orally available inhibitor of SARS-CoV-2, respiratory syncytial virus, and influenza virus replication. In this study, we assessed 4'-FlU's potency and breadth of inhibition against a panel of alphaviruses including CHIKV, and found that it broadly suppressed alphavirus production in cell culture. 4'-FlU acted on the viral RNA replication step, and the first 4 hours post-infection were the critical time for its antiviral effect. In vitro replication assays identified nsP4 as the target of inhibition. In vivo, treatment with 4'-FlU reduced disease signs, inflammatory responses, and viral tissue burden in mouse models of CHIKV and Mayaro virus infection. Treatment initiated at 2 hours post-infection was most effective; however, treatment initiated as late as 24-48 hours post-infection produced measurable antiviral effects in the CHIKV mouse model. 4'-FlU showed effective oral delivery in our mouse model and resulted in the accumulation of both 4'-FlU and its bioactive triphosphate form in tissues relevant to arthritogenic alphavirus pathogenesis. Together, our data indicate that 4'-FlU inhibits CHIKV infection in vitro and in vivo and is a promising oral therapeutic candidate against CHIKV infection.IMPORTANCEAlphaviruses including chikungunya virus (CHIKV) are mosquito-borne positive-strand RNA viruses that can cause various diseases in humans. Although compounds that inhibit CHIKV and other alphaviruses have been identified in vitro, there are no licensed antivirals against CHIKV. Here, we investigated a ribonucleoside analog, 4'-fluorouridine (4'-FlU), and demonstrated that it inhibited infectious virus production by several alphaviruses in vitro and reduced virus burden in mouse models of CHIKV and Mayaro virus infection. Our studies also indicated that 4'-FlU treatment reduced CHIKV-induced footpad swelling and reduced the production of pro-inflammatory cytokines. Inhibition in the mouse model correlated with effective oral delivery of 4'-FlU and accumulation of both 4'-FlU and its bioactive form in relevant tissues. In summary, 4'-FlU exhibits potential as a novel anti-alphavirus agent targeting the replication of viral RNA.


Asunto(s)
Alphavirus , Antivirales , Virus Chikungunya , Replicación Viral , Replicación Viral/efectos de los fármacos , Animales , Antivirales/farmacología , Antivirales/uso terapéutico , Ratones , Virus Chikungunya/efectos de los fármacos , Virus Chikungunya/fisiología , Alphavirus/efectos de los fármacos , Alphavirus/fisiología , Uridina/análogos & derivados , Uridina/farmacología , Humanos , Fiebre Chikungunya/tratamiento farmacológico , Fiebre Chikungunya/virología , Modelos Animales de Enfermedad , Línea Celular , Chlorocebus aethiops , Femenino , Células Vero
6.
Med Vet Entomol ; 38(2): 234-243, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38489505

RESUMEN

Mayaro virus (MAYV; Alphavirus: Togaviridae) is an emerging pathogen in Latin America, causing fever and polyarthritis. Sporadic outbreaks of MAYV have occurred in the region, with reported human cases being imported to Europe and North America. Although primarily a risk for those residing in the Amazon basin's tropical forests, recent reports highlight that urbanization would increase the risk of MAYV transmission in Latin America. Urban emergence depends on human susceptibility and the ability of mosquitos like Aedes aegypti  (Linnaeus, 1762) (Diptera: Culicidae) to transmit MAYV. Despite the absence of active MAYV transmission in Argentine, the risk of introduction is substantial due to human movement and the presence of Ae. aegypti in the region. This study aimed to evaluate the susceptibility of different Argentine Ae. aegypti populations to MAYV genotype L (MAYV-L) using dose-response assays and determine barriers to virus infection, dissemination and transmission. Immature mosquito stages were collected in Buenos Aires, Córdoba and Rosario cities. Female Ae. aegypti (F2) were orally infected by feeding on five concentrations of MAYV-L, ranging from 1.0 to 6.0 log10 PFU/mL. Abdomens, legs and saliva were analysed using viral plaque assays. Results revealed that MAYV-L between infection and dissemination were associated with viral doses rather than the population origin. Infection rates varied between 3% and 65%, with a 50% infectious dose >5.5 log10 PFU/mL. Dissemination occurred at 39%, with a 50% dissemination dose of ~6.0 log10 PFU/mL. Dissemination among infected mosquitoes ranged from 60% to 86%, and transmission from disseminated mosquitoes ranged from 11% to 20%. Argentine Ae. aegypti populations exhibited a need for higher viral doses of MAYV-L than those typically found in humans to become infected. In addition, only a small proportion of infected mosquitoes were capable of transmitting the virus. Understanding MAYV transmission in urban areas is crucial for public health interventions.


Asunto(s)
Aedes , Alphavirus , Mosquitos Vectores , Animales , Aedes/virología , Aedes/fisiología , Argentina , Mosquitos Vectores/virología , Mosquitos Vectores/fisiología , Alphavirus/fisiología , Femenino , Infecciones por Alphavirus/transmisión , Larva/virología , Larva/crecimiento & desarrollo
7.
J Fish Dis ; 47(7): e13950, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38555528

RESUMEN

Disease interactions between farmed and wild populations have been poorly documented for most aquaculture species, in part due to the complexities to study this. Here, we tested 567 farmed Atlantic salmon escapees, captured in a Norwegian river during 2014-2018, for five viral infections that are prevalent in global salmonid aquaculture. Over 90% of the escapees were infected with one or more viruses. Overall prevalences were: 75.7% for piscine orthoreovirus (PRV-1), 43.6% for salmonid alphavirus (SAV), 31.2% for piscine myocarditis virus (PMCV), 1.2% for infectious pancreatic necrosis virus (IPNV) and 0.4% for salmon anaemia virus (ISAV). A significantly higher prevalence of PMCV infection was observed in immature compared to mature individuals. The prevalence of both SAV and PMCV infections was higher in fish determined by fatty acid profiling to be 'recent' as opposed to 'early' escapees that had been in the wild for a longer period of time. This is the first study to establish a time-series of viral infection status of escapees entering a river with a native salmon population. Our results demonstrate that farmed escapees represent a continuous source of infectious agents which could potentially be transmitted to wild fish populations.


Asunto(s)
Acuicultura , Enfermedades de los Peces , Ríos , Salmo salar , Animales , Enfermedades de los Peces/virología , Enfermedades de los Peces/epidemiología , Noruega/epidemiología , Prevalencia , Alphavirus/aislamiento & purificación , Alphavirus/fisiología , Infecciones por Alphavirus/veterinaria , Infecciones por Alphavirus/epidemiología , Infecciones por Alphavirus/virología
8.
Viruses ; 16(2)2024 01 30.
Artículo en Inglés | MEDLINE | ID: mdl-38399981

RESUMEN

Alphaviruses can replicate in arthropods and in many vertebrate species including humankind, but only in vertebrate cells do infections with these viruses result in a strong inhibition of host translation and transcription. Translation shutoff by alphaviruses is a multifactorial process that involves both host- and virus-induced mechanisms, and some of them are not completely understood. Alphavirus genomes contain cis-acting elements (RNA structures and dinucleotide composition) and encode protein activities that promote the translational and transcriptional resistance to type I IFN-induced antiviral effectors. Among them, IFIT1, ZAP and PKR have played a relevant role in alphavirus evolution, since they have promoted the emergence of multiple viral evasion mechanisms at the translational level. In this review, we will discuss how the adaptations of alphaviruses to vertebrate hosts likely involved the acquisition of new features in viral mRNAs and proteins to overcome the effect of type I IFN.


Asunto(s)
Alphavirus , Interferón Tipo I , Animales , Alphavirus/fisiología , Línea Celular , Interferón Tipo I/genética , Vertebrados , Tropismo , Antivirales/farmacología , Replicación Viral
9.
Cell ; 187(2): 360-374.e19, 2024 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-38176410

RESUMEN

The very-low-density lipoprotein receptor (VLDLR) comprises eight LDLR type A (LA) domains and supports entry of distantly related alphaviruses, including Eastern equine encephalitis virus (EEEV) and Semliki Forest virus (SFV). Here, by resolving multiple cryo-electron microscopy structures of EEEV-VLDLR complexes and performing mutagenesis and functional studies, we show that EEEV uses multiple sites (E1/E2 cleft and E2 A domain) to engage more than one LA domain simultaneously. However, no single LA domain is necessary or sufficient to support efficient EEEV infection. Whereas all EEEV strains show conservation of two VLDLR-binding sites, the EEEV PE-6 strain and a few other EEE complex members feature a single amino acid substitution that enables binding of LA domains to an additional site on the E2 B domain. These structural and functional analyses informed the design of a minimal VLDLR decoy receptor that neutralizes EEEV infection and protects mice from lethal challenge.


Asunto(s)
Microscopía por Crioelectrón , Virus de la Encefalitis Equina del Este , Encefalomielitis Equina , Receptores de LDL , Animales , Ratones , Alphavirus/fisiología , Virus de la Encefalitis Equina del Este/fisiología , Virus de la Encefalitis Equina del Este/ultraestructura , Encefalomielitis Equina/metabolismo , Caballos , Unión Proteica , Receptores de LDL/ultraestructura
10.
J Virol ; 97(12): e0069523, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-38051046

RESUMEN

IMPORTANCE: Relative humidity (RH) is an environmental variable that affects mosquito physiology and can impact pathogen transmission. Low RH can induce dehydration in mosquitoes, leading to alterations in physiological and behavioral responses such as blood-feeding and host-seeking behavior. We evaluated the effects of a temporal drop in RH (RH shock) on mortality and Mayaro virus vector competence in Ae. aegypti. While dehydration induced by humidity shock did not impact virus infection, we detected a significant effect of dehydration on mosquito mortality and blood-feeding frequency, which could significantly impact transmission dynamics.


Asunto(s)
Aedes , Alphavirus , Mosquitos Vectores , Animales , Aedes/fisiología , Aedes/virología , Alphavirus/fisiología , Deshidratación
11.
J Virol ; 97(11): e0097923, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37902397

RESUMEN

IMPORTANCE: Our study highlights the mechanisms behind the cell's resistance to stress granule (SG) formation after infection with Old World alphaviruses. Shortly after infection, the replication of these viruses hinders the cell's ability to form SGs, even when exposed to chemical inducers such as sodium arsenite. This resistance is primarily attributed to virus-induced transcriptional and translational shutoffs, rather than interactions between the viral nsP3 and the key components of SGs, G3BP1/2, or the ADP-ribosylhydrolase activity of nsP3 macro domain. While interactions between G3BPs and nsP3 are essential for the formation of viral replication complexes, their role in regulating SG development appears to be small, if any. Cells harboring replicating viruses or replicons with lower abilities to inhibit transcription and/or translation, but expressing wild-type nsP3, retain the ability for SG development. Understanding these mechanisms of regulation of SG formation contributes to our knowledge of viral replication and the intricate relationships between alphaviruses and host cells.


Asunto(s)
Alphavirus , ADN Helicasas , Interacciones Microbiota-Huesped , Biosíntesis de Proteínas , Gránulos de Estrés , Transcripción Genética , Alphavirus/fisiología , ADN Helicasas/metabolismo , Proteínas de Unión a Poli-ADP-Ribosa/metabolismo , Replicón , ARN Helicasas/metabolismo , Proteínas con Motivos de Reconocimiento de ARN/metabolismo , Gránulos de Estrés/metabolismo , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/metabolismo , Replicación Viral
12.
J Virol ; 97(5): e0196022, 2023 05 31.
Artículo en Inglés | MEDLINE | ID: mdl-37098948

RESUMEN

Eilat virus (EILV) is an insect-specific alphavirus that has the potential to be developed into a tool to combat mosquito-borne pathogens. However, its mosquito host range and transmission routes are not well understood. Here, we fill this gap by investigating EILV's host competence and tissue tropism in five mosquito species: Aedes aegypti, Culex tarsalis, Anopheles gambiae, Anopheles stephensi, and Anopheles albimanus. Of the tested species, C. tarsalis was the most competent host for EILV. The virus was found in C. tarsalis ovaries, but no vertical or venereal transmission was observed. Culex tarsalis also transmitted EILV via saliva, suggesting the potential for horizontal transmission between an unknown vertebrate or invertebrate host. We found that reptile (turtle and snake) cell lines were not competent for EILV infection. We tested a potential invertebrate host (Manduca sexta caterpillars) but found they were not susceptible to EILV infection. Together, our results suggest that EILV could be developed as a tool to target pathogenic viruses that use Culex tarsalis as a vector. Our work sheds light on the infection and transmission dynamics of a poorly understood insect-specific virus and reveals it may infect a broader range of mosquito species than previously recognized. IMPORTANCE The recent discovery of insect-specific alphaviruses presents opportunities both to study the biology of virus host range and to develop them into tools against pathogenic arboviruses. Here, we characterize the host range and transmission of Eilat virus in five mosquito species. We find that Culex tarsalis-a vector of harmful human pathogens, including West Nile virus-is a competent host of Eilat virus. However, how this virus is transmitted between mosquitoes remains unclear. We find that Eilat virus infects the tissues necessary for both vertical and horizontal transmission-a crucial step in discerning how Eilat virus maintains itself in nature.


Asunto(s)
Alphavirus , Culex , Mosquitos Vectores , Animales , Humanos , Alphavirus/fisiología , Culex/virología
13.
Dev Comp Immunol ; 140: 104612, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36473548

RESUMEN

Salmon alphavirus (SAV) infection leads to severe pancreas disease (PD) with typical inflammatory responses in Atlantic salmon (Salmo salar) and rainbow trout (Oncorhynchus mykiss). Nsp2, an important nonstructural protein of SAV, can activate NF-κB signaling pathway to reduce inflammatory responses. However, the molecular mechanism remains unclear. In this study, the ML (279-421aa) of Nsp2 was revealed to be the key domain for activating NF-κB. We focused on a host protein, DEAD-box RNA helicase 3 (DDX3), that may interact with Nsp2 to regulate NF-κB-induced inflammatory. The interaction between DDX3 and Nsp2 was confirmed in vitro. Overexpression of DDX3 inhibited the activation of NF-κB by Nsp2. SAV Nsp2 relieves the inhibitory effect of DDX3 on NF-κB, thereby initiating the innate immune response. This study revealed the molecular mechanism of Nsp2-induced inflammatory response by targeting DDX3 to activate NF-κB, providing a theoretical basis for revealing the underlying infection mechanism and pathogenesis of SAV.


Asunto(s)
Infecciones por Alphavirus , Alphavirus , Enfermedades de los Peces , Oncorhynchus mykiss , Salmo salar , Animales , FN-kappa B , Alphavirus/fisiología , Transducción de Señal
14.
Fish Shellfish Immunol ; 129: 182-190, 2022 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-36058437

RESUMEN

Salmonid alphavirus (SAV) infection of Atlantic salmon (Salmo salar) and rainbow trout (Oncorhynchus mykiss) causes pancreas disease (PD) with typical inflammatory responses, such as necrosis of the exocrine pancreas, cardiomyopathy and skeletal myopathy. However, the pathogenic mechanism underlying SAV infection is still unclear. Inflammation may cause damage to the body, but it is a defense response against infection by pathogenic microorganisms, of which nuclear factor-kappa B (NF-κB) is the main regulator. This study revealed that SAV can activate NF-κB, of which the viral nonstructural protein Nsp2 is the major activating protein. SAV activates the NF-κB signaling pathway by simultaneously up-regulating TLR3, 7, 8 and then the expression of the signaling molecule myeloid differentiation factor 88 (Myd88) and tumor necrosis factor receptor-associated factor 6 (TRAF6). We found that Nsp2 can induce IκB degradation and p65 phosphorylation and transnucleation, and activate NF-κB downstream inflammatory cytokines. Nsp2 may simultaneously activate NF-κB through TLR3,7,8-dependent signaling pathways. Overexpression of Nsp2 can up-regulate mitochondrial antiviral signaling protein (MAVS) and then promote the expression of IFNa1 and antiviral protein Mx, which inhibits viral replication. This study shows that Nsp2 acts as a key activator protein for the NF-κB signaling pathway, which induces inflammation post-SAV infection. This study systematically analyzes the molecular mechanism of SAV activation of the NF-κB signaling pathway, and provides a theoretical basis for revealing the mechanism of innate immune response and inflammatory injury caused by SAV.


Asunto(s)
Infecciones por Alphavirus , Alphavirus , Enfermedades de los Peces , Oncorhynchus mykiss , Salmo salar , Alphavirus/fisiología , Infecciones por Alphavirus/veterinaria , Animales , Antivirales , Citocinas/metabolismo , Inflamación/veterinaria , Factor 88 de Diferenciación Mieloide/metabolismo , Proteínas de Resistencia a Mixovirus/metabolismo , FN-kappa B/metabolismo , Oncorhynchus mykiss/metabolismo , Salmo salar/genética , Salmo salar/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptor Toll-Like 3/metabolismo , Proteínas no Estructurales Virales
15.
Viruses ; 14(8)2022 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-36016409

RESUMEN

Mayaro virus is an emerging arbovirus that causes nonspecific febrile illness or arthralgia syndromes similar to the Chikungunya virus, a virus closely related from the Togaviridae family. MAYV outbreaks occur more frequently in the northern and central-western states of Brazil; however, in recent years, virus circulation has been spreading to other regions. Due to the undifferentiated initial clinical symptoms between MAYV and other endemic pathogenic arboviruses with geographic overlapping, identification of patients infected by MAYV might be underreported. Additionally, the lack of specific prophylactic approaches or antiviral drugs limits the pharmacological management of patients to treat symptoms like pain and inflammation, as is the case with most pathogenic alphaviruses. In this context, this review aims to present the state-of-the-art regarding the screening and development of compounds/molecules which may present anti-MAYV activity and infection inhibition.


Asunto(s)
Infecciones por Alphavirus , Alphavirus , Arbovirus , Virus Chikungunya , Alphavirus/fisiología , Infecciones por Alphavirus/tratamiento farmacológico , Infecciones por Alphavirus/epidemiología , Antivirales/farmacología , Antivirales/uso terapéutico , Virus Chikungunya/fisiología , Desarrollo de Medicamentos , Humanos
16.
Viruses ; 14(5)2022 04 23.
Artículo en Inglés | MEDLINE | ID: mdl-35632622

RESUMEN

Mayaro virus (MAYV) is an emerging mosquito-borne arbovirus and public health concern. We evaluated the influence of temperature on Aedes aegypti responses to MAYV oral infection and transmission at two constant temperatures (20 °C and 30 °C). Infection of mosquito tissues (bodies and legs) and salivary secretions with MAYV was determined at 3, 9, 15, 21, and 27 days post ingestion. At both temperatures, we observed a trend of increase in progression of MAYV infection and replication kinetics over time, followed by a decline during later periods. Peaks of MAYV infection, titer, and dissemination from the midgut were detected at 15 and 21 days post ingestion at 30 °C and 20 °C, respectively. Mosquitoes were able to transmit MAYV as early as day 3 at 30 °C, but MAYV was not detectable in salivary secretions until day 15 at 20 °C. Low rates of MAYV in salivary secretions collected from infected mosquitoes provided evidence supporting the notion that a substantial salivary gland barrier(s) in Florida Ae. aegypti can limit the risk of MAYV transmission. Our results provide insights into the effects of temperature and time on the progression of infection and replication of MAYV in Ae. aegypti vectors.


Asunto(s)
Aedes , Infecciones por Alphavirus , Alphavirus , Alphavirus/fisiología , Animales , Florida , Mosquitos Vectores , Temperatura
17.
Viruses ; 14(2)2022 02 05.
Artículo en Inglés | MEDLINE | ID: mdl-35215918

RESUMEN

Getah virus (GETV) is a member of the alphavirus genus, and it infects a variety of animal species, including horses, pigs, cattle, and foxes. Human infection with this virus has also been reported. The structure of GETV has not yet been determined. In this study, we report the cryo-EM structure of GETV at a resolution of 3.5 Å. This structure reveals conformational polymorphism of the envelope glycoproteins E1 and E2 at icosahedral 3-fold and quasi-3-fold axes, which is believed to be a necessary organization in forming a curvature surface of virions. In our density map, three extra densities are identified, one of which is believed a "pocket factor"; the other two are located by domain D of E2, and they may maintain the stability of E1/E2 heterodimers. We also identify three N-glycosylations at E1 N141, E2 N200, and E2 N262, which might be associated with receptor binding and membrane fusion. The resolving of the structure of GETV provides new insights into the structure and assembly of alphaviruses and lays a basis for studying the differences of biology and pathogenicity between arthritogenic and encephalitic alphaviruses.


Asunto(s)
Infecciones por Alphavirus/veterinaria , Infecciones por Alphavirus/virología , Alphavirus/fisiología , Alphavirus/ultraestructura , Ensamble de Virus , Alphavirus/clasificación , Alphavirus/genética , Animales , Bovinos/virología , Microscopía por Crioelectrón , Dimerización , Zorros/virología , Caballos/virología , Humanos , Modelos Moleculares , Filogenia , Porcinos/virología , Proteínas del Envoltorio Viral/química , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/metabolismo , Virión/clasificación , Virión/genética , Virión/fisiología , Virión/ultraestructura
18.
J Virol ; 96(6): e0006022, 2022 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-35107376

RESUMEN

The impact of the host microbiota on arbovirus infections is currently not well understood. Arboviruses are viruses transmitted through the bites of infected arthropods, predominantly mosquitoes or ticks. The first site of arbovirus inoculation is the biting site in the host skin, which is colonized by a complex microbial community that could possibly influence arbovirus infection. We demonstrated that preincubation of arboviruses with certain components of the bacterial cell wall, including lipopolysaccharides (LPS) of some Gram-negative bacteria and lipoteichoic acids or peptidoglycan of certain Gram-positive bacteria, significantly reduced arbovirus infectivity in vitro. This inhibitory effect was observed for arboviruses of different virus families, including chikungunya virus of the Alphavirus genus and Zika virus of the Flavivirus genus, showing that this is a broad phenomenon. A modest inhibitory effect was observed following incubation with a panel of heat-inactivated bacteria, including bacteria residing on the skin. No viral inhibition was observed after preincubation of cells with LPS. Furthermore, a virucidal effect of LPS on viral particles was noticed by electron microscopy. Therefore, the main inhibitory mechanism seems to be due to a direct effect on the virus particles. Together, these results suggest that bacteria are able to decrease the infectivity of alphaviruses and flaviviruses. IMPORTANCE During the past decades, the world has experienced a vast increase in epidemics of alphavirus and flavivirus infections. These viruses can cause severe diseases, such as hemorrhagic fever, encephalitis, and arthritis. Several alpha- and flaviviruses, such as chikungunya virus, Zika virus, and dengue virus, are significant global health threats because of their high disease burden, their widespread (re-)emergence, and the lack of (good) anti-arboviral strategies. Despite the clear health burden, alphavirus and flavivirus infection and disease are not fully understood. A knowledge gap in the interplay between the host and the arbovirus is the potential interaction with host skin bacteria. Therefore, we studied the effect of (skin) bacteria and bacterial cell wall components on alphavirus and flavivirus infectivity in cell culture. Our results show that certain bacterial cell wall components markedly reduced viral infectivity by interacting directly with the virus particle.


Asunto(s)
Alphavirus , Arbovirus , Pared Celular , Flavivirus , Alphavirus/patogenicidad , Alphavirus/fisiología , Animales , Arbovirus/patogenicidad , Arbovirus/fisiología , Bacterias , Virus Chikungunya , Flavivirus/patogenicidad , Flavivirus/fisiología , Lipopolisacáridos , Microbiota , Virus Zika
19.
J Virol ; 96(2): e0177421, 2022 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-34757841

RESUMEN

Alphaviruses and flaviviruses have class II fusion glycoproteins that are essential for virion assembly and infectivity. Importantly, the tip of domain II is structurally conserved between the alphavirus and flavivirus fusion proteins, yet whether these structural similarities between virus families translate to functional similarities is unclear. Using in vivo evolution of Zika virus (ZIKV), we identified several novel emerging variants, including an envelope glycoprotein variant in ß-strand c (V114M) of domain II. We have previously shown that the analogous ß-strand c and the ij loop, located in the tip of domain II of the alphavirus E1 glycoprotein, are important for infectivity. This led us to hypothesize that flavivirus E ß-strand c also contributes to flavivirus infection. We generated this ZIKV glycoprotein variant and found that while it had little impact on infection in mosquitoes, it reduced replication in human cells and mice and increased virus sensitivity to ammonium chloride, as seen for alphaviruses. In light of these results and given our alphavirus ij loop studies, we mutated a conserved alanine at the tip of the flavivirus ij loop to valine to test its effect on ZIKV infectivity. Interestingly, this mutation inhibited infectious virion production of ZIKV and yellow fever virus, but not West Nile virus. Together, these studies show that shared domains of the alphavirus and flavivirus class II fusion glycoproteins harbor structurally analogous residues that are functionally important and contribute to virus infection in vivo.IMPORTANCE Arboviruses are a significant global public health threat, yet there are no antivirals targeting these viruses. This problem is in part due to our lack of knowledge of the molecular mechanisms involved in the arbovirus life cycle. In particular, virus entry and assembly are essential processes in the virus life cycle and steps that can be targeted for the development of antiviral therapies. Therefore, understanding common, fundamental mechanisms used by different arboviruses for entry and assembly is essential. In this study, we show that flavivirus and alphavirus residues located in structurally conserved and analogous regions of the class II fusion proteins contribute to common mechanisms of entry, dissemination, and infectious-virion production. These studies highlight how class II fusion proteins function and provide novel targets for development of antivirals.


Asunto(s)
Alphavirus/fisiología , Flavivirus/fisiología , Proteínas Virales de Fusión/metabolismo , Virión/metabolismo , Replicación Viral , Células A549 , Alphavirus/efectos de los fármacos , Cloruro de Amonio/farmacología , Animales , Culicidae/virología , Flavivirus/efectos de los fármacos , Humanos , Interferón Tipo I/deficiencia , Ratones , Ratones Mutantes , Mutación , Dominios Proteicos , Proteínas Virales de Fusión/química , Proteínas Virales de Fusión/genética , Proteínas no Estructurales Virales/química , Proteínas no Estructurales Virales/genética , Proteínas no Estructurales Virales/metabolismo , Virión/genética , Ensamble de Virus/genética , Internalización del Virus/efectos de los fármacos , Replicación Viral/genética , Virus Zika/efectos de los fármacos , Virus Zika/fisiología , Infección por el Virus Zika/virología
20.
PLoS Negl Trop Dis ; 15(12): e0010016, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34898602

RESUMEN

Improving our understanding of Mayaro virus (MAYV) ecology is critical to guide surveillance and risk assessment. We conducted a PRISMA-adherent systematic review of the published and grey literature to identify potential arthropod vectors and non-human animal reservoirs of MAYV. We searched PubMed/MEDLINE, Embase, Web of Science, SciELO and grey-literature sources including PAHO databases and dissertation repositories. Studies were included if they assessed MAYV virological/immunological measured occurrence in field-caught, domestic, or sentinel animals or in field-caught arthropods. We conducted an animal seroprevalence meta-analysis using a random effects model. We compiled granular georeferenced maps of non-human MAYV occurrence and graded the quality of the studies using a customized framework. Overall, 57 studies were eligible out of 1523 screened, published between the years 1961 and 2020. Seventeen studies reported MAYV positivity in wild mammals, birds, or reptiles and five studies reported MAYV positivity in domestic animals. MAYV positivity was reported in 12 orders of wild-caught vertebrates, most frequently in the orders Charadriiformes and Primate. Sixteen studies detected MAYV in wild-caught mosquito genera including Haemagogus, Aedes, Culex, Psorophora, Coquillettidia, and Sabethes. Vertebrate animals or arthropods with MAYV were detected in Brazil, Panama, Peru, French Guiana, Colombia, Trinidad, Venezuela, Argentina, and Paraguay. Among non-human vertebrates, the Primate order had the highest pooled seroprevalence at 13.1% (95% CI: 4.3-25.1%). From the three most studied primate genera we found the highest seroprevalence was in Alouatta (32.2%, 95% CI: 0.0-79.2%), followed by Callithrix (17.8%, 95% CI: 8.6-28.5%), and Cebus/Sapajus (3.7%, 95% CI: 0.0-11.1%). We further found that MAYV occurs in a wide range of vectors beyond Haemagogus spp. The quality of evidence behind these findings was variable and prompts calls for standardization of reporting of arbovirus occurrence. These findings support further risk emergence prediction, guide field surveillance efforts, and prompt further in-vivo studies to better define the ecological drivers of MAYV maintenance and potential for emergence.


Asunto(s)
Infecciones por Alphavirus/veterinaria , Infecciones por Alphavirus/virología , Alphavirus/fisiología , Vectores Artrópodos/virología , Reservorios de Enfermedades/virología , Mosquitos Vectores/virología , Alphavirus/genética , Infecciones por Alphavirus/transmisión , Animales , Vectores Artrópodos/fisiología , Aves/virología , Humanos , Mamíferos/virología , Mosquitos Vectores/fisiología , Primates/virología , Reptiles/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...