Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Med J Aust ; 201(11): 647-9, 2014 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-25495308

RESUMEN

Australian bat lyssavirus (ABLV) infection in humans is rare but fatal, with no proven effective therapy. ABLV infection can be prevented by administration of a post-exposure prophylaxis regimen of human rabies immunoglobulin and rabies vaccine. All Australian bats (flying foxes and microbats) should be considered to be carrying ABLV unless proven otherwise. Any bat-related injury (bite, scratch or mucosal exposure to bat saliva or neural tissue) should be notified immediately to the relevant public health unit - no matter how small the injury or how long ago it occurred. Human-to-human transmission of ABLV has not been reported but is theoretically possible. Standard infection control precautions should be employed when managing patients with suspected or confirmed ABLV infection.


Asunto(s)
Quirópteros/virología , Lyssavirus , Infecciones por Rhabdoviridae/virología , Animales , Australia , Mordeduras y Picaduras/virología , Vectores de Enfermedades , Humanos , Salud Pública , Infecciones por Rhabdoviridae/etiología , Infecciones por Rhabdoviridae/prevención & control , Infecciones por Rhabdoviridae/terapia , Infecciones por Rhabdoviridae/transmisión
2.
Viruses ; 13(9)2021 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-34578350

RESUMEN

Pathogen discovery contributes to our knowledge of bat-borne viruses and is linked to the heightened interest globally in bats as recognised reservoirs of zoonotic agents. The transmission of lyssaviruses from bats-to-humans, domestic animals, or other wildlife species is uncommon, but interest in these pathogens remains due to their ability to cause an acute, progressive, invariably fatal encephalitis in humans. Consequently, the detection and characterisation of bat lyssaviruses continues to expand our knowledge of their phylogroup definition, viral diversity, host species association, geographical distribution, evolution, mechanisms for perpetuation, and the potential routes of transmission. Although the opportunity for lyssavirus cross-species transmission seems rare, adaptation in a new host and the possibility of onward transmission to humans requires continued investigation. Considering the limited efficacy of available rabies biologicals it is important to further our understanding of protective immunity to minimize the threat from these pathogens to public health. Hence, in addition to increased surveillance, the development of a niche pan-lyssavirus vaccine or therapeutic biologics for post-exposure prophylaxis for use against genetically divergent lyssaviruses should be an international priority as these emerging lyssaviruses remain a concern for global public health.


Asunto(s)
Salud Pública , Infecciones por Rhabdoviridae/terapia , Animales , Quirópteros/virología , Encefalitis/terapia , Encefalitis/virología , Humanos , Italia , Lyssavirus/clasificación , Rabia , Infecciones por Rhabdoviridae/epidemiología , Infecciones por Rhabdoviridae/virología , Zoonosis/virología
3.
Viruses ; 13(3)2021 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-33804519

RESUMEN

Australian bat lyssavirus (ABLV) is a rhabdovirus that circulates in four species of pteropid bats (ABLVp) and the yellow-bellied sheath-tailed bat (ABLVs) in mainland Australia. In the three confirmed human cases of ABLV, rabies illness preceded fatality. As with rabies virus (RABV), post-exposure prophylaxis (PEP) for potential ABLV infections consists of wound cleansing, administration of the rabies vaccine and injection of rabies immunoglobulin (RIG) proximal to the wound. Despite the efficacy of PEP, the inaccessibility of human RIG (HRIG) in the developing world and the high immunogenicity of equine RIG (ERIG) has led to consideration of human monoclonal antibodies (hmAbs) as a passive immunization option that offers enhanced safety and specificity. Using a recombinant vesicular stomatitis virus (rVSV) expressing the glycoprotein (G) protein of ABLVs and phage display, we identified two hmAbs, A6 and F11, which completely neutralize ABLVs/ABLVp, and RABV at concentrations ranging from 0.39 and 6.25 µg/mL and 0.19 and 0.39 µg/mL respectively. A6 and F11 recognize overlapping epitopes in the lyssavirus G protein, effectively neutralizing phylogroup 1 lyssaviruses, while having little effect on phylogroup 2 and non-grouped diverse lyssaviruses. These results suggest that A6 and F11 could be effective therapeutic and diagnostic tools for phylogroup 1 lyssavirus infections.


Asunto(s)
Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/aislamiento & purificación , Reacciones Cruzadas/inmunología , Lyssavirus/clasificación , Lyssavirus/inmunología , Filogenia , Animales , Anticuerpos Monoclonales/uso terapéutico , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Australia , Mordeduras y Picaduras , Técnicas de Visualización de Superficie Celular , Quirópteros/virología , Epítopos/inmunología , Células HEK293 , Caballos , Humanos , Lyssavirus/genética , Pruebas de Neutralización , Profilaxis Posexposición , Rabia/prevención & control , Vacunas Antirrábicas/inmunología , Virus de la Rabia/inmunología , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/prevención & control , Infecciones por Rhabdoviridae/terapia , Vesiculovirus/genética
4.
Viral Immunol ; 12(1): 35-40, 1999.
Artículo en Inglés | MEDLINE | ID: mdl-10333240

RESUMEN

Vesicular stomatitis virus (VSV) applied intranasally to mice initially infects the olfactory receptor neurons, and then spreads quickly to the rest of the central nervous system (CNS). Previously, we have shown that the cytokine interleukin-12 (IL-12) has a significant survival and recovery promoting effect in mice infected with VSV when administered at the time of infection. The question of whether IL-12 is efficacious under the more clinically relevant condition of post-infection administration was explored. The data show that when IL-12 is administered post-infection, it is as effective as at the time of infection.


Asunto(s)
Encefalitis Viral/terapia , Interleucina-12/uso terapéutico , Infecciones por Rhabdoviridae/terapia , Animales , Esquema de Medicación , Encefalitis Viral/inmunología , Encefalitis Viral/mortalidad , Inyecciones Intraperitoneales , Interleucina-12/administración & dosificación , Masculino , Ratones , Ratones Endogámicos BALB C , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/mortalidad , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Replicación Viral/inmunología
5.
Rev Sci Tech ; 19(1): 177-96, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-11189715

RESUMEN

Bats, which represent approximately 24% of all known mammalian species, frequently act as vectors of lyssaviruses. In particular, insectivorous bats play an important role in the epidemiology of rabies and some rabies-like viruses, while the haematophagous vampire bats are the major wildlife vector for rabies in Latin America. In contrast, the role of fruit bats (flying foxes) in the epidemiology of the recently discovered Australian bat lyssavirus is only just emerging. Information on the pathogenesis of lyssaviruses in bats is scarce. However, in general, mortality in bats infected via a natural route appears to be low, and seroconversion occurs in many of those that survive. While transmission of rabies from an infected bat may be via a bite, other routes are apparently also possible. Methods for the diagnosis of bat lyssavirus infections in bats and terrestrial mammals (including humans) are similar to the classical procedures for rabies. Measures for the prevention and control of these diseases are also similar to those for rabies, although additional innovative methods have been tested, specifically to control vampire bat rabies.


Asunto(s)
Quirópteros , Vectores de Enfermedades , Lyssavirus , Infecciones por Rhabdoviridae/epidemiología , Zoonosis/transmisión , Animales , Humanos , Lyssavirus/clasificación , Lyssavirus/genética , Lyssavirus/fisiología , Lyssavirus/ultraestructura , Rabia/diagnóstico , Rabia/epidemiología , Rabia/terapia , Rabia/transmisión , Infecciones por Rhabdoviridae/diagnóstico , Infecciones por Rhabdoviridae/terapia , Infecciones por Rhabdoviridae/transmisión , Zoonosis/epidemiología , Zoonosis/virología
6.
Am J Vet Res ; 55(8): 1093-100, 1994 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-7978648

RESUMEN

Recombinant equine interferon-beta 1 (reqIFN-beta 1) induces an antiviral state in blood mononuclear cells (BMC) of horses. Maximal protection against replication of vesicular stomatitis virus is achieved 6 hours after treatment with IFN in vitro and in vivo. Duration of the protective effect depends on the dose of IFN in vitro and in vivo. Availability of reqIFN-beta 1 in cultures of BMC for up to 48 hours does not prolong the antiviral state. The protective effect on BMC after treatment with IFN has similar duration in vivo and in vitro. Monitoring of the effect of IFN in vivo is, thus, simplified because the antiviral state may be recorded by testing cells twice (ie, before and 6 hours after application of interferon). All further tests may be performed in vitro. Multiple administration of reqIFN-beta 1 do not prolong duration of the protective phases after each administration. Duration of the antiviral state depends only on the dose of reqIFN-beta 1.


Asunto(s)
Interferón beta/farmacología , Virus de la Estomatitis Vesicular Indiana/efectos de los fármacos , Animales , Temperatura Corporal , Femenino , Enfermedades de los Caballos/terapia , Caballos , Técnicas In Vitro , Interferón beta/administración & dosificación , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/virología , Masculino , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Infecciones por Rhabdoviridae/terapia , Infecciones por Rhabdoviridae/veterinaria , Estomatitis/terapia , Estomatitis/veterinaria , Virus de la Estomatitis Vesicular Indiana/fisiología , Replicación Viral/efectos de los fármacos
7.
Pediatrics ; 133(4): e1063-7, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24590754

RESUMEN

Human infection with Australian Bat Lyssavirus is extremely rare and has not previously been reported in a child. We describe a fatal case of Australian Bat Lyssavirus in an 8-year-old child, and review the literature pertaining to the diagnosis and management of lyssavirus infection with consideration of its applicability to this emerging strain.


Asunto(s)
Lyssavirus , Infecciones por Rhabdoviridae , Australia , Niño , Resultado Fatal , Humanos , Masculino , Infecciones por Rhabdoviridae/diagnóstico , Infecciones por Rhabdoviridae/terapia
8.
Int Immunol ; 18(8): 1285-93, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16769750

RESUMEN

CD4 T cells are not thought to play a significant role in generating an effective primary CD8 T cell response to most viral infections. We have challenged this view by demonstrating that antigen-specific CD4 T cells can indeed suppress the proliferation of antigen-specific naive CD8 T cells in response to low doses of vesicular stomatitis virus. This finding is in contrast to the established observations that at high antigen loads CD4 T cells play little role in generating CD8 T cell responses, and that in non-infectious model systems CD4 T cells actually help the CD8 T cell response. Our results suggest that at low infectious doses, CD4 T cells play a much larger role in controlling infections than previously appreciated.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunoterapia Adoptiva/métodos , Infecciones por Rhabdoviridae/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Animales , Antígenos Virales/inmunología , Linfocitos B/inmunología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Rhabdoviridae/terapia , Infecciones por Rhabdoviridae/virología
9.
J Immunol ; 177(1): 492-500, 2006 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-16785546

RESUMEN

We have examined the role of dendritic cells (DCs) in the antiviral immune response and viral clearance using a transgenic mouse model (CD11c-diphtheria toxin (DT) receptor GFP) that allows for their conditional ablation in vivo. DT administration systemically ablated conventional and IFN-producing plasmacytoid DCs (pDCs) in transgenic, but not nontransgenic littermates, without elimination of splenic macrophages. Unexpectedly, early (12 and 48 h postinfection) viral clearance of vesicular stomatitis virus was normal in DC-depleted mice despite markedly reduced serum titers of type I IFN. DC-depleted mice remained virus-free with the exception of a subset (approximately 30%) that developed overwhelming and fatal brain infections 6 days postinfection. However, DT treatment profoundly inhibited clonal expansion of naive CD8+ vesicular stomatitis virus-specific T cells without altering the primary Th1 and Th2 cytokine response. Optimal clonal expansion required pDCs because selective elimination of these cells in vivo with a depleting Ab also suppressed expansion of tetramer+ cells, although Th1/Th2 cytokine production remained unaltered. Collectively, these data indicate that conventional DCs and to a lesser extent pDCs are critical for proliferation of naive antiviral T cells. However, other components of the primary adaptive immune response (Th1/Th2 cytokines) are essentially normal in the absence of DCs, which may account for the efficient viral clearance seen in DC-depleted mice. Thus, sufficient redundancy exists in the immune system to sustain efficient viral clearance despite loss of an APC considered essential for induction of a primary antiviral immune response.


Asunto(s)
Células Dendríticas/inmunología , Células Dendríticas/virología , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/virología , Virus de la Estomatitis Vesicular Indiana/inmunología , Carga Viral , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Proliferación Celular , Células Dendríticas/citología , Toxina Diftérica/administración & dosificación , Encefalitis Viral/genética , Encefalitis Viral/inmunología , Encefalitis Viral/virología , Femenino , Inmunosupresores/administración & dosificación , Interferón Tipo I/antagonistas & inhibidores , Interferón Tipo I/biosíntesis , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fase de Descanso del Ciclo Celular/genética , Fase de Descanso del Ciclo Celular/inmunología , Infecciones por Rhabdoviridae/genética , Infecciones por Rhabdoviridae/terapia , Bazo/citología , Bazo/inmunología , Subgrupos de Linfocitos T/citología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/virología , Virus de la Estomatitis Vesicular Indiana/crecimiento & desarrollo
10.
Vet Clin North Am Equine Pract ; 16(3): 457-69, viii-ix, 2000 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11219343

RESUMEN

Physical trauma, dietary factors, certain toxins, immune mediated disorders, and vesicular stomatitis virus (VSV) infection are known causes of stomatitis in horses. There is evidence that some outbreaks of equine stomatitis are caused by as yet unidentified infectious agents. It remains to be determined whether stomatitis is an emerging equine infectious disease, or if the increase in reported cases is simply the result of greater public awareness as a consequence of widespread outbreaks of VSV in the southwestern United States in recent years. Focused laboratory and epidemiological studies are necessary to more adequately define non-VS related infectious and noninfectious causes of equine stomatitis.


Asunto(s)
Enfermedades de los Caballos/etiología , Infecciones por Rhabdoviridae/veterinaria , Estomatitis/veterinaria , Virus de la Estomatitis Vesicular Indiana , Animales , Diagnóstico Diferencial , Enfermedades de los Caballos/diagnóstico , Enfermedades de los Caballos/epidemiología , Enfermedades de los Caballos/terapia , Caballos/virología , Infecciones por Rhabdoviridae/epidemiología , Infecciones por Rhabdoviridae/terapia , Estomatitis/epidemiología , Estomatitis/etiología , Estomatitis/terapia , Úlcera
11.
J Immunol ; 155(5): 2533-44, 1995 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-7650383

RESUMEN

Small resting B cells do not support a productive vesicular stomatitis virus (VSV) infection, but are induced by B cell activators to become fully permissive for VSV replication. Nonpermissive B cell populations restrict VSV expression at multiple points: transcript levels, translation, and maturation. Unstimulated resting G0 B cells can be infected by VSV and support the synthesis of all VSV mRNAs. Steady-state levels of viral transcripts are selectively enhanced by T cell-derived cytokines to an extent comparable with that seen for cytokine-regulated cellular mRNAs. However, viral proteins are not detected in immunoprecipitates from unstimulated or cytokine-stimulated B cells despite the fact that viral mRNAs are associated with polysomes and can be translated in vitro. This translational block is released by stimulation of infected B cells with mitogenic anti-lg or LPS, or non-mitogenic PMA. VSV virion maturation is also regulated by activation signals, because neither anti-lg nor PMA-stimulated B cells produce high levels of infectious VSV particles. Because anti-lg stimulation supports viral genome replication, maturational arrest is apparently at virus assembly or release. PMA and ionomycin induces changes beyond those seen with anti-lg, because these B cells produce PFUs at levels comparable with those seen with LPS-activated B cells and VSV-permissive cell lines. Activation-dependent regulation of virus expression provides a new paradigm for assessing activator-induced events in B cell differentiation not revealed by previous assessments of proliferation of Ab synthesis.


Asunto(s)
Linfocitos B/virología , Activación de Linfocitos/inmunología , Infecciones por Rhabdoviridae/terapia , Replicación Viral/inmunología , Animales , Femenino , Inmunoglobulinas/inmunología , Lipopolisacáridos/farmacología , Masculino , Ratones , Ratones Endogámicos A , Ratones Endogámicos CBA , Fitohemaglutininas/farmacología , Biosíntesis de Proteínas/efectos de los fármacos , Biosíntesis de Proteínas/inmunología , ARN Viral/análisis , Infecciones por Rhabdoviridae/inmunología , Infecciones por Rhabdoviridae/prevención & control , Linfocitos T Colaboradores-Inductores/inmunología , Virus de la Estomatitis Vesicular Indiana/genética , Ensayo de Placa Viral , Proteínas Virales/análisis , Replicación Viral/efectos de los fármacos
12.
J Immunol ; 155(12): 5684-9, 1995 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-7499854

RESUMEN

To investigate the role of a cytokine in host defense against the vesicular stomatitis virus (VSV) infection of the central nervous system (CNS), IL-12 was injected i.p. into groups of 10 BALB/c mice on days -1, 0, 1, 2, and 3 postinfection. Four days postinfection, mice were examined. IL-12 strongly enhanced immunity to VSV infection in the CNS as demonstrated by 1) decreased VSV titers in brain homogenate of IL-12-injected mice compared with those of controls; 2) increased expression of inducible nitric oxide synthase in the CNS; 3) enhanced expression of both MHC class I and class II Ags in the CNS; 4) increased T cell infiltration in the CNS, especially in the olfactory bulb; and 5) diminished VSV-induced apoptosis in olfactory bulb. No detrimental effect was observed even with the 200 ng/mouse dose of IL-12. Protective effects of IL-12 were dose dependent. Collectively, these results demonstrate that exogenously added IL-12, even when injected peripherally, significantly enhances recovery from VSV infection of the CNS.


Asunto(s)
Encefalopatías/virología , Interleucina-12/uso terapéutico , Infecciones por Rhabdoviridae/terapia , Virus de la Estomatitis Vesicular Indiana/aislamiento & purificación , Animales , Apoptosis/efectos de los fármacos , Encefalopatías/enzimología , Encefalopatías/inmunología , Antígenos H-2/biosíntesis , Antígenos de Histocompatibilidad Clase II/biosíntesis , Interleucina-12/farmacología , Masculino , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/efectos de los fármacos , Bulbo Olfatorio/inmunología , Infecciones por Rhabdoviridae/enzimología , Infecciones por Rhabdoviridae/inmunología , Linfocitos T/inmunología
13.
Mol Mar Biol Biotechnol ; 5(2): 114-22, 1996 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-8680524

RESUMEN

Plasmid vectors encoding the infectious hematopoietic necrosis virus (IHNV) nucleoprotein or glycoprotein gene under the control of a cytomegalovirus promoter were used to immunize rainbow trout (Oncorhynchus mykiss) against IHNV. The plasmid DNA was injected into the skeletal muscle of rainbow trout fry, and immunization was determined by the detection of virus-neutralizing and enzyme-linked immunosorbent assay antibody activity, and by protection against live virus challenge. Fish injected with the glycoprotein-encoding plasmid pCMV4-G, either alone or in combination with the nucleoprotein-encoding plasmid pCMV4-N, generated glycoprotein-specific and virus-neutralizing antibody responses. The vaccinated fish were also protected from subsequent IHNV challenge. Fish receiving pCMV4-N alone did not produce measurable virus-specific antibody and were killed by IHNV infection. These studies show that DNA vaccination will protect rainbow trout against the lethal effects of IHNV infection.


Asunto(s)
ADN Viral/genética , Enfermedades de los Peces/prevención & control , Oncorhynchus mykiss/virología , Infecciones por Rhabdoviridae/veterinaria , Rhabdoviridae/inmunología , Vacunas Virales/genética , Animales , Anticuerpos Antivirales/sangre , Especificidad de Anticuerpos , Cápside/biosíntesis , Cápside/genética , Cápside/inmunología , Línea Celular , Enfermedades de los Peces/terapia , Pruebas de Neutralización , Oncorhynchus mykiss/genética , Plásmidos/uso terapéutico , Rhabdoviridae/genética , Infecciones por Rhabdoviridae/prevención & control , Infecciones por Rhabdoviridae/terapia , Vacunación , Vacunas Sintéticas/genética , Proteínas del Núcleo Viral/biosíntesis , Proteínas del Núcleo Viral/genética , Proteínas del Núcleo Viral/inmunología , Proteínas del Envoltorio Viral/biosíntesis , Proteínas del Envoltorio Viral/genética , Proteínas del Envoltorio Viral/inmunología
14.
J Immunol ; 160(4): 1841-9, 1998 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-9469445

RESUMEN

A bispecific Ab (BsAb) that binds the TCR on T cells and the G protein of the vesicular stomatitis virus (VSV) can redirect staphylococcal enterotoxin B (SEB)-activated T cells to kill VSV-infected cells and to inhibit VSV replication in vitro. Inhibition of virus replication in our system is dependent upon the specificity of the Ab for the viral protein. IFN-gamma does not play a very important role in this phenomenon, which is mainly mediated by the release of Pfp from CD8+ T cells. We have used a Stat1 knockout mouse model in which VSV infection is lethal. Infusion of staphylococcal enterotoxin-activated B T cells and bispecific Ab significantly slowed virus progression and prolonged the survival of VSV-infected Stat1 knockout mice in vivo.


Asunto(s)
Anticuerpos Biespecíficos/farmacología , Infecciones por Rhabdoviridae/inmunología , Superantígenos/inmunología , Linfocitos T/inmunología , Virus de la Estomatitis Vesicular Indiana/inmunología , Replicación Viral/inmunología , Animales , Anticuerpos Biespecíficos/administración & dosificación , Anticuerpos Biespecíficos/biosíntesis , Antivirales/metabolismo , Antivirales/fisiología , Células Cultivadas , Cricetinae , Pruebas Inmunológicas de Citotoxicidad , Enterotoxinas/administración & dosificación , Enterotoxinas/genética , Enterotoxinas/inmunología , Inyecciones Intraperitoneales , Interferón gamma/metabolismo , Activación de Linfocitos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Rhabdoviridae/terapia , Infecciones por Rhabdoviridae/virología , Especificidad de la Especie , Staphylococcus aureus/inmunología , Estomatitis/inmunología , Estomatitis/terapia , Estomatitis/virología , Superantígenos/administración & dosificación , Superantígenos/genética , Linfocitos T/metabolismo , Linfocitos T/virología , Linfocitos T Citotóxicos/inmunología , Virus de la Estomatitis Vesicular Indiana/clasificación , Virus de la Estomatitis Vesicular Indiana/fisiología
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda