Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 4.042
Filtrar
1.
Vet Q ; 44(1): 1-12, 2024 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-38726839

RESUMEN

Duck plague (DP) is an acute, contagious and fatal disease, caused by duck enteritis virus (DEV), with worldwide distribution causing several outbreaks and posing severe economic losses. The present study was carried out with a goal of development of a live attenuated cell culture based DP vaccine using an Indian strain of DEV and evaluation of its safety, efficacy along with complete genome analysis. The live attenuated DP vaccine (DPvac/IVRI-19) was developed by serial propagation of a virulent isolate of DEV (DEV/India/IVRI-2016) in the chicken embryo fibroblast (CEF) primary cell culture. Adaptation of DEV in CEF cell culture was indicated by more rapid appearance of cytopathic effects (CPE) and gradual increase of virus titre, which reached up to 107.5 TCID50/mL after 41 passages. The safety, immunogenicity and efficacy of the vaccine were determined by immunization trials in ducklings. The DPvac/IVRI-19 was found to be avirulent and completely safe in the ducklings. Further, the vaccine induced both humoral and cell mediated immune responses and afforded 100% protection against the virulent DEV challenge. A comparison of the whole genome of DPvac/IVRI-19 (MZ911871) and DEV/India/IVRI-2016 (MZ824102) revealed significant number of mutations, which might be associated with viral attenuation. Phylogenetic tree of DEV/India/IVRI-2016 revealed its evolutionary relationship with other DEV isolates, but it formed a separate cluster with certain unique mutations. Thus, with the proven safety and 100% efficacy, the DPvac/IVRI-19 is suitable for large scale production with precisely pure form of vaccine and has potential utility at national and global levels.


Asunto(s)
Patos , Fibroblastos , Mardivirus , Enfermedades de las Aves de Corral , Vacunas Atenuadas , Vacunas Virales , Animales , Vacunas Atenuadas/inmunología , Patos/virología , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Fibroblastos/virología , Embrión de Pollo , Vacunas Virales/inmunología , Mardivirus/inmunología , Mardivirus/patogenicidad , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/virología , India
2.
J Gen Virol ; 105(5)2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38767608

RESUMEN

Herpesviruses establish a well-adapted balance with their host's immune system. Despite this co-evolutionary balance, infections can lead to severe disease including neurological disorders in their natural host. In horses, equine herpesvirus 1 (EHV-1) causes respiratory disease, abortions, neonatal foal death and myeloencephalopathy (EHM) in ~10 % of acute infections worldwide. Many aspects of EHM pathogenesis and protection from EHM are still poorly understood. However, it has been shown that the incidence of EHM increases to >70 % in female horses >20 years of age. In this study we used old mares as an experimental equine EHV-1 model of EHM to identify host-specific factors contributing to EHM. Following experimental infection with the neuropathogenic strain EHV-1 Ab4, old mares and yearling horses were studied for 21 days post-infection. Nasal viral shedding and cell-associated viremia were assessed by quantitative PCR. Cytokine/chemokine responses were evaluated in nasal secretions and cerebrospinal fluid (CSF) by Luminex assay and in whole blood by quantitative real-time PCR. EHV-1-specific IgG sub-isotype responses were measured by ELISA. All young horses developed respiratory disease and a bi-phasic fever post-infection, but only 1/9 horses exhibited ataxia. In contrast, respiratory disease was absent in old mares, but all old mares developed EHM that resulted in euthanasia in 6/9 old mares. Old mares also presented significantly decreased nasal viral shedding but higher viremia coinciding with a single fever peak at the onset of viremia. According to clinical disease manifestation, horses were sorted into an EHM group (nine old horses and one young horse) and a non-EHM group (eight young horses) for assessment of host immune responses. Non-EHM horses showed an early upregulation of IFN-α (nasal secretions), IRF7/IRF9, IL-1ß, CXCL10 and TBET (blood) in addition to an IFN-γ upregulation during viremia (blood). In contrast, IFN-α levels in nasal secretions of EHM horses were low and peak levels of IRF7, IRF9, CXCL10 and TGF-ß (blood) coincided with viremia. Moreover, EHM horses showed significantly higher IL-10 levels in nasal secretions, peripheral blood mononuclear cells and CSF and higher serum IgG3/5 antibody titres compared to non-EHM horses. These results suggest that protection from EHM depends on timely induction of type 1 IFN and upregulation cytokines and chemokines that are representative of cellular immunity. In contrast, induction of regulatory or TH-2 type immunity appeared to correlate with an increased risk for EHM. It is likely that future vaccine development for protection from EHM must target shifting this 'at-risk' immunophenotype.


Asunto(s)
Citocinas , Infecciones por Herpesviridae , Herpesvirus Équido 1 , Enfermedades de los Caballos , Animales , Caballos , Herpesvirus Équido 1/inmunología , Femenino , Enfermedades de los Caballos/virología , Enfermedades de los Caballos/inmunología , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Citocinas/sangre , Citocinas/inmunología , Anticuerpos Antivirales/sangre , Esparcimiento de Virus , Viremia/inmunología , Viremia/veterinaria , Inmunoglobulina G/sangre
3.
Sci Rep ; 14(1): 10651, 2024 05 09.
Artículo en Inglés | MEDLINE | ID: mdl-38724545

RESUMEN

Herpesviruses are large double-stranded DNA viruses that cause infections in animals and humans with a characteristic of latent infectious within specific tissues. Bats are natural hosts of variety human-infecting viruses and recently have been described as hosts for herpesviruses in several countries around the world. In this study we collected 140 insectivorous bats in the neighboring urban areas of Wuhan City, Hubei Province in the central China between 2020 and 2021. Nested PCR targeting the dpol gene sequence indicated that a total of 22 individuals (15.7% of the sample) tested positive for herpesvirus with 4 strains belonging to the genus Betaherpesvirus and the remaining 18 strains classified as Gammahersvirus. Furthermore, the herpesvirus prevalence in Rhinolophus pusillus was higher at 26.3%, compared to 8.4% in Myotis davidii. The RP701 strain from R. pusillus was the predominant gammaherpesvirus strain detected in bats, accounting for 94.4% (17/18) of all strains. The variations in γ-herpesviruses genomic sequences was evident in phylogenetic tree, where RP701 strain was clustered together with ruminant γ-herpesviruses, while MD704 strain formed a distinct clade with a hedgehog γ-herpesvirus. Four betaherpesviruses exclusively identified from M. davidii, with nucleotide identities ranging from 79.7 to 82.6% compared to known betaherpesviruses. Our study provided evidence that M. davidii can sever as natural host for ß-herpesviruses, which extended the host species range. In conclusion, we found that bats from central China harbored novel ß-herpesviruses and γ-herpesviruses which were phylogenetically related to ruminant γ-herpesvirus and hedgehog γ-herpesvirus. Our study indicates that bats are natural hosts of ß- and γ-herpesviruses and further studies are needed to determine whether there is cross-species transmission of herpesviruses between bats and other animals, or humans.


Asunto(s)
Betaherpesvirinae , Quirópteros , Gammaherpesvirinae , Infecciones por Herpesviridae , Filogenia , Animales , Quirópteros/virología , China/epidemiología , Gammaherpesvirinae/genética , Gammaherpesvirinae/aislamiento & purificación , Gammaherpesvirinae/clasificación , Betaherpesvirinae/genética , Betaherpesvirinae/aislamiento & purificación , Betaherpesvirinae/clasificación , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/epidemiología , Genoma Viral , ADN Viral/genética
4.
J Clin Invest ; 134(9)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38690731

RESUMEN

Herpesviruses establish latent infections, and most reactivate frequently, resulting in symptoms and virus shedding in healthy individuals. In immunocompromised patients, reactivating virus can cause severe disease. Persistent EBV has been associated with several malignancies in both immunocompromised and nonimmunocompromised persons. Reactivation and shedding occur with most herpesviruses, despite potent virus-specific antibodies and T cell immunity as measured in the blood. The licensure of therapeutic vaccines to reduce zoster indicates that effective therapeutic vaccines for other herpesviruses should be feasible. However, varicella-zoster virus is different from other human herpesviruses in that it is generally only shed during varicella and zoster. Unlike prophylactic vaccines, in which the correlate of immunity is antibody function, T cell immunity is the correlate of immunity for the only effective therapeutic herpesvirus vaccine-zoster vaccine. While most studies of therapeutic vaccines have measured immunity in the blood, cellular immunity at the site of reactivation is likely critical for an effective therapeutic vaccine for certain viruses. This Review summarizes the status of therapeutic vaccines for herpes simplex virus, cytomegalovirus, and Epstein-Barr virus and proposes approaches for future development.


Asunto(s)
Vacunas contra Herpesvirus , Humanos , Vacunas contra Herpesvirus/inmunología , Vacunas contra Herpesvirus/uso terapéutico , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/virología , Herpesvirus Humano 4/inmunología , Animales , Herpesviridae/inmunología , Activación Viral/inmunología , Citomegalovirus/inmunología
5.
J Med Virol ; 96(5): e29654, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38727099

RESUMEN

Human Herpesvirus 8 (HHV-8) has been classified by sequence analysis of open reading frame (ORF) K1, ORF K15, and variable sequence loci within the central constant region. The purpose of this study was to examine the molecular epidemiology of HHV-8 in an Irish population. This retrospective study included 30 patients who had HHV-8 DNA detected in plasma. Nested end-point PCR was used to characterise four regions of the HHV-8 genome, K1, T0.7 (K12), ORF 75, and K15. Sequencing data were obtained for 23 specimens from 19 patients. Phylogenetic analysis of ORF K1 demonstrated that subtypes A, B, C and F were present in 37%, 11%, 47% and 5%, respectively. For T0.7 and ORF 75, sequencing data were obtained for 12 patients. For T0.7, subtypes A/C, J, B, R and Q were present in 58%, 17%, 8%, 8%, and 8%, respectively. For ORF 75, subtypes A, B, C and D were present in 58%, 8%, 25%, and 8%, respectively. K15 sequences were determined for 13 patients. 69% had the P allele and 31% had the M allele. The data generated by this study demonstrate that a broad variety of HHV-8 subtypes are represented in patients exhibiting HHV-8-related disease in Ireland, a low prevalence country. The predominance of C and A K1 subtypes was as expected for a Western European population. The 31% prevalence for K15 subtype M was higher than expected for a Western European population. This may represent the changing and evolving epidemiology in Ireland due to altered migration patterns.


Asunto(s)
ADN Viral , Infecciones por Herpesviridae , Herpesvirus Humano 8 , Epidemiología Molecular , Filogenia , Análisis de Secuencia de ADN , Humanos , Irlanda/epidemiología , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Herpesvirus Humano 8/clasificación , Herpesvirus Humano 8/aislamiento & purificación , Masculino , Femenino , Estudios Retrospectivos , Persona de Mediana Edad , Adulto , ADN Viral/genética , Anciano , Adulto Joven , Reacción en Cadena de la Polimerasa , Genotipo , Adolescente , Sistemas de Lectura Abierta , Anciano de 80 o más Años , Niño , Datos de Secuencia Molecular
6.
Sci Rep ; 14(1): 10169, 2024 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-38702375

RESUMEN

Bovine viral diarrhea virus (BVDV) is considered to be the most common agent of severe diarrhea in cattle worldwide, causing fever, diarrhea, ulcers, and abortion. Bovine herpesvirus 1 (BoHV-1) is also a major bovine respiratory disease agent that spreads worldwide and causes extensive damage to the livestock industry. Recombinase polymerase amplification (RPA) is a novel nucleic acid amplification method with the advantages of high efficiency, rapidity and sensitivity, which has been widely used in the diagnosis of infectious diseases. A dual RPA assay was developed for the simultaneous detection of BVDV and BoHV-1. The assay was completed at a constant temperature of 37 °C for 30 min. It was highly sensitive and had no cross-reactivity with other common bovine viruses. The detection rate of BVDV RPA in clinical samples (36.67%) was higher than that of PCR (33.33%), the detection rate of BoHV-1 RPA and PCR were equal. Therefore, the established dual RPA assay for BVDV and BoHV-1 could be a potential candidate for use as an immediate diagnostic.


Asunto(s)
Virus de la Diarrea Viral Bovina , Herpesvirus Bovino 1 , Técnicas de Amplificación de Ácido Nucleico , Recombinasas , Animales , Bovinos , Herpesvirus Bovino 1/genética , Herpesvirus Bovino 1/aislamiento & purificación , Técnicas de Amplificación de Ácido Nucleico/métodos , Recombinasas/metabolismo , Virus de la Diarrea Viral Bovina/genética , Virus de la Diarrea Viral Bovina/aislamiento & purificación , Sensibilidad y Especificidad , Diarrea Mucosa Bovina Viral/virología , Diarrea Mucosa Bovina Viral/diagnóstico , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/diagnóstico , ADN Viral/genética
7.
J Virol ; 98(5): e0049324, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38578092

RESUMEN

CD4+ T cells play a key role in γ-herpesvirus infection control. However, the mechanisms involved are unclear. Murine herpesvirus type 4 (MuHV-4) allows relevant immune pathways to be dissected experimentally in mice. In the lungs, it colonizes myeloid cells, which can express MHC class II (MHCII), and type 1 alveolar epithelial cells (AEC1), which lack it. Nevertheless, CD4+ T cells can control AEC1 infection, and this control depends on MHCII expression in myeloid cells. Interferon-gamma (IFNγ) is a major component of CD4+ T cell-dependent MuHV-4 control. Here, we show that the action of IFNγ is also indirect, as CD4+ T cell-mediated control of AEC1 infection depended on IFNγ receptor (IFNγR1) expression in CD11c+ cells. Indirect control also depended on natural killer (NK) cells. Together, the data suggest that the activation of MHCII+ CD11c+ antigen-presenting cells is key to the CD4+ T cell/NK cell protection axis. By contrast, CD8+ T cell control of AEC1 infection appeared to operate independently. IMPORTANCE: CD4+ T cells are critical for the control of gamma-herpesvirus infection; they act indirectly, by recruiting natural killer (NK) cells to attack infected target cells. Here, we report that the CD4+ T cell/NK cell axis of gamma-herpesvirus control requires interferon-γ engagement of CD11c+ dendritic cells. This mechanism of CD4+ T cell control releases the need for the direct engagement of CD4+ T cells with virus-infected cells and may be a common strategy for host control of immune-evasive pathogens.


Asunto(s)
Linfocitos T CD4-Positivos , Infecciones por Herpesviridae , Interferón gamma , Células Asesinas Naturales , Receptores de Interferón , Rhadinovirus , Animales , Linfocitos T CD4-Positivos/inmunología , Interferón gamma/inmunología , Interferón gamma/metabolismo , Ratones , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Células Asesinas Naturales/inmunología , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Rhadinovirus/inmunología , Ratones Endogámicos C57BL , Receptor de Interferón gamma , Antígenos de Histocompatibilidad Clase II/inmunología , Antígenos de Histocompatibilidad Clase II/metabolismo , Células Epiteliales Alveolares/inmunología , Células Epiteliales Alveolares/virología , Linfocitos T CD8-positivos/inmunología , Antígeno CD11c/metabolismo , Antígeno CD11c/inmunología , Pulmón/inmunología , Pulmón/virología
8.
J Virol ; 98(5): e0198623, 2024 May 14.
Artículo en Inglés | MEDLINE | ID: mdl-38619272

RESUMEN

Human cytomegalovirus (hCMV) is a ubiquitous facultative pathogen, which establishes a characteristic latent and reactivating lifelong infection in immunocompetent hosts. Murine CMV (mCMV) infection is widely used as an experimental model of hCMV infection, employed to investigate the causal nature and extent of CMV's contribution to inflammatory, immunological, and health disturbances in humans. Therefore, mimicking natural human infection in mice would be advantageous to hCMV research. To assess the role of route and age at infection in modeling hCMV in mice, we infected prepubescent and young sexually mature C57BL/6 (B6) mice intranasally (i.n., a likely physiological route in humans) and intraperitoneally (i.p., a frequently used experimental route, possibly akin to transplant-mediated infection). In our hands, both routes led to comparable early viral loads and tissue spreads. However, they yielded differential profiles of innate and adaptive systemic immune activation. Specifically, the younger, prepubescent mice exhibited the strongest natural killer cell activation in the blood in response to i.p. infection. Further, the i.p. infected animals (particularly those infected at 12 weeks) exhibited larger anti-mCMV IgG and greater expansion of circulating CD8+ T cells specific for both acute (non-inflationary) and latent phase (inflationary) mCMV epitopes. By contrast, tissue immune responses were comparable between i.n. and i.p. groups. Our results illustrate a distinction in the bloodborne immune response profiles across infection routes and ages and are discussed in light of physiological parameters of interaction between CMV, immunity, inflammation, and health over the lifespan. IMPORTANCE: The majority of experiments modeling human cytomegalovirus (hCMV) infection in mice have been carried out using intraperitoneal infection in sexually mature adult mice, which stands in contrast to the large number of humans being infected with human CMV at a young age, most likely via bodily fluids through the nasopharyngeal/oral route. This study examined the impact of the choice of age and route of infection in modeling CMV infection in mice. By comparing young, prepubescent to older sexually mature counterparts, infected either via the intranasal or intraperitoneal route, we discovered substantial differences in deployment and response intensity of different arms of the immune system in systemic control of the virus; tissue responses, by contrast, appeared similar between ages and infection routes.


Asunto(s)
Inmunidad Adaptativa , Infecciones por Citomegalovirus , Modelos Animales de Enfermedad , Inmunidad Innata , Ratones Endogámicos C57BL , Muromegalovirus , Carga Viral , Animales , Ratones , Muromegalovirus/inmunología , Infecciones por Citomegalovirus/inmunología , Infecciones por Citomegalovirus/virología , Factores de Edad , Células Asesinas Naturales/inmunología , Anticuerpos Antivirales/sangre , Anticuerpos Antivirales/inmunología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Femenino , Humanos
9.
Viruses ; 16(4)2024 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-38675914

RESUMEN

Understanding the pathophysiology of myalgic encephalomyelitis/chronic fatigue syndrome (ME/CFS) is critical for advancing treatment options. This review explores the novel hypothesis that a herpesvirus infection of endothelial cells (ECs) may underlie ME/CFS symptomatology. We review evidence linking herpesviruses to persistent EC infection and the implications for endothelial dysfunction, encompassing blood flow regulation, coagulation, and cognitive impairment-symptoms consistent with ME/CFS and Long COVID. This paper provides a synthesis of current research on herpesvirus latency and reactivation, detailing the impact on ECs and subsequent systemic complications, including latent modulation and long-term maladaptation. We suggest that the chronicity of ME/CFS symptoms and the multisystemic nature of the disease may be partly attributable to herpesvirus-induced endothelial maladaptation. Our conclusions underscore the necessity for further investigation into the prevalence and load of herpesvirus infection within the ECs of ME/CFS patients. This review offers conceptual advances by proposing an endothelial infection model as a systemic mechanism contributing to ME/CFS, steering future research toward potentially unexplored avenues in understanding and treating this complex syndrome.


Asunto(s)
Células Endoteliales , Síndrome de Fatiga Crónica , Infecciones por Herpesviridae , Humanos , Células Endoteliales/virología , Síndrome de Fatiga Crónica/virología , Síndrome de Fatiga Crónica/fisiopatología , Herpesviridae/fisiología , Infecciones por Herpesviridae/virología , Latencia del Virus , Síndrome Post Agudo de COVID-19/patología , Síndrome Post Agudo de COVID-19/fisiopatología
10.
PLoS Pathog ; 20(4): e1011829, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38620036

RESUMEN

Viruses target mitochondria to promote their replication, and infection-induced stress during the progression of infection leads to the regulation of antiviral defenses and mitochondrial metabolism which are opposed by counteracting viral factors. The precise structural and functional changes that underlie how mitochondria react to the infection remain largely unclear. Here we show extensive transcriptional remodeling of protein-encoding host genes involved in the respiratory chain, apoptosis, and structural organization of mitochondria as herpes simplex virus type 1 lytic infection proceeds from early to late stages of infection. High-resolution microscopy and interaction analyses unveiled infection-induced emergence of rough, thin, and elongated mitochondria relocalized to the perinuclear area, a significant increase in the number and clustering of endoplasmic reticulum-mitochondria contact sites, and thickening and shortening of mitochondrial cristae. Finally, metabolic analyses demonstrated that reactivation of ATP production is accompanied by increased mitochondrial Ca2+ content and proton leakage as the infection proceeds. Overall, the significant structural and functional changes in the mitochondria triggered by the viral invasion are tightly connected to the progression of the virus infection.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Mitocondrias , Mitocondrias/metabolismo , Herpesvirus Humano 1/fisiología , Herpesvirus Humano 1/metabolismo , Humanos , Herpes Simple/metabolismo , Herpes Simple/virología , Herpes Simple/patología , Animales , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/patología , Progresión de la Enfermedad , Chlorocebus aethiops
11.
J Vet Intern Med ; 38(3): 1799-1807, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38609161

RESUMEN

BACKGROUND: Equine herpesvirus myeloencephalopathy (EHM) has severe impact on the sport horse population. OBJECTIVE: Study the influence of EHM on the likelihood of affected horses to return to their previous performance and investigate the association of clinical variables with prognosis. ANIMALS: Twenty-six horses positive for equine herpesvirus type 1 (EHV-1) were admitted to a veterinary teaching hospital (VTH) during a natural EHM outbreak at an international jumping event. METHODS: Data collected from the VTH, the International Equestrian Federation, and surveys completed by the riders and horse owners were retrospectively analyzed. RESULTS: Horses affected by EHM had 68% chance of returning to exercise, and 52.9% were able to achieve their preoutbreak performance level. Horses with an ataxia grade at admission ≥4/5 had an increased fatality rate (P < .05) and 10% chance of reaching their preoutbreak performance level. None of the horses with both vascular and urinary complications returned to their previous performance level. Finally, horses vaccinated against EHV-1 and those with urinary complications had a 71.4% and 43.7% fatality rate, respectively. CONCLUSIONS AND CLINICAL IMPORTANCE: Horses affected by EHM were able to return to their previous performance levels, but certain clinical variables were negatively associated with postoutbreak performance. Ataxia grade upon admission and the development of systemic signs of vasculitis and urinary complications were potential poor prognostic indicators in sport horses. Variables linked to fatality included prior vaccination against EHV-1, ataxia grade upon admission, and the development of urinary complications.


Asunto(s)
Ataxia , Infecciones por Herpesviridae , Herpesvirus Équido 1 , Enfermedades de los Caballos , Animales , Caballos , Enfermedades de los Caballos/virología , Ataxia/veterinaria , Ataxia/virología , Estudios Retrospectivos , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/complicaciones , Masculino , Femenino , Condicionamiento Físico Animal , Deportes
12.
BMC Infect Dis ; 24(1): 454, 2024 Apr 29.
Artículo en Inglés | MEDLINE | ID: mdl-38684967

RESUMEN

BACKGROUND: Clinically, most patients with Kaposi's sarcoma (KS) are male, and several direct and indirect mechanisms may underlie this increased susceptibility in men, Kaposi's sarcoma-associated herpesvirus (KSHV), also known as human herpesvirus 8 (HHV-8), is considered to be the primary etiological agent responsible for KS. Thus, we propose the hypothesis that men are more susceptible to HHV-8 infection, leading to a higher incidence of Kaposi's sarcoma among males. A meta-analysis was conducted to evaluate the association between gender and HHV-8 seropositivity in the general population. METHODS: A comprehensive literature search was performed using 6 online databases: PubMed, EMBASE, Cochrane library, Web of Science, CNKI, and Wanfang. Studies published before March 15, 2023, were included. RESULTS: In all, 33 articles including 41 studies were included in the meta-analysis. In the included adult population. men had a higher risk of HHV-8 infection than did women in adult populations from all over the world (odds ratio [OR]: 1.08, 95% confidence interval [CI]: 1.01-1.15), but no differences were found in child population from all over the world (OR: 0.90, 95% CI: 0.79-1.01). There was a significant difference in HHV-8 seroprevalence between men and women in sub-Saharan Africa (SSA) adult population (OR: 1.15, 95% CI: 1.05-1.26). However, no significant differences were observed in sub-Saharan Africa (SSA) child population (OR: 0.90, 95%CI 0.78-1.03). As for other continents, the results showed no significant difference, such as the Asian population (OR: 1.03, 95%CI: 0.92-1.16). or the European and American populations (OR 1.01, 95%CI 0.87-1.17). CONCLUSION: There was a slight gender disparity for HHV-8 infection in the adult population. Among the adult populations from SSA and globally, men were more likely to be infected with HHV-8 than were women. However, no statistical significance was observed in the child populations from SSA and globally. In the future, the inclusion of more standardized studies may strengthen the results of this study.


Asunto(s)
Infecciones por Herpesviridae , Herpesvirus Humano 8 , Sarcoma de Kaposi , Humanos , Masculino , Femenino , Infecciones por Herpesviridae/epidemiología , Infecciones por Herpesviridae/virología , Sarcoma de Kaposi/epidemiología , Sarcoma de Kaposi/virología , Factores Sexuales , Adulto , Incidencia , Factores de Riesgo , Niño
13.
Viruses ; 16(4)2024 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-38675960

RESUMEN

Reactivation and infection with cytomegalovirus (CMV) are frequently observed in recipients of solid organ transplants, bone marrow transplants, and individuals with HIV infection. This presents an increasing risk of allograft rejection, opportunistic infection, graft failure, and patient mortality. Among immunocompromised hosts, interstitial pneumonia is the most critical clinical manifestation of CMV infection. Recent studies have demonstrated the potential therapeutic benefits of exosomes derived from mesenchymal stem cells (MSC-exos) in preclinical models of acute lung injury, including pneumonia, ARDS, and sepsis. However, the role of MSC-exos in the pathogenesis of infectious viral diseases, such as CMV pneumonia, remains unclear. In a mouse model of murine CMV-induced pneumonia, we observed that intravenous administration of mouse MSC (mMSC)-exos reduced lung damage, decreased the hyperinflammatory response, and shifted macrophage polarization from the M1 to the M2 phenotype. Treatment with mMSC-exos also significantly reduced the infiltration of inflammatory cells and pulmonary fibrosis. Furthermore, in vitro studies revealed that mMSC-exos reversed the hyperinflammatory phenotype of bone marrow-derived macrophages infected with murine CMV. Mechanistically, mMSC-exos treatment decreased activation of the NF-κB/NLRP3 signaling pathway both in vivo and in vitro. In summary, our findings indicate that mMSC-exo treatment is effective in severe CMV pneumonia by reducing lung inflammation and fibrosis through the NF-κB/NLRP3 signaling pathway, thus providing promising therapeutic potential for clinical CMV infection.


Asunto(s)
Modelos Animales de Enfermedad , Exosomas , Células Madre Mesenquimatosas , Muromegalovirus , FN-kappa B , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Animales , Exosomas/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , FN-kappa B/metabolismo , Muromegalovirus/fisiología , Ratones Endogámicos C57BL , Macrófagos/inmunología , Infecciones por Citomegalovirus/terapia , Infecciones por Citomegalovirus/virología , Pulmón/virología , Pulmón/patología , Neumonía Viral/terapia , Neumonía Viral/virología , Infecciones por Herpesviridae/terapia , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/inmunología , Neumonía/terapia , Neumonía/virología
14.
Am J Vet Res ; 85(5)2024 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-38457927

RESUMEN

OBJECTIVE: Compare immune responses induced by 2 commercial intranasal (IN) modified-live viral (MLV) vaccines given individually or coadministered and evaluate prevention of infection and lung pathology following bovine herpesvirus-1 (BHV-1) challenge. ANIMALS: 36 male Holstein calves (ages, 5 to 12 days). METHODS: In a randomized complete block design, each calf received an IN injection of either vaccine diluent (Placebo), an MLV vaccine containing bovine herpesvirus-1 (BHV-1; N3), bovine coronavirus vaccine (BC), or both N3 and BC (BC + N3) with a booster 4 weeks later. Nasal secretions and blood were collected weekly. Three weeks after the booster, the calves were challenged with BHV-1, sampled for virus shedding, and euthanized 10 days later to quantify lung pathology. The study period was September 7, 2020, to April 6, 2021. RESULTS: Calves were seropositive for BHV-1 and BC before vaccination. No significant difference in BC-specific serum immunoglobin G and nasal immunoglobin A antibody responses in the BC versus BC + N3 group or BHV-1-specific serum immunoglobin G and nasal immunoglobin A antibody responses in the N3 versus BC + N3 group. Cytokine responses to BHV-1 and BC did not differ among groups. BHV-1 shedding after challenge was significantly reduced in N3 groups versus Placebo and BC. There was a significant reduction in lung pathology in the N3 + BC group versus Placebo. CLINICAL RELEVANCE: This study provides evidence an MLV vaccine containing BHV-1 and an MLV BC vaccine can be coadministered to neonatal calves without significantly altering immune responses to the 2 viruses or compromising the prevention of BHV-1 respiratory disease. Calves receiving the BC + N3 vaccine had a significant reduction in lung pathology after BHV-1 aerosol challenge.


Asunto(s)
Administración Intranasal , Animales Recién Nacidos , Enfermedades de los Bovinos , Infecciones por Coronavirus , Coronavirus Bovino , Infecciones por Herpesviridae , Herpesvirus Bovino 1 , Vacunas Atenuadas , Vacunas Virales , Animales , Bovinos , Herpesvirus Bovino 1/inmunología , Administración Intranasal/veterinaria , Masculino , Vacunas Virales/inmunología , Vacunas Virales/administración & dosificación , Vacunas Atenuadas/administración & dosificación , Vacunas Atenuadas/inmunología , Coronavirus Bovino/inmunología , Enfermedades de los Bovinos/prevención & control , Enfermedades de los Bovinos/virología , Enfermedades de los Bovinos/inmunología , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/virología , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/virología , Rinotraqueítis Infecciosa Bovina/prevención & control , Rinotraqueítis Infecciosa Bovina/inmunología , Esparcimiento de Virus , Anticuerpos Antivirales/sangre , Distribución Aleatoria
15.
J Neurovirol ; 30(1): 86-99, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38453879

RESUMEN

Simian varicella virus (SVV) produces peripheral inflammatory responses during varicella (primary infection) and zoster (reactivation) in rhesus macaques (RM). However, it is unclear if peripheral measures are accurate proxies for central nervous system (CNS) responses. Thus, we analyzed cytokine and Aß42/Aß40 changes in paired serum and cerebrospinal fluid (CSF) during the course of infection. During varicella and zoster, every RM had variable changes in serum and CSF cytokine and Aß42/Aß40 levels compared to pre-inoculation levels. Overall, peripheral infection appears to affect CNS cytokine and Aß42/Aß40 levels independent of serum responses, suggesting that peripheral disease may contribute to CNS disease.


Asunto(s)
Péptidos beta-Amiloides , Citocinas , Macaca mulatta , Animales , Péptidos beta-Amiloides/líquido cefalorraquídeo , Péptidos beta-Amiloides/sangre , Citocinas/líquido cefalorraquídeo , Citocinas/sangre , Activación Viral , Fragmentos de Péptidos/líquido cefalorraquídeo , Fragmentos de Péptidos/sangre , Varicellovirus/genética , Varicellovirus/inmunología , Herpesvirus Humano 3/patogenicidad , Herpesvirus Humano 3/inmunología , Infecciones por Herpesviridae/líquido cefalorraquídeo , Infecciones por Herpesviridae/virología , Infecciones por Herpesviridae/sangre , Infecciones por Herpesviridae/inmunología , Masculino , Herpes Zóster/líquido cefalorraquídeo , Herpes Zóster/virología , Herpes Zóster/sangre , Herpes Zóster/inmunología , Enfermedades de los Monos/virología , Enfermedades de los Monos/líquido cefalorraquídeo , Enfermedades de los Monos/sangre
16.
J Vet Intern Med ; 38(3): 1290-1299, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38497217

RESUMEN

Equine herpesvirus-1 (EHV-1) is a highly prevalent and frequently pathogenic infection of equids. The most serious clinical consequences of infection are abortion and equine herpesvirus myeloencephalopathy (EHM). The previous consensus statement was published in 2009 and considered pathogenesis, strain variation, epidemiology, diagnostic testing, vaccination, outbreak prevention and control, and treatment. A recent survey of American College of Veterinary Internal Medicine large animal diplomates identified the need for a revision to this original consensus statement. This updated consensus statement is underpinned by 4 systematic reviews that addressed key questions concerning vaccination, pharmaceutical treatment, pathogenesis, and diagnostic testing. Evidence for successful vaccination against, or effective treatment of EHV-1 infection was limited, and improvements in experimental design and reporting of results are needed in future studies of this important disease. This consensus statement also updates the topics considered previously in 2009.


Asunto(s)
Infecciones por Herpesviridae , Herpesvirus Équido 1 , Enfermedades de los Caballos , Animales , Caballos , Enfermedades de los Caballos/virología , Enfermedades de los Caballos/prevención & control , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/virología , Embarazo , Femenino
17.
J Virol ; 98(2): e0176423, 2024 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-38193709

RESUMEN

Herpes simplex virus-1 (HSV-1) establishes a latent infection in peripheral neurons and periodically reactivates to permit transmission, which can result in clinical manifestations. Viral transactivators required for lytic infection are largely absent during latent infection, and therefore, HSV-1 relies on the co-option of neuronal host signaling pathways to initiate its gene expression. The activation of the neuronal c-Jun N-terminal kinase (JNK) cell stress pathway is central to initiating biphasic reactivation in response to multiple stimuli. However, how host factors work with JNK to stimulate the initial wave of gene expression (known as Phase I) or the progression to full Phase II reactivation remains unclear. Here, we found that c-Jun, the primary target downstream of neuronal JNK cell stress signaling, functions during reactivation but not during the JNK-mediated initiation of Phase I gene expression. Instead, c-Jun was required to transition from Phase I to full HSV-1 reactivation and was detected in viral replication compartments of reactivating neurons. Interestingly, we also identified a role for both c-Jun and enhanced neuronal stress during initial neuronal infection in promoting a more reactivation-competent form of HSV-1 latency. Therefore, c-Jun functions at multiple stages during the HSV latent infection of neurons to promote reactivation but not during the initial JNK-dependent Phase I. Importantly, by demonstrating that initial infection conditions can contribute to later reactivation abilities, this study highlights the potential for latently infected neurons to maintain a molecular scar of previous exposure to neuronal stressors.IMPORTANCEThe molecular mechanisms that regulate the reactivation of herpes simplex virus-1 (HSV-1) from latent infection are unknown. The host transcription and pioneer factor c-Jun is the main target of the JNK cell stress pathway that is known to be important in exit of HSV from latency. Surprisingly, we found that c-Jun does not act with JNK during exit from latency but instead promotes the transition to full reactivation. Moreover, c-Jun and enhanced neuronal stress during initial neuronal infection promoted a more reactivation-competent form of HSV-1 latency. c-Jun, therefore, functions at multiple stages during HSV-1 latent infection of neurons to promote reactivation. Importantly, this study contributes to a growing body of evidence that de novo HSV-1 infection conditions can modulate latent infection and impact future reactivation events, raising important questions on the clinical impact of stress during initial HSV-1 acquisition on future reactivation events and consequences.


Asunto(s)
Herpes Simple , Herpesvirus Humano 1 , Infección Latente , Transducción de Señal , Humanos , Herpes Simple/metabolismo , Herpes Simple/virología , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología , Herpesvirus Humano 1/fisiología , Activación Viral , Latencia del Virus , Animales , Ratones
18.
J Virol ; 97(12): e0100823, 2023 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-37962378

RESUMEN

IMPORTANCE: The human gammaherpesviruses Epstein-Barr virus and Kaposi's sarcoma-associated herpesvirus are etiologic agents of numerous B cell lymphomas. A hallmark of gammaherpesvirus infection is their ability to establish lifelong latency in B cells. However, the specific mechanisms that mediate chronic infection in B cells in vivo remain elusive. Cellular E3 ubiquitin ligases regulate numerous biological processes by catalyzing ubiquitylation and modifying protein location, function, or half-life. Many viruses hijack host ubiquitin ligases to evade antiviral host defense and promote viral fitness. Here, we used the murine gammaherpesvirus 68 in vivo system to demonstrate that the E3 ligase Cul4b is essential for this virus to establish latency in germinal center B cells. These findings highlight an essential role for this E3 ligase in promoting chronic gammaherpesvirus infection in vivo and suggest that targeted inhibition of E3 ligases may provide a novel and effective intervention strategy against gammaherpesvirus-associated diseases.


Asunto(s)
Linfocitos B , Gammaherpesvirinae , Infecciones por Herpesviridae , Infección Persistente , Animales , Ratones , Linfocitos B/enzimología , Linfocitos B/metabolismo , Linfocitos B/virología , Proteínas Cullin/metabolismo , Gammaherpesvirinae/fisiología , Centro Germinal/citología , Centro Germinal/virología , Infecciones por Herpesviridae/enzimología , Infecciones por Herpesviridae/virología , Infección Persistente/enzimología , Infección Persistente/virología , Ubiquitinas/metabolismo , Latencia del Virus
19.
J Virol ; 97(11): e0132223, 2023 Nov 30.
Artículo en Inglés | MEDLINE | ID: mdl-37882519

RESUMEN

IMPORTANCE: Chickens immunized with the infectious laryngotracheitis chicken embryo origin (CEO) vaccine (Medivac, PT Medion Farma Jaya) experience adverse reactions, hindering its safety and effective use in poultry flocks. To improve the effect of the vaccine, we sought to find a strategy to alleviate the respiratory reactions associated with the vaccine. Here, we confirmed that co-administering the CEO vaccine with chIL-2 by oral delivery led to significant alleviation of the vaccine reactions in chickens after immunization. Furthermore, we found that the co-administration of chIL-2 with the CEO vaccine reduced the clinical signs of the CEO vaccine while enhancing natural killer cells and cytotoxic T lymphocyte response to decrease viral loads in their tissues, particularly in the trachea and conjunctiva. Importantly, we demonstrated that the chIL-2 treatment can ameliorate the replication of the CEO vaccine without compromising its effectiveness. This study provides new insights into further applications of chIL-2 and a promising strategy for alleviating the adverse reaction of vaccines.


Asunto(s)
Pollos , Infecciones por Herpesviridae , Herpesvirus Gallináceo 1 , Interleucina-2 , Células Asesinas Naturales , Linfocitos T Citotóxicos , Vacunas Virales , Animales , Administración Oral , Pollos/inmunología , Pollos/virología , Conjuntiva/virología , Infecciones por Herpesviridae/inmunología , Infecciones por Herpesviridae/prevención & control , Infecciones por Herpesviridae/veterinaria , Infecciones por Herpesviridae/virología , Herpesvirus Gallináceo 1/inmunología , Interleucina-2/administración & dosificación , Interleucina-2/inmunología , Células Asesinas Naturales/inmunología , Enfermedades de las Aves de Corral/inmunología , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/virología , Enfermedades Respiratorias/inmunología , Enfermedades Respiratorias/prevención & control , Enfermedades Respiratorias/veterinaria , Enfermedades Respiratorias/virología , Linfocitos T Citotóxicos/inmunología , Tráquea/virología , Carga Viral , Vacunas Virales/administración & dosificación , Vacunas Virales/efectos adversos , Vacunas Virales/biosíntesis , Vacunas Virales/inmunología
20.
J Virol ; 97(6): e0050623, 2023 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-37191529

RESUMEN

Oncogenic virus infections are estimated to cause ~15% of all cancers. Two prevalent human oncogenic viruses are members of the gammaherpesvirus family: Epstein-Barr virus (EBV) and Kaposi's sarcoma herpesvirus (KSHV). We use murine herpesvirus 68 (MHV-68), which shares significant homology with KSHV and EBV, as a model system to study gammaherpesvirus lytic replication. Viruses implement distinct metabolic programs to support their life cycle, such as increasing the supply of lipids, amino acids, and nucleotide materials necessary to replicate. Our data define the global changes in the host cell metabolome and lipidome during gammaherpesvirus lytic replication. Our metabolomics analysis found that MHV-68 lytic infection induces glycolysis, glutaminolysis, lipid metabolism, and nucleotide metabolism. We additionally observed an increase in glutamine consumption and glutamine dehydrogenase protein expression. While both glucose and glutamine starvation of host cells decreased viral titers, glutamine starvation led to a greater loss in virion production. Our lipidomics analysis revealed a peak in triacylglycerides early during infection and an increase in free fatty acids and diacylglyceride later in the viral life cycle. Furthermore, we observed an increase in the protein expression of multiple lipogenic enzymes during infection. Interestingly, pharmacological inhibitors of glycolysis or lipogenesis resulted in decreased infectious virus production. Taken together, these results illustrate the global alterations in host cell metabolism during lytic gammaherpesvirus infection, establish essential pathways for viral production, and recommend targeted mechanisms to block viral spread and treat viral induced tumors. IMPORTANCE Viruses are intracellular parasites which lack their own metabolism, so they must hijack host cell metabolic machinery in order to increase the production of energy, proteins, fats, and genetic material necessary to replicate. Using murine herpesvirus 68 (MHV-68) as a model system to understand how similar human gammaherpesviruses cause cancer, we profiled the metabolic changes that occur during lytic MHV-68 infection and replication. We found that MHV-68 infection of host cells increases glucose, glutamine, lipid, and nucleotide metabolic pathways. We also showed inhibition or starvation of glucose, glutamine, or lipid metabolic pathways results in an inhibition of virus production. Ultimately, targeting changes in host cell metabolism due to viral infection can be used to treat gammaherpesvirus-induced cancers and infections in humans.


Asunto(s)
Infecciones por Herpesviridae , Interacciones Microbiota-Huesped , Lipidómica , Metaboloma , Rhadinovirus , Replicación Viral , Animales , Ratones , Glucosa/metabolismo , Glutamina/metabolismo , Nucleótidos/metabolismo , Rhadinovirus/fisiología , Replicación Viral/fisiología , Ácidos Grasos/metabolismo , Infecciones por Herpesviridae/metabolismo , Infecciones por Herpesviridae/virología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA