Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
1.
Vet Anaesth Analg ; 50(3): 238-244, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-36781322

RESUMO

OBJECTIVE: To compare PaO2 and PaCO2 in horses recovering from general anesthesia maintained with either apneustic anesthesia ventilation (AAV) or conventional mechanical ventilation (CMV). STUDY DESIGN: Randomized, crossover design. ANIMALS: A total of 10 healthy adult horses from a university-owned herd. METHODS: Dorsally recumbent horses were anesthetized with isoflurane in oxygen [inspired oxygen fraction = 0.3 initially, with subsequent titration to maintain PaO2 ≥ 85 mmHg (11.3 kPa)] and ventilated with AAV or CMV according to predefined criteria [10 mL kg-1 tidal volume, PaCO2 40-45 mmHg (5.3-6.0 kPa) during CMV and < 60 mmHg (8.0 kPa) during AAV]. Horses were weaned from ventilation using a predefined protocol and transferred to a stall for unassisted recovery. Arterial blood samples were collected and analyzed at predefined time points. Tracheal oxygen insufflation at 15 L minute-1 was provided if PaO2 < 60 mmHg (8.0 kPa) on any analysis. Time to oxygen insufflation, first movement, sternal recumbency and standing were recorded. Data were analyzed using repeated measures anova, paired t tests and Fisher's exact test with significance defined as p < 0.05. RESULTS: Data from 10 horses were analyzed. Between modes, PaO2 was significantly higher immediately after weaning from ventilation and lower at sternal recumbency for AAV than for CMV. No PaCO2 differences were noted between ventilation modes. All horses ventilated with CMV required supplemental oxygen, whereas three horses ventilated with AAV did not. Time to first movement was shorter with AAV. Time to oxygen insufflation was not different between ventilation modes. CONCLUSIONS: Although horses ventilated with AAV entered the recovery period with higher PaO2, this advantage was not sustained during recovery. Whereas fewer horses required supplemental oxygen after AAV, the use of AAV does not preclude the need for routine supplemental oxygen administration in horses recovering from general anesthesia.


Assuntos
Infecções por Citomegalovirus , Doenças dos Cavalos , Cavalos , Animais , Respiração Artificial/veterinária , Respiração Artificial/métodos , Estudos Prospectivos , Anestesia Geral/veterinária , Oxigênio , Infecções por Citomegalovirus/veterinária
2.
Sci Rep ; 12(1): 21545, 2022 12 13.
Artigo em Inglês | MEDLINE | ID: mdl-36513687

RESUMO

Porcine cytomegalovirus (PCMV), that is actually a porcine roseolovirus (PRV), is a common herpesvirus in domestic pigs and wild boars. In xenotransplantation, PCMV/PRV has been shown to significantly reduce the survival time of pig kidneys and hearts in preclinical trials with different non-human primates. Furthermore, PCMV/PRV has been transmitted in the first pig to human heart xenotransplantation and contributed to the death of the patient. Although transmitted to the recipient, there is no evidence that PCMV/PRV can infect primate cells including human cells. PCMV/PRV is closely related to the human herpesviruses 6 and 7, and only distantly related to the human CMV (HCMV). Antiviral drugs used for the treatment of HCMV are less effective against PCMV/PRV. However, there are well described strategies to eliminate the virus from pig facilities. In order to detect the virus and to eliminate it, highly sensitive detection methods and the knowledge of how, where and when to screen the donor pigs is required. Here, a comparative testing of organs from pigs of different ages using polymerase chain reaction (PCR)-based and immunological methods was performed. Testing young piglets, PCMV/PRV was detected effectively by PCR in blood, bronchoalveolar lavage fluid, tonsils and heart. In adult animals, detection by PCR was not successful in most cases, because the virus load was below the detection limit or the virus was in its latent stage. Therefore, detection of antibodies against selected recombinant proteins corresponding to epitopes detected by nearly all infected animals in a Western blot assay is advantageous. By contrast, immunological testing is not beneficial in young animals as piglets might have PCMV/PRV-specific antibodies obtained from their infected mother via the colostrum. Using a thoughtful combination of PCR-based and immunological methods, detection of PCMV/PRV in donor pigs for xenotransplantation is feasible and a controlled elimination of the virus by early weaning or other methods is possible.


Assuntos
Infecções por Citomegalovirus , Doenças dos Suínos , Animais , Suínos , Humanos , Transplante Heterólogo , Citomegalovirus , Infecções por Citomegalovirus/diagnóstico , Infecções por Citomegalovirus/veterinária , Doadores de Tecidos , Primatas
3.
Xenotransplantation ; 29(5): e12775, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36082418

RESUMO

The potential for a donor-derived transmission of porcine cytomegalovirus/porcine roseolovirus (PCMV/PRV) to the recipient has been recognized since pigs were considered candidate donors for xenotransplantation. This review gives a short description of the viral properties and summarizes the current evidence of the effects of PCMV/PRV transmission in preclinical xenotransplantation. Despite evidence that PCMV/PRV does not infect human and non-human primate cells, activation in the transplanted organ and detrimental systemic complications have been described. As PCMV/PRV is a herpesvirus able to establish latency, the importance of adequate screening of donor pigs is emphasized, as no efficient treatment is available. Furthermore, easy and successful ways of elimination of PCMV/PRV from pig herds are indicated.


Assuntos
Infecções por Citomegalovirus , Roseolovirus , Animais , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/veterinária , Humanos , Primatas , Suínos , Doadores de Tecidos , Transplante Heterólogo
4.
Vet Anaesth Analg ; 49(4): 372-381, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35644741

RESUMO

OBJECTIVE: To compare the cardiopulmonary effects of apneustic anesthesia ventilation (AAV) and conventional mechanical ventilation (CMV) in dorsally recumbent anesthetized horses. STUDY DESIGN: Randomized, crossover design. ANIMALS: A total of 10 healthy adult horses from a university-owned herd. METHODS: Following xylazine, midazolam and ketamine administration, horses were orotracheally intubated and positioned in dorsal recumbency. Anesthesia was maintained with isoflurane in oxygen [inspired oxygen fraction (FiO2) = 0.3 initially, with subsequent titration to maintain PaO2 ≥ 85 mmHg (11.3 kPa)]. Horses were instrumented and ventilated with AAV or CMV for 1 hour according to predefined criteria [10 mL kg-1 tidal volume (VT), PaCO2 of 40-45 mmHg (5.3-6.0 kPa) during CMV and <60 mmHg (8.0 kPa) during AAV]. Dobutamine was administered to maintain mean arterial pressure (MAP) >65 mmHg. Cardiopulmonary data were collected at baseline, 30 and 60 minutes. The effects of ventilation mode and time were analyzed using repeated-measures anova with significance defined as p < 0.05. RESULTS: Data from nine horses were analyzed. A significant effect of mode at one or more time points was found for respiratory rate, arterial and end-tidal CO2 tensions, arterial pH, mean airway pressure (Paw), respiratory system dynamic compliance index (CrsI), venous admixture (Q˙s/Q˙t), mean pulmonary artery pressure and systemic vascular resistance. No significant differences between modes were found for VT, FiO2, PaO2, arterial hemoglobin saturation, alveolar dead space, heart rate, MAP, cardiac index, stroke volume index, oxygen delivery index, oxygen extraction ratio and dobutamine administration. CONCLUSIONS AND CLINICAL RELEVANCE: In dorsally recumbent anesthetized horses, both ventilation modes supported adequate oxygenation with minimal supplemental oxygen. Compared with CMV, AAV resulted in higher CrsI and lower Q˙s/Q˙t. Despite higher mean Paw with AAV, the cardiovascular effects of each mode were not different. Further trials of AAV in anesthetized horses are warranted.


Assuntos
Anestesia , Infecções por Citomegalovirus , Doenças dos Cavalos , Anestesia/veterinária , Animais , Infecções por Citomegalovirus/veterinária , Dobutamina , Cavalos/cirurgia , Humanos , Oxigênio , Respiração Artificial/métodos , Respiração Artificial/veterinária
5.
J Virol ; 96(3): e0165321, 2022 02 09.
Artigo em Inglês | MEDLINE | ID: mdl-34788083

RESUMO

Rhesus cytomegalovirus (RhCMV) infection of rhesus macaques (Macaca mulatta) is a valuable nonhuman primate model of human CMV (HCMV) persistence and pathogenesis. In vivo studies predominantly use tissue culture-adapted variants of RhCMV that contain multiple genetic mutations compared to wild-type (WT) RhCMV. In many studies, animals have been inoculated by nonnatural routes (e.g., subcutaneous, intravenous) that do not recapitulate disease progression via the normative route of mucosal exposure. Accordingly, the natural history of RhCMV would be more accurately reproduced by infecting macaques with strains of RhCMV that reflect the WT genome using natural routes of mucosal transmission. Here, we tested two WT-like RhCMV strains, UCD52 and UCD59, and demonstrated that systemic infection and frequent, high-titer viral shedding in bodily fluids occurred following oral inoculation. RhCMV disseminated to a broad range of tissues, including the central nervous system and reproductive organs. Commonly infected tissues included the thymus, spleen, lymph nodes, kidneys, bladder, and salivary glands. Histological examination revealed prominent nodular hyperplasia in spleens and variable levels of lymphoid lymphofollicular hyperplasia in lymph nodes. One of six inoculated animals had limited viral dissemination and shedding, with commensurately weak antibody responses to RhCMV antigens. These data suggest that long-term RhCMV infection parameters might be restricted by local innate factors and/or de novo host immune responses in a minority of primary infections. Together, we have established an oral RhCMV infection model that mimics natural HCMV infection. The virological and immunological parameters characterized in this study will greatly inform HCMV vaccine designs for human immunization. IMPORTANCE Human cytomegalovirus (HCMV) is globally ubiquitous with high seroprevalence rates in all communities. HCMV infections can occur vertically following mother-to-fetus transmission across the placenta and horizontally following shedding of virus in bodily fluids in HCMV-infected hosts and subsequent exposure of susceptible individuals to virus-laden fluids. Intrauterine HCMV has long been recognized as an infectious threat to fetal growth and development. Since vertical HCMV infections occur following horizontal HCMV transmission to the pregnant mother, the nonhuman primate model of HCMV pathogenesis was used to characterize the virological and immunological parameters of infection following primary mucosal exposures to rhesus cytomegalovirus.


Assuntos
Infecções por Citomegalovirus/veterinária , Citomegalovirus/fisiologia , Suscetibilidade a Doenças , Interações Hospedeiro-Patógeno , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , Animais , Anticorpos Neutralizantes/imunologia , Anticorpos Antivirais/imunologia , Biópsia , DNA Viral , Suscetibilidade a Doenças/imunologia , Interações Hospedeiro-Patógeno/imunologia , Imunoglobulina G/imunologia , Imuno-Histoquímica , Macaca mulatta , Doenças dos Macacos/patologia , Doenças dos Macacos/transmissão , Fases de Leitura Aberta , Especificidade de Órgãos , Carga Viral , Viremia , Eliminação de Partículas Virais
6.
Sci Immunol ; 6(57)2021 03 25.
Artigo em Inglês | MEDLINE | ID: mdl-33766849

RESUMO

Simian immunodeficiency virus (SIV) insert-expressing, 68-1 rhesus cytomegalovirus (RhCMV/SIV) vectors elicit major histocompatibility complex E (MHC-E)- and MHC-II-restricted, SIV-specific CD8+ T cell responses, but the basis of these unconventional responses and their contribution to demonstrated vaccine efficacy against SIV challenge in the rhesus monkeys (RMs) have not been characterized. We show that these unconventional responses resulted from a chance genetic rearrangement in 68-1 RhCMV that abrogated the function of eight distinct immunomodulatory gene products encoded in two RhCMV genomic regions (Rh157.5/Rh157.4 and Rh158-161), revealing three patterns of unconventional response inhibition. Differential repair of these genes with either RhCMV-derived or orthologous human CMV (HCMV)-derived sequences (UL128/UL130; UL146/UL147) leads to either of two distinct CD8+ T cell response types-MHC-Ia-restricted only or a mix of MHC-II- and MHC-Ia-restricted CD8+ T cells. Response magnitude and functional differentiation are similar to RhCMV 68-1, but neither alternative response type mediated protection against SIV challenge. These findings implicate MHC-E-restricted CD8+ T cell responses as mediators of anti-SIV efficacy and indicate that translation of RhCMV/SIV vector efficacy to humans will likely require deletion of all genes that inhibit these responses from the HCMV/HIV vector.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Reprogramação Celular/imunologia , Infecções por Citomegalovirus/veterinária , Citomegalovirus/imunologia , Doenças dos Macacos/prevenção & controle , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vacinas Virais/imunologia , Animais , Antígenos Virais/genética , Antígenos Virais/imunologia , Linfócitos T CD8-Positivos/metabolismo , Reprogramação Celular/genética , Engenharia Genética/métodos , Vetores Genéticos/genética , Imunogenicidade da Vacina , Memória Imunológica , Macaca mulatta , Doenças dos Macacos/imunologia , Doenças dos Macacos/virologia , Fases de Leitura Aberta/genética , Fases de Leitura Aberta/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/prevenção & controle , Vírus da Imunodeficiência Símia/imunologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Eficácia de Vacinas
7.
Comp Med ; 69(1): 55-62, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-30704552

RESUMO

Cytomegalovirus (CMV) is a common chronic herpesvirus found in humans and numerous other mammalian species. In people, chronic viruses like CMV can alter overall health and immunity and pose a serious risk for those with an inadequate immune system. In addition, CMV plays an important role in animal health, and could affect the health of vulnerable populations, like endangered species. Previous studies found a high rate of CMV seropositivity among adult baboons (Papio anubis), and results from our laboratory revealed that baboon CMV (BaCMV) seropositivity was correlated with altered immune cell populations. In the current study, we further characterized BaCMV infection in normal, adult baboons. Analysis of blood samples from baboons (age, 6 to 26 y) revealed a low overall prevalence of detectable of BaCMV DNA, with a higher detection rate in aged baboons (older than 15 y). Furthermore, data suggest that individual baboons maintain similar rates of recurrence and levels of BaCMV shedding in saliva over time. Finally, we evaluated multiple commercially available assays for antihuman CMV IgG and IgM for use with baboon sera. Results of this study will improve our understanding of BaCMV and may be directly relevant to other closely related species.


Assuntos
Infecções por Citomegalovirus/veterinária , Doenças dos Macacos/patologia , Papio anubis , Fatores Etários , Animais , Anticorpos Antivirais/sangue , Infecções por Citomegalovirus/sangue , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/patologia , DNA Viral/sangue , Doenças dos Macacos/sangue , Doenças dos Macacos/imunologia , Saliva/virologia , Eliminação de Partículas Virais
8.
J Gen Virol ; 99(11): 1509-1514, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30226462

RESUMO

Human cytomegalovirus (HCMV) colonizes blood-borne dendritic cells (DCs). They express US28, a viral G protein-coupled receptor (GPCR). In vitro functions have been described for US28, but how it contributes to host colonization has been unclear. The murine CMV (MCMV) M33 GPCR promotes DC recirculation. We show that US28 shares this function. Thus, DC recirculation is also available to HCMV via US28, and inhibiting US28 G protein-dependent signalling has the potential to reduce systemic infection. We show that M33 also promotes systemic infection through infected DC extravasation.


Assuntos
Movimento Celular , Infecções por Citomegalovirus/virologia , Citomegalovirus/patogenicidade , Células Dendríticas/virologia , Interações Hospedeiro-Patógeno , Linfonodos/virologia , Receptores de Quimiocinas/metabolismo , Proteínas Virais/metabolismo , Estruturas Animais/virologia , Animais , Células Cultivadas , Citomegalovirus/crescimento & desenvolvimento , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/veterinária , Células Dendríticas/imunologia , Humanos , Linfonodos/imunologia , Camundongos Endogâmicos BALB C , Muromegalovirus/crescimento & desenvolvimento
9.
Med Microbiol Immunol ; 207(5-6): 307-318, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-30032349

RESUMO

CD200 is a membrane protein that interacts with CD200R on the surface of immune cells and delivers an inhibitory signal. In this study, we characterized the distribution of inhibitory CD200R in rats. In addition, we investigated if e127, a homologue of rat CD200 expressed by rat cytomegalovirus (RCMV), can suppress immune functions in vitro. RT-PCR analysis was carried out to test the expression of CD200R in different rat tissues and flow cytometry was performed to characterize CD200R at the cellular level. To test the inhibitory functions of e127, a co-culture system was utilized in which immune cells were incubated with e127-expressing cells. The strongest CD200R expression was detected in lymphoid organs such as bone marrow and spleen. Flow cytometry analyses showed that CD200R+ cells were mainly CD4- dendritic cells (DC) and CD4+ T cells in the spleen. In blood, nearly all monocytes and granulocytes expressed CD200R and in bone marrow the NKRP1low subset of natural killer cells highly expressed CD200R. In addition, both peritoneal macrophages and the NR8383 macrophage cell line carried CD200R. At the functional level, viral e127 conferred an inhibitory signal on TNFα and IL6 cytokine release from IFNγ-stimulated macrophages. However, e127 did not affect the cytotoxic activity of DC. CD200R in the rat is mainly expressed on myeloid cells but also on non-myeloid cell subsets, and RCMV e127 can deliver inhibitory signals to immune cells by engaging CD200R. The RCMV model provides a useful tool to study potential immune evasion mechanisms of the herpesviridae and opens new avenues for understanding and controlling herpesvirus infections.


Assuntos
Interações Hospedeiro-Patógeno , Evasão da Resposta Imune , Muromegalovirus/fisiologia , Receptores Imunológicos/metabolismo , Proteínas Virais/metabolismo , Animais , Medula Óssea/patologia , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/veterinária , Citometria de Fluxo , Perfilação da Expressão Gênica , Leucócitos/imunologia , Mapeamento de Interação de Proteínas , Ratos Endogâmicos Lew , Reação em Cadeia da Polimerase em Tempo Real , Baço/patologia
10.
J Virol ; 92(13)2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29669841

RESUMO

Subclinical viral infections (SVI), including cytomegalovirus (CMV), are highly prevalent in humans, resulting in lifelong persistence. However, the impact of SVI on the interplay between the host immunity and gut microbiota in the context of environmental exposures is not well defined. We utilized the preclinical nonhuman primate (NHP) model consisting of SVI-free (specific-pathogen-free [SPF]) rhesus macaques and compared them to the animals with SVI (non-SPF) acquired through natural exposure and investigated the impact of SVI on immune cell distribution and function, as well as on gut microbiota. These changes were examined in animals housed in the outdoor environment compared to the controlled indoor environment. We report that SVI are associated with altered immune cell subsets and gut microbiota composition in animals housed in the outdoor environment. Non-SPF animals harbored a higher proportion of potential butyrate-producing Firmicutes and higher numbers of lymphocytes, effector T cells, and cytokine-producing T cells. Surprisingly, these differences diminished following their transfer to the controlled indoor environment, suggesting that non-SPFs had increased responsiveness to environmental exposures. An experimental infection of indoor SPF animals with CMV resulted in an increased abundance of butyrate-producing bacteria, validating that CMV enhanced colonization of butyrate-producing commensals. Finally, non-SPF animals displayed lower antibody responses to influenza vaccination compared to SPF animals. Our data show that subclinical CMV infection heightens host immunity and gut microbiota changes in response to environmental exposures. This may contribute to the heterogeneity in host immune response to vaccines and environmental stimuli at the population level.IMPORTANCE Humans harbor several latent viruses that modulate host immunity and commensal microbiota, thus introducing heterogeneity in their responses to pathogens, vaccines, and environmental exposures. Most of our understanding of the effect of CMV on the immune system is based on studies of children acquiring CMV or of immunocompromised humans with acute or reactivated CMV infection or in ageing individuals. The experimental mouse models are genetically inbred and are completely adapted to the indoor laboratory environment. In contrast, nonhuman primates are genetically outbred and are raised in the outdoor environment. Our study is the first to report the impact of long-term subclinical CMV infection on host immunity and gut microbiota, which is evident only in the outdoor environment but not in the indoor environment. The significance of this study is in highlighting the impact of SVI on enhancing host immune susceptibility to environmental exposures and immune heterogeneity.


Assuntos
Bactérias/classificação , Infecções por Citomegalovirus/veterinária , Citomegalovirus/patogenicidade , Doenças dos Macacos/imunologia , Doenças dos Macacos/microbiologia , Animais , Bactérias/isolamento & purificação , Citocinas/metabolismo , Infecções por Citomegalovirus/imunologia , Modelos Animais de Doenças , Microbioma Gastrointestinal , Abrigo para Animais , Linfócitos/metabolismo , Macaca mulatta , Filogenia , Organismos Livres de Patógenos Específicos , Linfócitos T/imunologia
11.
Vet Immunol Immunopathol ; 197: 58-62, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29475507

RESUMO

Porcine cytomegalovirus (PCMV) causes lifelong latent infections in swine. The pathogen is occasionally associated with inclusion body rhinitis and pneumonia in piglets, reproductive disorders in pregnant sows and respiratory disease complex in older pigs. Immunosuppressive potential of PCMV infection is discussed. Macrophages were recognised as one of target cell types where propagation of virus occurs. The aim of present study was to set up model PCMV infection of monocyte derived macrophages (MDMs) in vitro for PCMV immunobiology research. Obtained results showed that PCMV is able to infect and propagate in MDMs. Possible immunosuppressive effect of PCMV on infected macrophages was evaluated by measurement of immune relevant gene expression in MDMs. Infection decreased expression of IL-8 and TNF-α (pro-inflammatory cytokines) and increased expression of IL-10 (anti-inflammatory cytokine) on mRNA transcription level. Obtained data support hypothesis that higher sensitivity of animals to coinfection with other swine pathogens and its more severe clinical manifestations could potentially be the consequence of PCMV infection.


Assuntos
Citocinas/imunologia , Infecções por Citomegalovirus/veterinária , Macrófagos/imunologia , Macrófagos/virologia , Doenças dos Suínos/virologia , Animais , Citomegalovirus/fisiologia , Infecções por Citomegalovirus/imunologia , Expressão Gênica/imunologia , Imunidade Inata , Interleucina-10/imunologia , Interleucina-8/imunologia , Suínos , Doenças dos Suínos/imunologia , Fator de Necrose Tumoral alfa/imunologia
12.
Viruses ; 10(2)2018 02 06.
Artigo em Inglês | MEDLINE | ID: mdl-29415517

RESUMO

The porcine cytomegalovirus (PCMV) is a herpesvirus that may pose a risk for xenotransplantation using pig cells, tissues, or organs. Here, three orthotopic pig heart transplantations into baboons were studied. To detect PCMV, a real-time PCR and a Western blot assay based on four PCMV protein sequences, including two tegument proteins, were used. The transmission of PCMV from the donor pig to the recipient baboon was found in two cases, despite PCMV not being detected in the blood of the donor pigs by real-time PCR. Although it was not in the blood, PCMV was detected in different organs of the donor pigs, and in sibling animals. Immunohistochemistry using an antiserum that is specific for PCMV detected virus protein-expressing cells in all of the organs of the recipient baboon, most likely representing disseminated pig cells. Therefore, for the first time, the distribution of PCMV in organs of the donor pigs and the recipient baboons was described. In addition, baboon cytomegalovirus (BaCMV) was found activated in the recipient, and a screening for hepatitis E virus (HEV) and porcine lymphotropic herpesviruses (PLHV) was performed. For the first time, a cross-reactivity between antibodies directed against PCMV and BaCMV was found.


Assuntos
Infecções por Citomegalovirus/veterinária , Citomegalovirus/genética , Transplante de Coração/efeitos adversos , Doenças dos Macacos/virologia , Doenças dos Suínos/virologia , Doadores de Tecidos , Transplantados , Animais , Animais Geneticamente Modificados , Citomegalovirus/imunologia , DNA Viral , Feminino , Humanos , Leucócitos Mononucleares/metabolismo , Masculino , Doenças dos Macacos/imunologia , Especificidade de Órgãos , Papio , Prevalência , Suínos , Doenças dos Suínos/imunologia , Carga Viral
13.
Virol J ; 15(1): 16, 2018 01 18.
Artigo em Inglês | MEDLINE | ID: mdl-29347945

RESUMO

BACKGROUND: Porcine cytomegalovirus (PCMV) is an immunosuppressive virus that mainly inhibits T-lymphocyte and macrophage immune functions; it has significantly damaged the farming industry. Although recent studies have shown that miRNAs play important roles in immune responses, the regulatory mechanisms of miRNAs during immunosuppressive virus infection remain unclear. METHODS: In this study, porcine small-RNA transcriptomes of PCMV-infected and uninfected vital organs were first characterised by high-throughput sequencing. miRDeep2 software was used to predict novel pig-encoded miRNAs. To verify the accuracy of the high-throughput sequencing results, stem-loop qRT-PCR was performed on 12 significantly DE miRNAs. The physical and functional interactions between the immune-related target genes of the DE miRNAs in PCMV-infected organs were analysed using the STRING database. RESULTS: In total, 306 annotated and 295 novel miRNAs were identified from PCMV-infected and uninfected porcine organs, respectively, through alignment with known Sus scrofa pre-miRNAs. Overall, 92, 107, 95, 77 and 111 miRNAs were significantly differentially expressed in lung, liver, spleen, kidney and thymus after PCMV infection, respectively. According to Gene Ontology enrichment analysis, target genes of the differentially expressed miRNAs associated with immune system processes, regulation of biological processes and metabolic processes were enriched in every sample. Integrated expression analysis of the differentially expressed miRNAs and their target mRNAs in PCMV-infected thymus showed that the significant differential expression of specific miRNAs under the pressure of PCMV infection in central immune organs interfered with the expression of genes involved in important immune-related signalling pathways, thus promoting the viral infection. CONCLUSIONS: This is the first comprehensive analysis of the responses of host small-RNA transcriptomes to PCMV infection in vital porcine organs. It provides new insights into the regulatory mechanisms of miRNAs during infection by immunosuppressive viruses.


Assuntos
Infecções por Citomegalovirus/veterinária , Citomegalovirus , Interações Hospedeiro-Patógeno/genética , MicroRNAs/genética , Doenças dos Suínos/genética , Doenças dos Suínos/virologia , Transcriptoma , Animais , Biologia Computacional/métodos , Citomegalovirus/imunologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Redes Reguladoras de Genes , Sequenciamento de Nucleotídeos em Larga Escala , Hospedeiro Imunocomprometido , Anotação de Sequência Molecular , Especificidade de Órgãos/genética , Especificidade de Órgãos/imunologia , Interferência de RNA , Reprodutibilidade dos Testes , Análise de Sequência de DNA , Suínos , Doenças dos Suínos/imunologia , Doenças dos Suínos/patologia
14.
Viruses ; 10(1)2017 12 29.
Artigo em Inglês | MEDLINE | ID: mdl-29286318

RESUMO

Cytomegaloviruses (CMVs) are known to infect many mammals, including a number of nonhuman primates (NHPs). However, most data available arose from studies led on captive individuals and little is known about CMV diversity in wild NHPs. Here, we analyzed a community of wild nonhuman primates (seven species) in Taï National Park (TNP), Côte d'Ivoire, with two PCR systems targeting betaherpesviruses. CMV DNA was detected in 17/87 primates (4/7 species). Six novel CMVs were identified in sooty mangabeys, Campbell's monkeys and Diana monkeys, respectively. In 3/17 positive individuals (from three NHP species), different CMVs were co-detected. A major part of the glycoprotein B coding sequences of the novel viruses was amplified and sequenced, and phylogenetic analyses were performed that included three previously discovered CMVs of western red colobus from TNP and published CMVs from other NHP species and geographic locations. We find that, despite this locally intensified sampling, NHP CMVs from TNP are completely host-specific, pinpointing the absence or rarity of cross-species transmission. We also show that on longer timescales the evolution of CMVs is characterized by frequent co-divergence with their hosts, although other processes, including lineage duplication and host switching, also have to be invoked to fully explain their evolutionary relationships.


Assuntos
Animais Selvagens/virologia , Infecções por Citomegalovirus/veterinária , Citomegalovirus/classificação , Citomegalovirus/genética , Doenças dos Primatas/virologia , Primatas/virologia , Sequência de Aminoácidos , Animais , Sequência de Bases , Coevolução Biológica , Coinfecção , Côte d'Ivoire , Infecções por Citomegalovirus/virologia , DNA Viral/genética , Variação Genética , Especificidade de Hospedeiro , Parques Recreativos , Filogenia , Doenças dos Primatas/epidemiologia , Proteínas do Envelope Viral/genética
15.
J Comp Pathol ; 157(4): 299-302, 2017 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29169627

RESUMO

Cytomegalic inclusion disease (CID) in the salivary gland of African hedgehogs (Atelerix arbiventris) has been reported before, and is suspected to reflect a cytomegalovirus infection. However, a recent ultrastructural study reported that African hedgehog CID reflected oncocytic metaplasia, mimicking a cytomegalovirus infection. We examined the submandibular and sublingual salivary glands of a 1-year-old male African hedgehog. Histologically, there were multiple foci composed of cytomegalic cells with intranuclear inclusion bodies. Ultrastructurally, viral particles (109-118 nm in diameter) were observed in the nuclei of the cytomegalic cells. There were numerous vesicles containing various numbers of enveloped viruses in the cytoplasm. We also attempted to detect viral DNA fragments by degenerate polymerase chain reaction and obtained amplicons of a predicted size. Phylogenetic analysis indicated that the virus is a betaherpesvirus, comparatively related to human and rodent cytomegaloviruses. The present study suggested that African hedgehog CIDs also include those caused by the cytomegalovirus.


Assuntos
Infecções por Citomegalovirus/veterinária , Ouriços , Animais , Masculino
17.
Viruses ; 9(1)2017 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-28085106

RESUMO

Shortage of human organs forced the development of xenotransplantation using cells, tissues, and organs from pigs. Xenotransplantation may be associated with the transmission of porcine zoonotic microorganisms, among them the porcine cytomegalovirus (PCMV). To prevent virus transmission, pigs have to be screened using sensitive methods. In order to perform regular follow-ups and further breeding of the animals, samples for testing should be collected by low-invasive or non-invasive methods. Sera, ear biopsies, as well as oral and anal swabs were collected from ten 10-day-old Aachen minipigs (AaMP) and tested for PCMV using sensitive nested polymerase chain reaction (PCR) as well as uniplex and duplex real-time PCR. Porcine cytomegalovirus DNA was detected most frequently in oral and anal swabs. Comparison of duplex and uniplex real-time PCR systems for PCMV detection demonstrated a lower sensitivity of duplex real-time PCR when the copy numbers of the target genes were low (less 200). Therefore, to increase the efficacy of PCMV detection in piglets, early testing of oral and anal swabs by uniplex real-time PCR is recommended.


Assuntos
Infecções por Citomegalovirus/veterinária , Citomegalovirus/isolamento & purificação , Reação em Cadeia da Polimerase/métodos , Manejo de Espécimes/métodos , Doenças dos Suínos/diagnóstico , Doenças dos Suínos/virologia , Animais , Estudos de Coortes , Citomegalovirus/genética , Infecções por Citomegalovirus/virologia , Suínos
18.
J Infect Dev Ctries ; 10(11): 1268-1270, 2016 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-27886042

RESUMO

INTRODUCTION: Porcine cytomegalovirus (PCMV) causes rhinitis in both young and older pigs. The present study describes the detection and characterization of shedding profiles of PCMV in nine farrow-to-finish Brazilian swine herds. METHODOLOGY: Tonsil swabs from sows, nursery and grow-finish pigs of nine farrow-to-finish commercial herds (n = 756) were evaluated for the presence of PCMV by PCR. RESULTS: The virus was detected in all herds. Positive samples were concentrated in piglets of ages varying from 40 to 60 days (nursery phase), while none of the sows were positive for PCMV detection. CONCLUSIONS: These findings corroborate the literature regarding PCMV worldwide distribution, and introduce the first report of PCMV shedding profile in Brazilian pig farms.


Assuntos
Infecções por Citomegalovirus/veterinária , Citomegalovirus/isolamento & purificação , Doenças dos Suínos/epidemiologia , Doenças dos Suínos/virologia , Eliminação de Partículas Virais , Animais , Brasil/epidemiologia , Infecções por Citomegalovirus/epidemiologia , Infecções por Citomegalovirus/virologia , Tonsila Palatina/virologia , Suínos
19.
J Gen Virol ; 97(11): 2957-2972, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27638684

RESUMO

Cytomegaloviruses (CMVs) produce chemokines (vCXCLs) that have both sequence and functional homology to host chemokines. Assessment of vCXCL-1's role in CMV infection is limited to in vitro and in silico analysis due to CMVs species specificity. In this study, we used the murine CMV (MCMV) mouse model to evaluate the function of vCXCL-1 in vivo. Recombinant MCMVs expressing chimpanzee CMV vCXCL-1 (vCXCL-1CCMV) or host chemokine, mCXCL1, underwent primary dissemination to the popliteal lymph node, spleen and lung similar to the parental MCMV. However, neither of the recombinants expressing chemokines was recovered from the salivary gland (SG) at any time post-infection although viral DNA was detected. This implies that the virus does not grow in the SG or the overexpressed chemokine induces an immune response that leads to suppressed growth. Pointing to immune suppression of virus replication, recombinant viruses were isolated from the SG following infection of immune-ablated mice [i.e. SCID (severe combined immunodeficiency), NSG (non-obese diabetic SCID gamma) or cyclophosphamide treated]. Depletion of neutrophils or NK cells does not rescue the recovery of chemokine-expressing recombinants in the SG. Surprisingly we found that co-infection of parental virus and chemokine-expressing virus leads to the recovery of the recombinants in the SG. We suggest that parental virus reduces the levels of chemokine expression leading to a decrease in inflammatory monocytes and subsequent SG growth. Therefore, aberrant expression of the chemokines induces cells of the innate and adaptive immune system that curtail the growth and dissemination of the recombinants in the SG.


Assuntos
Quimiocinas CXC/imunologia , Infecções por Citomegalovirus/veterinária , Muromegalovirus/imunologia , Glândulas Salivares/virologia , Proteínas Virais/imunologia , Imunidade Adaptativa , Animais , Quimiocinas CXC/genética , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Interações Hospedeiro-Patógeno , Imunidade Inata , Camundongos , Camundongos SCID , Muromegalovirus/genética , Pan troglodytes , Glândulas Salivares/imunologia , Proteínas Virais/genética
20.
mBio ; 7(2): e00251-16, 2016 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-27118588

RESUMO

UNLABELLED: Viruses transmit via the environmental and social interactions of their hosts. Herpesviruses have colonized mammals since their earliest origins, suggesting that they exploit ancient, common pathways. Cytomegaloviruses (CMVs) are assumed to enter new hosts orally, but no site has been identified. We show by live imaging that murine CMV (MCMV) infects nasally rather than orally, both after experimental virus uptake and during natural transmission. Replication-deficient virions revealed the primary target as olfactory neurons. Local, nasal replication by wild-type MCMV was not extensive, but there was rapid systemic spread, associated with macrophage infection. A long-term, transmissible infection was then maintained in the salivary glands. The viral m131/m129 chemokine homolog, which influences tropism, promoted salivary gland colonization after nasal entry but was not required for entry per se The capacity of MCMV to transmit via olfaction, together with previous demonstrations of experimental olfactory infection by murid herpesvirus 4 (MuHV-4) and herpes simplex virus 1 (HSV-1), suggest that this is a common, conserved route of mammalian herpesvirus entry. IMPORTANCE: Cytomegaloviruses (CMVs) infect most mammals. Human CMV (HCMV) harms people with poor immune function and can damage the unborn fetus. It infects approximately 1% of live births. We lack a good vaccine. One problem is that how CMVs first enter new hosts remains unclear. Oral entry is often assumed, but the evidence is indirect, and no infection site is known. The difficulty of analyzing HCMV makes related animal viruses an important source of insights. Murine CMV (MCMV) infected not orally but nasally. Specifically, it targeted olfactory neurons. Viral transmission was also a nasal infection. Like HCMV, MCMV infected cells by binding to heparan, and olfactory surfaces display heparan to incoming viruses, whereas most other mucosal surfaces do not. These data establish a new understanding of CMV infections and a basis for infection control.


Assuntos
Infecções por Citomegalovirus/veterinária , Muromegalovirus/fisiologia , Nariz/virologia , Doenças dos Roedores/virologia , Animais , Infecções por Citomegalovirus/virologia , Humanos , Camundongos , Muromegalovirus/genética , Glândulas Salivares/virologia , Olfato , Internalização do Vírus
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...