Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(9)2021 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-33946994

RESUMO

The gastrointestinal lumen is a rich source of eukaryotic and prokaryotic viruses which, together with bacteria, fungi and other microorganisms comprise the gut microbiota. Pathogenic viruses inhabiting this niche have the potential to induce local as well as systemic complications; among them, the viral ability to disrupt the mucosal barrier is one mechanism associated with the promotion of diarrhea and tissue invasion. This review gathers recent evidence showing the contributing effects of diet, gut microbiota and the enteric nervous system to either support or impair the mucosal barrier in the context of viral attack.


Assuntos
Bacteriófagos/fisiologia , Dieta , Sistema Nervoso Entérico/fisiologia , Mucosa Gástrica/virologia , Microbioma Gastrointestinal , Interações entre Hospedeiro e Microrganismos/fisiologia , Mucosa Intestinal/virologia , Vírus , Defensinas/fisiologia , Digestão , Suscetibilidade a Doenças , Sistema Nervoso Entérico/virologia , Alimentos/virologia , Mucosa Gástrica/imunologia , Mucosa Gástrica/inervação , Mucosa Gástrica/metabolismo , Gastroenterite/virologia , Interações entre Hospedeiro e Microrganismos/imunologia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/inervação , Mucosa Intestinal/metabolismo , Desnutrição/virologia , Muco/metabolismo , Muco/virologia , Neurônios/virologia , Infecções Oportunistas/virologia , Vírus de Plantas , Viroses/microbiologia , Viroses/fisiopatologia
3.
Neurogastroenterol Motil ; 33(3): e14104, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33591607

RESUMO

BACKGROUND: Coronavirus disease 2019 (COVID-19) is associated with gastrointestinal and hepatic manifestation in up to one fifth of patients. Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent of COVID-19, infects gastrointestinal epithelial cells expressing angiotensin-converting enzyme 2 (ACE2) receptors triggering a cascade of events leading to mucosal and systemic inflammation. Symptomatic patients display changes in gut microbiota composition and function which may contribute to intestinal barrier dysfunction and immune activation. Evidence suggests that SARS-CoV-2 infection and related mucosal inflammation impact on the function of the enteric nervous system and the activation of sensory fibers conveying information to the central nervous system, which, may at least in part, contribute symptom generation such as vomiting and diarrhea described in COVID-19. Liver and pancreas dysfunctions have also been described as non-respiratory complications of COVID-19 and add further emphasis to the common view of SARS-CoV-2 infection as a systemic disease with multiorgan involvement. PURPOSE: The aim of this review was to highlight the current knowledge on the pathophysiology of gastrointestinal SARS-CoV-2 infection, including the crosstalk with the gut microbiota, the fecal-oral route of virus transmission, and the potential interaction of the virus with the enteric nervous system. We also review the current available data on gastrointestinal and liver manifestations, management, and outcomes of patients with COVID-19.


Assuntos
COVID-19/complicações , COVID-19/fisiopatologia , Gastroenteropatias/etiologia , Gastroenteropatias/fisiopatologia , Microbioma Gastrointestinal/fisiologia , Trato Gastrointestinal/fisiopatologia , Animais , Diarreia/etiologia , Diarreia/fisiopatologia , Diarreia/virologia , Disbiose/etiologia , Disbiose/fisiopatologia , Disbiose/virologia , Sistema Nervoso Entérico/fisiopatologia , Sistema Nervoso Entérico/virologia , Gastroenteropatias/virologia , Trato Gastrointestinal/virologia , Humanos , Hepatopatias/etiologia , Hepatopatias/fisiopatologia , Hepatopatias/virologia , Pancreatopatias/etiologia , Pancreatopatias/fisiopatologia , Pancreatopatias/virologia
4.
Neurogastroenterol Motil ; 33(8): e14014, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33094876

RESUMO

BACKGROUND: Intravenous administration of adeno-associated virus (AAV) can be used as a noninvasive approach to trace neuronal morphology and links. AAV-PHP.S is a variant of AAV9 that effectively transduces the peripheral nervous system. The objective was to label randomly and sparsely enteric plexus in the mouse colon using AAV-PHP.S with a tunable two-component multicolor vector system and digitally trace individual neurons and nerve fibers within microcircuits in three dimensions (3D). METHODS: A vector system including a tetracycline inducer with a tet-responsive element driving three separate fluorophores was packaged in the AAV-PHP.S capsid. The vectors were injected retro-orbitally in mice, and the colon was harvested 3 weeks after. Confocal microscopic images of enteric plexus were digitally segmented and traced in 3D using Neurolucida 360, neuTube, or Imaris software. KEY RESULTS: The transduction of multicolor AAV vectors induced random sparse spectral labeling of soma and neurites primarily in the myenteric plexus of the proximal colon, while neurons in the submucosal plexus were occasionally transduced. Digital tracing in 3D showed various types of wiring, including multiple conjunctions of one neuron with other neurons, neurites en route, and endings; clusters of neurons in close apposition between each other; axon-axon parallel conjunctions; and intraganglionic nerve endings consisting of multiple nerve endings and passing fibers. Most of digitally traced neuronal somas were of small or medium in size. CONCLUSIONS & INFERENCES: The multicolor AAV-PHP.S-packaged vectors enabled random sparse spectral labeling and revealed complexities of enteric microcircuit in the mouse proximal colon. The techniques can facilitate digital modeling of enteric micro-circuitry.


Assuntos
Colo/metabolismo , Sistema Nervoso Entérico/metabolismo , Plexo Submucoso/metabolismo , Animais , Colo/inervação , Dependovirus , Sistema Nervoso Entérico/virologia , Feminino , Técnicas de Transferência de Genes , Proteínas de Fluorescência Verde , Masculino , Camundongos , Plexo Submucoso/virologia
5.
J Infect Dis ; 218(suppl_2): S113-S119, 2018 09 22.
Artigo em Inglês | MEDLINE | ID: mdl-30247599

RESUMO

Varicella zoster virus (VZV) infects and becomes latent in sensory, enteric, and other autonomic neurons during the viremia of varicella. Reactivation of VZV in neurons that project to the skin causes the rash of zoster; however, reactivation of VZV in enteric neurons can cause a painful gastrointestinal disorder ("enteric zoster") without cutaneous manifestations. Detection of VZV DNA in saliva of patients with gastrointestinal symptoms may suggest enteric zoster. This diagnosis is reinforced by observing a response to antiviral therapy and can be confirmed by detecting VZV gene products in intestinal mucosal biopsies. We developed an in vivo guinea pig model that may be useful in studies of VZV latency and reactivation. VZV-infected lymphocytes are used to induce latent infection in sensory and enteric neurons; evidence suggests that exosomes and stimulator of interferon genes (STING) may, by preventing proliferation play roles in the establishment of neuronal latency.


Assuntos
Sistema Nervoso Entérico/virologia , Gastroenteropatias/virologia , Herpesvirus Humano 3/fisiologia , Leucócitos Mononucleares/virologia , Neurônios/virologia , Animais , Sistema Nervoso Entérico/citologia , Cobaias , Humanos , Leucócitos Mononucleares/fisiologia , Ativação Viral , Latência Viral
6.
Viruses ; 10(4)2018 03 28.
Artigo em Inglês | MEDLINE | ID: mdl-29597335

RESUMO

The pathogenesis of enteric zoster, a rare debilitating complication of reactivation of latent varicella-zoster virus (VZV) in the enteric nervous system (ENS), is largely unknown. Infection of monkeys with the closely related Varicellovirus simian varicella virus (SVV) mimics VZV disease in humans. In this study, we determined the applicability of the SVV nonhuman primate model to study Varicellovirus infection of the ENS. We confirmed VZV infection of the gut in latently infected adults and demonstrated that SVV DNA was similarly present in gut of monkeys latently infected with SVV using quantitative real-time PCR. In situ analyses showed that enteric neurons expressed SVV open reading frame (ORF) 63 RNA, but not viral nucleocapsid proteins, suggestive of latent ENS infection. During primary infection, SVV-infected T-cells were detected in gut-draining mesenteric lymph nodes and located in close vicinity to enteric nerves in the gut. Furthermore, flow cytometric analysis of blood from acutely SVV-infected monkeys demonstrated that virus-infected T-cells expressed the gut-homing receptor α4ß7 integrin. Collectively, the data demonstrate that SVV infects ENS neurons during primary infection and supports the role of T-cells in virus dissemination to the gut. Because SVV reactivation can be experimentally induced, the SVV nonhuman primate model holds great potential to study the pathogenesis of enteric zoster.


Assuntos
Expressão Gênica , Integrinas/genética , Neurônios/metabolismo , Neurônios/virologia , Linfócitos T/fisiologia , Linfócitos T/virologia , Varicellovirus/fisiologia , Adulto , Idoso , Animais , Biomarcadores , Biópsia , Sistema Nervoso Entérico/virologia , Feminino , Imunofluorescência , Infecções por Herpesviridae/veterinária , Herpesvirus Humano 3/fisiologia , Humanos , Integrinas/metabolismo , Linfonodos/imunologia , Linfonodos/metabolismo , Linfonodos/patologia , Linfonodos/virologia , Macaca mulatta , Masculino , Pessoa de Meia-Idade , Doenças dos Macacos/genética , Doenças dos Macacos/imunologia , Doenças dos Macacos/patologia , Doenças dos Macacos/virologia , Nódulos Linfáticos Agregados/virologia , Infecção pelo Vírus da Varicela-Zoster/genética , Infecção pelo Vírus da Varicela-Zoster/imunologia , Infecção pelo Vírus da Varicela-Zoster/patologia , Infecção pelo Vírus da Varicela-Zoster/virologia , Carga Viral
7.
Artigo em Inglês | MEDLINE | ID: mdl-29600197

RESUMO

Herpes Simplex Virus type 1 (HSV-1), a neurotropic pathogen widespread in human population, infects the enteric nervous system (ENS) in humans and rodents and causes intestinal neuromuscular dysfunction in rats. Although infiltration of inflammatory cells in the myenteric plexus and neurodegeneration of enteric nerves are common features of patients suffering from functional intestinal disorders, the proof of a pathogenic link with HSV-1 is still unsettled mainly because the underlying mechanisms are largely unknown. In this study we demonstrated that following intragastrical administration HSV-1 infects neurons within the myenteric plexus resulting in functional and structural alterations of the ENS. By infecting mice with HSV-1 replication-defective strain we revealed that gastrointestinal neuromuscular anomalies were however independent of viral replication. Indeed, enteric neurons exposed to UV-inactivated HSV-1 produced monocyte chemoattractant protein-1 (MCP-1/CCL2) to recruit activated macrophages in the longitudinal muscle myenteric plexus. Infiltrating macrophages produced reactive oxygen and nitrogen species and directly harmed enteric neurons resulting in gastrointestinal dysmotility. In HSV-1 infected mice intestinal neuromuscular dysfunctions were ameliorated by in vivo administration of (i) liposomes containing dichloromethylene bisphosphonic acid (clodronate) to deplete tissue macrophages, (ii) CCR2 chemokine receptor antagonist RS504393 to block the CCL2/CCR2 pathway, (iii) Nω-Nitro-L-arginine methyl ester hydrochloride (L-NAME) and AR-C 102222 to quench production of nitrogen reactive species produced via iNOS. Overall these data demonstrate that HSV-1 infection makes enteric neurons recruit macrophages via production of a specific chemoattractant factor. The resulting inflammatory reaction is mandatory for intestinal dysmotility. These findings provide insights into the neuro-immune communication that occurs in the ENS following HSV-1 infection and allow recognition of an original pathophysiologic mechanism underlying gastrointestinal diseases as well as identification of novel therapeutic targets.


Assuntos
Sistema Nervoso Entérico/efeitos dos fármacos , Sistema Nervoso Entérico/virologia , Motilidade Gastrointestinal/efeitos dos fármacos , Herpes Simples/metabolismo , Herpesvirus Humano 1/patogenicidade , Macrófagos/metabolismo , Neurônios/efeitos dos fármacos , Imunidade Adaptativa , Animais , Arginina/análogos & derivados , Arginina/metabolismo , Quimiocina CCL2/metabolismo , Ácido Clodrônico , Modelos Animais de Doenças , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/patologia , Herpes Simples/imunologia , Herpes Simples/patologia , Herpes Simples/virologia , Íleo/imunologia , Íleo/patologia , Íleo/virologia , Inflamação/metabolismo , Lipossomos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Plexo Mientérico/efeitos dos fármacos , Plexo Mientérico/metabolismo , Plexo Mientérico/patologia , Plexo Mientérico/virologia , NG-Nitroarginina Metil Éster/metabolismo , Neurônios/virologia , Ratos , Espécies Reativas de Nitrogênio/metabolismo , Espécies Reativas de Nitrogênio/toxicidade , Espécies Reativas de Oxigênio/metabolismo , Espécies Reativas de Oxigênio/toxicidade , Receptores de Quimiocinas , Internalização do Vírus , Replicação Viral
8.
PLoS One ; 12(7): e0181863, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28732069

RESUMO

BACKGROUND: We evaluated the effect of Saccharomyces boulardii CNCM I-745 on intestinal neuromuscular anomalies in an IBS-type mouse model of gastrointestinal motor dysfunctions elicited by Herpes Simplex Virus type 1 (HSV-1) exposure. METHODS: Mice were inoculated intranasally with HSV-1 (102 PFU) or vehicle at time 0 and 4 weeks later by the intragastric (IG) route (108 PFU). Six weeks after IG inoculum, mice were randomly allocated to receive oral gavage with either S. boulardii (107 CFU/day) or vehicle. After 4 weeks the following were determined: a) intestinal motility using fluorescein-isothiocyanate dextran distribution in the gut, fecal pellet expulsion, stool water content, and distal colonic transit of glass beads; b) integrity of the enteric nervous system (ENS) by immunohistochemistry on ileal whole-mount preparations and western blot of protein lysates from ileal longitudinal muscle and myenteric plexus; c) isometric muscle tension with electric field and pharmacological (carbachol) stimulation of ileal segments; and d) intestinal inflammation by levels of tumor necrosis factor α, interleukin(IL)-1ß, IL-10 and IL-4. RESULTS: S. boulardii CNCM I-745 improved HSV-1 induced intestinal dysmotility and alteration of intestinal transit observed ten weeks after IG inoculum of the virus. Also, the probiotic yeast ameliorated the structural alterations of the ENS induced by HSV-1 (i.e., reduced peripherin immunoreactivity and expression, increased glial S100ß protein immunoreactivity and neuronal nitric oxide synthase level, reduced substance P-positive fibers). Moreover, S. boulardii CNCM I-745 diminished the production of HSV-1 associated pro-inflammatory cytokines in the myenteric plexus and increased levels of anti-inflammatory interleukins. CONCLUSIONS: S. boulardii CNCM I-745 ameliorated gastrointestinal neuromuscular anomalies in a mouse model of gut dysfunctions typically observed with irritable bowel syndrome.


Assuntos
Motilidade Gastrointestinal/efeitos dos fármacos , Síndrome do Intestino Irritável/microbiologia , Síndrome do Intestino Irritável/terapia , Probióticos/farmacologia , Saccharomyces boulardii/crescimento & desenvolvimento , Animais , Colo/metabolismo , Colo/microbiologia , Colo/virologia , Citocinas/metabolismo , Diarreia/metabolismo , Diarreia/microbiologia , Diarreia/virologia , Modelos Animais de Doenças , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/microbiologia , Sistema Nervoso Entérico/virologia , Herpes Simples/metabolismo , Herpes Simples/microbiologia , Herpes Simples/virologia , Herpesvirus Humano 1/patogenicidade , Íleo/metabolismo , Íleo/microbiologia , Íleo/virologia , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/virologia , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Interleucina-4/metabolismo , Síndrome do Intestino Irritável/metabolismo , Síndrome do Intestino Irritável/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Músculos/metabolismo , Músculos/microbiologia , Músculos/virologia , Plexo Mientérico/metabolismo , Plexo Mientérico/microbiologia , Plexo Mientérico/virologia , Fator de Necrose Tumoral alfa/metabolismo
9.
Cell Host Microbe ; 19(6): 788-99, 2016 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-27281569

RESUMO

Herpes simplex virus 1 (HSV-1), a leading cause of genital herpes, infects oral or genital mucosal epithelial cells before infecting the peripheral sensory nervous system. The spread of HSV-1 beyond the sensory nervous system and the resulting broader spectrum of disease are not well understood. Using a mouse model of genital herpes, we found that HSV-1-infection-associated lethality correlated with severe fecal and urinary retention. No inflammation or infection of the brain was evident. Instead, HSV-1 spread via the dorsal root ganglia to the autonomic ganglia of the enteric nervous system (ENS) in the colon. ENS infection led to robust viral gene transcription, pathological inflammatory responses, and neutrophil-mediated destruction of enteric neurons, ultimately resulting in permanent loss of peristalsis and the development of toxic megacolon. Laxative treatment rescued mice from lethality following genital HSV-1 infection. These results reveal an unexpected pathogenesis of HSV associated with ENS infection.


Assuntos
Sistema Nervoso Entérico/virologia , Herpes Genital/virologia , Herpesvirus Humano 1/patogenicidade , Megacolo Tóxico/virologia , Neurônios/virologia , Doenças Vaginais/virologia , Animais , Modelos Animais de Doenças , Sistema Nervoso Entérico/patologia , Feminino , Gânglios/patologia , Gânglios/ultraestrutura , Gânglios/virologia , Gânglios Espinais/patologia , Gânglios Espinais/virologia , Genoma Viral , Herpes Genital/patologia , Herpesvirus Humano 1/genética , Herpesvirus Humano 1/fisiologia , Intestinos/virologia , Megacolo Tóxico/patologia , Camundongos , Camundongos Endogâmicos C57BL , Neurônios/patologia , Neutrófilos/virologia , Nociceptores/virologia , Vagina/virologia , Doenças Vaginais/patologia , Replicação Viral/fisiologia
10.
Trans Am Clin Climatol Assoc ; 127: 282-299, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-28066065

RESUMO

Varicella zoster virus (VZV) gives rise to two diseases, a primary infection, varicella, and a secondary infection, zoster. Morbidity and mortality from VZV in the United States has decreased by 80% to 90% due to the effective use of attenuated live viral vaccines. Because latent VZV continues to reactivate, however, serious VZV-induced disease persists. Newly developed molecular analyses have revealed that zoster is more common than previously realized; moreover, the establishment of VZV latency in neurons, such as those of the enteric nervous system, which do not project to the skin, leads to unexpected, serious, and clandestine manifestations of disease, including perforating gastrointestinal ulcers and intestinal pseudo-obstruction. The development of the first animal model of zoster, in guinea pigs, now enables the pathophysiology of latency and reactivation to be analyzed.


Assuntos
Varicela/fisiopatologia , Herpes Zoster/fisiopatologia , Herpesvirus Humano 3 , Latência Viral , Animais , Modelos Animais de Doenças , Sistema Nervoso Entérico/virologia , Cobaias , Humanos
11.
Neurogastroenterol Motil ; 27(4): 449-54, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25817054

RESUMO

Human immune deficient virus (HIV) is an immunosuppressive virus that targets CD4(+) T-lymphocytes. HIV infections cause increased susceptibility to opportunistic infections and cancer. HIV infection can also alter central nervous system (CNS) function causing cognitive impairment. HIV does not infect neurons but it does infect astrocytes and microglia in the CNS. HIV can also infect enteric glia initiating an intestinal inflammatory response which causes enteric neural injury and gut dysfunction. Part of the inflammatory response is HIV induced production of proteins including, Transactivator of transcription (Tat) which contribute to neuronal injury after release from HIV infected glial cells. A risk factor for HIV infection is intravenous drug use with contaminated needles and chronic opiate use can exacerbate neural injury in the nervous system. While most research focuses on the actions of Tat and other HIV related proteins and opiates on the brain, recent data indicate that Tat can cause intestinal inflammation and disruption of enteric neuron function, including alteration of Na(+) channel activity and action potential generation. A paper published in this issue of Neurogastroenterology and Motility extends these findings by identifying an interaction between Tat and morphine on enteric neuron Na(+) channels and on intestinal motility in vivo using a Tat expressing transgenic mouse model. These new data show that Tat protein can enhance the inhibitory actions of morphine on action potential generation and propulsive motility. These findings are important to our understanding of how HIV causes diarrhea in infected patients and for the use of opioid drugs to treat HIV-induced diarrhea.


Assuntos
Sistema Nervoso Entérico/virologia , Infecções por HIV/complicações , Morfina/efeitos adversos , Neuroglia/virologia , Neurônios/virologia , Animais , Diarreia/virologia , Sistema Nervoso Entérico/fisiopatologia , Humanos , Canais de Sódio/metabolismo , Produtos do Gene rev do Vírus da Imunodeficiência Humana/metabolismo
12.
J Neurovirol ; 20(5): 442-56, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24965252

RESUMO

Latent wild-type (WT) and vaccine (vOka) varicella zoster virus (VZV) are found in the human enteric nervous system (ENS). VZV also infects guinea pig enteric neurons in vitro, establishes latency and can be reactivated. We therefore determined whether lymphocytes infected in vitro with VZV secrete infectious virions and can transfer infection in vivo to the ENS of recipient guinea pigs. T lymphocytes (CD3-immunoreactive) were preferentially infected following co-culture of guinea pig or human peripheral blood mononuclear cells with VZV-infected HELF. VZV proliferated in the infected T cells and expressed immediate early and late VZV genes. Electron microscopy confirmed that VZV-infected T cells produced encapsulated virions. Extracellular virus, however, was pleomorphic, suggesting degradation occurred prior to release, which was confirmed by the failure of VZV-infected T cells to secrete infectious virions. Intravenous injection of WT- or vOka-infected PBMCs, nevertheless, transmitted VZV to recipient animals (guinea pig > human lymphocytes). Two days post-inoculation, lung and liver, but not gut, contained DNA and transcripts encoding ORFs 4, 40, 66 and 67. Twenty-eight days after infection, gut contained DNA and transcripts encoding ORFs 4 and 66 but neither DNA nor transcripts could any longer be found in lung or liver. In situ hybridization revealed VZV DNA in enteric neurons, which also expressed ORF63p (but not ORF68p) immunoreactivity. Observations suggest that VZV infects T cells, which can transfer VZV to and establish latency in enteric neurons in vivo. Guinea pigs may be useful for studies of VZV pathogenesis in the ENS.


Assuntos
Sistema Nervoso Entérico/virologia , Infecções por Herpesviridae/virologia , Herpesvirus Humano 3 , Linfócitos T/virologia , Ativação Viral/fisiologia , Latência Viral/fisiologia , Animais , Técnicas de Cocultura , Modelos Animais de Doenças , Cobaias , Humanos , Imuno-Histoquímica , Hibridização In Situ , Técnicas In Vitro , Leucócitos Mononucleares/virologia , Reação em Cadeia da Polimerase
13.
Trans Am Clin Climatol Assoc ; 123: 17-33; discussion 33-5, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23303966

RESUMO

Despite vaccination, varicella-zoster virus (VZV) remains an important pathogen. We investigated VZV latency in autopsy specimens from vaccinees, in gastrointestinal tissue removed surgically, and in a guinea pig model. We propose that retrograde transport from infected skin and viremia deliver VZV to neurons in which it becomes latent. Wild type (WT) VZV was found to be latent in many ganglia of vaccinated children with no history of varicella, suggesting that subclinical infection with WT-VZV occurs with subsequent viremic dissemination. The 30% to 40% rate of WT-VZV zoster reported in vaccinees and occasional trigeminal zoster due to vaccine type VZV (vOka) are consistent with viremic delivery of VZV to multiple ganglia. Most human intestinal specimens contained latent VZV within neurons of the enteric nervous system (ENS). Induction of viremia in guinea pigs led to VZV latency throughout the ENS. The possibility VZV reactivation in the ENS is an unsuspected cause of gastrointestinal disease requires future investigation.


Assuntos
Vacina contra Varicela/administração & dosagem , Varicela/prevenção & controle , Nervos Cranianos/virologia , Sistema Nervoso Entérico/virologia , Gânglios Espinais/virologia , Herpesvirus Humano 3/fisiologia , Latência Viral/fisiologia , Adolescente , Animais , Autopsia , Varicela/sangue , Varicela/virologia , Criança , Pré-Escolar , Nervos Cranianos/patologia , Modelos Animais de Doenças , Sistema Nervoso Entérico/patologia , Feminino , Gânglios Espinais/patologia , Gastroenteropatias/fisiopatologia , Gastroenteropatias/virologia , Cobaias , Humanos , Lactente , Recém-Nascido , Masculino , RNA Viral/sangue , Estudos Retrospectivos , Viremia/sangue , Viremia/virologia
14.
J Neurovirol ; 17(6): 578-89, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22190254

RESUMO

Case reports have linked varicella-zoster virus (VZV) to gastrointestinal disorders, including severe abdominal pain preceding fatal varicella and acute colonic pseudoobstruction (Ogilvie's syndrome). Because we had previously detected DNA and transcripts encoding latency-associated VZV gene products in the human gut, we sought to determine whether latent VZV is present in the human enteric nervous system (ENS) and, if so, to identify the cells in which it is located and its route to the bowel. Neither DNA, nor transcripts encoding VZV gene products, could be detected in resected gut from any of seven control children (<1 year old) who had not received the varicella vaccine or experienced varicella; however, VZV DNA and transcripts were each found to be present in resected bowel from 6/6 of children with a past history of varicella and in that of 6/7 of children who received the varicella vaccine. Both wild-type (WT) and vaccine-type (vOka) VZV thus establish latent infection in human gut. To determine routes by which VZV might gain access to the bowel, we injected guinea pigs with human or guinea pig lymphocytes expressing green fluorescent protein (GFP) under the control of the VZV ORF66 gene (VZV(OKA66.GFP)). GFP-expressing enteric neurons were found throughout the bowel within 2 days and continued to be present for greater than 6 weeks. DNA encoding VZV gene products also appeared in enteric and dorsal root ganglion (DRG) neurons following intradermal administration of WT-VZV and in enteric neurons after intradermal injection of VZV(OKA66.GFP); moreover, a small number of guinea pig DRG neurons were found to project both to the skin and the intraperitoneal viscera. Viremia, in which lymphocytes carry VZV, or axonal transport from DRG neurons infected through their epidermal projections are thus each potential routes that enable VZV to gain access to the ENS.


Assuntos
Pseudo-Obstrução do Colo/prevenção & controle , Sistema Nervoso Entérico/virologia , Infecções por Herpesviridae/prevenção & controle , Herpesvirus Humano 3/fisiologia , Intestinos/virologia , Células Receptoras Sensoriais/virologia , Proteínas do Envelope Viral/genética , Latência Viral/fisiologia , Adolescente , Animais , Vacina contra Varicela/administração & dosagem , Criança , Pré-Escolar , Pseudo-Obstrução do Colo/etiologia , Pseudo-Obstrução do Colo/imunologia , Pseudo-Obstrução do Colo/virologia , Sistema Nervoso Entérico/patologia , Gânglios Espinais/virologia , Genes Reporter , Cobaias , Infecções por Herpesviridae/complicações , Infecções por Herpesviridae/imunologia , Infecções por Herpesviridae/virologia , Humanos , Lactente , Recém-Nascido , Intestinos/citologia , Transfusão de Linfócitos , Linfócitos/virologia , Camundongos , Células Receptoras Sensoriais/patologia , Proteínas do Envelope Viral/metabolismo
15.
Avian Pathol ; 40(5): 453-61, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21780967

RESUMO

In the present paper, we report the unexpected discovery of a new virus in samples from chicken and turkey flocks with clinical disorders such as tenosynovitis, enteric problems, or runting and/or stunting-like conditions. Since 1987, several virus isolation attempts on samples from these flocks resulted in the same macroscopic characteristic lesions in embryonated specific pathogen free eggs, being mortality with bright-red discolouration of legs and wing-tips, a swollen dark-red liver and oedema. Initial work suggested the presence of an agent with characteristics of a non-enveloped RNA virus. Further work, which is described in this paper, showed that the isolated strains formed a new group of avian nephritis viruses, which is genetically and antigenically distinct from known avian astroviruses. Inoculation of a representative strain (isolate 19) of this new group of avian nephritis viruses, provisionally named avian nephritis virus-3, in specific pathogen free layer chicks resulted in diarrhoea, runting and stunting, and even mortality.


Assuntos
Infecções por Astroviridae/veterinária , Avastrovirus/genética , Galinhas , Sistema Nervoso Entérico/patologia , Transtornos Neurológicos da Marcha/veterinária , Doenças das Aves Domésticas/patologia , Doenças das Aves Domésticas/virologia , Perus , Sequência de Aminoácidos , Animais , Infecções por Astroviridae/epidemiologia , Infecções por Astroviridae/patologia , Sequência de Bases , Primers do DNA/genética , Sistema Nervoso Entérico/virologia , Imunofluorescência , Transtornos Neurológicos da Marcha/patologia , Transtornos Neurológicos da Marcha/virologia , Genoma Viral/genética , Dados de Sequência Molecular , Países Baixos/epidemiologia , Doenças das Aves Domésticas/epidemiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Alinhamento de Sequência , Análise de Sequência de DNA , Estudos Soroepidemiológicos , Especificidade da Espécie
16.
Gastroenterology ; 138(5): 1790-801, 2010 May.
Artigo em Inglês | MEDLINE | ID: mdl-20102717

RESUMO

BACKGROUND & AIMS: Infectious agents, such as neurotropic viruses, are proposed to disrupt the enteric neuromuscular system, leading to dysmotility, although the mechanisms are unknown. Our purpose was to assess whether herpes simplex virus type-1 (HSV-1) establishes an enteric-neuronal infection and induces gut dysmotility. METHODS: Rats were inoculated with HSV-1 intranasally and after 4 weeks intragastrically. After 1-10 weeks, infection was determined by molecular analysis whereas neuromuscular function was evaluated by pharmacologic/electrical stimulation of longitudinal ileal segments and by gastrointestinal transit and by [(3)H]acetylcholine release measurements. Inflammation in the neuromuscular layer was assessed by myeloperoxidase and cytokine levels and by anti-CD3(+) immunohistochemistry. RESULTS: After 1-10 weeks of intragastric inoculation, HSV-1 latency-associated messenger RNA transcripts were detected in the brain and in ileal neurons with no signs of illness or histologic gut abnormalities. By using a recombinant HSV-1 carrying the lacZ gene, HSV-1 virions were localized in myenteric ganglia by in situ X-gal staining. Interleukin-2 and IFN-gamma levels were increased significantly 1 and 6 weeks after inoculation. CD3(+) cells were found around the myenteric ganglia 6 weeks after inoculation. Smooth muscle responses to carbachol, CaCl(2), and gut transit were increased significantly after 1 and 6 weeks, whereas KCl- and electrical field stimulation-mediated contractions were modified significantly only 1-2 weeks after HSV-1 administration. The release of [(3)H]acetylcholine was reduced significantly in ileum segments after 1 and 6 weeks. CONCLUSIONS: After intragastric inoculation, HSV-1 establishes a latent infection in the rat myenteric ganglia, which leads to gut dysmotility.


Assuntos
Sistema Nervoso Entérico/fisiopatologia , Motilidade Gastrointestinal , Herpes Simples/fisiopatologia , Herpesvirus Humano 1/patogenicidade , Íleo/inervação , Junção Neuromuscular/fisiopatologia , Acetilcolina/metabolismo , Animais , Complexo CD3/análise , Cloreto de Cálcio/farmacologia , Carbacol/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Estimulação Elétrica , Sistema Nervoso Entérico/imunologia , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/virologia , Trânsito Gastrointestinal , Herpes Simples/imunologia , Herpes Simples/metabolismo , Herpes Simples/virologia , Herpesvirus Humano 1/genética , Mediadores da Inflamação/metabolismo , Interferon gama/metabolismo , Interleucina-2/metabolismo , Masculino , MicroRNAs/metabolismo , Contração Muscular , Junção Neuromuscular/imunologia , Junção Neuromuscular/metabolismo , Junção Neuromuscular/virologia , Peroxidase/metabolismo , Cloreto de Potássio/farmacologia , Ratos , Ratos Wistar , Fatores de Tempo , Latência Viral
17.
J Virol ; 83(21): 11367-71, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19706702

RESUMO

A canary bird (Serinus canaria) died with nonsuppurative ganglioneuritis of the proventriculus and gizzard and encephalitis, lesions comparable to proventricular dilatation disease (PDD) of psittacine birds. Recently, several genotypes of a novel avian bornavirus have been linked to PDD. In the canary, bornaviral antigen was detected by immunohistochemistry in both neural and extraneural tissues. The widespread viral dissemination was confirmed by reverse transcription-PCR. Sequence analysis revealed a unique genotype of avian bornavirus. This observation suggests that bornaviruses are natural pathogens of several avian species and that the family Bornaviridae comprises more viral genotypes (or viral species) than previously assumed.


Assuntos
Doenças das Aves , Bornaviridae/patogenicidade , Canários/virologia , Encefalite , Sistema Nervoso Entérico , Gânglios , Neurite (Inflamação) , Animais , Doenças das Aves/patologia , Doenças das Aves/fisiopatologia , Doenças das Aves/virologia , Bornaviridae/classificação , Bornaviridae/genética , Encéfalo/patologia , Encéfalo/virologia , Encefalite/fisiopatologia , Encefalite/veterinária , Encefalite/virologia , Sistema Nervoso Entérico/patologia , Sistema Nervoso Entérico/fisiopatologia , Sistema Nervoso Entérico/virologia , Gânglios/patologia , Gânglios/fisiopatologia , Gânglios/virologia , Moela das Aves/patologia , Moela das Aves/virologia , Dados de Sequência Molecular , Neurite (Inflamação)/fisiopatologia , Neurite (Inflamação)/veterinária , Neurite (Inflamação)/virologia , Filogenia , Proventrículo/patologia , Proventrículo/virologia , Alinhamento de Sequência
18.
J Infect Dis ; 197 Suppl 2: S61-5, 2008 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-18419411

RESUMO

Because human primary afferent neurons are not readily obtained, we sought to develop a model in which the lytic, latent, and reactivating phases of varicella-zoster virus (VZV) infection were recapitulated in neurons from an animal source. Enteric neurons were obtained from the small intestine of adult guinea pigs and from the bowel of fetal mice. Latency was established when these neurons were infected by cell-free VZV in the absence of fibroblasts or other cells of mesodermal origin. In contrast, lytic infection ensued when fibroblasts were present or when the enteric neurons were infected by cell-associated VZV. Latency was associated with the expression of a limited subset of viral genes, the products of which were restricted to the cytoplasm. Lysis was associated with the expression of viral glycoproteins, nuclear translocation of latency-associated gene products, and rapid cell death. Reactivation was accomplished by expressing VZV open reading frame (ORF) 61p or herpes simplex virus ICP0 in latently infected neurons. Isolated enteric neurons from guinea pigs and mice recapitulate latent gene expression in human cranial nerve and dorsal root ganglia. Expression of latency-associated VZV gene products was detected in 88% of samples of adult human intestine, suggesting that VZV not only infects enteric neurons but also is latent in the human enteric nervous system. This in vitro model should facilitate further understanding of latency and reactivation of VZV.


Assuntos
Modelos Animais de Doenças , Sistema Nervoso Entérico/citologia , Herpesvirus Humano 3/patogenicidade , Neurônios/virologia , Ativação Viral , Latência Viral , Animais , Células Cultivadas , Sistema Nervoso Entérico/virologia , Cobaias , Herpes Zoster/virologia , Herpesvirus Humano 3/fisiologia , Humanos , Camundongos , Neurônios/citologia , Proteínas Virais/genética , Proteínas Virais/metabolismo
19.
J Virol ; 81(12): 6265-75, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17392357

RESUMO

Gastrointestinal (GI) disease is a debilitating feature of human immunodeficiency virus (HIV) infection that can occur in the absence of histopathological abnormalities or identifiable enteropathogens. However, the mechanisms of GI dysfunction are poorly understood. The present study was undertaken to characterize changes in resident and inflammatory cells in the enteric nervous system (ENS) of macaques during the acute stage of simian immunodeficiency virus (SIV) infection to gain insight into potential pathogenic mechanisms of GI disease. Ganglia from duodenum, ileum, and colon were examined in healthy and acutely infected macaques by using a combination of routine histology, double-label immunofluorescence and in situ hybridization. Evaluation of tissues from infected macaques showed progressive infiltration of myenteric ganglia by CD3+ T cells and IBA1+ macrophages beginning as early as 8 days postinfection. Quantitative image analysis revealed that the severity of myenteric ganglionitis increased with time after SIV infection and, in general, was more severe in ganglia from the small intestine than in ganglia from the colon. Despite an abundance of inflammatory cells in myenteric ganglia during acute infection, the ENS was not a target for virus infection. This study provides evidence that the ENS may be playing a role in the pathogenesis of GI disease and enteropathy in HIV-infected people.


Assuntos
Sistema Nervoso Entérico/patologia , Sistema Nervoso Entérico/virologia , Gânglios/imunologia , Gastroenteropatias/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/diagnóstico , Vírus da Imunodeficiência Símia/metabolismo , Animais , Complexo CD3/biossíntese , Feminino , Gastroenteropatias/complicações , Infecções por HIV , Processamento de Imagem Assistida por Computador , Hibridização in Situ Fluorescente , Macaca mulatta , Masculino , Microscopia de Fluorescência , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Linfócitos T/imunologia
20.
J Pediatr Surg ; 41(3): e29-31, 2006 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-16516611

RESUMO

BACKGROUND AND PURPOSE: In this report, we present the first case of an immunologically impaired child surviving a lytic varicella-zoster virus infection affecting the enteric nervous system. In histological findings, myenteric and submucous enteric ganglia were nearly completely absent owing to virus infection. METHODS: A 3-year-old girl with acute lymphoblastic leukemia and generalized varicella-zoster infection developed an ileus. She underwent multiple laparotomies in which histological sections of the entire small intestine could be obtained. RESULTS: The histological evaluation of these samples showed a generalized aganglionosis with inflammatory residuals. A more detailed immunohistochemical analysis using neuronal (PGP, enolase), glial (S100), and lymphocytic (LCA) antibodies demonstrated a nearly complete neuronal loss. CONCLUSION: To our knowledge, this is the first case of a secondary intestinal aganglionosis after varicella-zoster virus infection.


Assuntos
Sistema Nervoso Entérico/patologia , Sistema Nervoso Entérico/virologia , Herpes Zoster/complicações , Enteropatias/etiologia , Intestino Delgado/inervação , Pré-Escolar , Feminino , Herpes Zoster/etiologia , Humanos , Íleus/etiologia , Hospedeiro Imunocomprometido , Leucemia-Linfoma Linfoblástico de Células Precursoras/complicações
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA