Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Glycobiology ; 34(6)2024 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-38590172

RESUMO

Human noroviruses, globally the main cause of viral gastroenteritis, show strain specific affinity for histo-blood group antigens (HBGA) and can successfully be propagated ex vivo in human intestinal enteroids (HIEs). HIEs established from jejunal stem cells of individuals with different ABO, Lewis and secretor geno- and phenotypes, show varying susceptibility to such infections. Using bottom-up glycoproteomic approaches we have defined and compared the N-linked glycans of glycoproteins of seven jejunal HIEs. Membrane proteins were extracted, trypsin digested, and glycopeptides enriched by hydrophilic interaction liquid chromatography and analyzed by nanoLC-MS/MS. The Byonic software was used for glycopeptide identification followed by hands-on verifications and interpretations. Glycan structures and attachment sites were identified from MS2 spectra obtained by higher-energy collision dissociation through analysis of diagnostic saccharide oxonium ions (B-ions), stepwise glycosidic fragmentation of the glycans (Y-ions), and peptide sequence ions (b- and y-ions). Altogether 694 unique glycopeptides from 93 glycoproteins were identified. The N-glycans encompassed pauci- and oligomannose, hybrid- and complex-type structures. Notably, polyfucosylated HBGA-containing glycopeptides of the four glycoproteins tetraspanin-8, carcinoembryonic antigen-related cell adhesion molecule 5, sucrose-isomaltase and aminopeptidase N were especially prominent and were characterized in detail and related to donor ABO, Lewis and secretor types of each HIE. Virtually no sialylated N-glycans were identified for these glycoproteins suggesting that terminal sialylation was infrequent compared to fucosylation and HBGA biosynthesis. This approach gives unique site-specific information on the structural complexity of N-linked glycans of glycoproteins of human HIEs and provides a platform for future studies on the role of host glycoproteins in gastrointestinal infectious diseases.


Assuntos
Antígenos de Grupos Sanguíneos , Infecções por Caliciviridae , Fucose , Glicoproteínas , Antígenos de Histocompatibilidade , Jejuno , Organoides , Glicômica , Proteômica , Genótipo , Fenótipo , Glicoproteínas/química , Glicoproteínas/genética , Glicoproteínas/metabolismo , Fucose/metabolismo , Glicosilação , Antígenos de Grupos Sanguíneos/química , Antígenos de Grupos Sanguíneos/genética , Antígenos de Grupos Sanguíneos/metabolismo , Antígenos de Histocompatibilidade/química , Antígenos de Histocompatibilidade/genética , Antígenos de Histocompatibilidade/metabolismo , Humanos , Glicopeptídeos/química , Infecções por Caliciviridae/sangue , Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/metabolismo , Organoides/metabolismo , Jejuno/metabolismo , Jejuno/virologia
2.
Vet Res ; 52(1): 93, 2021 Jun 23.
Artigo em Inglês | MEDLINE | ID: mdl-34162433

RESUMO

Porcine reproductive and respiratory syndrome (PRRS) induces respiratory disease and reproductive failure accompanied by gastroenteritis-like symptoms. The mechanism of intestinal barrier injury caused by PRRSV infection in piglets has yet to be investigated. An in vivo PRRSV-induced model was established in 30-day-old piglets by the intramuscular injection of 2 mL of 104 TCID50/mL PRRSV for 15 days. Observations of PRRSV replication and histology were conducted in the lungs and intestine, and goblet cell counts, relative MUC2 mRNA expression, and tight junction protein, proinflammatory cytokine, TLR4, MyD88, IκB and p-IκB expression were measured. PRRSV replicated in the lungs and small intestine, as demonstrated by absolute RT-qPCR quantification, and the PRRSV N protein was detected in the lung interstitium and jejunal mucosa. PRRSV infection induced both lung and gut injury, markedly decreased villus height and the villus to crypt ratio in the small intestine, and obviously increased the number of goblet cells and the relative expression of MUC2 mRNA in the jejunum. PRRSV infection aggravated the morphological depletion of tight junction proteins and increased IL-1ß, IL-6, IL-8 and TNF-α expression by activating the NF-κB signalling pathway in the jejunum. PRRSV infection impaired intestinal integrity by damaging physical and immune barriers in the intestine by inducing inflammation, which may be related to the regulation of the gut-lung axis. This study also provides a new hypothesis regarding the pathogenesis of PRRSV-induced diarrhoea.


Assuntos
Expressão Gênica , Células Caliciformes/virologia , Jejuno/virologia , Síndrome Respiratória e Reprodutiva Suína/fisiopatologia , Proteínas de Junções Íntimas/genética , Replicação Viral , Animais , Vírus da Síndrome Respiratória e Reprodutiva Suína/fisiologia , Sus scrofa , Suínos , Proteínas de Junções Íntimas/metabolismo
3.
Science ; 370(6519)2020 11 20.
Artigo em Inglês | MEDLINE | ID: mdl-33214249

RESUMO

Rotavirus causes severe diarrheal disease in children by broadly dysregulating intestinal homeostasis. However, the underlying mechanism(s) of rotavirus-induced dysregulation remains unclear. We found that rotavirus-infected cells produce paracrine signals that manifested as intercellular calcium waves (ICWs), observed in cell lines and human intestinal enteroids. Rotavirus ICWs were caused by the release of extracellular adenosine 5'-diphosphate (ADP) that activated P2Y1 purinergic receptors on neighboring cells. ICWs were blocked by P2Y1 antagonists or CRISPR-Cas9 knockout of the P2Y1 receptor. Blocking the ADP signal reduced rotavirus replication, inhibited rotavirus-induced serotonin release and fluid secretion, and reduced diarrhea severity in neonatal mice. Thus, rotavirus exploited paracrine purinergic signaling to generate ICWs that amplified the dysregulation of host cells and altered gastrointestinal physiology to cause diarrhea.


Assuntos
Difosfato de Adenosina/metabolismo , Sinalização do Cálcio/fisiologia , Cálcio/metabolismo , Infecções por Rotavirus/metabolismo , Rotavirus/fisiologia , Animais , Sinalização do Cálcio/efeitos dos fármacos , Sinalização do Cálcio/genética , Feminino , Células HEK293 , Humanos , Jejuno/metabolismo , Jejuno/virologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Comunicação Parácrina , Antagonistas do Receptor Purinérgico P2Y/farmacologia , Receptores Purinérgicos P2Y1/genética , Receptores Purinérgicos P2Y1/metabolismo
4.
Vet Pathol ; 57(5): 642-652, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32880235

RESUMO

In the small intestine, localized innate mucosal immunity is critical for intestinal homeostasis. Porcine epidemic diarrhea virus (PEDV) infection induces villus injury and impairs digestive function. Moreover, the infection might comprise localized innate mucosal immunity. This study investigated specific enterocyte subtypes and innate immune components of weaned pigs during PEDV infection. Four-week-old pigs were orally inoculated with PEDV IN19338 strain (n = 40) or sham-inoculated (n = 24). At day post inoculation (DPI) 2, 4, and 6, lysozyme expression in Paneth cells, cellular density of villous and Peyer's patch microfold (M) cells, and the expression of polymeric immunoglobulin receptor (pIgR) were assessed in the jejunum and ileum by immunohistochemistry, and interleukin (IL)-1ß and tumor necrosis factor (TNF)-α were measured in the jejunum by ELISA. PEDV infection led to a decrease in the ratios of villus height to crypt depth (VH-CD) in jejunum at DPI 2, 4, and 6 and in ileum at DPI 4. The number of villous M cells was reduced in jejunum at DPI 4 and 6 and in ileum at DPI 6, while the number of Peyer's patch M cells in ileum increased at DPI 2 and then decreased at DPI 6. PEDV-infected pigs also had reduced lysozyme expression in ileal Paneth cells at DPI 2 and increased ileal pIgR expression at DPI 4. There were no significant changes in IL-1ß and TNF-α expression in PEDV-infected pigs compared to controls. In conclusion, PEDV infection affected innate mucosal immunity of weaned pigs through alterations in Paneth cells, villous and Peyer's patch M cells, and pIgR expression.


Assuntos
Infecções por Coronavirus/veterinária , Imunidade Inata , Mucosa Intestinal/imunologia , Vírus da Diarreia Epidêmica Suína , Animais , Infecções por Coronavirus/imunologia , Infecções por Coronavirus/patologia , Citocinas/análise , Íleo/imunologia , Íleo/patologia , Íleo/virologia , Mucosa Intestinal/química , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Jejuno/imunologia , Jejuno/patologia , Jejuno/virologia , Receptores de Imunoglobulina Polimérica/metabolismo , Suínos , Desmame
5.
Vet Microbiol ; 235: 220-228, 2019 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-31383305

RESUMO

The highly virulent porcine epidemic diarrhea virus (PEDV) variants cause the death of mainly neonatal piglets, but how the viruses spread within the gastro-intestinal tract in a temporal and spatial manner has remained poorly characterized but is critical to understand the viral pathogenesis. In this study, we used the Chinese PEDV epidemic strain BJ2011C as a model organism and took advantage of the newly developed RNAscope in situ hybridization technology to investigate the tempo-spatial infection dynamics in neonatal piglets. We found that the PEDV strain BJ2011C could quickly colonize the small intestine, which occurred in just 6 h post infection, with virus shedding starting at 6 hpi and peaking at 24 hpi. Jejunum was the first target tissue for infection and then ileum, followed by infrequent infection of duodenum. In these tissues, the virus nucleic acids were mainly present in the villous epithelial cells but not in crypt cells. Interestingly, the viral RNAs were not detectable by RNAscope in large intestines although tissue damages could be discerned by H & E staining. Overall, our results provide useful information about spread dynamics and tissue preference of PEDV epidemic strain BJ2011C.


Assuntos
Infecções por Coronavirus/patologia , Intestino Delgado/virologia , Vírus da Diarreia Epidêmica Suína/patogenicidade , RNA Viral/isolamento & purificação , Doenças dos Suínos/patologia , Animais , Animais Recém-Nascidos , Diarreia/veterinária , Diarreia/virologia , Células Epiteliais/virologia , Hibridização in Situ Fluorescente , Jejuno/citologia , Jejuno/virologia , Suínos , Doenças dos Suínos/virologia , Eliminação de Partículas Virais
6.
Poult Sci ; 96(10): 3550-3558, 2017 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-28938792

RESUMO

Hemorrhagic enteritis (HE) is an acute viral disease that affects avian species, particularly turkeys, compromising their commercial production and having a negative effect on animal welfare. Turkey adenovirus 3 (TAdV-3), is the main causal agent of the disease. In this study, we considered 3 groups of turkeys to achieve 2 purposes: 1) A preliminary investigation on the microbiota content in the 4 parts of healthy turkey's intestine (group A), namely duodenum, jejunum, ileum, and ceca was done; 2) an investigation on the relationship between natural infections with TAdV-3 and the intestinal microbiota in the jejunum, where HE mostly develops, comparing group A with animals with molecular positivity for the virus and with clinical signs of HE (group B) and animals with molecular positivity for the virus but without clinical signs (group C). Massive sequencing of the hypervariable V1-V2 regions of 16S rRNA gene and QIIME 1.9.1 software analysis was performed, and operation taxonomic units (OTUs) were classified into 4 abundant phyla: Actinobacteria, Bacteroidetes, Firmicutes, and Proteobacteria. The microbial population of small intestine was distributed almost homogeneously in the healthy turkeys, and Firmicutes was the prevalent phylum (79.85% in duodenum, 89.57% in jejunum and 99.28% in ileum). As compared with small intestine, ceca microbial community was much more heterogeneous: Firmicutes (48.03%), Bacteroidetes (33.60%) and Proteobacteria (12.32%). In the natural infections of HEV, the main bacterial families were Bacteroidaceae (Bacteroidetes) and Peptostreptococcaceae (Firmicutes), uniquely detected in group B and C. Also Clostridiaceae (Firmicutes) was detected, uniquely in group B.


Assuntos
Infecções por Adenoviridae/veterinária , Microbioma Gastrointestinal , Doenças das Aves Domésticas/virologia , Siadenovirus/fisiologia , Perus , Infecções por Adenoviridae/virologia , Animais , Trato Gastrointestinal/microbiologia , Jejuno/microbiologia , Jejuno/virologia
7.
Mucosal Immunol ; 9(1): 68-82, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25899688

RESUMO

In-depth phenotyping of human intestinal antibody secreting cells (ASCs) and their precursors is important for developing improved mucosal vaccines. We used single-cell mass cytometry to simultaneously analyze 34 differentiation and trafficking markers on intestinal and circulating B cells. In addition, we labeled rotavirus (RV) double-layered particles with a metal isotope and characterized B cells specific to the RV VP6 major structural protein. We describe the heterogeneity of the intestinal B-cell compartment, dominated by ASCs with some phenotypic and transcriptional characteristics of long-lived plasma cells. Using principal component analysis, we visualized the phenotypic relationships between major B-cell subsets in the intestine and blood, and revealed that IgM(+) memory B cells (MBCs) and naive B cells were phenotypically related as were CD27(-) MBCs and switched MBCs. ASCs in the intestine and blood were highly clonally related, but associated with distinct trajectories of phenotypic development. VP6-specific B cells were present among diverse B-cell subsets in immune donors, including naive B cells, with phenotypes representative of the overall B-cell pool. These data provide a high dimensional view of intestinal B cells and the determinants regulating humoral memory to a ubiquitous, mucosal pathogen at steady-state.


Assuntos
Antígenos Virais/imunologia , Subpopulações de Linfócitos B/imunologia , Proteínas do Capsídeo/imunologia , Linhagem da Célula/imunologia , Citocinas/imunologia , Regulação da Expressão Gênica/imunologia , Imunidade nas Mucosas , Animais , Antígenos Virais/genética , Subpopulações de Linfócitos B/patologia , Subpopulações de Linfócitos B/virologia , Proteínas do Capsídeo/genética , Diferenciação Celular , Linhagem Celular , Linhagem da Célula/genética , Movimento Celular , Chlorocebus aethiops , Citocinas/genética , Células Epiteliais/imunologia , Células Epiteliais/virologia , Perfilação da Expressão Gênica , Interações Hospedeiro-Patógeno , Humanos , Imunoglobulina M/genética , Imunoglobulina M/imunologia , Memória Imunológica , Imunofenotipagem , Jejuno/imunologia , Jejuno/patologia , Jejuno/virologia , Análise de Componente Principal , Rotavirus/imunologia , Coloração e Rotulagem/métodos , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/deficiência , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia
8.
Vet Pathol ; 50(4): 715-21, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23051916

RESUMO

Opportunistic viral infections are common in simian immunodeficiency virus-infected rhesus macaques and include simian polyomavirus 40 (SV40), which causes interstitial nephritis, pneumonia, meningoencephalitis, and progressive multifocal leukoencephalopathy and rhesus cytomegalovirus (Macacine herpesvirus-3), which is associated with many pathologic manifestations, including the formation of neutrophil-rich gastrointestinal masses. Herein we report the findings of a simian immunodeficiency virus-infected rhesus macaque that presented to necropsy with multiple nodular masses restricted to the proximal jejunum. Histologically, the masses within the lamina propria were composed of abundant, loosely organized, mesenchymal tissue forming broad interlacing whorls and sheets admixed with variable numbers of neutrophils. Cells within the mesenchymoproliferative nodules contained numerous basophilic, intranuclear inclusion bodies with only scattered cytomegalic cells. Immunohistochemistry for rhesus cytomegalovirus and SV40 demonstrated variable numbers of immunopositive cells within the affected nodules. This report is the first description of SV40-associated pathology in the small intestine of a rhesus macaque and highlights the role that opportunistic viral infections can have on gastrointestinal pathology in immunosuppressed rhesus macaques.


Assuntos
Infecções por Citomegalovirus/veterinária , Macaca mulatta , Doenças dos Macacos/patologia , Infecções por Polyomavirus/veterinária , Vírus 40 dos Símios/isolamento & purificação , Infecções Tumorais por Vírus/veterinária , Animais , Proliferação de Células , Citomegalovirus/isolamento & purificação , Infecções por Citomegalovirus/complicações , Infecções por Citomegalovirus/patologia , Infecções por Citomegalovirus/virologia , Hospedeiro Imunocomprometido , Imuno-Histoquímica , Intestino Delgado/patologia , Intestino Delgado/virologia , Jejuno/patologia , Jejuno/virologia , Mesoderma/patologia , Mesoderma/virologia , Doenças dos Macacos/virologia , Mucosa/patologia , Mucosa/virologia , Infecções Oportunistas/complicações , Infecções Oportunistas/patologia , Infecções Oportunistas/veterinária , Infecções Oportunistas/virologia , Infecções por Polyomavirus/complicações , Infecções por Polyomavirus/patologia , Infecções por Polyomavirus/virologia , Síndrome de Imunodeficiência Adquirida dos Símios/complicações , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Vírus da Imunodeficiência Símia/fisiologia , Infecções Tumorais por Vírus/complicações , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia
9.
J Gen Virol ; 93(Pt 9): 2008-2016, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22739061

RESUMO

We have developed a porcine intestine epithelial cell line, designated SD-PJEC for the propagation of influenza viruses. The SD-PJEC cell line is a subclone of the IPEC-J2 cell line, which was originally derived from newborn piglet jejunum. Our results demonstrate that SD-PJEC is a cell line of epithelial origin that preferentially expresses receptors of oligosaccharides with Sia2-6Gal modification. This cell line is permissive to infection with human and swine influenza A viruses and some avian influenza viruses, but poorly support the growth of human-origin influenza B viruses. Propagation of swine-origin influenza viruses in these cells results in a rapid growth rate within the first 24 h post-infection and the titres ranged from 4 to 8 log(10) TCID(50) ml(-1). The SD-PJEC cell line was further tested as a potential alternative cell line to Madin-Darby canine kidney (MDCK) cells in conjunction with 293T cells for rescue of swine-origin influenza viruses using the reverse genetics system. The recombinant viruses A/swine/North Carolina/18161/02 (H1N1) and A/swine/Texas/4199-2/98 (H3N2) were rescued with virus titres of 7 and 8.25 log(10) TCID(50) ml(-1), respectively. The availability of this swine-specific cell line represents a more relevant substrate for studies and growth of swine-origin influenza viruses.


Assuntos
Linhagem Celular/virologia , Células Epiteliais/virologia , Vírus da Influenza A/crescimento & desenvolvimento , Vírus da Influenza B/crescimento & desenvolvimento , Jejuno/virologia , Cultura de Vírus/instrumentação , Animais , Animais Recém-Nascidos , Aves , Cães , Humanos , Vírus da Influenza A/fisiologia , Vírus da Influenza B/fisiologia , Influenza Aviária/virologia , Influenza Humana/virologia , Jejuno/citologia , Suínos , Doenças dos Suínos/virologia , Cultura de Vírus/métodos , Replicação Viral
10.
Mucosal Immunol ; 5(6): 658-69, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22669579

RESUMO

Innate lymphoid cells (ILCs) are an emerging subset of lymphocytes involved in surveillance against virally infected cells. Here, we show CD3(-)CD8(high) lymphocytes in macaque blood include major subsets of ILCs including natural killer (NK) cells expressing CD16, NKp46, and NKG2A, but also populations of ILCs in mucosal tissues having different properties. One ILC subset secreted interleukin (IL)-17 (ILC17), but these were restricted to mucosal tissues. Some mucosal ILC17 cells expressed classical NK-cell markers, but little NKG2A or NKG2D. Some ILC17 cells secreted IL-22 and tumor necrosis factor-α, but few produced interferon (IFN)-γ or contained granzyme B. IL-17 production by ILCs was induced by IL-6, transforming growth factor-ß, and IL-23. Further, simian immunodeficiency virus (SIV) infection resulted in a significant loss of ILC17 cells, especially in the jejunum, which persisted throughout SIV infection. These findings indicate that ILC17 cells may be involved in innate mucosal immune responses, and their loss may contribute to loss of intestinal mucosal integrity and disease progression in human immunodeficiency virus (HIV)/SIV infection.


Assuntos
Imunidade nas Mucosas , Mucosa Intestinal/imunologia , Jejuno/imunologia , Células Matadoras Naturais/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/imunologia , Vírus da Imunodeficiência Símia/fisiologia , Células Th17/imunologia , Animais , Antígenos CD/imunologia , Linhagem da Célula , Granzimas/imunologia , Granzimas/metabolismo , Interferon gama/imunologia , Interferon gama/metabolismo , Interleucina-17/deficiência , Interleucina-17/genética , Interleucina-17/imunologia , Interleucinas/deficiência , Interleucinas/genética , Interleucinas/imunologia , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Jejuno/patologia , Jejuno/virologia , Células Matadoras Naturais/patologia , Células Matadoras Naturais/virologia , Contagem de Linfócitos , Depleção Linfocítica , Macaca mulatta , Subfamília C de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília K de Receptores Semelhantes a Lectina de Células NK/imunologia , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Síndrome de Imunodeficiência Adquirida dos Símios/virologia , Células Th17/patologia , Células Th17/virologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo , Interleucina 22
11.
PLoS Pathog ; 7(7): e1002115, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21779163

RESUMO

Rotavirus (RV) is the major cause of severe gastroenteritis in young children. A virus-encoded enterotoxin, NSP4 is proposed to play a major role in causing RV diarrhoea but how RV can induce emesis, a hallmark of the illness, remains unresolved. In this study we have addressed the hypothesis that RV-induced secretion of serotonin (5-hydroxytryptamine, 5-HT) by enterochromaffin (EC) cells plays a key role in the emetic reflex during RV infection resulting in activation of vagal afferent nerves connected to nucleus of the solitary tract (NTS) and area postrema in the brain stem, structures associated with nausea and vomiting. Our experiments revealed that RV can infect and replicate in human EC tumor cells ex vivo and in vitro and are localized to both EC cells and infected enterocytes in the close vicinity of EC cells in the jejunum of infected mice. Purified NSP4, but not purified virus particles, evoked release of 5-HT within 60 minutes and increased the intracellular Ca²âº concentration in a human midgut carcinoid EC cell line (GOT1) and ex vivo in human primary carcinoid EC cells concomitant with the release of 5-HT. Furthermore, NSP4 stimulated a modest production of inositol 1,4,5-triphosphate (IP3), but not of cAMP. RV infection in mice induced Fos expression in the NTS, as seen in animals which vomit after administration of chemotherapeutic drugs. The demonstration that RV can stimulate EC cells leads us to propose that RV disease includes participation of 5-HT, EC cells, the enteric nervous system and activation of vagal afferent nerves to brain structures associated with nausea and vomiting. This hypothesis is supported by treating vomiting in children with acute gastroenteritis with 5-HT3 receptor antagonists.


Assuntos
Encéfalo/metabolismo , Células Enterocromafins/metabolismo , Náusea/metabolismo , Infecções por Rotavirus/metabolismo , Rotavirus/metabolismo , Serotonina/metabolismo , Vômito/metabolismo , Animais , Encéfalo/patologia , Cálcio/metabolismo , Linhagem Celular Tumoral , Criança , Pré-Escolar , Células Enterocromafins/patologia , Células Enterocromafins/virologia , Regulação da Expressão Gênica/efeitos dos fármacos , Glicoproteínas/metabolismo , Humanos , Jejuno/metabolismo , Jejuno/patologia , Jejuno/virologia , Camundongos , Camundongos Endogâmicos BALB C , Náusea/patologia , Náusea/virologia , Proteínas Proto-Oncogênicas c-fos/biossíntese , Infecções por Rotavirus/tratamento farmacológico , Infecções por Rotavirus/patologia , Antagonistas da Serotonina/uso terapêutico , Toxinas Biológicas/metabolismo , Nervo Vago/metabolismo , Nervo Vago/patologia , Proteínas não Estruturais Virais/metabolismo , Vômito/patologia , Vômito/virologia
12.
J Vet Med Sci ; 72(4): 489-92, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19959885

RESUMO

A 7-week-old SPF chicken inoculated at 4 weeks of age with chicken anemia virus was puffed up depressed and had ruffled feathers and a good body condition. Intestinal volvulus involving the jejunum and part of the duodenum forming two loops with one knob was observed. Microscopically, venous infarction of the obstructed loops, periportal and sublobular multifocal coagulative hepatic necrosis and granulomatous inflammation of the cecal tonsils were observed. Gram staining revealed no bacteria in hepatic tissue; however, gram-positive bacilli were detected in the necrotic debris in the intestinal lumen. Immunosuppression might have predisposed the chicken to intestinal and cecal tonsil infection that then progressed to volvulus. Loss of the mucosal barrier in infarction might allow bacterial toxins and vasoactive factors to escape into the systemic circulation (toxemia) and be responsible for the hepatic necrosis.


Assuntos
Vírus da Anemia da Galinha/isolamento & purificação , Infecções por Circoviridae/veterinária , Volvo Intestinal/veterinária , Necrose Hepática Massiva/veterinária , Animais , Galinhas , Infecções por Circoviridae/diagnóstico , Progressão da Doença , Duodeno/patologia , Duodeno/virologia , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Volvo Intestinal/complicações , Volvo Intestinal/patologia , Jejuno/patologia , Jejuno/virologia , Masculino , Necrose Hepática Massiva/patologia
13.
J Virol ; 82(4): 1777-86, 2008 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-18045944

RESUMO

We previously characterized the pathogenesis of two host-specific bovine enteric caliciviruses (BEC), the GIII.2 norovirus (NoV) strain CV186-OH and the phylogenetically unassigned NB strain, in gnotobiotic (Gn) calves. In this study we evaluated the Gn calf as an alternative animal model to study the pathogenesis and host immune responses to the human norovirus (HuNoV) strain GII.4-HS66. The HuNoV HS66 strain caused diarrhea (five/five calves) and intestinal lesions (one/two calves tested) in the proximal small intestine (duodenum and jejunum) of Gn calves, with lesions similar to, but less severe than, those described for the Newbury agent 2 (NA-2) and NB BEC. Viral capsid antigen was also detected in the jejunum of the proximal small intestine of one of two calves tested by immunohistochemistry. All inoculated calves shed virus in feces (five/five calves), and one/five had viremia. Antibodies and cytokine (proinflammatory, tumor necrosis factor alpha [TNF-alpha]; Th1, interleukin-12 [IL-12] and gamma interferon [IFN-gamma]; Th2, IL-4; Th2/T-regulatory, IL-10) profiles were determined in serum, feces, and intestinal contents (IC) of the HuNoV-HS66-inoculated calves (n = 5) and controls (n = 4) by enzyme-linked immunosorbent assay in the acute (postinoculation day 3 [PID 3]) and convalescent (PID 28) stages of infection. The HuNoV-HS66-specific antibody and cytokine-secreting cells (CSCs) were quantitated by ELISPOT in mononuclear cells of local and systemic tissues at PID 28. Sixty-seven percent of the HuNoV-HS66-inoculated calves seroconverted, and 100% coproconverted with immunoglobulin A (IgA) and/or IgG antibodies to HuNoV-HS66, at low titers. The highest numbers of antibody-secreting cells (ASC), both IgA and IgG, were detected locally in intestine, but systemic IgA and IgG ASC responses also occurred in the HuNoV-HS66-inoculated calves. In serum, HuNoV-HS66 induced higher peaks of TNF-alpha and IFN-gamma at PIDs 2, 7, and 10; of IL-4 and IL-10 at PID 4; and of IL-12 at PIDs 7 and 10, compared to controls. In feces, cytokines increased earlier (PID 1) than in serum and TNF-alpha and IL-10 were elevated acutely in the IC of the HS66-inoculated calves. Compared to controls, at PID 28 higher numbers of IFN-gamma and TNF-alpha CSCs were detected in mesenteric lymph nodes (MLN) or spleen and Th2 (IL-4) CSCs were elevated in intestine; IL-10 CSCs were highest in spleen. Our study provides new data confirming HuNoV-HS66 replication and enteropathogenicity in Gn calves and reveals important and comprehensive aspects of the host's local (intestine and MLN) and systemic (spleen and blood) immune responses to HuNoV-HS66.


Assuntos
Infecções por Caliciviridae/imunologia , Infecções por Caliciviridae/virologia , Gastroenterite/imunologia , Gastroenterite/virologia , Vida Livre de Germes/imunologia , Norovirus/genética , Norovirus/imunologia , Animais , Anticorpos Antivirais/análise , Células Produtoras de Anticorpos/imunologia , Antígenos Virais/análise , Bovinos , Citocinas/metabolismo , Diarreia/virologia , Fezes/virologia , Gastroenterite/patologia , Humanos , Imunoglobulina A/análise , Imunoglobulina G/análise , Jejuno/patologia , Jejuno/virologia , Norovirus/isolamento & purificação , Células Th2/imunologia , Viremia/imunologia , Viremia/virologia , Eliminação de Partículas Virais
14.
Am J Pathol ; 171(6): 1952-65, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18055558

RESUMO

Gastrointestinal disease and inflammation are common sequelae of human and simian immunodeficiency virus (SIV) infection. Nevertheless, the molecular mechanisms that lead to gastrointestinal dysfunction remain unclear. We investigated regulation of the interleukin (IL)-6-JAK-STAT3 pathway in jejunum and colon, collected at necropsy, from 10 SIV-infected macaques with diarrhea (group 1), 10 non-SIV-infected macaques with diarrhea (group 2), and 7 control uninfected macaques (group 3). All group 1 and 2 macaques had chronic diarrhea, wasting, and colitis, but group 1 animals had more frequent and severe lesions in the jejunum. A significant increase in IL-6 and SOCS-3 gene expression along with constitutive STAT3 activation was observed in the colon of all group 1 and 2 macaques and in the jejunum of only group 1 macaques compared to controls. Further, in colon, histopathology severity scores correlated significantly with IL-6 (groups 1 and 2) and SOCS-3 (group 2) gene expression. In jejunum, a similar correlation was observed only in group 1 animals. Phosphorylated STAT3 (p-STAT3) was localized to lymphocytes (CD3+) and macrophages (CD68+), with fewer CD3+ lymphocytes expressing p-STAT3 in group 1 macaques. Despite high SOCS-3 expression, STAT3 remained constitutively active, providing a possible explanation for persistent intestinal inflammation and immune activation that may favor viral replication and disease pro-gression.


Assuntos
Enterocolite/metabolismo , Interleucina-6/metabolismo , Janus Quinases/metabolismo , Macaca mulatta/virologia , Doenças dos Macacos/metabolismo , Fator de Transcrição STAT3/metabolismo , Síndrome de Imunodeficiência Adquirida dos Símios/metabolismo , Animais , Contagem de Linfócito CD4 , Colo/metabolismo , Colo/patologia , Colo/virologia , Diarreia/metabolismo , Diarreia/virologia , Enterocolite/patologia , Enterocolite/virologia , Expressão Gênica , Interleucina-6/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Janus Quinases/genética , Jejuno/metabolismo , Jejuno/patologia , Jejuno/virologia , Leucócitos Mononucleares , Doenças dos Macacos/patologia , Doenças dos Macacos/virologia , Mucosa/metabolismo , Fosforilação , Fator de Transcrição STAT3/genética , Síndrome de Imunodeficiência Adquirida dos Símios/patologia , Carga Viral
15.
J Feline Med Surg ; 9(3): 202-13, 2007 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-17363313

RESUMO

This paper reports the first genomic RNA sequence of a field strain feline coronavirus (FCoV). Viral RNA was isolated at post mortem from the jejunum and liver of a cat with feline infectious peritonitis (FIP). A consensus sequence of the jejunum-derived genomic RNA (FCoV C1Je) was determined from overlapping cDNA fragments produced by reverse transcriptase polymerase chain reaction (RT-PCR) amplification. RT-PCR products were sequenced by a reiterative sequencing strategy and the genomic RNA termini were determined using a rapid amplification of cDNA ends PCR strategy. The FCoV C1Je genome was found to be 29,255 nucleotides in length, excluding the poly(A) tail. Comparison of the FCoV C1Je genomic RNA sequence with that of the laboratory strain FCoV FIP virus (FIPV) 79-1146 showed that both viruses have a similar genome organisation and predictions made for the open reading frames and cis-acting elements of the FIPV 79-1146 genome hold true for FCoV C1Je. In addition, the sequence of the 3'-proximal third of the liver derived genomic RNA (FCoV C1Li), which encompasses the structural and accessory protein genes of the virus, was also determined. Comparisons of the enteric (jejunum) and non-enteric (liver) derived viral RNA sequences revealed 100% nucleotide identity, a finding that questions the well accepted 'internal mutation theory' of FIPV pathogenicity.


Assuntos
Coronavirus Felino/genética , Peritonite Infecciosa Felina/virologia , Variação Genética , Genoma Viral , RNA Viral/genética , Animais , Gatos , Coronavirus Felino/patogenicidade , DNA Viral , Jejuno/virologia , Fígado/virologia , Dados de Sequência Molecular , RNA Polimerase Dependente de RNA/genética
16.
J Clin Pathol ; 57(9): 1001-3, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15333670

RESUMO

Human astrovirus infection often causes outbreaks of self limiting diarrhoea, but may also infect patients who are immunodeficient or immunocompromised. Although there are previous publications relating to various aspects of astroviruses, there is a minimal amount of literature on the histopathological features of gastrointestinal astrovirus infection in humans. We report the histopathological findings, including immunohistochemical and electron microscopic features, of astrovirus infection in a bone marrow transplant recipient aged 4 years with diarrhoea. The appearance of a small intestinal biopsy did not suggest graft versus host disease, but demonstrated villous blunting, irregularity of surface epithelial cells, and an increase in lamina propria inflammatory cell density. Immunohistochemical staining with a murine astrovirus group specific monoclonal antibody demonstrated progressively more extensive staining in the duodenal and jejunal biopsies, predominantly restricted to the luminal surface and cytoplasm of surface epithelial cells, most marked at the villus tips. Electron microscopic examination demonstrated viral particles within the cytoplasm of enterocytes, focally forming paracrystalline arrays.


Assuntos
Infecções por Astroviridae/patologia , Diarreia/virologia , Intestino Delgado/virologia , Mamastrovirus , Transplante de Medula Óssea , Pré-Escolar , Citoplasma/virologia , Diagnóstico Diferencial , Diarreia/patologia , Enterócitos/virologia , Doença Enxerto-Hospedeiro/diagnóstico , Humanos , Íleo/virologia , Hospedeiro Imunocomprometido , Intestino Delgado/patologia , Jejuno/virologia , Masculino , Microscopia Imunoeletrônica
17.
Biotechnol Bioeng ; 79(2): 211-6, 2002 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-12115437

RESUMO

The goal of this study was to evaluate the efficacy of a virus-inactivating process for use during the preparation of porcine-derived extracellular matrix biomaterials for human clinical implantation. Porcine small intestine, the source material for the tissue-engineered, small intestinal submucosa (SIS) biomaterial, was evaluated. Relevant enveloped, non-enveloped, and model viruses representative of different virus families were included in the investigation: porcine parvovirus (PPV), porcine reovirus, murine leukemia retrovirus (LRV), and porcine pseudorabies (herpes) virus (PRV). Samples of small intestine were deliberately inoculated with approximately 1 x 10(7) plaque-forming units (PFU) of virus which were thereafter exposed to a 0.18% peracetic acid/4.8% aqueous ethanol mixture for time periods ranging from 5 minutes to 2 hours. Enveloped viruses were more easily inactivated than non-enveloped viruses, but material processed for 30 minutes or longer inactivated all of the viruses. D(10) values were calculated and used to extrapolate the extent of inactivation after 2 hours. Viral titers were reduced by more than 14.0 log(10) PPV, 21.0 log(10) reovirus, 40.0 log(10) PRV, and 27.0 log(10) LRV, meeting international standards for viral sterility. These results demonstrate that treatment of porcine small intestine with a peracetic acid/ethanol solution leads to a virus-free, non-crosslinked biomaterial safe for xenotransplantation into humans.


Assuntos
Bioprótese/virologia , Etanol/farmacologia , Jejuno/virologia , Ácido Peracético/farmacologia , Esterilização/métodos , Vírus/efeitos dos fármacos , Animais , Materiais Biocompatíveis , Desinfetantes/farmacologia , Herpesvirus Suídeo 1/efeitos dos fármacos , Técnicas In Vitro , Jejuno/metabolismo , Jejuno/transplante , Camundongos , Parvovirus Suíno/efeitos dos fármacos , Valores de Referência , Reoviridae/efeitos dos fármacos , Sensibilidade e Especificidade , Suínos
18.
J Infect Dis ; 185(5): 584-90, 2002 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-11865414

RESUMO

To elucidate the role of mucosal macrophages in intestinal human cytomegalovirus (HCMV) disease, primary lamina propria macrophages (LPM) were isolated from normal human jejunum, infected with HCMV, and studied for their cytokine responses. HCMV infection of LPM was confirmed by the presence of HCMV IE72 (UL123), pp65 (UL83), and glycoprotein B (UL55) proteins, which were detected by immunofluorescence, beginning at postinfection (pi) day 3, and were sustained through pi day 12 in 0.1%-0.5% of LPM. The late protein pp28 (UL99) was also detected up to pi day 12, consistent with productive infection. HCMV infection in LPM was characterized by quantitative competitive polymerase chain reaction, with maximum levels of HCMV DNA detected at pi day 7. HCMV infection of the LPM augmented lipopolysaccharide-inducible chemokine (interleukin [IL]-8 and macrophage inflammatory protein-1alpha) and cytokine (IL-6) production. These findings suggest that mucosal macrophages, via enhanced mediator production, play an important role in intestinal inflammation associated with HCMV infection.


Assuntos
Quimiocinas/biossíntese , Citomegalovirus/imunologia , Jejuno/imunologia , Lipopolissacarídeos/farmacologia , Macrófagos/imunologia , Infecções Oportunistas Relacionadas com a AIDS/virologia , Citomegalovirus/metabolismo , Infecções por Citomegalovirus/imunologia , Infecções por Citomegalovirus/virologia , Enterite/imunologia , Enterite/virologia , Humanos , Jejuno/patologia , Jejuno/virologia , Macrófagos/efeitos dos fármacos , Macrófagos/virologia
19.
Vet Pathol ; 38(4): 359-71, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11467470

RESUMO

Cats with feline leukemia virus (FeLV)-associated enteritis (FAE), enteritis of other known viral etiology (parvovirus [PV], enteric coronavirus [CoV]), and enteritis of unknown etiology with histologic features similar to those of FAE and PV enteritis (EUE) and FeLV-negative and FeLV-positive cats without enterocyte alterations were examined. Amount and types of infiltrating leukocytes in the jejunum and activity and cellular constituents of mesenteric lymph nodes, spleen, and bone marrow were determined. PV and CoV infections were confirmed by immunohistologic demonstration of PV and CoV antigen, ultrastructural demonstration of viral particles in the intestinal content, and in situ hybridization for PV genome. FeLV infection was detected by immunohistology for gp70, p27, and p15E. Latent FeLV infection was excluded by polymerase chain reaction methods for exogenous FeLV DNA. Enterocyte lesions involved the crypts in cats with PV enteritis, FAE, and EUE and the villous tips in cats with CoV enteritis. Inflammatory infiltration was generally dominated by mononuclear cells and was moderate in the unaltered intestine and in cats with PV enteritis and marked in cats with FAE, CoV enteritis, and EUE. In cats with EUE, myeloid/histiocyte antigen-positive macrophages were relatively numerous, suggesting recruitment of peripheral blood monocytes. Lymphoid tissues were depleted in cats with PV enteritis and with EUE but were normal or hyperplastic in cats with FAE. Bone marrow activity was decreased in cats with PV enteritis; in cats with FAE or EUE and in FeLV-positive cats without enterocyte alterations, activity was slightly increased. In cats with FAE and PV enteritis, a T-cell-dominated response prevailed. EUE showed some parallels to human inflammatory bowel disease, indicating a potential harmful effect of infiltrating macrophages on the intestinal epithelium.


Assuntos
Doenças do Gato/virologia , Enterite/veterinária , Vírus da Leucemia Felina/isolamento & purificação , Infecções por Retroviridae/veterinária , Animais , Medula Óssea/patologia , Medula Óssea/virologia , Doenças do Gato/patologia , Gatos , Coronavirus/isolamento & purificação , Coronavirus/ultraestrutura , Infecções por Coronavirus/veterinária , Infecções por Coronavirus/virologia , DNA Viral/química , DNA Viral/isolamento & purificação , Enterite/patologia , Enterite/virologia , Vírus da Panleucopenia Felina/isolamento & purificação , Vírus da Panleucopenia Felina/ultraestrutura , Feminino , Imuno-Histoquímica/veterinária , Hibridização In Situ/veterinária , Jejuno/patologia , Jejuno/virologia , Vírus da Leucemia Felina/genética , Vírus da Leucemia Felina/ultraestrutura , Linfonodos/patologia , Linfonodos/virologia , Masculino , Microscopia Eletrônica/veterinária , Infecções por Parvoviridae/patologia , Infecções por Parvoviridae/veterinária , Infecções por Parvoviridae/virologia , Reação em Cadeia da Polimerase/veterinária , Estudos Retrospectivos , Infecções por Retroviridae/patologia , Infecções por Retroviridae/virologia , Baço/patologia , Baço/virologia , Infecções Tumorais por Vírus/veterinária , Infecções Tumorais por Vírus/virologia
20.
Vet Pathol ; 38(4): 441-6, 2001 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-11467479

RESUMO

An approximately 8-week-old pet Syrian hamster (Mesocricetus auratus) with a 1-week history of dyspnea, hyporexia, and ataxia was submitted for necropsy. On gross examination, the hamster had multiple abdominal adhesions and enlargement of the mesenteric lymph node. Histologic evaluation revealed multicentric lymphoma of the liver, jejunum, mesenteric lymph node, testicular fat pad, and epididymis. Based on the hamster's age and the type and distribution of the lymphoma, a presumptive diagnosis of hamster polyomavirus-induced lymphoma was made. A specific polymerase chain reaction (PCR) was developed, which confirmed the diagnosis. An in situ PCR demonstrated hamster polyomavirus DNA within lymphocytes of the multicentric lymphoma and renal tubular epithelial cells and within clusters of enterocytes in the jejunum. These data are consistent with environmental dissemination of hamster polyomavirus virions through the renal tubular epithelium and into the urine and with fecal shedding of hamster polyomavirus virions; however, additional studies will be needed to confirm these observations.


Assuntos
Linfoma/veterinária , Mesocricetus , Infecções por Polyomavirus/veterinária , Polyomavirus/isolamento & purificação , Doenças dos Roedores/virologia , Infecções Tumorais por Vírus/veterinária , Animais , Cricetinae , DNA Viral/química , DNA Viral/isolamento & purificação , Evolução Fatal , Histocitoquímica/veterinária , Jejuno/patologia , Jejuno/virologia , Rim/patologia , Rim/virologia , Linfoma/patologia , Linfoma/virologia , Masculino , Reação em Cadeia da Polimerase/veterinária , Polyomavirus/genética , Infecções por Polyomavirus/patologia , Infecções por Polyomavirus/virologia , Doenças dos Roedores/patologia , Infecções Tumorais por Vírus/patologia , Infecções Tumorais por Vírus/virologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA