Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
1.
Gene ; 912: 148365, 2024 Jun 20.
Artigo em Inglês | MEDLINE | ID: mdl-38485033

RESUMO

BACKGROUND: Hirschsprung's-associated enterocolitis (HAEC) is a prevalent complication of Hirschsprung's disease (HSCR). Zinc finger E-box binding homeobox 2 (ZEB2) and Notch-1/Jagged-2 are dysregulated in HSCR, but their role in HAEC progression remains poorly understood. We aimed to explore the role and underlying mechanism of enteric neural precursor cells (ENPCs) and the ZEB2/Notch-1/Jagged-2 pathway in HAEC development. METHODS: Colon tissues were collected from HSCR and HAEC patients. ENPCs were isolated from the HAEC group and stimulated by lipopolysaccharide (LPS). The expressions of ZEB2/Notch-1/Jagged-2 were measured using RT-qPCR and Western blot. Immunofluorescence and cell counting kit-8 assays were performed to assess the differentiation and proliferation of ENPCs. Inflammatory factors were measured by ELISA kits. Co-immunoprecipitation and bioinformatic analysis were used to explore the interaction between ZEB2 and Notch-1. Small interfering RNA and overexpression vectors were used to investigate the role and mechanism of ZEB2 and Notch-1 in regulating ENPCs' proliferation and differentiation during HAEC progression. RESULTS: We observed increased LPS in the colon tissues of HAEC, with downregulated ZEB2 expression and upregulated Notch-1/Jagged-2 expression. ZEB2 interacts with Notch-1. LPS treatment downregulated ZEB2 expression, upregulated Notch-1/Jagged-2 expression, and induced proliferation and differentiation disorders in ENPCs, which were reversed by the knockdown of Notch-1. Furthermore, overexpression of ZEB2 inhibited Notch-1/Jagged-2 signaling and ameliorated inflammation and dysfunction in LPS-induced ENPCs. Notch-1 overexpression enhanced LPS-induced dysfunction, but this effect was antagonized by the overexpression of ZEB2. CONCLUSION: Overexpression of ZEB2 ameliorates LPS-induced ENPCs' dysfunction via the Notch-1/Jagged-2 pathway, thus playing a role in HAEC.


Assuntos
Enterocolite , Doença de Hirschsprung , Células-Tronco Neurais , Humanos , Proliferação de Células , Colo/metabolismo , Enterocolite/complicações , Enterocolite/metabolismo , Doença de Hirschsprung/genética , Lipopolissacarídeos/farmacologia , Lipopolissacarídeos/metabolismo , Células-Tronco Neurais/metabolismo , Homeobox 2 de Ligação a E-box com Dedos de Zinco/genética , Homeobox 2 de Ligação a E-box com Dedos de Zinco/metabolismo
2.
Development ; 150(4)2023 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-36691900

RESUMO

Intestinal homeostasis depends on interactions between the intestinal epithelium, the immune system and the microbiota. Because of these complicated connections, there are many problems that need to be solved. Current research has indicated that genes targeted by Wnt signaling are responsible for controlling intestinal stem cell fate and for modulating intestinal homeostasis. Our data show that loss of frizzled 7 (Fzd7), an important element in Wnt signaling, interrupts the differentiation of mouse intestinal stem cells into absorptive progenitors instead of secretory progenitors (precursors of goblet and Paneth cells). The alteration in canonical Wnt and Notch signaling pathways interrupts epithelial homeostasis, resulting in a decrease in physical protection in the intestine. Several phenotypes in our Fzd7-deleted model were similar to the features of enterocolitis, such as shortened intestines, decreased numbers of goblet cells and Paneth cells, and severe inflammation. Additionally, loss of Fzd7 exacerbated the defects in a chemical-induced colitis model and could initiate tumorigenesis. These findings may provide important information for the discovery of efficient therapeutic methods to treat enterocolitis and related cancers in the intestines.


Assuntos
Enterocolite , Celulas de Paneth , Animais , Camundongos , Diferenciação Celular , Enterocolite/metabolismo , Células Caliciformes/metabolismo , Homeostase , Mucosa Intestinal/metabolismo , Intestinos , Via de Sinalização Wnt
3.
J Crohns Colitis ; 17(4): 565-579, 2023 Apr 19.
Artigo em Inglês | MEDLINE | ID: mdl-36322638

RESUMO

BACKGROUND: Proton pump inhibitors [PPIs] are widely used to treat a number of gastro-oesophageal disorders. PPI-induced elevation in intragastric pH may alter gastrointestinal physiology. The tight junctions [TJs] residing at the apical intercellular contacts act as a paracellular barrier. TJ barrier dysfunction is an important pathogenic factor in inflammatory bowel disease [IBD]. Recent studies suggest that PPIs may promote disease flares in IBD patients. The role of PPIs in intestinal permeability is not clear. AIM: The aim of the present study was to study the effect of PPIs on the intestinal TJ barrier function. METHODS: Human intestinal epithelial cell culture and organoid models and mouse IBD models of dextran sodium sulphate [DSS] and spontaneous enterocolitis in IL-10-/- mice were used to study the role of PPIs in intestinal permeability. RESULTS: PPIs increased TJ barrier permeability via an increase in a principal TJ regulator, myosin light chain kinase [MLCK] activity and expression, in a p38 MAPK-dependent manner. The PPI-induced increase in extracellular pH caused MLCK activation via p38 MAPK. Long-term PPI administration in mice exaggerated the increase in intestinal TJ permeability and disease severity in two independent models of DSS colitis and IL-10-/- enterocolitis. The TJ barrier disruption by PPIs was prevented in MLCK-/- mice. Human database studies revealed increased hospitalizations associated with PPI use in IBD patients. CONCLUSIONS: Our results suggest that long-term use of PPIs increases intestinal TJ permeability and exaggerates experimental colitis via an increase in MLCK expression and activity.


Assuntos
Colite , Enterocolite , Doenças Inflamatórias Intestinais , Humanos , Camundongos , Animais , Inibidores da Bomba de Prótons/farmacologia , Interleucina-10/metabolismo , Mucosa Intestinal/metabolismo , Junções Íntimas/metabolismo , Células CACO-2 , Colite/patologia , Doenças Inflamatórias Intestinais/metabolismo , Enterocolite/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/farmacologia , Permeabilidade
4.
Mol Med Rep ; 25(5)2022 May.
Artigo em Inglês | MEDLINE | ID: mdl-35302172

RESUMO

Hirschsprung­associated enterocolitis (HAEC) is characterized by intestinal mucosal damage and an imbalance in the intestinal microbiota. Recent studies have indicated that the TLR4/p­p38/NF­κB signaling pathway in the intestine is of great importance to intestinal mucosal integrity. The present study aimed to investigate the role of TLR4/phosphorylated (p­)38/NF­κB signaling in the pathogenesis of HAEC in E. coli JM83­infected endothelin receptor B (Ednrb)­/­ mice. Ednrb­/­ mice were infected with E. coli JM83 by oral gavage to establish the HAEC model. Wild­type and Ednrb­/­ mice were randomly divided into uninfected and E. coli groups. The role of TLR4/p­p38/NF­κB signaling was further evaluated by in vivo and in vitro analyses. The activation of the TLR4/p­p38/NF­κB signaling pathway induced by E. coli JM83 resulted in HAEC in Ednrb­/­ mice, which was evidenced by a significant increase in the expression of TNF­α, TGF­ß and IL­10, and a decreased density of F­actin protein expression. TLR4 knockdown reduced the severity of enterocolitis and attenuated the expression of IL­10, TNF­α and TGF­ß, whilst increasing the density of F­actin protein in Ednrb­/­ mice after E. coli infection. These results indicated that E. coli JM83 activates TLR4/p­p38/NF­κB signaling in Ednrb­/­ to promote the development of HAEC. Thus, inhibition of this signaling pathway may benefit the treatment and prevention of HAEC.


Assuntos
Enterocolite , NF-kappa B , Animais , Modelos Animais de Doenças , Enterocolite/genética , Enterocolite/metabolismo , Escherichia coli/metabolismo , Mucosa Intestinal/metabolismo , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Receptor de Endotelina B/metabolismo , Receptores de Endotelina/metabolismo , Transdução de Sinais , Receptor 4 Toll-Like/genética , Receptor 4 Toll-Like/metabolismo
5.
Sci Rep ; 11(1): 16167, 2021 08 09.
Artigo em Inglês | MEDLINE | ID: mdl-34373532

RESUMO

Hirschsprung's disease (HD) is a congenital structural abnormality of the colon seen in approximately 1 to 5000 live births. Despite surgical correction shortly after presentation, up to 60% of patients will express long-term gastrointestinal complaints, including potentially life-threatening Hirschsprung-associated enterocolitis (HAEC). In this study fecal samples from postoperative HD patients (n = 38) and their healthy siblings (n = 21) were analysed using high-resolution liquid chromatography-mass spectrometry aiming to further unravel the nature of the chronic gastrointestinal disturbances. Furthermore, within the patient group, we compared the faecal metabolome between patients with and without a history of HAEC as well as those diagnosed with short or long aganglionic segment. Targeted analysis identified several individual metabolites characteristic for all HD patients as well as those with a history of HAEC and long segment HD. Moreover, multivariate models based on untargeted data established statistically significant (p < 0.05) differences in comprehensive faecal metabolome in the patients' cohort as a whole and in patients with a history of HAEC. Pathway analysis revealed the most impact on amino sugar, lysine, sialic acid, hyaluronan and heparan sulphate metabolism in HD, as well as impaired tyrosine metabolism in HAEC group. Those changes imply disruption of intestinal mucosal barrier due to glycosaminoglycan breakdown and dysbiosis as major metabolic changes in patients' group and should be further explored for potential diagnostic or treatment targets.


Assuntos
Doença de Hirschsprung/metabolismo , Metaboloma , Estudos de Casos e Controles , Criança , Pré-Escolar , Enterocolite/etiologia , Enterocolite/metabolismo , Fezes/química , Feminino , Gânglios/anormalidades , Doença de Hirschsprung/complicações , Doença de Hirschsprung/cirurgia , Humanos , Mucosa Intestinal/metabolismo , Intestino Grosso/anormalidades , Intestino Grosso/inervação , Masculino , Redes e Vias Metabólicas , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/metabolismo , Período Pós-Operatório
6.
Int J Mol Sci ; 22(8)2021 Apr 07.
Artigo em Inglês | MEDLINE | ID: mdl-33917126

RESUMO

Hirschsprung (HSCR) Associated Enterocolitis (HAEC) is a common life-threatening complication in HSCR. HAEC is suggested to be due to a loss of gut homeostasis caused by impairment of immune system, barrier defense, and microbiome, likely related to genetic causes. No gene has been claimed to contribute to HAEC occurrence, yet. Genetic investigation of HAEC by Whole-Exome Sequencing (WES) on 24 HSCR patients affected (HAEC) or not affected (HSCR-only) by enterocolitis and replication of results on a larger panel of patients allowed the identification of the HAEC susceptibility variant p.H187Q in the Oncostatin-M receptor (OSMR) gene (14.6% in HAEC and 5.1% in HSCR-only, p = 0.0024). Proteomic analysis on the lymphoblastoid cell lines from one HAEC patient homozygote for this variant and one HAEC patient not carrying the variant revealed two well distinct clusters of proteins significantly up or downregulated upon OSM stimulation. A marked enrichment in immune response pathways (q < 0.0001) was shown in the HAEC H187 cell line, while proteins upregulated in the HAEC Q187 lymphoblasts sustained pathways likely involved in pathogen infection and inflammation. In conclusion, OSMR p.H187Q is an HAEC susceptibility variant and perturbates the downstream signaling cascade necessary for the gut immune response and homeostasis maintenance.


Assuntos
Suscetibilidade a Doenças , Enterocolite/etiologia , Enterocolite/metabolismo , Doença de Hirschsprung/complicações , Doença de Hirschsprung/genética , Subunidade beta de Receptor de Oncostatina M/genética , Transdução de Sinais , Alelos , Enterocolite/patologia , Expressão Gênica , Frequência do Gene , Variação Genética , Genótipo , Doença de Hirschsprung/diagnóstico , Humanos , Modelos Moleculares , Subunidade beta de Receptor de Oncostatina M/química , Subunidade beta de Receptor de Oncostatina M/metabolismo , Conformação Proteica , Proteômica/métodos , Relação Estrutura-Atividade , Sequenciamento do Exoma , Sequenciamento Completo do Genoma
7.
Infect Immun ; 88(12)2020 11 16.
Artigo em Inglês | MEDLINE | ID: mdl-33020211

RESUMO

Stimulator of interferon genes (STING) acts as a cytoplasmic signaling hub of innate immunity that is activated by host-derived or bacterially derived cyclic dinucleotides. Listeria monocytogenes is a foodborne, facultative intracellular pathogen that secretes c-di-AMP and activates STING, yet the in vivo role of the STING pathway during bacterial pathogenesis remains unclear. In this study, we found that STING-deficient mice had increased weight loss and roughly 10-fold-increased systemic bacterial burden during L. monocytogenes-induced enterocolitis. Infection with a L. monocytogenes mutant impaired in c-di-AMP secretion failed to elicit a protective response, whereas a mutant with increased c-di-AMP secretion triggered enhanced protection. Type I interferon (IFN) is a major output of STING signaling; however, disrupting IFN signaling during L. monocytogenes-induced enterocolitis did not recapitulate STING deficiency. In the absence of STING, the intestinal immune response was associated with a reduced influx of inflammatory monocytes. These studies suggest that in barrier sites such as the intestinal tract, where pathogen-associated molecular patterns are abundant, cytosolic surveillance systems such as STING are well positioned to detect pathogenic bacteria.


Assuntos
Fosfatos de Dinucleosídeos/metabolismo , Enterocolite/imunologia , Enterocolite/microbiologia , Listeria monocytogenes/metabolismo , Listeriose/imunologia , Proteínas de Membrana/metabolismo , Animais , Enterocolite/metabolismo , Citometria de Fluxo , Imunidade Inata , Interferon Tipo I/genética , Interferon Tipo I/metabolismo , Listeria monocytogenes/genética , Listeria monocytogenes/patogenicidade , Listeriose/metabolismo , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/metabolismo , Transdução de Sinais/genética , Transdução de Sinais/imunologia
8.
Trends Mol Med ; 26(11): 971-974, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32958405

RESUMO

G protein-coupled receptors (GPCRs) are crucial for establishing the resolution phase following an intestinal inflammatory episode. Because current treatments for intestinal inflammation have a high percentage of failure and lead to immunosuppression, repair GPCRs have promising therapeutic potential because they trigger resolution pathways without compromising the immune response.


Assuntos
Mucosa Intestinal/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Cicatrização/fisiologia , Animais , Enterocolite/etiologia , Enterocolite/metabolismo , Enterocolite/patologia , Enterocolite/terapia , Humanos , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Regeneração , Medicina Regenerativa , Transdução de Sinais
9.
Yonsei Med J ; 61(10): 891-894, 2020 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-32975064

RESUMO

The cellular entry of severe respiratory syndrome coronavirus-2 (SARS-CoV-2) is mediated by interaction with the human angiotensin-converting enzyme 2 (ACE2), a receptor that is expressed on both lung and intestinal epithelial cells. We performed a quantitative proteomic analysis to investigate the expression of possible receptors for SARS-CoV-2 in the intestinal mucosa of 23 patients with chronic colitis. ACE2 expression was low and remained unaltered in the gut of patients with ulcerative colitis (UC), Crohn's disease (CD), intestinal Behcet's disease (BD), and intestinal tuberculosis (TB), when compared with that of healthy individuals. Additionally, the expression levels of some probable co-receptors, including dipeptidyl peptidase 4 (DPP4), aminopeptidase N (AMPN), and glutamyl aminopeptidase (AMPE), were unchanged in the affected UC, CD, intestinal BD, and intestinal TB colon mucosa samples. In conclusion, gut inflammation associated with chronic colitis does not mediate a further increase in the cellular entry of SARS-CoV-2.


Assuntos
Infecções por Coronavirus , Enterocolite , Peptidil Dipeptidase A/metabolismo , Pneumonia Viral , Proteômica , Enzima de Conversão de Angiotensina 2 , Betacoronavirus , COVID-19 , Dipeptidil Peptidase 4/metabolismo , Enterocolite/metabolismo , Microbioma Gastrointestinal , Humanos , Pulmão/metabolismo , Pandemias , SARS-CoV-2
10.
J Clin Invest ; 130(12): 6443-6456, 2020 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-32809970

RESUMO

Interstitial cells of Cajal (ICCs) are pacemaker cells in the intestine, and their function can be compromised by loss of C-KIT expression. Macrophage activation has been identified in intestine affected by Hirschsprung disease-associated enterocolitis (HAEC). In this study, we examined proinflammatory macrophage activation and explored the mechanisms by which it downregulates C-KIT expression in ICCs in colon affected by HAEC. We found that macrophage activation and TNF-α production were dramatically increased in the proximal dilated colon of HAEC patients and 3-week-old Ednrb-/- mice. Moreover, ICCs lost their C-KIT+ phenotype in the dilated colon, resulting in damaged pacemaker function and intestinal dysmotility. However, macrophage depletion or TNF-α neutralization led to recovery of ICC phenotype and restored their pacemaker function. In isolated ICCs, TNF-α-mediated phosphorylation of p65 induced overexpression of microRNA-221 (miR-221), resulting in suppression of C-KIT expression and pacemaker currents. We also identified a TNF-α/NF-κB/miR-221 pathway that downregulated C-KIT expression in ICCs in the colon affected by HAEC. These findings suggest the important roles of proinflammatory macrophage activation in a phenotypic switch of ICCs, representing a promising therapeutic target for HAEC.


Assuntos
Colo/metabolismo , Enterocolite/metabolismo , Doença de Hirschsprung/metabolismo , Células Intersticiais de Cajal/metabolismo , Ativação de Macrófagos , Transdução de Sinais , Animais , Colo/patologia , Enterocolite/genética , Enterocolite/patologia , Feminino , Doença de Hirschsprung/patologia , Humanos , Células Intersticiais de Cajal/patologia , Macrófagos , Masculino , Camundongos , Camundongos Knockout , MicroRNAs/genética , MicroRNAs/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Receptor de Endotelina B/genética , Receptor de Endotelina B/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
11.
Lancet Gastroenterol Hepatol ; 5(7): 679-697, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32553146

RESUMO

Immune checkpoint inhibitors are a novel class of cancer treatment that have improved outcomes for a subset of cancer patients. They work by antagonising inhibitory immune pathways, thereby augmenting immune-mediated antitumour responses. However, immune activation is not cancer-specific and often results in the activation of immune cells in non-cancer tissues, resulting in off-target immune-mediated injury and organ dysfunction. Diarrhoea and gastrointestinal tract inflammation are common and sometimes serious side-effects of this type of therapy. Prompt recognition of gastrointestinal toxicity and, in many cases, rapid institution of anti-inflammatory or biologic therapy (or both) is required to reverse these complications. Management of organ-specific complications benefits from multidisciplinary input, including engagement with gastroenterologists for optimal management of immune checkpoint inhibitor-induced enterocolitis. In this British Society of Gastroenterology endorsed guidance document, we have developed a consensus framework for the investigation and management of immune checkpoint inhibitor-induced enterocolitis.


Assuntos
Anticorpos Monoclonais Humanizados/efeitos adversos , Antineoplásicos Imunológicos/efeitos adversos , Enterocolite/induzido quimicamente , Neoplasias/tratamento farmacológico , Sociedades Médicas/organização & administração , Corticosteroides/uso terapêutico , Anticorpos Monoclonais Humanizados/uso terapêutico , Anticorpos Monoclonais Humanizados/toxicidade , Antineoplásicos Imunológicos/uso terapêutico , Antineoplásicos Imunológicos/toxicidade , Consenso , Endoscopia/métodos , Endoscopia do Sistema Digestório/métodos , Enterocolite/tratamento farmacológico , Enterocolite/metabolismo , Gastroenterologia/organização & administração , Gastroenteropatias/induzido quimicamente , Gastroenteropatias/diagnóstico por imagem , Gastroenteropatias/patologia , Guias como Assunto , Humanos , Infliximab/uso terapêutico , Lactoferrina/metabolismo , Complexo Antígeno L1 Leucocitário/metabolismo , Administração dos Cuidados ao Paciente/métodos , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Reino Unido/epidemiologia
12.
Nat Immunol ; 21(6): 626-635, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32424362

RESUMO

The inflammasome NLRP6 plays a crucial role in regulating inflammation and host defense against microorganisms in the intestine. However, the molecular mechanisms by which NLRP6 function is inhibited to prevent excessive inflammation remain unclear. Here, we demonstrate that the deubiquitinase Cyld prevents excessive interleukin 18 (IL-18) production in the colonic mucosa by deubiquitinating NLRP6. We show that deubiquitination inhibited the NLRP6-ASC inflammasome complex and regulated the maturation of IL-18. Cyld deficiency in mice resulted in elevated levels of active IL-18 and severe colonic inflammation following Citrobacter rodentium infection. Further, in patients with ulcerative colitis, the concentration of active IL-18 was inversely correlated with CYLD expression. Thus, we have identified a novel regulatory mechanism that inhibits the NLRP6-IL-18 pathway in intestinal inflammation.


Assuntos
Enzima Desubiquitinante CYLD/metabolismo , Enterocolite/etiologia , Enterocolite/metabolismo , Inflamassomos/metabolismo , Interleucina-18/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Citrobacter rodentium , Enzima Desubiquitinante CYLD/genética , Modelos Animais de Doenças , Suscetibilidade a Doenças , Infecções por Enterobacteriaceae/imunologia , Infecções por Enterobacteriaceae/metabolismo , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Enterocolite/patologia , Expressão Gênica , Humanos , Interleucina-18/antagonistas & inibidores , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Knockout , Ligação Proteica/imunologia , Ubiquitinação
13.
J Virol ; 94(14)2020 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-32376622

RESUMO

Porcine deltacoronavirus (PDCoV) is an economically important enteropathogen of swine with worldwide distribution. PDCoV primarily infects the small intestine instead of the large intestine in vivo However, the underlying mechanism of PDCoV tropism to different intestinal segments remains poorly understood as a result of the lack of a suitable in vitro intestinal model that recapitulates the cellular diversity and complex functions of the gastrointestinal tract. Here, we established the PDCoV infection model of crypt-derived enteroids from different intestinal segments. Enteroids were susceptible to PDCoV, and multiple types of different functional intestinal epithelia were infected by PDCoV in vitro and in vivo We further found that PDCoV favorably infected the jejunum and ileum and restrictedly replicated in the duodenum and colon. Mechanistically, enteroids from different intestinal regions displayed a distinct gene expression profile, and the differential expression of primary viral receptor host aminopeptidase N (APN) instead of the interferon (IFN) responses determined the susceptibility of different intestinal segments to PDCoV, although PDCoV substantially elicited antiviral genes production in enteroids after infection. Additional studies showed that PDCoV infection significantly induced the expression of type I and III IFNs at the late stage of infection, and exogenous IFN inhibited PDCoV replication in enteroids. Hence, our results provide critical inputs to further dissect the molecular mechanisms of PDCoV-host interactions and pathogenesis.IMPORTANCE The zoonotic potential of the PDCoV, a coronavirus efficiently infecting cells from a broad range species, including porcine, chicken, and human, emphasizes the urgent need to further study the cell and tissue tropism of PDCoV in its natural host. Herein, we generated crypt stem cell-derived enteroids from porcine different intestinal regions, which well recapitulated the events in vivo of PDCoV infection that PDCoV targeted multiple types of intestinal epithelia and preferably infected the jejunum and ileum over the duodenum and colon. Mechanistically, we demonstrated that the expression of APN receptor rather than the IFN responses determined the susceptibility of different regions of the intestines to PDCoV infection, though PDCoV infection markedly elicited the IFN responses. Our findings provide important insights into how the distinct gene expression profiles of the intestinal segments determine the cell and tissue tropism of PDCoV.


Assuntos
Antígenos CD13/genética , Infecções por Coronavirus/veterinária , Coronavirus/fisiologia , Regulação Viral da Expressão Gênica , Interações Hospedeiro-Patógeno , Doenças dos Suínos/metabolismo , Doenças dos Suínos/virologia , Tropismo Viral , Animais , Enterocolite/metabolismo , Enterocolite/patologia , Enterocolite/virologia , Interferons/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Mucosa Intestinal/virologia , Suínos , Doenças dos Suínos/patologia , Replicação Viral
14.
Sci Rep ; 10(1): 4883, 2020 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-32184453

RESUMO

Loss-of-function mutations in the solute carrier organic anion transporter family, member 2a1 gene (SLCO2A1), which encodes a prostaglandin (PG) transporter, have been identified as causes of chronic nonspecific multiple ulcers in the small intestine; however, the underlying mechanisms have not been revealed. We, therefore, evaluated the effects of systemic knockout of Slco2a1 (Slco2a1-/-) and conditional knockout in intestinal epithelial cells (Slco2a1ΔIEC) and macrophages (Slco2a1ΔMP) in mice with dextran sodium sulphate (DSS)-induced acute colitis. Slco2a-/- mice were more susceptible to DSS-induced colitis than wild-type (WT) mice, but did not spontaneously develop enteritis or colitis. The nucleotide-binding domain, leucine-rich repeats containing family, pyrin domain-containing-3 (NLRP3) inflammasome was more strongly upregulated in colon tissues of Slco2a-/- mice administered DSS and in macrophages isolated from Slco2a1-/- mice than in the WT counterparts. Slco2a1ΔMP, but not Slco2a1ΔIEC mice, were more susceptible to DSS-induced colitis than WT mice, partly phenocopying Slco2a-/- mice. Concentrations of PGE2 in colon tissues and macrophages from Slco2a1-/- mice were significantly higher than those of WT mice. Blockade of inflammasome activation suppressed the exacerbation of colitis. These results indicated that Slco2a1-deficiency increases the PGE2 concentration, resulting in NLRP3 inflammasome activation in macrophages, thus exacerbating intestinal inflammation.


Assuntos
Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Transportadores de Ânions Orgânicos/deficiência , Transportadores de Ânions Orgânicos/metabolismo , Animais , Western Blotting , Células Cultivadas , Colite/genética , Sulfato de Dextrana/toxicidade , Enterocolite/induzido quimicamente , Enterocolite/genética , Enterocolite/metabolismo , Enterocolite/patologia , Ensaio de Imunoadsorção Enzimática , Inflamassomos/imunologia , Inflamassomos/metabolismo , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos , Modelos Teóricos , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Transportadores de Ânions Orgânicos/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa
15.
J Immunol Res ; 2019: 1325181, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31781671

RESUMO

Innate lymphoid cells (ILCs) comprise a recently identified subset of innate immune cells that are mainly localized to mucosa-associated tissues. Although they have not yet been fully characterized, they can generally be divided into ILC1s, ILC2s, and ILC3s. ILCs and their corresponding cytokines act as important mediators of the early stages of the immune response during inflammation, tissue repair, and the maintenance of epithelial integrity. Consequently, the dysregulation of ILC subsets might promote inflammation and cancer. Numerous studies have demonstrated that these cells play an important role in maintaining the microecological balance of the small intestine; however, their specific roles in mediating inflammation in this tissue and tumorigenesis remain unclear and controversial. In this review, we focus on recent progress that has helped to gain a better understanding of the role of ILCs in intestinal homeostasis, chronic inflammation, and cancer. Further focused research on the regulation and role of ILCs in intestinal homeostasis and pathology will help to reveal valuable diagnostic and therapeutic targets for the treatment of intestinal diseases.


Assuntos
Enterocolite/etiologia , Enterocolite/metabolismo , Imunidade Inata , Neoplasias Intestinais/etiologia , Neoplasias Intestinais/metabolismo , Linfócitos/imunologia , Linfócitos/metabolismo , Animais , Doença Crônica , Suscetibilidade a Doenças , Enterocolite/patologia , Microbioma Gastrointestinal/imunologia , Humanos , Imunidade nas Mucosas , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Neoplasias Intestinais/patologia
16.
Clin Exp Allergy ; 49(9): 1178-1190, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31468626

RESUMO

Food protein-induced enterocolitis syndrome (FPIES) is a non-IgE-mediated gastrointestinal food allergic disorder that has gained a major interest the past decade. FPIES prevalence, which still needs to be accurately determine in different populations, appears to be higher than previously thought (ie up to 0.7% in infants in the 1st year of life). FPIES to seafood in adults is also increasingly reported; limited data suggest that adult FPIES is most commonly triggered by shellfish, tends to affect females more than men, is characterized by a significant delay in diagnosis and a prolonged course. The first international consensus guidelines on diagnosis and management of FPIES have been published in 2017, proposing new diagnostic criteria as well as new criteria for a positive oral food challenge. However, there is a need to develop new biomarkers to improve the diagnosis and management of FPIES patients, and this requires a better understanding of the pathophysiology. Recently, the role of T cells has been questioned and a major role of innate immune cells has been suggested in acute FPIES. Regarding the treatment of acute FPIES reaction, ondansetron has emerged as an adjunct to intravenous rehydration in moderate-severe reactions and as a first-line treatment in mild reactions. Important information regarding the nutritional management of FPIES patients that might be complex has also been provided in the international guidelines. In this review, we discuss recent advances regarding all those different aspects.


Assuntos
Proteínas Alimentares/efeitos adversos , Enterocolite , Hipersensibilidade Alimentar , Ondansetron/uso terapêutico , Adulto , Biomarcadores/metabolismo , Enterocolite/diagnóstico , Enterocolite/metabolismo , Enterocolite/terapia , Feminino , Hipersensibilidade Alimentar/diagnóstico , Hipersensibilidade Alimentar/metabolismo , Hipersensibilidade Alimentar/terapia , Humanos , Lactente , Recém-Nascido , Masculino
17.
FASEB J ; 33(6): 7615-7624, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30908942

RESUMO

Hirschsprung disease (HSCR) is a common cause of intestinal obstruction in the newborn. Hirschsprung-associated enterocolitis (HAEC) is a significant and life-threatening complication of HSCR, affecting up to 60% of patients. Animal models of endothelin receptor B (EdnrB) mutation reliably model human HSCR and HAEC. We previously demonstrated intestinal dysbiosis and a gut-specific deficiency of B-lymphocyte-produced secretory IgA (sIgA), the primary effector molecule of mucosal immunity, in mice with homozygous neural crest cell-conditional deletion of EdnrB (EdnrBNCC-/-). To determine mechanisms for sIgA deficiency, we examined intrinsic and extrinsic aspects of B-lymphocyte development and function. Expression of the endothelin axis components [endothelin-1 (ET-1), endothelin-3 (ET-3), endothelin receptor A (EdnrA), EdnrB] were determined over a developmental time course. B-lymphocyte survival and Ig production were assayed in vitro. Polymeric Ig receptor (pIgR)-mediated IgA transport into the intestinal lumen was interrogated. We found endothelin axis component (EdnrA, EdnrB, ET-1, ET-3) expression in developing extramedullary hematopoietic organs and that some splenic B lymphocytes express EdnrB. Splenic B lymphocytes from EdnrBNCC-/- mice showed no intrinsic defect in survival vs. wild-type (WT) B lymphocytes. In vitro stimulation of splenic B lymphocytes demonstrated decreased IgA, IgG, and IgM production in EdnrBNCC-/-vs. WT mice. Additionally, small intestinal pIgR was decreased ∼50% in EdnrBNCC-/- mice. These results suggest an intrinsic B-lymphocyte defect in antibody production as well as an extrinsic defect in IgA transport in the EdnrBNCC-/- model of HAEC. Our results are consistent with human HAEC observations of decreased luminal sIgA and mouse models of other inflammatory bowel diseases, in which decreased pIgR is seen in concert with a dysregulated microbiota. Finally, our results suggest targeting the dysbiotic microbiome and pIgR-mediated sIgA transport as potential therapeutic approaches in prevention and treatment of HAEC.-Medrano, G., Cailleux, F., Guan, P., Kuruvilla, K., Barlow-Anacker, A. J., Gosain, A. B-lymphocyte-intrinsic and -extrinsic defects in secretory immunoglobulinA production in the neural crest-conditional deletion of endothelin receptor B model of Hirschsprung-associated enterocolitis.


Assuntos
Linfócitos B/metabolismo , Enterocolite/metabolismo , Doença de Hirschsprung/metabolismo , Imunoglobulina A Secretora/biossíntese , Crista Neural/metabolismo , Receptor de Endotelina B/genética , Deleção de Sequência , Animais , Modelos Animais de Doenças , Feminino , Masculino , Camundongos , Camundongos Knockout , Receptor de Endotelina B/metabolismo , Baço/metabolismo
18.
Shock ; 52(2): 215-223, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30148762

RESUMO

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in premature infants, and is associated with the development of severe lung inflammation. The pathogenesis of NEC-induced lung injury remains unknown, yet infiltrating immune cells may play a role. In support of this possibility, we now show that NEC in mice and humans was associated with the development of profound lung injury that was characterized by an influx of Th17 cells and a reduction in T regulatory lymphocytes (Tregs). Importantly, the adoptive transfer of CD4 T cells isolated from lungs of mice with NEC into the lungs of immune incompetent mice (Rag1 mice) induced profound inflammation in the lung, while the depletion of Tregs exacerbated NEC induced lung injury, demonstrating that imbalance of Th17/Treg in the lung is required for the induction of injury. In seeking to define the mechanisms involved, the selective deletion of toll-like receptor 4 (TLR4) from the Sftpc1 pulmonary epithelial cells reversed lung injury, while TLR4 activation induced the Th17 recruiting chemokine (C-C motif) ligand 25 (CCL25) in the lungs of mice with NEC. Strikingly, the aerosolized inhibition of both CCL25 and TLR4 and the administration of all trans retinoic acid restored Tregs attenuated NEC-induced lung injury. In summary, we show that TLR4 activation in Surfactant protein C-1 (Sftpc1) cells disrupts the Treg/Th17 balance in the lung via CCL25 leading to lung injury after NEC and reveal that inhibition of TLR4 and stabilization of Th17/Treg balance in the neonatal lung may prevent this devastating complication of NEC.


Assuntos
Lesão Pulmonar/metabolismo , Linfócitos/metabolismo , Receptor 4 Toll-Like/metabolismo , Animais , Células Cultivadas , Quimiocinas CC/metabolismo , Enterocolite/metabolismo , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Humanos , Imuno-Histoquímica , Interleucina-17/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Linfócitos T Reguladores/metabolismo
19.
Cell Mol Gastroenterol Hepatol ; 7(3): 655-678, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30594740

RESUMO

BACKGROUND & AIMS: RET, the receptor for the glial cell line-derived neurotrophic factor (GDNF) family ligands, is the most frequently mutated gene in congenital aganglionic megacolon or Hirschsprung's disease (HSCR). The leading cause of mortality in HSCR is HSCR-associated enterocolitis (HAEC), which is characterized by altered mucin composition, mucin retention, bacterial adhesion to enterocytes, and epithelial damage, although the order of these events is obscure. In mice, loss of GDNF signaling leads to a severely underdeveloped enteric nervous system and neonatally fatal kidney agenesis, thereby precluding the use of these mice for modeling postnatal HSCR and HAEC. Our aim was to generate a postnatally viable mouse model for HSCR/HAEC and analyze HAEC etiology. METHODS: GDNF family receptor alpha-1 (GFRa1) hypomorphic mice were generated by placing a selectable marker gene in the sixth intron of the Gfra1 locus using gene targeting in mouse embryonic stem cells. RESULTS: We report that 70%-80% reduction in GDNF co-receptor GFRa1 expression levels in mice results in HSCR and HAEC, leading to death within the first 25 postnatal days. These mice mirror the disease progression and histopathologic findings in children with untreated HSCR/HAEC. CONCLUSIONS: In GFRa1 hypomorphic mice, HAEC proceeds from goblet cell dysplasia, with abnormal mucin production and retention, to epithelial damage. Microbial enterocyte adherence and tissue invasion are late events and therefore unlikely to be the primary cause of HAEC. These results suggest that goblet cells may be a potential target for preventative treatment and that reduced expression of GFRa1 may contribute to HSCR susceptibility.


Assuntos
Enterocolite/complicações , Enterocolite/metabolismo , Receptores de Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Doença de Hirschsprung/complicações , Doença de Hirschsprung/metabolismo , Animais , Proteínas Sanguíneas/metabolismo , Neurônios Colinérgicos/metabolismo , Colo/inervação , Colo/patologia , Citocinas/genética , Citocinas/metabolismo , Sistema Nervoso Entérico/metabolismo , Sistema Nervoso Entérico/patologia , Enterocolite/sangue , Genótipo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Células Caliciformes/patologia , Doença de Hirschsprung/sangue , Homozigoto , Hipertrofia , Mucosa Intestinal/patologia , Camundongos Endogâmicos C57BL , Mucinas/metabolismo , Células-Tronco Neurais/metabolismo , Proteínas Proto-Oncogênicas c-ret , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
20.
Front Immunol ; 9: 2318, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30356663

RESUMO

Escherichia coli is a facultative anaerobic symbiont found widely among mammalian gastrointestinal tracts. Several human studies have reported increased commensal E. coli abundance in the intestine during inflammation; however, host immunological responses toward commensal E. coli during inflammation are not well-defined. Here, we show that colonization of gnotobiotic mice with different genotypes of commensal E. coli isolated from healthy conventional microbiota mice and representing distinct populations of E. coli elicited strain-specific disease phenotypes and immunopathological changes following treatment with the inflammatory stimulus, dextran sulfate sodium (DSS). Production of the inflammatory cytokines GM-CSF, IL-6, and IFN-γ was a hallmark of the severe inflammation induced by E. coli strains of Sequence Type 129 (ST129) and ST375 following DSS administration. In contrast, colonization with E. coli strains ST150 and ST468 caused mild intestinal inflammation and triggered only low levels of pro-inflammatory cytokines, a response indistinguishable from that of E. coli-free control mice treated with DSS. The disease development observed with ST129 and ST375 colonization was not directly associated with their abundance in the GI tract as their levels did not change throughout DSS treatment, and no major differences in bacterial burden in the gut were observed among the strains tested. Data mining and in vivo neutralization identified IL-6 as a key cytokine responsible for the observed differential disease severity. Collectively, our results show that the capacity to exacerbate acute intestinal inflammation is a strain-specific trait that can potentially be overcome by blocking the pro-inflammatory immune responses that mediate intestinal tissue damage.


Assuntos
Suscetibilidade a Doenças , Enterocolite/etiologia , Enterocolite/metabolismo , Escherichia coli , Microbioma Gastrointestinal , Interleucina-6/biossíntese , Animais , Biópsia , Citocinas/biossíntese , Modelos Animais de Doenças , Progressão da Doença , Enterocolite/patologia , Escherichia coli/classificação , Escherichia coli/genética , Feminino , Microbioma Gastrointestinal/imunologia , Imunomodulação , Interleucina-6/metabolismo , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Masculino , Camundongos , Tipagem de Sequências Multilocus , Filogenia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...