Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
J Vet Med Sci ; 86(3): 277-284, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38267031

RESUMEN

The mechanism by which the neonicotinoid pesticide clothianidin (CLO) disrupts the intestinal microbiota of experimental animals is unknown. We focused on α-defensins, which are regulators of the intestinal microbiota. Subchronic exposure to CLO induced dysbiosis and reduced short-chain fatty acid-producing bacteria in the intestinal microbiota of mice. Levels of cryptdin-1 (Crp1, a major α-defensin in mice) in feces and cecal contents were lower in the CLO-exposed groups than in control. In Crp1 immunostaining, Paneth cells in the jejunum and ileum of the no-observed-adverse-effect-level CLO-exposed group showed a stronger positive signal than control, likely due to the suppression of Crp1 release. Our results showed that CLO exposure suppresses α-defensin secretion from Paneth cells as part of the mechanism underlying CLO-induced dysbiosis.


Asunto(s)
Microbioma Gastrointestinal , Guanidinas , Plaguicidas , Enfermedades de los Roedores , Tiazoles , alfa-Defensinas , Ratones , Animales , Plaguicidas/toxicidad , Disbiosis/inducido químicamente , Disbiosis/microbiología , Disbiosis/veterinaria , Neonicotinoides/toxicidad , Células de Paneth/microbiología
2.
Proc Natl Acad Sci U S A ; 118(4)2021 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-33483420

RESUMEN

RNA helicases play roles in various essential biological processes such as RNA splicing and editing. Recent in vitro studies show that RNA helicases are involved in immune responses toward viruses, serving as viral RNA sensors or immune signaling adaptors. However, there is still a lack of in vivo data to support the tissue- or cell-specific function of RNA helicases owing to the lethality of mice with complete knockout of RNA helicases; further, there is a lack of evidence about the antibacterial role of helicases. Here, we investigated the in vivo role of Dhx15 in intestinal antibacterial responses by generating mice that were intestinal epithelial cell (IEC)-specific deficient for Dhx15 (Dhx15 f/f Villin1-cre, Dhx15ΔIEC). These mice are susceptible to infection with enteric bacteria Citrobacter rodentium (C. rod), owing to impaired α-defensin production by Paneth cells. Moreover, mice with Paneth cell-specific depletion of Dhx15 (Dhx15 f/f Defensinα6-cre, Dhx15ΔPaneth) are more susceptible to DSS (dextran sodium sulfate)-induced colitis, which phenocopy Dhx15ΔIEC mice, due to the dysbiosis of the intestinal microbiota. In humans, reduced protein levels of Dhx15 are found in ulcerative colitis (UC) patients. Taken together, our findings identify a key regulator of Wnt-induced α-defensins in Paneth cells and offer insights into its role in the antimicrobial response as well as intestinal inflammation.


Asunto(s)
Colitis/inmunología , Defensinas/genética , Infecciones por Enterobacteriaceae/inmunología , Células de Paneth/inmunología , ARN Helicasas/genética , Vía de Señalización Wnt , Animales , Citrobacter rodentium/inmunología , Citrobacter rodentium/patogenicidad , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Defensinas/inmunología , Sulfato de Dextran/administración & dosificación , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Microbioma Gastrointestinal/inmunología , Regulación de la Expresión Génica , Humanos , Ratones , Ratones Transgénicos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/inmunología , Células de Paneth/microbiología , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , ARN Helicasas/inmunología
3.
JCI Insight ; 5(20)2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-33055426

RESUMEN

High-fat feeding (HFF) leads to gut dysbiosis through unclear mechanisms. We hypothesize that bile acids secreted in response to high-fat diets (HFDs) may act on intestinal Paneth cells, leading to gut dysbiosis. We found that HFF resulted in widespread taxonomic shifts in the bacteria of the ileal mucosa, characterized by depletion of Lactobacillus and enrichment of Akkermansia muciniphila, Clostridium XIVa, Ruminococcaceae, and Lachnospiraceae, which were prevented by the bile acid binder cholestyramine. Immunohistochemistry and in situ hybridization studies showed that G protein-coupled bile acid receptor (TGR5) expressed in Paneth cells was upregulated in the rats fed HFD or normal chow supplemented with cholic acid. This was accompanied by decreased lysozyme+ Paneth cells and α-defensin 5 and 6 and increased expression of XBP-1. Pretreatment with ER stress inhibitor 4PBA or with cholestyramine prevented these changes. Ileal explants incubated with deoxycholic acid or cholic acid caused a decrease in α-defensin 5 and 6 and an increase in XBP-1, which was prevented by TGR5 antibody or 4PBA. In conclusion, this is the first demonstration to our knowledge that TGR5 is expressed in Paneth cells. HFF resulted in increased bile acid secretion and upregulation of TGR5 expression in Paneth cells. Bile acid toxicity in Paneth cells contributes to gut dysbiosis induced by HFF.


Asunto(s)
Ácidos y Sales Biliares/metabolismo , Disbiosis/genética , Microbioma Gastrointestinal/genética , Receptores Acoplados a Proteínas G/genética , Proteína 1 de Unión a la X-Box/genética , Akkermansia/genética , Akkermansia/patogenicidad , Animales , Ácidos y Sales Biliares/efectos adversos , Ácidos y Sales Biliares/biosíntesis , Clostridium/genética , Clostridium/patogenicidad , Dieta Alta en Grasa/efectos adversos , Modelos Animales de Enfermedad , Disbiosis/inducido químicamente , Disbiosis/metabolismo , Disbiosis/patología , Microbioma Gastrointestinal/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Lactobacillus/genética , Lactobacillus/metabolismo , Masculino , Células de Paneth/metabolismo , Células de Paneth/microbiología , Células de Paneth/patología , Ratas , alfa-Defensinas/genética
4.
Gut Microbes ; 12(1): 1782156, 2020 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-32584650

RESUMEN

Although the oral administration of Bifidobacterium longum (B. longum) relieves the signs of irritable bowel syndrome (IBS) in clinical settings, the mechanisms underlying its effects are unclear. In this study, we evaluated the precise effects of B. longum on IBS via regulation of Paneth cell function. We confirmed the beneficial effects of B. longum on defecation habits and visceral hypersensitivity in WAS rats. Further analysis revealed that B. longum enhanced mucosal repair, promoted lysozyme production, and ameliorated dysbiosis of the microbiota in WAS rats. These processes are closely correlated with Paneth cell functions. In vitro, we incubated primary cultured enteroids with B. longum and found that B. longum promoted the proliferation of these organoids; this may be attributed to the upregulation of the stem niche factors WNT3A and TGF-ß, which are secreted by Paneth cells. Based on our findings, we propose that B. longum relieves IBS by restoring the antimicrobial activity and stem niche maintenance function of Paneth cells.


Asunto(s)
Bifidobacterium longum/metabolismo , Disbiosis/microbiología , Microbioma Gastrointestinal/fisiología , Síndrome del Colon Irritable/terapia , Células de Paneth/metabolismo , Animales , Proliferación Celular/fisiología , Síndrome del Colon Irritable/microbiología , Síndrome del Colon Irritable/patología , Masculino , Células de Paneth/microbiología , Ratas , Ratas Sprague-Dawley , Factor de Crecimiento Transformador beta1/metabolismo , Regulación hacia Arriba , Proteína Wnt3A/metabolismo
5.
Front Immunol ; 10: 2289, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31608070

RESUMEN

The main environmental risk factor associated with the development of Crohn's disease (CD) is cigarette smoking. Although the mechanism is still unknown, some studies have shown that cigarette exposure affects the intestinal barrier of the small bowel. Among the factors that may be involved in this process are Paneth cells. These specialized epithelial cells are located into the small intestine, and they are able to secrete antimicrobial peptides, having an essential role in the control of the growth of microorganisms. Alterations in its function are associated with inflammatory processes, such as CD. To study how cigarette components impact ileum homeostasis and Paneth cells integrity, we used intragastric administration of cigarette smoke condensate (CSC) in mice. Our results showed that inflammation was triggered after mucosal exposure of CSC, which induced particular alterations in Paneth cells granules, antimicrobial peptide production, and a reduction of bactericidal capacity. In fact, exposure to CSC generated an imbalance in the fecal bacterial population and increased the susceptibility of mice to develop ileal damage in response to bacterial infection. Moreover, our results obtained in mice unable to produce interleukin 10 (IL-10-/- mice) suggest that CSC treatment can induce a symptomatic enterocolitis with a pathological inflammation in genetically susceptible individuals.


Asunto(s)
Íleon/inmunología , Inflamación/inmunología , Mucosa Intestinal/inmunología , Productos de Tabaco/efectos adversos , Animales , Enfermedad de Crohn/inmunología , Enfermedad de Crohn/microbiología , Íleon/microbiología , Inflamación/microbiología , Interleucina-10/inmunología , Mucosa Intestinal/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Células de Paneth/inmunología , Células de Paneth/microbiología
6.
J Cell Physiol ; 234(11): 19406-19419, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31020664

RESUMEN

The intestinal mucosal barrier is the first line to defense against luminal content penetration and performs numerous biological functions. The intestinal epithelium contains a huge surface that is lined by a monolayer of intestinal epithelial cells (IECs). IECs are dominant mediators in maintaining intestinal homeostasis that drive diverse functions including nutrient absorption, physical segregation, secretion of antibacterial peptides, and modulation of immune responses. Autophagy is a cellular self-protection mechanism in response to various stresses, and accumulating studies have revealed its importance in participating physiological processes of IECs. The regulatory effects of autophagy depend on the specific IEC types. This review aims to elucidate the myriad roles of autophagy in regulating the functions of different IECs (stem cells, enterocytes, goblet cells, and Paneth cells), and present the progress of autophagy-targeting therapy in intestinal diseases. Understanding the involved mechanisms can provide new preventive and therapeutic strategies for gastrointestinal dysfunction and diseases.


Asunto(s)
Autofagia/genética , Células Epiteliales/metabolismo , Enfermedades Gastrointestinales/genética , Mucosa Intestinal/metabolismo , Péptidos Catiónicos Antimicrobianos , Colitis/genética , Colitis/microbiología , Colitis/patología , Células Epiteliales/microbiología , Células Epiteliales/patología , Enfermedades Gastrointestinales/metabolismo , Enfermedades Gastrointestinales/microbiología , Células Caliciformes/metabolismo , Humanos , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Intestinos/microbiología , Intestinos/patología , Células de Paneth/metabolismo , Células de Paneth/microbiología , Células de Paneth/patología
7.
Life Sci Alliance ; 2(2)2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30948494

RESUMEN

Clostridium difficile impairs Paneth cells, driving intestinal inflammation that exaggerates colitis. Besides secreting bactericidal products to restrain C. difficile, Paneth cells act as guardians that constitute a niche for intestinal epithelial stem cell (IESC) regeneration. However, how IESCs are sustained to specify Paneth-like cells as their niche remains unclear. Cytokine-JAK-STATs are required for IESC regeneration. We investigated how constitutive STAT5 activation (Ca-pYSTAT5) restricts IESC differentiation towards niche cells to restrain C. difficile infection. We generated inducible transgenic mice and organoids to determine the effects of Ca-pYSTAT5-induced IESC lineages on C. difficile colitis. We found that STAT5 absence reduced Paneth cells and predisposed mice to C. difficile ileocolitis. In contrast, Ca-pYSTAT5 enhanced Paneth cell lineage tracing and restricted Lgr5 IESC differentiation towards pYSTAT5+Lgr5-CD24+Lyso+ or cKit+ niche cells, which imprinted Lgr5hiKi67+ IESCs. Mechanistically, pYSTAT5 activated Wnt/ß-catenin signaling to determine Paneth cell fate. In conclusion, Ca-pYSTAT5 gradients control niche differentiation. Lack of pYSTAT5 reduces the niche cells to sustain IESC regeneration and induces C. difficile ileocolitis. STAT5 may be a transcription factor that regulates Paneth cells to maintain niche regeneration.


Asunto(s)
Clostridioides difficile , Colitis/metabolismo , Colitis/microbiología , Células de Paneth/metabolismo , Células de Paneth/microbiología , Factor de Transcripción STAT5/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Humanos , Células Madre Pluripotentes Inducidas/metabolismo , Células Madre Pluripotentes Inducidas/microbiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Organoides/metabolismo , Organoides/microbiología , Nicho de Células Madre/fisiología , Vía de Señalización Wnt , beta Catenina/metabolismo
8.
Semin Cell Dev Biol ; 88: 138-146, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-29355606

RESUMEN

The gut is the biggest immune organ in the body that encloses commensal microbiota which aids in food digestion. Paneth cells, positioned at the frontline of host-microbiota interphase, can modulate the composition of microbiota. Paneth cells achieve this via the delivery of microbicidal substances, among which enteric α-defensins play the primary role. If microbiota is dysregulated, it can impact the function of the local mucosal immune system. Importantly, this system is also exposed to an enormous number of antigens which are derived from the gut-resident microbiota and processed food, and may potentially trigger undesirable local inflammatory responses. To understand the intricate regulations and liaisons between Paneth cells, microbiota and the immune system in this intestinal-specific setting, one must consider their mode of interaction in a wider context of regulatory processes which impose immune tolerance not only to self, but also to microbiota and food-derived antigens. These include, but are not limited to, tolerogenic mechanisms of central tolerance in the thymus and peripheral tolerance in the secondary lymphoid organs, and the intestine itself. Defects in these processes can compromise homeostasis in the intestinal mucosal immunity. In this review, which is focused on tolerance to intestinal antigens and its relevance for the pathogenesis of gut immune diseases, we provide an outline of such multilayered immune control mechanisms and highlight functional links that underpin their cooperative nature.


Asunto(s)
Disbiosis/prevención & control , Tracto Gastrointestinal/inmunología , Células de Paneth/inmunología , Tolerancia Periférica , alfa-Defensinas/inmunología , Animales , Tolerancia Central , Disbiosis/inmunología , Disbiosis/microbiología , Microbioma Gastrointestinal/inmunología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Expresión Génica/inmunología , Homeostasis/inmunología , Humanos , Inmunidad Mucosa/efectos de los fármacos , Inflamación , Células de Paneth/efectos de los fármacos , Células de Paneth/microbiología , Simbiosis/inmunología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/microbiología , alfa-Defensinas/biosíntesis , alfa-Defensinas/farmacología
9.
Br J Haematol ; 182(6): 887-894, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30004111

RESUMEN

Host genetics shape the gut microbiota, and gut dysbiosis increases the risk of acute graft-versus-host disease (aGVHD). Paneth cells and microbiota have interactions that contribute to immune regulation. α-defensin-5 (HD5) and regenerating islet-derived protein 3 alpha (Reg3A) are the most abundant Paneth cell antimicrobial peptides (AMPs). We hypothesized that single nucleotide polymorphisms (SNPs) in the genes for HD5 (DEFA5) and Reg3A (REG3A) predict aGVHD risk. We analysed pre-transplant recipient peripheral blood mononuclear cell samples from randomized Blood and Marrow Transplant Clinical Trials Network (BMT CTN) studies 0201 (94 patients with bone marrow and 93 with peripheral blood grafts) and 0901 (86 patients with myeloablative and 77 with reduced-intensity conditioning; all using peripheral blood grafts). In multivariable analysis (with a SNP × graft source interaction term in CTN-0201 and a SNP × conditioning intensity term in CTN-0901), DEFA5 rs4415345 and rs4610776 were associated with altered incidence of aGVHD grade II-IV [rs4415345 G vs. C: hazard ratio (HR) 0·58, 95% confidence interval (95% CI) 0·37-0·92, P = 0·02; rs4610776 T vs. A: HR 1·53, 95% CI 1·01-2·32, P = 0·05] in CTN-0201, but not CTN-0901, suggesting a stronger effect in bone marrow allografts. REG3A SNP was not associated with aGVHD. Host genetics may influence aGVHD risk by modulating Paneth cell function.


Asunto(s)
Péptidos Catiónicos Antimicrobianos/genética , Enfermedad Injerto contra Huésped/etiología , Células de Paneth/química , Polimorfismo de Nucleótido Simple , Enfermedad Aguda , Recolección de Muestras de Sangre , Trasplante de Médula Ósea/efectos adversos , Ensayos Clínicos como Asunto , Enfermedad Injerto contra Huésped/microbiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Interacciones Microbiota-Huesped/genética , Humanos , Microbiota , Proteínas Asociadas a Pancreatitis/genética , Células de Paneth/microbiología , Pronóstico , alfa-Defensinas/genética
10.
Eur J Gastroenterol Hepatol ; 30(8): 828-837, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29787419

RESUMEN

BACKGROUND: Approximately 5% of patients with celiac disease (CeD) do not respond to a gluten-free diet and progress to refractory celiac disease (RCD), a severe progression that is characterized by infiltration of intraepithelial T lymphocytes. Patients with RCD type II (RCDII) show clonal expansions of intraepithelial T lymphocytes that result in a poor prognosis and a high mortality rate through development of aggressive enteropathy-associated T-cell lymphoma. It is not known whether genetic variations play a role in severe progression of CeD to RCDII. PATIENTS AND METHODS: We performed the first genome-wide association study to identify the causal genes for RCDII and the molecular pathways perturbed in RCDII. The genome-wide association study was performed in 38 Dutch patients with RCDII, and the 15 independent top-associated single nucleotide polymorphism (SNP) variants (P<5×10) were replicated in 56 independent French and Dutch patients with RCDII. RESULTS: After replication, SNP rs2041570 on chromosome 7 was significantly associated with progression to RCDII (P=2.37×10, odds ratio=2.36) but not with CeD susceptibility. SNP rs2041570 risk allele A was associated with lower levels of FAM188B expression in blood and small intestinal biopsies. Stratification of RCDII biopsies based on rs2041570 genotype showed differential expression of innate immune and antibacterial genes that are expressed in Paneth cells. CONCLUSION: We have identified a novel SNP associated with the severe progression of CeD to RCDII. Our data suggest that genetic susceptibility to CeD might be distinct from the progression to RCDII and suggest a role for Paneth cells in RCDII progression.


Asunto(s)
Enfermedad Celíaca/genética , Cromosomas Humanos Par 7/genética , Polimorfismo de Nucleótido Simple , Biopsia , Estudios de Casos y Controles , Enfermedad Celíaca/diagnóstico , Enfermedad Celíaca/dietoterapia , Enfermedad Celíaca/inmunología , Dieta Sin Gluten , Progresión de la Enfermedad , Femenino , Francia , Microbioma Gastrointestinal/genética , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Inmunidad Innata/genética , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Intestino Delgado/patología , Masculino , Proteínas de la Membrana/genética , Análisis Multivariante , Países Bajos , Oportunidad Relativa , Células de Paneth/inmunología , Células de Paneth/microbiología , Células de Paneth/patología , Fenotipo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Insuficiencia del Tratamiento
11.
Autophagy ; 14(4): 719-721, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29388875

RESUMEN

Secretion of antimicrobial proteins is an important host defense mechanism against bacteria, yet how secretory cells maintain function during bacterial invasion has been unclear. We discovered that Paneth cells, specialized secretory cells in the small intestine, react to bacterial invasion by rerouting a critical secreted antibacterial protein through a macroautophagy/autophagy-based secretion system termed secretory autophagy. Mice harboring a mutation in an essential autophagy gene, a mutation which is common in Crohn disease patients, cannot reroute their antimicrobial cargo during bacterial invasion and thus have compromised innate immunity. We showed that this alternative secretion system is triggered by both a cell-intrinsic mechanism, involving the ER stress response, and a cell-extrinsic mechanism, involving subepithelial innate immune cells. Our findings uncover a new role for secretory autophagy in host defense and suggest how a mutation in an autophagy gene can predispose individuals to Crohn disease.


Asunto(s)
Autofagia/fisiología , Estrés del Retículo Endoplásmico/genética , Intestino Delgado/microbiología , Células de Paneth/metabolismo , Proteínas Relacionadas con la Autofagia/metabolismo , Transporte Biológico/fisiología , Proteínas Portadoras/metabolismo , Enfermedad de Crohn/genética , Enfermedad de Crohn/microbiología , Muramidasa/metabolismo , Mutación/genética , Células de Paneth/microbiología
12.
Microbiome ; 6(1): 9, 2018 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-29321057

RESUMEN

BACKGROUND: Microbiota integrity is essential for a growing number of physiological processes. Consequently, disruption of microbiota homeostasis correlates with a variety of pathological states. Importantly, commensal microbiota provide a shield against invading bacterial pathogens, probably by direct competition. The impact of viral infections on host microbiota composition and dynamics is poorly understood. Influenza A viruses (IAV) are common respiratory pathogens causing acute infections. Here, we show dynamic changes in respiratory and intestinal microbiota over the course of a sublethal IAV infection in a mouse model. RESULTS: Using a combination of 16S rRNA gene-specific next generation sequencing and qPCR as well as culturing of bacterial organ content, we found body site-specific and transient microbiota responses. In the lower respiratory tract, we observed only minor qualitative changes in microbiota composition. No quantitative impact on bacterial colonization after IAV infection was detectable, despite a robust antimicrobial host response and increased sensitivity to bacterial super infection. In contrast, in the intestine, IAV induced robust depletion of bacterial content, disruption of mucus layer integrity, and higher levels of antimicrobial peptides in Paneth cells. As a functional consequence of IAV-mediated microbiota depletion, we demonstrated that the small intestine is rendered more susceptible to bacterial pathogen invasion, in a Salmonella typhimurium super infection model. CONCLUSION: We show for the first time the consequences of IAV infection for lower respiratory tract and intestinal microbiobiota in a qualitative and quantitative fashion. The discrepancy of relative 16S rRNA gene next-generation sequencing (NGS) and normalized 16S rRNA gene-specific qPCR stresses the importance of combining qualitative and quantitative approaches to correctly analyze composition of organ associated microbial communities. The transiently induced dysbiosis underlines the overall stability of microbial communities to effects of acute infection. However, during a short-time window, specific ecological niches might lose their microbiota shield and remain vulnerable to bacterial invasion.


Asunto(s)
Bacterias/clasificación , Infecciones por Orthomyxoviridae/microbiología , Células de Paneth/microbiología , ARN Ribosómico 16S/genética , Animales , Bacterias/genética , Bacterias/aislamiento & purificación , ADN Bacteriano/genética , ADN Ribosómico/genética , Modelos Animales de Enfermedad , Disbiosis/microbiología , Femenino , Microbioma Gastrointestinal , Virus de la Influenza A/patogenicidad , Ratones , Análisis de Secuencia de ADN
13.
Gastroenterology ; 153(6): 1594-1606.e2, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28865734

RESUMEN

BACKGROUND & AIMS: Separation of newborn rats from their mothers induces visceral hypersensitivity and impaired epithelial secretory cell lineages when they are adults. Little is known about the mechanisms by which maternal separation causes visceral hypersensitivity or its relationship with defects in epithelial secretory cell lineages. METHODS: We performed studies with C3H/HeN mice separated from their mothers as newborns and mice genetically engineered (Sox9flox/flox-vil-cre on C57BL/6 background) to have deficiencies in Paneth cells. Paneth cell deficiency was assessed by lysozyme staining of ileum tissues and lysozyme activity in fecal samples. When mice were 50 days old, their abdominal response to colorectal distension was assessed by electromyography. Fecal samples were collected and microbiota were analyzed using Gut Low-Density Array quantitative polymerase chain reaction. RESULTS: Mice with maternal separation developed visceral hypersensitivity and defects in Paneth cells, as reported from rats, compared with mice without maternal separation. Sox9flox/flox-vil-Cre mice also had increased visceral hypersensitivity compared with control littermate Sox9flox/flox mice. Fecal samples from mice with maternal separation and from Sox9flox/flox-vil-cre mice had evidence for intestinal dysbiosis of the microbiota, characterized by expansion of Escherichia coli. Daily gavage of conventional C3H/HeN adult mice with 109 commensal E coli induced visceral hypersensitivity. Conversely, daily oral administration of lysozyme prevented expansion of E coli during maternal separation and visceral hypersensitivity. CONCLUSIONS: Mice with defects in Paneth cells (induced by maternal separation or genetically engineered) have intestinal expansion of E coli leading to visceral hypersensitivity. These findings provide evidence that Paneth cell function and intestinal dysbiosis are involved in visceral sensitivity.


Asunto(s)
Ansiedad de Separación/complicaciones , Escherichia coli/crecimiento & desarrollo , Microbioma Gastrointestinal , Hiperalgesia/etiología , Células de Paneth/microbiología , Dolor Visceral/etiología , Factores de Edad , Animales , Animales Recién Nacidos , Ansiedad de Separación/metabolismo , Ansiedad de Separación/microbiología , Ansiedad de Separación/fisiopatología , Modelos Animales de Enfermedad , Disbiosis , Heces/microbiología , Femenino , Predisposición Genética a la Enfermedad , Hiperalgesia/metabolismo , Hiperalgesia/microbiología , Hiperalgesia/fisiopatología , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Noqueados , Muramidasa/administración & dosificación , Muramidasa/metabolismo , Células de Paneth/metabolismo , Fenotipo , Factor de Transcripción SOX9/genética , Factor de Transcripción SOX9/metabolismo , Dolor Visceral/metabolismo , Dolor Visceral/microbiología , Dolor Visceral/fisiopatología
14.
PLoS One ; 12(4): e0176583, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28441432

RESUMEN

OBJECTIVES: Intestinal barrier dysfunction plays an important role in acute necrotizing pancreatitis (ANP) and intestinal microbiota dysbiosis was involved in intestinal barrier failure. Paneth cells protect intestinal barrier and are associated with intestinal microbiota. Here, we investigated changes in intestinal microbiota and antimicrobial peptides of Paneth cells in ileum during ANP. METHODS: Rats with ANP were established by retrograde injection of 3.5% sodium taurocholate into biliopancreatic duct and sacrificed at 24h and 48h, respectively. Injuries of pancreas and distal ileum were evaluated by histopathological score. Intestinal barrier function was assessed by plasma diamine oxidase activity (DAO) and D-lactate. Systemic and intestinal inflammation was evaluated by TNFα, IL-1ß and IL-17A concentration by ELISA, respectively. 16S rRNA high throughput sequencing on fecal samples was used to investigate the changes in intestinal microbiota in the ANP group at 48h. Lysozyme and α-defensin5 were measured by real-time PCR, western blot and immunofluoresence. RESULTS: ANP rats had more severe histopathological injuries in pancreas and distal ileum, injured intestinal barrier and increased expression of TNFα, IL-1ß and IL-17A in plasma and distal ileum compared with those of the sham-operated (SO) group. Principal component analysis (PCA) showed structural segregation between the SO and ANP groups. Operational taxonomic unit (OTU) number and ACE index revealed decreased microbiota diversity in the ANP group. Taxonomic analysis showed dysbiosis of intestinal microbiota structure. At phyla level, Saccharibacteria and Tenericutes decreased significantly. At genus level, Escherichia-Shigella and Phascolarctobacterium increased significantly, while Candidatus_Saccharimonas, Prevotellaceae_UCG-001, Lachnospiraceae_UCG-001, Ruminiclostridium_5 and Ruminococcaceae_UCG-008 decreased significantly. Lysozyme and α-defensin5 mRNA expression levels decreased significantly in ANP group at 48h. Protein expression of lysozyme decreased in ANP groups at 24h and 48h. Meanwhile, the relative abundance of Escherichia-Shigella correlated inversely with the decrease in lysozyme. CONCLUSION: The disorder in intestinal microbiota and decreases of Paneth cell antimicrobial peptides might participate in the pathogenesis of intestinal barrier dysfunction during ANP.


Asunto(s)
Disbiosis/microbiología , Microbioma Gastrointestinal , Íleon/microbiología , Pancreatitis Aguda Necrotizante/microbiología , Células de Paneth/microbiología , Animales , Disbiosis/metabolismo , Disbiosis/patología , Íleon/metabolismo , Íleon/patología , Interleucina-17/metabolismo , Masculino , Pancreatitis Aguda Necrotizante/inducido químicamente , Pancreatitis Aguda Necrotizante/metabolismo , Células de Paneth/metabolismo , Células de Paneth/patología , Ratas , Ratas Sprague-Dawley , Ácido Taurocólico , Factor de Necrosis Tumoral alfa/metabolismo
15.
Sci Rep ; 7(1): 83, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28250440

RESUMEN

The strong age dependency of neonatal systemic infection with Escherichia coli K1 can be replicated in the neonatal rat. Gastrointestinal (GI) colonization of two-day-old (P2) rats leads to invasion of the blood within 48 h of initiation of colonization; pups become progressively less susceptible to infection over the P2-P9 period. We show that, in animals colonized at P2 but not at P9, E. coli K1 bacteria gain access to the enterocyte surface in the mid-region of the small intestine and translocate through the epithelial cell monolayer by an intracellular pathway to the submucosa. In this region of the GI tract, the protective mucus barrier is poorly developed but matures to full thickness over P2-P9, coincident with the development of resistance to invasion. At P9, E. coli K1 bacteria are physically separated from villi by the mucus layer and their numbers controlled by mucus-embedded antimicrobial peptides, preventing invasion of host tissues.


Asunto(s)
Infecciones por Escherichia coli/microbiología , Escherichia coli/fisiología , Mucosa Intestinal/crecimiento & desarrollo , Intestino Delgado/citología , Animales , Animales Recién Nacidos , Sangre/microbiología , Células Cultivadas , Modelos Animales de Enfermedad , Mucosa Intestinal/microbiología , Intestino Delgado/crecimiento & desarrollo , Intestino Delgado/microbiología , Especificidad de Órganos , Células de Paneth/citología , Células de Paneth/microbiología , Ratas
16.
Infect Immun ; 85(6)2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28348052

RESUMEN

In addition to their chemical antimicrobial nature, bile acids are thought to have other functions in the homeostatic control of gastrointestinal immunity. However, those functions have remained largely undefined. In this work, we used ileal explants and mouse models of bile acid administration to investigate the role of bile acids in the regulation of the intestinal antimicrobial response. Mice fed on a diet supplemented with 0.1% chenodeoxycholic acid (CDCA) showed an upregulated expression of Paneth cell α-defensins as well as an increased synthesis of the type-C lectins Reg3b and Reg3g by the ileal epithelium. CDCA acted on several epithelial cell types, by a mechanism independent from farnesoid X receptor (FXR) and not involving STAT3 or ß-catenin activation. CDCA feeding did not change the relative abundance of major commensal bacterial groups of the ileum, as shown by 16S analyses. However, administration of CDCA increased the expression of ileal Muc2 and induced a change in the composition of the mucosal immune cell repertoire, decreasing the proportion of Ly6G+ and CD68+ cells, while increasing the relative amount of IgGκ+ B cells. Oral administration of CDCA to mice attenuated infections with the bile-resistant pathogens Salmonella enterica serovar Typhimurium and Citrobacter rodentium, promoting lower systemic colonization and faster bacteria clearance, respectively. Our results demonstrate that bile acid signaling in the ileum triggers an antimicrobial program that can be potentially used as a therapeutic option against intestinal bacterial infections.


Asunto(s)
Ácido Quenodesoxicólico/administración & dosificación , Infecciones por Enterobacteriaceae/inmunología , Íleon/microbiología , Inmunidad Mucosa , Infecciones por Salmonella/inmunología , alfa-Defensinas/inmunología , Animales , Carga Bacteriana , Citrobacter rodentium/efectos de los fármacos , Íleon/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Células de Paneth/inmunología , Células de Paneth/microbiología , Salmonella typhimurium/efectos de los fármacos
17.
J Immunol ; 198(5): 2133-2146, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-28130498

RESUMEN

Intact ATG16L1 plays an essential role in Paneth cell function and intestinal homeostasis. However, the functional consequences of ATG16L1 deficiency in myeloid cells, particularly macrophages, are not fully characterized. We generated mice with Atg16l1 deficiency in myeloid and dendritic cells and showed that mice with myeloid Atg16l1 deficiency had exacerbated colitis in two acute and one chronic model of colitis with increased proinflammatory to anti-inflammatory macrophage ratios, production of proinflammatory cytokines, and numbers of IgA-coated intestinal microbes. Mechanistic analyses using primary murine macrophages showed that Atg16l1 deficiency led to increased reactive oxygen species production, impaired mitophagy, reduced microbial killing, impaired processing of MHC class II Ags, and altered intracellular trafficking to the lysosomal compartments. Increased production of reactive oxygen species and reduced microbial killing may be general features of the myeloid compartment, as they were also observed in Atg16l1-deficient primary murine neutrophils. A missense polymorphism (Thr300Ala) in the essential autophagy gene ATG16L1 is associated with Crohn disease (CD). Previous studies showed that this polymorphism leads to enhanced cleavage of ATG16L1 T300A protein and thus reduced autophagy. Similar findings were shown in primary human macrophages from controls and a population of CD patients carrying the Atg16l1 T300A risk variant and who were controlled for NOD2 CD-associated variants. This study revealed that ATG16L1 deficiency led to alterations in macrophage function that contribute to the severity of CD.


Asunto(s)
Proteínas Relacionadas con la Autofagia/metabolismo , Autofagia , Colitis/inmunología , Enfermedad de Crohn/inmunología , Intestinos/inmunología , Células Mieloides/fisiología , Proteína Adaptadora de Señalización NOD2/genética , Células de Paneth/inmunología , Infecciones por Salmonella/inmunología , Salmonella typhimurium/inmunología , Animales , Autofagia/genética , Autofagia/inmunología , Células Cultivadas , Enfermedad de Crohn/genética , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Predisposición Genética a la Enfermedad , Genotipo , Homeostasis , Interacciones Huésped-Patógeno , Humanos , Intestinos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Células de Paneth/microbiología , Polimorfismo Genético , Riesgo
18.
J Clin Gastroenterol ; 51(9): 814-817, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27636409

RESUMEN

BACKGROUND: We have previously shown a reduction of gastrointestinal symptoms after the oral administration of Bifidobacterium infantis Natren Life Start super strain (NLS-SS) in untreated celiac disease (CD) patients. The symptomatic improvement was not associated with changes in intestinal permeability or serum levels of cytokines, chemokines, or growth factors. Therefore, we hypothesized that the beneficial symptomatic effect observed previously in patients with CD treated with B. infantis may be related to the modulation of innate immunity. GOALS: To investigate the potential mechanisms of a probiotic B. infantis Natren Life Start super strain on the mucosal expression of innate immune markers in adult patients with active untreated CD compared with those treated with B. infantis×6 weeks and after 1 year of gluten-free diet (GFD). METHODS: Numbers of macrophages and Paneth cells and α-defensin-5 expression were assessed by immunohistochemistry in duodenal biopsies. RESULTS: We showed that GFD decreases duodenal macrophage counts in CD patients more effectively than B. infantis. In contrast, B. infantis decreases Paneth cell counts and expression of α-defensin-5 in CD (P<0.001). CONCLUSIONS: The results identify differential innate immune effects of treatment with B. infantis compared with 1 year of GFD. Further studies are needed to investigate synergistic effects of GFD and B. infantis supplementation in CD.


Asunto(s)
Bifidobacterium longum subspecies infantis/crecimiento & desarrollo , Enfermedad Celíaca/terapia , Dieta Sin Gluten , Duodeno/metabolismo , Inmunidad Innata , Inmunidad Mucosa , Mucosa Intestinal/metabolismo , Probióticos/uso terapéutico , alfa-Defensinas/metabolismo , Adulto , Biomarcadores/metabolismo , Biopsia , Enfermedad Celíaca/inmunología , Enfermedad Celíaca/metabolismo , Enfermedad Celíaca/microbiología , Regulación hacia Abajo , Duodeno/inmunología , Duodeno/microbiología , Femenino , Microbioma Gastrointestinal , Humanos , Inmunohistoquímica , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Masculino , Persona de Mediana Edad , Células de Paneth/inmunología , Células de Paneth/metabolismo , Células de Paneth/microbiología , Probióticos/efectos adversos , Factores de Tiempo , Resultado del Tratamiento , Adulto Joven
19.
Med Sci Monit ; 22: 3062-72, 2016 Aug 30.
Artículo en Inglés | MEDLINE | ID: mdl-27572949

RESUMEN

BACKGROUND Type 1 diabetes mellitus (T1DM) is associated with increased risks of enteric infection. Paneth cells constitute the first line of the gut defense. Little is known about the impact of T1DM on the bactericidal function of intestinal Paneth cells. MATERIAL AND METHODS A T1DM mouse model was induced by intraperitoneal injection of streptozocin. The analysis of intestinal microbiota and the mucosal bactericidal assay were conducted to evaluate intestinal innate defense. Numbers of Paneth cells and their expression of related antimicrobial peptides were analyzed. Expression of total insulin receptor (IR) mRNA and relative levels of IR-A/IR-B were analyzed. The primary mouse small intestinal crypt culture was used to analyze the effect of insulin and glucose on the expression of related antimicrobial peptides of Paneth cells. RESULTS In T1DM mice, bacterial loads were increased and there was an alteration in the composition of the intestinal microflora. Exogenous bacteria had better survival in the small bowel of the T1DM mice. The expression of Paneth cell-derived antimicrobial peptides was significantly decreased in the T1DM mice, although the number of Paneth cells was increased. Relative levels of IR-A/IR-B in Paneth cells of diabetic mice were elevated, but the total IR mRNA did not change. Insulin treatment restored the expression of antimicrobial peptides and normalized the microbiota in the gut of T1DM mice. Subsequently, in vitro culture assay demonstrated that insulin rather than glucose was essential for the optimal expression of Paneth cell-derived antimicrobial peptides. CONCLUSIONS The bactericidal function of intestinal Paneth cells was impaired in STZ-induced diabetic mice, resulting in the altered intestinal flora, and insulin was essential for the optimal expression of Paneth cell-derived antimicrobial peptides.


Asunto(s)
Diabetes Mellitus Experimental/inmunología , Insulina/deficiencia , Células de Paneth/inmunología , Animales , Péptidos Catiónicos Antimicrobianos/biosíntesis , Péptidos Catiónicos Antimicrobianos/inmunología , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/microbiología , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/microbiología , Inmunidad Innata , Insulina/administración & dosificación , Insulina/sangre , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Ratones , Ratones Endogámicos C57BL , Microbiota , Células de Paneth/microbiología , Distribución Aleatoria , Receptor de Insulina/biosíntesis , Receptor de Insulina/deficiencia , Receptor de Insulina/metabolismo
20.
Gut Microbes ; 7(5): 414-23, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27459363

RESUMEN

Doxorubicin (DOXO) induces significant, but transient, increases in apoptosis in the stem cell zone of the jejunum, followed by mucosal damage involving a decrease in crypt proliferation, crypt number, and villus height. The gastrointestinal tract is home to a vast population of commensal bacteria and numerous studies have demonstrated a symbiotic relationship between intestinal bacteria and intestinal epithelial cells (IEC) in maintaining homeostatic functions of the intestine. However, whether enteric bacteria play a role in DOXO-induced damage is not well understood. We hypothesized that enteric bacteria are necessary for induction of apoptosis and damage associated with DOXO treatment. Conventionally raised (CONV) and germ free (GF) mice were given a single injection of DOXO, and intestinal tissue was collected at 6, 72, and 120 h after treatment and from no treatment (0 h) controls. Histology and morphometric analyses quantified apoptosis, mitosis, crypt depth, villus height, and crypt density. Immunostaining for muc2 and lysozyme evaluated Paneth cells, goblet cells or dual stained intermediate cells. DOXO administration induced significant increases in apoptosis in jejunal epithelium regardless of the presence of enteric bacteria; however, the resulting injury, as demonstrated by statistically significant changes in crypt depth, crypt number, and proliferative cell number, was dependent upon the presence of enteric bacteria. Furthermore, we observed expansion of Paneth and goblet cells and presence of intermediate cells only in CONV and not GF mice. These findings provide evidence that manipulation and/or depletion of the enteric microbiota may have clinical significance in limiting chemotherapy-induced mucositis.


Asunto(s)
Antibióticos Antineoplásicos/efectos adversos , Apoptosis/efectos de los fármacos , Bacterias/efectos de los fármacos , Doxorrubicina/efectos adversos , Microbioma Gastrointestinal/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/microbiología , Animales , Antibióticos Antineoplásicos/administración & dosificación , Bacterias/genética , Bacterias/aislamiento & purificación , Doxorrubicina/administración & dosificación , Femenino , Vida Libre de Gérmenes , Células Caliciformes/efectos de los fármacos , Células Caliciformes/microbiología , Intestinos/citología , Ratones , Ratones Endogámicos C57BL , Células de Paneth/efectos de los fármacos , Células de Paneth/microbiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...