Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 435
Filtrar
1.
Nat Cardiovasc Res ; 3(5): 525-540, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-39195931

RESUMEN

Post-injury dysfunction of humoral immunity accounts for infections and poor outcomes in cardiovascular diseases. Among immunoglobulins (Ig), IgA, the most abundant mucosal antibody, is produced by plasma B cells in intestinal Peyer's patches (PP) and lamina propria. Here we show that patients with stroke and myocardial ischemia (MI) had strongly reduced IgA blood levels. This was phenocopied in experimental mouse models where decreased plasma and fecal IgA were accompanied by rapid loss of IgA-producing plasma cells in PP and lamina propria. Reduced plasma IgG was detectable in patients and experimental mice 3-10 d after injury. Stroke/MI triggered the release of neutrophil extracellular traps (NETs). Depletion of neutrophils, NET degradation or blockade of NET release inhibited the loss of IgA+ cells and circulating IgA in experimental stroke and MI and in patients with stroke. Our results unveil how tissue-injury-triggered systemic NET release disrupts physiological Ig secretion and how this can be inhibited in patients.


Asunto(s)
Trampas Extracelulares , Infarto del Miocardio , Neutrófilos , Trampas Extracelulares/metabolismo , Trampas Extracelulares/inmunología , Humanos , Animales , Infarto del Miocardio/inmunología , Infarto del Miocardio/patología , Infarto del Miocardio/metabolismo , Masculino , Neutrófilos/inmunología , Neutrófilos/metabolismo , Femenino , Modelos Animales de Enfermedad , Ratones Endogámicos C57BL , Accidente Cerebrovascular/inmunología , Accidente Cerebrovascular/patología , Accidente Cerebrovascular/metabolismo , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/patología , Ganglios Linfáticos Agregados/metabolismo , Inmunoglobulina A/metabolismo , Inmunoglobulina A/inmunología , Inmunoglobulina A/sangre , Anciano , Persona de Mediana Edad , Inmunoglobulina G/inmunología , Inmunoglobulina G/metabolismo , Inmunidad Humoral , Estudios de Casos y Controles , Ratones , Células Plasmáticas/inmunología , Células Plasmáticas/metabolismo
2.
Wiad Lek ; 75(5 pt 2): 1309-1312, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35758449

RESUMEN

OBJECTIVE: The aim: Based on the above cytological signs of M-cells, we set the goal of more detailed clarification of some of their topological relationships with other enterocytes in the follicle-associated epithelium of Peyer's patches of albino rat small intestine. PATIENTS AND METHODS: Materials and methods: 10 mature albino male rats weighted 200,0±20,0 g were involved into the study. Anatomical dissection with the sampling of the sections of the small intestine containing Peyer's patches was carried out with subsequent embedment of the latter into paraffin blocks and making of serial histological sections of 4 µm thick in the cross-section of the small intestine, followed with hematoxylin-eosin staining. The specimens were studied and documented on the "Konus" light microscope equipped. Morphometric characteristics of the specimen tissue structures were studied using the Sigeta X 1 mm/100 Div.x0.01mm stage micrometer. RESULTS: Results: The findings of the study revealed enterocytes with phagocytic properties found in the lymphoid-associated epithelium of Peyer's patches of the small intestine of albino rats. Moreover, if they are clearly visualized at the light-optical level, then M-cells are poorly recognizable, which is consistent with a similar assessment made by other authors. CONCLUSION: Conclusions: Given this, the issue on the topology and functional purpose of M-cells remains uncertain to date and, thereby, the prospect of further research is being outlined, which, in our opinion, can be successful using the method of stereomorphological analysis. For this purpose, multilayer plastic reconstruction methods can be used for serial semi-thin sections of Peyer's patches embedded in epoxy resin, according to the requirements of transmission electron microscopy.


Asunto(s)
Intestino Delgado , Ganglios Linfáticos Agregados , Epitelio , Humanos , Ganglios Linfáticos Agregados/química , Ganglios Linfáticos Agregados/patología , Ratas , Coloración y Etiquetado
3.
Front Immunol ; 12: 697725, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34804004

RESUMEN

The intestinal mucosal immune environment requires multiple immune cells to maintain homeostasis. Although intestinal B cells are among the most important immune cells, little is known about the mechanism that they employ to regulate immune homeostasis. In this study, we found that CD11b+ B cells significantly accumulated in the gut lamina propria and Peyer's patches in dextran sulfate sodium-induced colitis mouse models and patients with ulcerative colitis. Adoptive transfer of CD11b+ B cells, but not CD11b-/- B cells, effectively ameliorated colitis and exhibited therapeutic effects. Furthermore, CD11b+ B cells were found to produce higher levels of IgA than CD11b- B cells. CD11b deficiency in B cells dampened IgA production, resulting in the loss of their ability to ameliorate colitis. Mechanistically, CD11b+ B cells expressed abundant TGF-ß and TGF-ß receptor II, as well as highly activate phosphorylated Smad2/3 signaling pathway, consequently promoting the class switch to IgA. Collectively, our findings demonstrate that CD11b+ B cells are essential intestinal suppressive immune cells and the primary source of intestinal IgA, which plays an indispensable role in maintaining intestinal homeostasis.


Asunto(s)
Linfocitos B/inmunología , Antígeno CD11b/inmunología , Colitis Ulcerosa/inmunología , Colitis/inmunología , Inmunoglobulina A Secretora/inmunología , Ganglios Linfáticos Agregados/inmunología , Traslado Adoptivo , Animales , Linfocitos B/patología , Antígeno CD11b/genética , Colitis/inducido químicamente , Colitis/patología , Colitis Ulcerosa/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Humanos , Cambio de Clase de Inmunoglobulina , Mucosa Intestinal/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ganglios Linfáticos Agregados/patología , Transducción de Señal , Proteína Smad2/metabolismo
4.
Virology ; 552: 43-51, 2021 01 02.
Artículo en Inglés | MEDLINE | ID: mdl-33059319

RESUMEN

This study focused on intestinal restitution including phenotype switching of absorptive enterocytes and the abundance of different enterocyte subtypes in weaned pigs after porcine epidemic diarrhea virus (PEDV) infection. At 10 days post-PEDV-inoculation, the ratio of villus height to crypt depth in both jejunum and ileum had restored, and the PEDV antigen was not detectable. However, enterocytes at the villus tips revealed epithelial-mesenchymal transition (EMT) in the jejunum in which E-cadherin expression decreased while expression of N-cadherin, vimentin, and Snail increased. Additionally, there was reduced expression of actin in microvilli and Zonula occludens-1 (ZO-1) in tight junctions. Moreover, the protein concentration of transforming growth factor ß1 (TGFß1), which mediates EMT and cytoskeleton alteration, was increased. We also found a decreased number of Peyer's patch M cells in the ileum. These results reveal incomplete restitution of enterocytes in the jejunum and potentially impaired immune surveillance in the ileum after PEDV infection.


Asunto(s)
Infecciones por Coronavirus/veterinaria , Enterocitos/patología , Transición Epitelial-Mesenquimal , Gastroenteritis Porcina Transmisible/patología , Ganglios Linfáticos Agregados/patología , Virus de la Diarrea Epidémica Porcina/patogenicidad , Animales , Cadherinas/metabolismo , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Infecciones por Coronavirus/virología , Gastroenteritis Porcina Transmisible/inmunología , Gastroenteritis Porcina Transmisible/virología , Íleon/inmunología , Íleon/patología , Mucosa Intestinal/patología , Yeyuno/inmunología , Yeyuno/patología , Microvellosidades/patología , Porcinos , Uniones Estrechas/patología , Factor de Crecimiento Transformador beta1/metabolismo , Destete
5.
Nutrients ; 12(11)2020 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-33158132

RESUMEN

There is no effective therapy for milk allergy. The role of lactic acid bacteria (LAB) and probiotics in protection against allergy-related outcomes is still under investigation. The aim of the study was to evaluate the immunomodulative and therapeutic potential of yogurt drinks in cow's milk allergy (CMA) management. We compared immunoreactivity of α-casein (α-CN), ß-casein (ß-CN), κ-casein (κ-CN), α-lactalbumin (α-LA), and ß-lactoglobulin (ß-LG) in 27 yogurt drinks fermented with different basic yogurt cultures, or yogurt cultures enriched with Lactobacillus plantarum and/or Bifidobacterium lactis strains, by competitive ELISA assay. Drinks with the lowest antigenic potential were used as allergoids for CMA therapy. BALB/c mice were sensitized via intraperitoneal injection of α-CN + ß-LG mixture with aluminum adjuvant, and gavaged with increasing doses of selected low-immunogenic drinks (YM-basic, or YM-LB-enriched with L. plantarum and B. lactis) to induce tolerance. Milk- or phosphate-buffered saline (PBS)-dosed mice served as controls. Compared to milk, the immunoreactivity of proteins in drinks increased or decreased, depending on the bacterial sets applied for fermentation. Only a few sets acted synergistically in reducing immunoreactivity. The selected low-immunogenic drinks stimulated allergic mice for profiling Th2 to Th1 response and acquire tolerance, and the effect was greater with YM-LB drink, which during long-lasting interventional feeding strongly increased the secretion of regulatory cytokines, i.e., IL-10 and TGF-ß, and IgA and decreased IL-4, IgE, and anti-(α-CN + ß-LG) IgG1. The studies revealed variations in the potency of yogurt bacteria to change allergenicity of milk proteins and the need for their strict selection to obtain a safe product for allergy sufferers. The YM-LB drink with reduced antigenic potential may be a source of allergoids used in the immunotherapy of IgE mediated CMA, but further clinical or volunteer studies are required.


Asunto(s)
Bacterias/inmunología , Tolerancia Inmunológica , Proteínas de la Leche/inmunología , Leche/microbiología , Probióticos/farmacología , Yogur/microbiología , Animales , Peso Corporal , Caseínas/inmunología , Ciego/microbiología , Citocinas/metabolismo , Conducta Alimentaria , Femenino , Fermentación , Microbioma Gastrointestinal , Inmunidad Humoral , Ratones Endogámicos BALB C , Ganglios Linfáticos Agregados/patología , Bazo/patología , Proteína de Suero de Leche/inmunología
7.
J Immunol ; 204(10): 2697-2711, 2020 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-32238457

RESUMEN

The adaptive immune system of all jawed vertebrates relies on the presence of B and T cell lymphocytes that aggregate in specific body sites to form primary and secondary lymphoid structures. Secondary lymphoid organs include organized MALT (O-MALT) such as the tonsils and Peyer patches. O-MALT became progressively organized during vertebrate evolution, and the TNF superfamily of genes has been identified as essential for the formation and maintenance of O-MALT and other secondary and tertiary lymphoid structures in mammals. Yet, the molecular drivers of O-MALT structures found in ectotherms and birds remain essentially unknown. In this study, we provide evidence that TNFSFs, such as lymphotoxins, are likely not a universal mechanism to maintain O-MALT structures in adulthood of teleost fish, sarcopterygian fish, or birds. Although a role for TNFSF2 (TNF-α) cannot be ruled out, transcriptomics suggest that maintenance of O-MALT in nonmammalian vertebrates relies on expression of diverse genes with shared biological functions in neuronal signaling. Importantly, we identify that expression of many genes with olfactory function is a unique feature of mammalian Peyer patches but not the O-MALT of birds or ectotherms. These results provide a new view of O-MALT evolution in vertebrates and indicate that different genes with shared biological functions may have driven the formation of these lymphoid structures by a process of convergent evolution.


Asunto(s)
Linfocitos B/inmunología , Tejido Linfoide/inmunología , Membrana Mucosa/patología , Tonsila Palatina/patología , Ganglios Linfáticos Agregados/patología , Linfocitos T/inmunología , Factores de Necrosis Tumoral/metabolismo , Inmunidad Adaptativa , Animales , Evolución Biológica , Inmunidad Mucosa , Conducción Nerviosa , Tonsila Palatina/inmunología , Ganglios Linfáticos Agregados/inmunología , Transcriptoma , Factor de Necrosis Tumoral alfa/metabolismo , Factores de Necrosis Tumoral/genética , Vertebrados
8.
Nutrients ; 12(3)2020 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-32183266

RESUMEN

Some lactic acid bacteria (LAB) are known to improve atopic dermatitis (AD) through the regulation and stimulation of the host immune system. In this study, we found that ingestion of yogurt containing Lactococcus lactis 11/19-B1 strain (L. lactis 11/19-B1) daily for 8 weeks significantly improved the severity scoring of atopic dermatitis (SCORAD) system score from 38.8 ± 14.4 to 24.2 ± 12.0 in children suffering from AD. We tried to identify which LAB species among the five species contained in the test yogurt contributed to the improvement in AD pathology using an AD mouse model induced by repeated application of 1-fluoro-2, 4-dinitrobenzene (DNFB). AD-like skin lesions on the dorsal skin and ear were most improved by L. lactis 11/19-B1 intake among the five LAB species. In addition, analysis of CD4+ T cell subsets in Peyer's patches (PPs) and cervical lymph nodes (CLNs) indicated that the intake of L. lactis 11/19-B1 generally suppressed all subsets related to inflammation, i.e., Th1, Th2 and Th17, instead of activating the suppressive system, Treg, in the AD mouse model. Histological observations showed ingestion of L. lactis 11/19-B1 significantly suppressed severe inflammatory findings, such as inflammatory cell filtration, epidermal erosion and eosinophil infiltration. These results suggest that the immunomodulatory effects of L. lactis 11/19-B1 contribute to improvements in AD pathology.


Asunto(s)
Dermatitis Atópica , Lactococcus lactis/inmunología , Piel , Yogur , Adolescente , Animales , Niño , Preescolar , Dermatitis Atópica/dietoterapia , Dermatitis Atópica/inmunología , Dermatitis Atópica/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/patología , Piel/inmunología , Piel/patología , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/patología
9.
J Immunol ; 204(5): 1075-1083, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-32071090

RESUMEN

The mammalian intestine is a complex environment that is constantly exposed to Ags derived from food, microbiota, and metabolites. Intestinal dendritic cells (DC) have the responsibility of establishing oral tolerance against these Ags while initiating immune responses against mucosal pathogens. We now know that DC are a heterogeneous population of innate immune cells composed of classical and monocyte-derived DC, Langerhans cells, and plasmacytoid DC. In the intestine, DC are found in organized lymphoid tissues, such as the mesenteric lymph nodes and Peyer's patches, as well as in the lamina propria. In this Brief Review, we review recent work that describes a division of labor between and collaboration among gut DC subsets in the context of intestinal homeostasis and inflammation. Understanding relationships between DC subtypes and their biological functions will rationalize oral vaccine design and will provide insights into treatments that quiet pathological intestinal inflammation.


Asunto(s)
Inmunidad Mucosa , Mucosa Intestinal/inmunología , Células de Langerhans/inmunología , Ganglios Linfáticos Agregados/inmunología , Animales , Humanos , Inflamación/inmunología , Inflamación/patología , Mucosa Intestinal/patología , Células de Langerhans/patología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Mesenterio/inmunología , Mesenterio/patología , Ganglios Linfáticos Agregados/patología
10.
J Gastroenterol Hepatol ; 35(7): 1143-1149, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-31734952

RESUMEN

BACKGROUND AND AIM: Peyer's patches (PPs) play a major role in intestinal mucosal immunity; however, their role in ulcerative colitis (UC) is not well investigated. We evaluated endoscopic features of PPs on narrow-band imaging with magnifying endoscopy (NBI-ME) and investigated their association with clinical factors. METHODS: We prospectively recruited 105 patients with UC, 18 with Crohn's disease, 16 with disease control, and 33 healthy control subjects at three institutions from 2014 to 2017. NBI-ME images of the villi of PPs were evaluated according to the Villi Index, and patients were divided into the Villi Index low (L) and high (H) types. The 1-year sustained clinical remission rate was evaluated between L-type and H-type PPs in patients with UC. RESULTS: The proportions of patients with H-type PPs were significantly higher among UC, Crohn's disease, and disease control patients than among healthy control patients (P = 0.0125, 0.018, 0.0007). In UC, age, gender, endoscopic score, and extent of disease involvement were not significantly different between L-type and H-type PPs, whereas the sustained clinical remission rate was significantly higher in L-type PPs than in H-type PPs (88% [57/65] vs 65% [17/26], P = 0.019). Multivariate analysis revealed that the L type of PPs was a significant factor for sustained clinical remission (odds ratio 3.8, 95% confidence interval 1.1-12.9, P = 0.033). CONCLUSIONS: Patients with UC showed endoscopic alterations in PPs on NBI-ME, and highly altered appearance of PPs can be associated with a high risk of clinical relapse in patients with UC.


Asunto(s)
Colitis Ulcerosa/diagnóstico por imagen , Colitis Ulcerosa/patología , Endoscopía Gastrointestinal/métodos , Ganglios Linfáticos Agregados/diagnóstico por imagen , Ganglios Linfáticos Agregados/patología , Adolescente , Adulto , Niño , Femenino , Humanos , Masculino , Imagen de Banda Estrecha/métodos , Estudios Prospectivos , Recurrencia , Inducción de Remisión , Riesgo , Adulto Joven
11.
J Allergy Clin Immunol ; 145(3): 982-992, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-31816409

RESUMEN

BACKGROUND: Kabuki syndrome (KS) is commonly caused by mutations in the histone-modifying enzyme lysine methyltransferase 2D (KMT2D). Immune dysfunction is frequently observed in individuals with KS, but the role of KMT2D in immune system function has not been identified. OBJECTIVE: We sought to understand the mechanisms driving KS-associated immune deficiency (hypogammaglobulinemia [low IgA], splenomegaly, and diminished immunization responses). METHODS: We performed a comprehensive evaluation of humoral immunity and secondary lymphoid tissues in an established KS (Kmt2d+/ßGeo) mouse model and validated select findings in a patient with KS. RESULTS: Compared with wild-type littermates, Kmt2d+/ßGeo mice demonstrated deficiencies in multiple B-cell lineages and reduced serum IgA and elevated IgM levels across multiple ages. The bone marrow, spleen, and intestine of Kmt2d+/ßGeo mice contained diminished numbers of IgA-secreting cells, while elevated germinal center B cells were found in the mesenteric lymph node and Peyer patches. Kmt2d+/ßGeo mice have decreased size and numbers of Peyer patches, a finding confirmed in human samples. We identified deficiency of Itgb7 RNA and protein expression, a gene encoding an adhesion protein that mediates intestinal homing, and we demonstrated KMT2D-dependent control of ITGB7 expression in a human cell line. CONCLUSIONS: Kmt2d haploinsufficiency has broad deleterious effects on B-cell differentiation, specifically hampering gut lymphocyte homing and IgA+ plasma cell differentiation. Intestinal lymphoid defects caused by ITGB7 deficiency have not previously been recognized in KS, and these results provide new mechanistic insights into the pathogenesis of KS-associated immune deficiency.


Asunto(s)
Anomalías Múltiples/inmunología , Anomalías Múltiples/patología , Linfocitos B/patología , Cara/anomalías , Enfermedades Hematológicas/inmunología , Enfermedades Hematológicas/patología , Ganglios Linfáticos Agregados/patología , Enfermedades Vestibulares/inmunología , Enfermedades Vestibulares/patología , Animales , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Movimiento Celular/inmunología , Proteínas de Unión al ADN/genética , Cara/patología , N-Metiltransferasa de Histona-Lisina/genética , Humanos , Deficiencia de IgA/genética , Deficiencia de IgA/inmunología , Cadenas beta de Integrinas/metabolismo , Intestinos/inmunología , Ratones , Mutación , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Ganglios Linfáticos Agregados/inmunología
12.
Front Immunol ; 10: 2345, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31649668

RESUMEN

Microfold (M) cells are located in the epithelium covering mucosa-associated lymphoid tissues, such as the Peyer's patches (PPs) of the small intestine. M cells actively transport luminal antigens to the underlying lymphoid follicles to initiate an immune response. The molecular machinery of M-cell differentiation and function has been vigorously investigated over the last decade. Studies have shed light on the role of M cells in the mucosal immune system and have revealed that antigen uptake by M cells contributes to not only mucosal but also systemic immune responses. However, M-cell studies usually focus on infectious diseases; the contribution of M cells to autoimmune diseases has remained largely unexplored. Accumulating evidence suggests that dysbiosis of the intestinal microbiota is implicated in multiple systemic diseases, including autoimmune diseases. This implies that the uptake of microorganisms by M cells in PPs may play a role in the pathogenesis of autoimmune diseases. We provide an outline of the current understanding of M-cell biology and subsequently discuss the potential contribution of M cells and PPs to the induction of systemic autoimmunity, beyond the mucosal immune response.


Asunto(s)
Enfermedades Autoinmunes , Microbioma Gastrointestinal/inmunología , Inmunidad Mucosa , Ganglios Linfáticos Agregados , Animales , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/microbiología , Enfermedades Autoinmunes/patología , Disbiosis/inmunología , Disbiosis/microbiología , Disbiosis/patología , Humanos , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/microbiología , Ganglios Linfáticos Agregados/patología
13.
Cell ; 178(5): 1072-1087.e14, 2019 08 22.
Artículo en Inglés | MEDLINE | ID: mdl-31442401

RESUMEN

Nutritional status potentially influences immune responses; however, how nutritional signals regulate cellular dynamics and functionality remains obscure. Herein, we report that temporary fasting drastically reduces the number of lymphocytes by ∼50% in Peyer's patches (PPs), the inductive site of the gut immune response. Subsequent refeeding seemingly restored the number of lymphocytes, but whose cellular composition was conspicuously altered. A large portion of germinal center and IgA+ B cells were lost via apoptosis during fasting. Meanwhile, naive B cells migrated from PPs to the bone marrow during fasting and then back to PPs during refeeding when stromal cells sensed nutritional signals and upregulated CXCL13 expression to recruit naive B cells. Furthermore, temporal fasting before oral immunization with ovalbumin abolished the induction of antigen-specific IgA, failed to induce oral tolerance, and eventually exacerbated food antigen-induced diarrhea. Thus, nutritional signals are critical in maintaining gut immune homeostasis.


Asunto(s)
Linfocitos B/fisiología , Inmunidad Mucosa , Animales , Antígenos/inmunología , Linfocitos B/inmunología , Linfocitos B/metabolismo , Médula Ósea/inmunología , Médula Ósea/metabolismo , Quimiocina CXCL13/genética , Quimiocina CXCL13/metabolismo , Ayuno , Regulación de la Expresión Génica , Glucólisis , Inmunoglobulina A/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Estado Nutricional , Ovalbúmina/inmunología , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/metabolismo , Ganglios Linfáticos Agregados/patología , Receptores CXCR5/genética , Receptores CXCR5/metabolismo , Transducción de Señal , Células del Estroma/citología , Células del Estroma/metabolismo , Serina-Treonina Quinasas TOR/metabolismo
14.
JCI Insight ; 52019 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-31162138

RESUMEN

Excess dietary salt contributes to inflammation and hypertension via poorly understood mechanisms. Antigen presenting cells including dendritic cells (DCs) play a key role in regulating intestinal immune homeostasis in part by surveying the gut epithelial surface for pathogens. Previously, we found that highly reactive γ-ketoaldehydes or isolevuglandins (IsoLGs) accumulate in DCs and act as neoantigens, promoting an autoimmune-like state and hypertension. We hypothesized that excess dietary salt alters the gut microbiome leading to hypertension and this is associated with increased immunogenic IsoLG-adduct formation in myeloid antigen presenting cells. To test this hypothesis, we performed fecal microbiome analysis and measured blood pressure of healthy human volunteers with salt intake above or below the American Heart Association recommendations. We also performed 16S rRNA analysis on cecal samples of mice fed normal or high salt diets. In humans and mice, high salt intake was associated with changes in the gut microbiome reflecting an increase in Firmicutes, Proteobacteria and genus Prevotella bacteria. These alterations were associated with higher blood pressure in humans and predisposed mice to vascular inflammation and hypertension in response to a sub-pressor dose of angiotensin II. Mice fed a high salt diet exhibited increased intestinal inflammation including the mesenteric arterial arcade and aorta, with a marked increase in the B7 ligand CD86 and formation of IsoLG-protein adducts in CD11c+ myeloid cells. Adoptive transfer of fecal material from conventionally housed high salt-fed mice to germ-free mice predisposed them to increased intestinal inflammation and hypertension. These findings provide novel insight into the mechanisms underlying inflammation and hypertension associated with excess dietary salt and may lead to interventions targeting the microbiome to prevent and treat this important disease.


Asunto(s)
Células Dendríticas/efectos de los fármacos , Células Dendríticas/metabolismo , Disbiosis , Hipertensión/metabolismo , Cloruro de Sodio Dietético/efectos adversos , Cloruro de Sodio/efectos adversos , Adolescente , Traslado Adoptivo , Adulto , Angiotensina II , Animales , Aorta/metabolismo , Bacterias/clasificación , Bacterias/genética , Presión Sanguínea , Antígeno CD11c/inmunología , Colon/microbiología , Colon/patología , Citocinas/metabolismo , Células Dendríticas/patología , Modelos Animales de Enfermedad , Femenino , Microbioma Gastrointestinal , Humanos , Inflamación/metabolismo , Lípidos , Ganglios Linfáticos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Células Mieloides/metabolismo , Ganglios Linfáticos Agregados/microbiología , Ganglios Linfáticos Agregados/patología , ARN Ribosómico 16S/genética , Cloruro de Sodio/administración & dosificación , Cloruro de Sodio Dietético/administración & dosificación , Adulto Joven
15.
Mycopathologia ; 184(4): 461-478, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31230200

RESUMEN

The details of how gut-associated lymphoid tissues such as Peyer's patches (PPs) in the small intestine play a role in immune surveillance, microbial differentiation and the mucosal barrier protection in response to fungal organisms such as Candida albicans are still unclear. We particularly focus on PPs as they are the immune sensors and inductive sites of the gut that influence inflammation and tolerance. We have previously demonstrated that CD11c+ phagocytes that include dendritic cells and macrophages are located in the sub-epithelial dome within PPs sample C. albicans. To gain insight on how specific cells within PPs sense and respond to the sampling of fungi, we gavaged naïve mice with C. albicans strains ATCC 18804 and SC5314 as well as Saccharomyces cerevisiae. We measured the differential gene expression of sorted CD45+ B220+ B-cells, CD3+ T-cells and CD11c+ DCs within the first 24 h post-gavage using nanostring nCounter® technology. The results reveal that at 24 h, PP phagocytes were the cell type that displayed differential gene expression. These phagocytes were able to sample C. albicans and discriminate between strains. In particular, strain ATCC 18804 upregulated fungal-specific pro-inflammatory genes pertaining to innate and adaptive immune responses. Interestingly, PP CD11c+ phagocytes also differentially expressed genes in response to C. albicans that were important in the protection of the mucosal barrier. These results highlight that the mucosal barrier not only responds to C. albicans, but also aids in the protection of the host.


Asunto(s)
Candida albicans/inmunología , Perfilación de la Expresión Génica , Interacciones Huésped-Patógeno , Inflamación/patología , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/patología , Administración Oral , Animales , Antígenos CD/análisis , Linfocitos B/química , Linfocitos B/inmunología , Células Dendríticas/química , Células Dendríticas/inmunología , Femenino , Ratones , Saccharomyces cerevisiae/inmunología , Linfocitos T/química , Linfocitos T/inmunología
16.
Sci Rep ; 9(1): 6674, 2019 04 30.
Artículo en Inglés | MEDLINE | ID: mdl-31040320

RESUMEN

The early replication of some orally-acquired prion strains upon stromal-derived follicular dendritic cells (FDC) within the small intestinal Peyer's patches is essential to establish host infection, and for the disease to efficiently spread to the brain. Factors that influence the early accumulation of prions in Peyer's patches can directly influence disease pathogenesis. The host's immune response to a gastrointestinal helminth infection can alter susceptibility to co-infection with certain pathogenic bacteria and viruses. Here we used the natural mouse small intestine-restricted helminth pathogen Heligmosomoides polygyrus to test the hypothesis that pathology specifically within the small intestine caused by a helminth co-infection would influence oral prion disease pathogenesis. When mice were co-infected with prions on d 8 after H. polygyrus infection the early accumulation of prions within Peyer's patches was reduced and survival times significantly extended. Natural prion susceptible hosts such as sheep, deer and cattle are regularly exposed to gastrointestinal helminth parasites. Our data suggest that co-infections with small intestine-restricted helminth pathogens may be important factors that influence oral prion disease pathogenesis.


Asunto(s)
Coinfección , Susceptibilidad a Enfermedades , Helmintiasis Animal/complicaciones , Helmintiasis Animal/parasitología , Helmintos , Parasitosis Intestinales/complicaciones , Parasitosis Intestinales/parasitología , Enfermedades por Prión/etiología , Animales , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades/inmunología , Helmintiasis Animal/patología , Inmunidad Mucosa , Inmunohistoquímica , Parasitosis Intestinales/patología , Ratones , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/patología
17.
Front Immunol ; 10: 361, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30891036

RESUMEN

The intestinal mucosa in inflammatory bowel disease (IBD) contains increased frequencies of lymphocytes and a disproportionate increase in plasma cells secreting immunoglobulin (Ig)G relative to other isotypes compared to healthy controls. Despite consistent evidence of B lineage cells in the mucosa in IBD, little is known of B cell recruitment to the gut in IBD. Here we analyzed B cells in blood of patients with Crohn's disease (CD) and ulcerative colitis (UC) with a range of disease activities. We analyzed the frequencies of known B cell subsets in blood and observed a consistent reduction in the proportion of CD27-IgD- B cells expressing all Ig isotypes in the blood in IBD (independent of severity of disease and treatment) compared to healthy controls. Successful treatment of patients with biologic therapies did not change the profile of B cell subsets in blood. By mass cytometry we demonstrated that CD27-IgD- B cells were proportionately enriched in the gut-associated lymphoid tissue (GALT) in IBD. Since production of TNFα is a feature of IBD relevant to therapies, we sought to determine whether B cells in GALT or the CD27-IgD- subset in particular could contribute to pathology by secretion of TNFα or IL-10. We found that donor matched GALT and blood B cells are capable of producing TNFα as well as IL-10, but we saw no evidence that CD27-IgD- B cells from blood expressed more TNFα compared to other subsets. The reduced proportion of CD27-IgD- B cells in blood and the increased proportion in the gut implies that CD27-IgD- B cells are recruited from the blood to the gut in IBD. CD27-IgD- B cells have been implicated in immune responses to intestinal bacteria and recruitment to GALT, and may contribute to the intestinal inflammatory milieu in IBD.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Colitis Ulcerosa/sangre , Colitis Ulcerosa/inmunología , Enfermedad de Crohn/sangre , Enfermedad de Crohn/inmunología , Inmunoglobulina D/metabolismo , Mucosa Intestinal/inmunología , Ganglios Linfáticos Agregados/inmunología , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Adulto , Anticuerpos Monoclonales Humanizados/uso terapéutico , Biopsia , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/patología , Enfermedad de Crohn/tratamiento farmacológico , Enfermedad de Crohn/patología , Femenino , Fármacos Gastrointestinales/uso terapéutico , Humanos , Infliximab/uso terapéutico , Interleucina-10/metabolismo , Mucosa Intestinal/patología , Masculino , Ganglios Linfáticos Agregados/patología , Índice de Severidad de la Enfermedad , Factor de Necrosis Tumoral alfa/metabolismo , Ustekinumab/uso terapéutico
18.
Radiother Oncol ; 132: 8-15, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30825974

RESUMEN

OBJECTIVE: Exposure to lethal doses of radiation has severe effects on normal tissues. Exposed individuals experience a plethora of symptoms in different organ systems including the gastrointestinal (GI) tract, summarized as Acute Radiation Syndrome (ARS). There are currently no approved drugs for mitigating GI-ARS. A recent high-throughput screen performed at the UCLA Center for Medical Countermeasures against Radiation identified compounds containing sulfonylpiperazine groups with radiation mitigation properties to the hematopoietic system and the gut. Among these 1-[(4-Nitrophenyl)sulfonyl]-4-phenylpiperazine (Compound #5) efficiently mitigated gastrointestinal ARS. However, the mechanism of action and target cells of this drug is still unknown. In this study we examined if Compound #5 affects gut-associated lymphoid tissue (GALT) with its subepithelial domes called Peyer's patches. METHODS: C3H mice were irradiated with 0 or 12 Gy total body irradiation (TBI). A single dose of Compound #5 or solvent was administered subcutaneously 24 h later. 48 h after irradiation the mice were sacrificed, and the guts examined for changes in the number of visible Peyer's patches. In some experiments the mice received 4 daily injections of treatment and were sacrificed 96 h after TBI. For immune histochemistry gut tissues were fixed in formalin and embedded in paraffin blocks. Sections were stained with H&E, anti-Ki67 or a TUNEL assay to assess the number of regenerating crypts, mitotic and apoptotic indices. Cells isolated from Peyer's patches were subjected to immune profiling using flow cytometry. RESULTS: Compound #5 significantly increased the number of visible Peyer's patches when compared to its control in non-irradiated and irradiated mice. Additionally, assessment of total cells per Peyer's patch isolated from these mice demonstrated an overall increase in the total number of Peyer's patch cells per mouse in Compound #5-treated mice. In non-irradiated animals the number of CD11bhigh in Peyer's patches increased significantly. These Compound #5-driven increases did not coincide with a decrease in apoptosis or an increase in proliferation in the germinal centers inside Peyer's patches 24 h after drug treatment. A single dose of Compound #5 significantly increased the number of CD45+ cells after 12 Gy TBI. Importantly, 96 h after 12 Gy TBI Compound #5 induced a significant rise in the number of visible Peyer's patches and the number of Peyer's patch-associated regenerating crypts. CONCLUSION: In summary, our study provides evidence that Compound #5 leads to an influx of immune cells into GALT, thereby supporting crypt regeneration preferentially in the proximity of Peyer's patches.


Asunto(s)
Intestino Delgado/efectos de los fármacos , Nitrobencenos/farmacología , Ganglios Linfáticos Agregados/efectos de los fármacos , Piperazinas/farmacología , Traumatismos por Radiación/tratamiento farmacológico , Protectores contra Radiación/farmacología , Regeneración/efectos de los fármacos , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Femenino , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Intestino Delgado/patología , Intestino Delgado/efectos de la radiación , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/patología , Ganglios Linfáticos Agregados/efectos de la radiación , Traumatismos por Radiación/patología , Distribución Aleatoria , Regeneración/efectos de la radiación , Irradiación Corporal Total
19.
Nat Commun ; 9(1): 4870, 2018 11 19.
Artículo en Inglés | MEDLINE | ID: mdl-30451821

RESUMEN

RORγt controls the differentiation of TH17 cells, which are mediators of autoimmune conditions such as experimental autoimmune encephalomyelitis (EAE). RORγt also regulates thymocyte development and lymph node genesis. Here we show that the function of RORγt is regulated by its sumoylation. Loss of Sumo3, but not Sumo1, dampens TH17 differentiation and delays the progression of thymic CD8+ immature single-positive cells (ISPs). RORγt is SUMO3-modified by E3 ligase PIAS4 at lysine 31 (K31), and the mutation of K31 to arginine in mice prevents RORγt sumoylation, leading to impaired TH17 differentiation, resistance to TH17-mediated EAE, accumulation of thymic ISPs, and a lack of Peyer's patches. Mechanistically, sumoylation of RORγt-K31 recruits histone acetyltransferase KAT2A, which stabilizes the binding of SRC1 to enhance RORγt transcription factor activity. This study thus demonstrates that sumoylation is a critical mechanism for regulating RORγt function, and reveals new drug targets for preventing TH17-mediated autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Procesamiento Proteico-Postraduccional , Células Th17/inmunología , Timocitos/microbiología , Timo/inmunología , Ubiquitinas/genética , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Diferenciación Celular , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Femenino , Hematopoyesis/genética , Hematopoyesis/inmunología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/patología , Masculino , Ratones , Ratones Transgénicos , Coactivador 1 de Receptor Nuclear/genética , Coactivador 1 de Receptor Nuclear/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/patología , Proteína SUMO-1/deficiencia , Proteína SUMO-1/genética , Proteína SUMO-1/inmunología , Sumoilación , Células Th17/patología , Timocitos/inmunología , Timocitos/patología , Timo/patología , Ubiquitinas/deficiencia , Ubiquitinas/inmunología , Factores de Transcripción p300-CBP/genética , Factores de Transcripción p300-CBP/inmunología
20.
J Exp Med ; 215(11): 2936-2954, 2018 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-30355616

RESUMEN

The foodborne pathogen Listeria monocytogenes (Lm) crosses the intestinal villus epithelium via goblet cells (GCs) upon the interaction of Lm surface protein InlA with its receptor E-cadherin. Here, we show that Lm infection accelerates intestinal villus epithelium renewal while decreasing the number of GCs expressing luminally accessible E-cadherin, thereby locking Lm portal of entry. This novel innate immune response to an enteropathogen is triggered by the infection of Peyer's patch CX3CR1+ cells and the ensuing production of IL-23. It requires STAT3 phosphorylation in epithelial cells in response to IL-22 and IL-11 expressed by lamina propria gp38+ stromal cells. Lm-induced IFN-γ signaling and STAT1 phosphorylation in epithelial cells is also critical for Lm-associated intestinal epithelium response. GC depletion also leads to a decrease in colon mucus barrier thickness, thereby increasing host susceptibility to colitis. This study unveils a novel innate immune response to an enteropathogen, which implicates gp38+ stromal cells and locks intestinal villus invasion, but favors colitis.


Asunto(s)
Colitis/inmunología , Mucosa Intestinal/inmunología , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Glicoproteínas de Membrana/inmunología , Células Mieloides/inmunología , Ganglios Linfáticos Agregados/inmunología , Animales , Colitis/genética , Colitis/microbiología , Colitis/patología , Citocinas/genética , Citocinas/inmunología , Inmunidad Innata/genética , Inmunidad Mucosa/genética , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Listeriosis/genética , Listeriosis/patología , Glicoproteínas de Membrana/genética , Ratones , Ratones Noqueados , Células Mieloides/microbiología , Células Mieloides/patología , Ganglios Linfáticos Agregados/microbiología , Ganglios Linfáticos Agregados/patología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Células del Estroma/inmunología , Células del Estroma/microbiología , Células del Estroma/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA