Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Brain Behav Immun ; 122: 604-616, 2024 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-39187048

RESUMEN

The prevalence of bacterial infections significantly increases among patients with severe traumatic brain injury (STBI), leading to a notable rise in mortality rates. While immune dysfunctions are linked to the incidence of pneumonia, our observations indicate that endogenous pathogens manifest in the lungs post-STBI due to the migration of gut commensal bacteria. This translocation involves gut-innervating nociceptor sensory neurons, which are crucial for host defense. Following STBI, the expression of transient receptor potential vanilloid 1 (TRPV1) in dorsal root ganglion (DRG) neurons significantly decreases, despite an initial brief increase. The timing of TRPV1 defects coincides with the occurrence of pulmonary infections post-STBI. This alteration in TRPV1+ neurons diminishes their ability to signal bacterial injuries, weakens defense mechanisms against intestinal bacteria, and increases susceptibility to pulmonary infections via bacterial translocation. Experimental evidence demonstrates that pulmonary infections can be successfully replicated through the chemical ablation and gene interference of TRPV1+ nociceptors, and that these infections can be mitigated by TRPV1 activation, thereby confirming the crucial role of nociceptor neurons in controlling intestinal bacterial migration. Furthermore, TRPV1+ nociceptors regulate the immune response of microfold cells by releasing calcitonin gene-related peptide (CGRP), thereby influencing the translocation of gut bacteria to the lungs. Our study elucidates how changes in nociceptive neurons post-STBI impact intestinal pathogen defense. This new understanding of endogenous risk factors within STBI pathology offers novel insights for preventing and treating pulmonary infections.


Asunto(s)
Lesiones Traumáticas del Encéfalo , Nociceptores , Canales Catiónicos TRPV , Animales , Lesiones Traumáticas del Encéfalo/metabolismo , Lesiones Traumáticas del Encéfalo/microbiología , Canales Catiónicos TRPV/metabolismo , Nociceptores/metabolismo , Ratones , Masculino , Ganglios Espinales/metabolismo , Traslocación Bacteriana , Intestinos/microbiología , Ratones Endogámicos C57BL , Microbioma Gastrointestinal/fisiología , Pulmón/metabolismo , Pulmón/microbiología
2.
J Neuroinflammation ; 20(1): 282, 2023 Nov 27.
Artículo en Inglés | MEDLINE | ID: mdl-38012646

RESUMEN

BACKGROUND: The gut microbiota has recently attracted attention as a pathogenic factor in Alzheimer's disease (AD). Microfold (M) cells, which play a crucial role in the gut immune response against external antigens, are also exploited for the entry of pathogenic bacteria and proteins into the body. However, whether changes in M cells can affect the gut environments and consequently change brain pathologies in AD remains unknown. METHODS: Five familial AD (5xFAD) and 5xFAD-derived fecal microbiota transplanted (5xFAD-FMT) naïve mice were used to investigate the changes of M cells in the AD environment. Next, to establish the effect of M cell depletion on AD environments, 5xFAD mice and Spib knockout mice were bred, and behavioral and histological analyses were performed when M cell-depleted 5xFAD mice were six or nine months of age. RESULTS: In this study, we found that M cell numbers were increased in the colons of 5xFAD and 5xFAD-FMT mice compared to those of wild-type (WT) and WT-FMT mice. Moreover, the level of total bacteria infiltrating the colons increased in the AD-mimicked mice. The levels of M cell-related genes and that of infiltrating bacteria showed a significant correlation. The genetic inhibition of M cells (Spib knockout) in 5xFAD mice changed the composition of the gut microbiota, along with decreasing proinflammatory cytokine levels in the colons. M cell depletion ameliorated AD symptoms including amyloid-ß accumulation, microglial dysfunction, neuroinflammation, and memory impairment. Similarly, 5xFAD-FMT did not induce AD-like pathologies, such as memory impairment and excessive neuroinflammation in Spib-/- mice. CONCLUSION: Therefore, our findings provide evidence that the inhibiting M cells can prevent AD progression, with therapeutic implications.


Asunto(s)
Enfermedad de Alzheimer , Ratones , Animales , Enfermedad de Alzheimer/patología , Microglía/metabolismo , Células M , Enfermedades Neuroinflamatorias , Péptidos beta-Amiloides/metabolismo , Trastornos de la Memoria , Ratones Noqueados , Fenotipo , Modelos Animales de Enfermedad , Ratones Transgénicos
3.
Vet Pathol ; 57(5): 642-652, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32880235

RESUMEN

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


Asunto(s)
Infecciones por Coronavirus/veterinaria , Inmunidad Innata , Mucosa Intestinal/inmunología , Virus de la Diarrea Epidémica Porcina , Animales , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/patología , Citocinas/análisis , Íleon/inmunología , Íleon/patología , Íleon/virología , Mucosa Intestinal/química , Mucosa Intestinal/patología , Mucosa Intestinal/virología , Yeyuno/inmunología , Yeyuno/patología , Yeyuno/virología , Receptores de Inmunoglobulina Polimérica/metabolismo , Porcinos , Destete
4.
J Nanobiotechnology ; 17(1): 70, 2019 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-31113462

RESUMEN

BACKGROUND: Copper oxide nanomaterials (CuO NMs) are exploited in many products including inks, cosmetics, textiles, wood preservatives and food contact materials. Their incorporation into these products may enhance oral exposure in consumer, environmental and occupational settings. Undifferentiated and differentiated monocultures of Caco-2 cells are commonly used to assess NM toxicity to the intestine in vitro. However, the integration of other cell types into Caco-2 in vitro models increases their physiological relevance. Therefore, the aim of this study is to evaluate the toxicity of CuO NMs and copper sulphate (CuSO4) to intestinal microfold (M) cell (Caco-2/Raji B) and mucus secreting (Caco-2/HT29-MTX) co-culture in vitro models via assessment of their impact on barrier integrity, viability and interleukin (IL)-8 secretion. The translocation of CuO NMs and CuSO4 across the intestinal barrier was also investigated in vitro. RESULTS: CuO NMs and CuSO4 impaired the function of the intestinal barrier in the co-culture models [as indicated by a reduction in transepithelial electrical resistance (TEER) and Zonular occludens (ZO-1) staining intensity]. Cu translocation was observed in both models but was greatest in the Caco-2/Raji B co-culture. CuO NMs and CuSO4 stimulated an increase in IL-8 secretion, which was greatest in the Caco-2/HT29-MTX co-culture model. CuO NMs and CuSO4 did not stimulate a loss of cell viability, when assessed using light microscopy, nuclei counts and scanning electron microscopy. CuO NMs demonstrated a relatively similar level of toxicity to CuO4 in both Caco-2/Raji B and Caco-2/HT29-MTX co- culture models. CONCLUSIONS: The Caco-2/Raji B co-culture model was more sensitive to CuO NM and CuSO4 toxicity than the Caco-2/HT29-MTX co-culture model. However, both co-culture models were less sensitive to CuO NM and CuSO4 toxicity than simple monocultures of undifferentiated and differentiated Caco-2 cells, which are more routinely used to investigate NM toxicity to the intestine. Obtained data can therefore feed into the design of future studies which assess the toxicity of substances (e.g. NMs) and pathogens to the intestine (e.g. by informing model and endpoint selection). However, more testing with a wider panel of NMs would be beneficial in order to help select which in vitro models and endpoints to prioritise when screening the safety of ingested NMs. Comparisons with in vivo findings will also be essential to identify the most suitable in vitro model to screen the safety of ingested NMs.


Asunto(s)
Cobre/toxicidad , Tracto Gastrointestinal/efectos de los fármacos , Nanoestructuras/toxicidad , Transporte Biológico , Células CACO-2 , Diferenciación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Técnicas de Cocultivo/métodos , Cobre/química , Sulfato de Cobre/química , Sulfato de Cobre/toxicidad , Humanos , Interleucina-8/metabolismo , Absorción Intestinal , Mucosa Intestinal , Moco/citología , Nanoestructuras/química , Permeabilidad
5.
J Allergy Clin Immunol ; 137(1): 214-222.e2, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-26414879

RESUMEN

BACKGROUND: Transmission of mucosal pathogens relies on their ability to bind to the surfaces of epithelial cells, to cross this thin barrier, and to gain access to target cells and tissues, leading to systemic infection. This implies that pathogen-specific immunity at mucosal sites is critical for the control of infectious agents using these routes to enter the body. Although mucosal delivery would ensure the best onset of protective immunity, most of the candidate vaccines are administered through the parenteral route. OBJECTIVE: The present study evaluates the feasibility of delivering the chemically bound p24gag (referred to as p24 in the text) HIV antigen through secretory IgA (SIgA) in nasal mucosae in mice. RESULTS: We show that SIgA interacts specifically with mucosal microfold cells present in the nasal-associated lymphoid tissue. p24-SIgA complexes are quickly taken up in the nasal cavity and selectively engulfed by mucosal dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin-positive dendritic cells. Nasal immunization with p24-SIgA elicits both a strong humoral and cellular immune response against p24 at the systemic and mucosal levels. This ensures effective protection against intranasal challenge with recombinant vaccinia virus encoding p24. CONCLUSION: This study represents the first example that underscores the remarkable potential of SIgA to serve as a carrier for a protein antigen in a mucosal vaccine approach targeting the nasal environment.


Asunto(s)
Proteína p24 del Núcleo del VIH/inmunología , Inmunización/métodos , Inmunoglobulina A Secretora/inmunología , Tejido Linfoide/inmunología , Mucosa Nasal/inmunología , Administración Intranasal , Animales , Citocinas/inmunología , Células Dendríticas , Femenino , Inmunidad Mucosa , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Tejido Linfoide/citología , Ratones Endogámicos BALB C , Ratones Noqueados , Mucosa Nasal/citología , Bazo/inmunología , Vagina/inmunología
6.
Eur J Immunol ; 45(3): 773-9, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25412898

RESUMEN

HIV transmission and spread in the host are based on the survival of the virus or infected cells present in mucosal secretions, and the virus' ability to cross the epithelial barrier and access immune target cells, which leads to systemic infection. Therefore, HIV-specific immunity at mucosal sites is critical for control of infection. Although mucosal delivery would ensure the best onset of protective immunity, most candidate vaccines are administered through the parenteral route. Remarkably, secretory IgA (SIgA) interacts specifically with mucosal microfold (M) cells present in gut-associated lymphoid tissues. Here we evaluate the feasibility of delivering chemically bound p24HIV antigen via SIgA into the intestinal mucosae in mice. After oral administration, p24-SIgA complexes are quickly delivered into the tissue and selectively captured by CX3CR1(+) dendritic cells. Oral immunization with p24gag linked to SIgA (p24-SIgA) adjuvanted with E. coli heat labile enterotoxin (HLT) elicits both humoral and cellular immune responses against p24 at the systemic and mucosal levels and induces efficient protection against rectal challenge with a recombinant vaccinia virus encoding gag. This is the first study which underscores the remarkable potential of SIgA to serve as a vaccine carrier for an HIV antigen in mucosal administration targeting the gastrointestinal environment.


Asunto(s)
Vacunas contra el SIDA/farmacología , Portadores de Fármacos/farmacología , Proteína p24 del Núcleo del VIH/farmacología , VIH-1/inmunología , Inmunoglobulina A Secretora/farmacología , Mucosa Intestinal/inmunología , Vacunas contra el SIDA/inmunología , Animales , Proteína p24 del Núcleo del VIH/inmunología , Inmunidad Celular/efectos de los fármacos , Inmunidad Humoral/efectos de los fármacos , Inmunoglobulina A Secretora/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos
7.
Cell Tissue Res ; 364(1): 175-84, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26553655

RESUMEN

Murine nasopharynx-associated lymphoid tissue (NALT), located at the base of the nasal cavity, serves as a major site for the induction of mucosal immune responses against airway antigens. The follicle-associated epithelium (FAE) covering the luminal surface of NALT is characterized by the presence of microfold cells (M cells), which take up and transport luminal antigens to lymphocytes. Glycoprotein 2 (GP2) has recently been identified as a reliable marker for M cells in Peyer's patches of the intestine. However, the expression of GP2 and other functional molecules in the M cells of NALT has not yet been examined. We have immunohistochemically detected GP2-expressing cells in the FAE of NALT and the simultaneous expression of other intestinal M-cell markers, namely Tnfaip2, CCL9, and Spi-B. These cells have been further identified as M cells because of their higher uptake capacity of luminal microbeads. Electron microscopic observations have shown that GP2-expressing cells on the FAE display morphological features typical of M cells: they possess short microvilli and microfolds on the luminal surface and are closely associated with intraepithelial lymphocytes. We have also found that the receptor activator of nuclear factor kappa-B ligand (RANKL) is expressed by stromal cells underneath the FAE, which provides its receptor RANK. The administration of RANKL markedly increases the number of GP2(+)Tnfaip2(+) cells on the NALT FAE and that of intestinal M cells. These results suggest that GP2(+)Tnfaip2(+) cells in NALT are equivalent to intestinal M cells, and that RANKL-RANK signaling induces their differentiation.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Tejido Linfoide/inmunología , Faringe/inmunología , Ligando RANK/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Antígenos de Diferenciación/inmunología , Diferenciación Celular/inmunología , Proteínas Ligadas a GPI/inmunología , Regulación de la Expresión Génica/inmunología , Tejido Linfoide/citología , Ratones , Ratones Endogámicos BALB C , Faringe/citología , Ligando RANK/inmunología , Transducción de Señal/inmunología
8.
Carbohydr Polym ; 346: 122586, 2024 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-39245482

RESUMEN

Lentinan (LNT), a natural polysaccharide, has been reported to exhibit immunomodulatory effects in the intestine after oral administration. Herein, we aimed to investigate the lymphatic transport of LNT in Peyer's patches (PPs) by traceable fluorescent labeling and to explore whether/how LNT contacts related immune cells. Near-infrared imaging confirmed the absorption of LNT in the small intestinal segment and its accumulation within PPs after oral administration. Subsequently, tissue imaging confirmed that M cells are the main cells responsible for transporting LNT to PPs, and an M cell model was established to explore the involvement of Dectin-1 in the absorption process. Systematic in vitro and in vivo studies revealed that the Dectin-1 further mediates the uptake of LNT by mononuclear phagocytes in PPs. Moreover, LNT can promote the proliferation and differentiation of mononuclear phagocytes, thereby activating immune responses. In summary, this study elucidates the pharmacokinetic mechanisms by which LNT exerts oral immunomodulatory effects, providing a theoretical basis for the development and application of other polysaccharides.


Asunto(s)
Lectinas Tipo C , Lentinano , Ganglios Linfáticos Agregados , Ganglios Linfáticos Agregados/inmunología , Ganglios Linfáticos Agregados/efectos de los fármacos , Ganglios Linfáticos Agregados/metabolismo , Animales , Lentinano/farmacología , Lentinano/química , Lectinas Tipo C/metabolismo , Ratones , Administración Oral , Fagocitos/efectos de los fármacos , Fagocitos/metabolismo , Fagocitos/inmunología , Inmunomodulación/efectos de los fármacos , Masculino , Ratones Endogámicos BALB C , Células M
9.
Immunol Lett ; 270: 106925, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-39260525

RESUMEN

Vitamin D receptor (VDR) is involved in the pathogenesis of inflammatory bowel disease (IBD). However, the mechanism of VDR in IBD is still unclear. Microfold cells (M cells) mediated antigen-sampling pathway is central in developing immune responses to pathogenic and commensal bacteria and related to IBD. We found that Intestinal epithelial cell-specific knockdown of VDR(VDRIEC-KO) increases the susceptibility of mice to experimental colitis induced by sodium dextran sulfate(DSS) by producing more M cells. Knockdown VDR in intestinal epithelial cells increased RANKL-induced microfold cells and promoted the ability of microfold cells to uptake S. Typhimurium (S. T.). Mechanistically, we demonstrated that knockdown VDR promoted the differentiation and maturation of M cells via the Spi-B-dependent pathway. We conclude that M cells may be a potential target of VDR for treating intestinal mucosal barrier dysfunction in IBD.

10.
Int J Biol Macromol ; 266(Pt 1): 131096, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38522695

RESUMEN

Polysaccharides of vinegar-baked Radix Bupleuri (VBCP) have been reported to exhibit liver-targeting and immunomodulatory activities through oral administration, but the absorption behavior and mechanism of VBCPs have not been extensively studied. In this study, a novel HG type pectin polysaccharide, VBCP1-4, with a high molecular weight of 2.94 × 106 Da, was separated from VBCP. VBCP1-4 backbone was contained 1,4-α-D-GalpA, 1,4-α-D-GalpA6OMe, 1,3,4-α-D-GalpA and 1,2,4-α-D-Rhap. The branches were mainly contained 1,5-α-L-Araf, 1,3,5-α-L-Araf, t-α-L-Araf and t-α-D-Galp, which linked to the 3 position of 1,3,4-α-D-GalpA and the 4 position of 1,2,4-α-D-Rhap. VBCP1-4 could self-assemble to nanoparticles in water, with CMC values of 106.41 µg/mL, particle sizes of 178.20 ± 2.82 nm and zeta potentials of -23.19 ± 1.44 mV. The pharmacokinetic study of VBCP1-4, which detected by marking with FITC, revealed that it could be partially absorbed into the body through Peyer's patches of the ileum. In vitro absorption study demonstrated that VBCP1-4 was difficult to be absorbed by Caco-2 cell monolayer, but could be absorbed by M cells in a time and concentration dependent manner. The absorption mechanism was elucidated that VBCP1-4 entered M cells through clathrin-mediated endocytosis in the form of nanoparticles. These findings provide valuable insights into the absorption behavior of VBCP and contribute to its further development.


Asunto(s)
Ácido Acético , Bupleurum , Nanopartículas , Pectinas , Pectinas/química , Bupleurum/química , Ácido Acético/química , Nanopartículas/química , Humanos , Animales , Células CACO-2 , Tamaño de la Partícula , Peso Molecular , Células M
11.
Front Immunol ; 15: 1373766, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39359724

RESUMEN

Food components suppressing small intestinal tumorigenesis are not well-defined partly because of the rarity of this tumor type compared to colorectal tumors. Using Apcmin/+ mice, a mouse model for intestinal tumorigenesis, and antigen-free diet, we report here that food antigens serve this function in the small intestine. By depleting Peyer's patches (PPs), immune inductive sites in the small intestine, we found that PPs have a role in the suppression of small intestinal tumors and are important for the induction of small intestinal T cells by food antigens. On the follicle-associated epithelium (FAE) of PPs, microfold (M) cells pass food antigens from lumen to the dendritic cells to induce T cells. Single-cell RNA-seq (scRNA-seq) analysis of immune cells in PPs revealed a significant impact of food antigens on the induction of the PP T cells and the antigen presentation capacity of dendritic cells. These data demonstrate the role of food antigens in the suppression of small intestinal tumorigenesis by PP-mediated immune cell induction.


Asunto(s)
Células Dendríticas , Neoplasias Intestinales , Intestino Delgado , Ganglios Linfáticos Agregados , Animales , Ratones , Intestino Delgado/inmunología , Intestino Delgado/patología , Neoplasias Intestinales/inmunología , Neoplasias Intestinales/patología , Neoplasias Intestinales/genética , Ganglios Linfáticos Agregados/inmunología , Células Dendríticas/inmunología , Carcinogénesis/inmunología , Antígenos/inmunología , Ratones Endogámicos C57BL , Linfocitos T/inmunología , Presentación de Antígeno/inmunología , Modelos Animales de Enfermedad , Alimentos
12.
Methods Mol Biol ; 2650: 3-16, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37310619

RESUMEN

The intestine is a prime example of self-renewal where stem cells give rise to progenitor cells called transit-amplifying cells which differentiate into more specialized cells. There are two intestinal lineages: the absorptive (enterocytes and microfold cells) and the secretory (Paneth cells, enteroendocrine, goblet cells, and tuft cells). Each of these differentiated cell types has a role in creating an "ecosystem" to maintain intestinal homeostasis. Here, we summarize the main roles of each cell type.


Asunto(s)
Enterocitos , Células Epiteliales , Diferenciación Celular , Células M , Células Madre
13.
Metallomics ; 15(6)2023 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-37209082

RESUMEN

This study aimed to investigate the transportation and absorption mechanism of lanthanum carbonate [La2(CO3)3] through the gastrointestinal (GI) tract using in vitro and in vivo models. The results demonstrated that La2(CO3)3 can be dissolved in gastric fluids and precipitated into lanthanum phosphate as the main transformed specie in intestinal fluid. Using Caco-2 cell monoculture and Caco-2/Raji B cell coculture models to simulate the intestinal epithelium and microfold (M) cells, it was found that the amount of lanthanum transported in Caco-2/Raji B coculture model was significantly higher than that in Caco-2 monoculture model (about 50 times higher), indicating that M cells play an important role in the intestinal absorption of La2(CO3)3. Furthermore, oral administration of La2(CO3)3 to Balb/c mice demonstrated that lanthanum can be absorbed by both Peyer's patches (PPs) and non-PPs intestinal epithelium, with a higher amount of absorption in the PPs per unit weight. This finding further confirmed that the lanthanum absorption in GI tract could be mainly due to the contribution of M cells. Meanwhile, the administration of La2(CO3)3 caused a marked lanthanum accumulation in liver, accompanied by the activation of Kupffer cells. This study clarified how La2(CO3)3 is absorbed through the GI tract to enter the body and would be helpful to evaluate its potential biological consequences of accumulation in human beings.


Asunto(s)
Lantano , Células M , Ratones , Animales , Humanos , Células CACO-2 , Fosfatos , Tracto Gastrointestinal
14.
Front Med (Lausanne) ; 9: 868508, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35530046

RESUMEN

Ulcerative Colitis (UC) is a chronic inflammatory disease of the intestinal tract for which a definitive etiology is yet unknown. Both genetic and environmental factors have been implicated in the development of UC. Recently, single cell RNA sequencing (scRNA-seq) technology revealed cell subpopulations contributing to the pathogenesis of UC and brought new insight into the pathways that connect genome to pathology. This review describes key scRNA-seq findings in two major studies by Broad Institute and University of Oxford, investigating the transcriptomic landscape of epithelial cells in UC. We focus on five major findings: (1) the identification of BEST4 + cells, (2) colonic microfold (M) cells, (3) detailed comparison of the transcriptomes of goblet cells, and (4) colonocytes and (5) stem cells in health and disease. In analyzing the two studies, we identify the commonalities and differences in methodologies, results, and conclusions, offering possible explanations, and validated several cell cluster markers. In systematizing the results, we hope to offer a framework that the broad scientific GI community and GI clinicians can use to replicate or corroborate the extensive new findings that RNA-seq offers.

15.
Cells ; 11(21)2022 10 25.
Artículo en Inglés | MEDLINE | ID: mdl-36359753

RESUMEN

Increasing interest is being addressed to the development of a reliable, reproducible and relevant in vitro model of intestinal barrier, mainly for engineered nanomaterials hazard and risk assessment, in order to meet regulatory and scientific demands. Starting from the consolidated Caco-2 cell model, widely used for determining translocation of drugs and chemicals, the establishment of an advanced intestinal barrier model with different level of complexity is important for overcoming Caco-2 monoculture limitations. For this purpose, a tri-culture model, consisting of two human intestinal epithelial cells (Caco-2 and HT29-MTX) and a human lymphocyte B cell (Raji B), was developed by several research groups to mimic the in vivo intestinal epithelium, furnishing appropriate tools for nanotoxicological studies. However, tri-culture model shows high levels of variability in ENM uptake/translocation studies. With the aim of implementing the standardization and optimization of this tri-culture for ENM translocation studies, the present paper intends to identify and discuss such relevant parameters involved in model establishment as: tri-culture condition set-up, barrier integrity evaluation, mucus characterization, M-cell induction. SiO2 fluorescent nanoparticles were used to compare the different models. Although a low level of SiO2 translocation is reported for all the different culture conditions. a relevant role of mucus and M-cells in NPs uptake/translocation has been highlighted.


Asunto(s)
Nanopartículas , Dióxido de Silicio , Humanos , Células CACO-2 , Permeabilidad , Células HT29 , Técnicas de Cocultivo , Estándares de Referencia
16.
Cell Mol Gastroenterol Hepatol ; 12(3): 873-889, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34058415

RESUMEN

BACKGROUND & AIMS: Microfold cells (M cells) are immunosurveillance epithelial cells located in the Peyer's patches (PPs) in the intestine and are responsible for monitoring and transcytosis of antigens, microorganisms, and pathogens. Mature M cells use the receptor glycoprotein 2 (GP2) to aid in transcytosis. Recent studies have shown transcription factors, Spi-B and SRY-Box Transcription Factor 8 (Sox8). are necessary for M-cell differentiation, but not sufficient. An exhaustive set of factors sufficient for differentiation and development of a mature GP2+ M cell remains elusive. Our aim was to understand the role of polycomb repressive complex 2 (PRC2) as an epigenetic regulator of M-cell development. Estrogen-related-receptor γ (Esrrg), identified as a PRC2-regulated gene, was studied in depth, in addition to its relationship with Spi-B and Sox8. METHODS: Comparative chromatin immunoprecipitation and global run-on sequencing analysis of mouse intestinal organoids were performed in stem condition, enterocyte conditions, and receptor activator of nuclear factor κ B ligand-induced M-cell condition. Esrrg, which was identified as one of the PRC2-regulated transcription factors, was studied in wild-type mice and knocked out in intestinal organoids using guide RNA's. Sox8 null mice were used to study Esrrg and its relation to Sox8. RESULTS: chromatin immunoprecipitation and global run-on sequencing analysis showed 12 novel PRC2 regulated transcription factors, PRC2-regulated Esrrg is a novel M-cell-specific transcription factor acting on a receptor activator of nuclear factor κB ligand-receptor activator of nuclear factor κB-induced nuclear factor-κB pathway, upstream of Sox8, and necessary but not sufficient for a mature M-cell marker of Gp2 expression. CONCLUSIONS: PRC2 regulates a significant set of genes in M cells including Esrrg, which is critical for M-cell development and differentiation. Loss of Esrrg led to an immature M-cell phenotype lacking in Sox8 and Gp2 expression. Transcript profiling: the data have been deposited in the NCBI Gene Expression Omnibus database (GSE157629).


Asunto(s)
Células Epiteliales/metabolismo , Regulación de la Expresión Génica , Mucosa Intestinal/citología , Mucosa Intestinal/metabolismo , Ganglios Linfáticos Agregados/citología , Ganglios Linfáticos Agregados/metabolismo , Complejo Represivo Polycomb 2/metabolismo , Animales , Biomarcadores , Diferenciación Celular/genética , Perfilación de la Expresión Génica , Mucosa Intestinal/inmunología , Ratones , FN-kappa B/metabolismo , Ganglios Linfáticos Agregados/inmunología , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal
17.
Int J Pharm ; 594: 120167, 2021 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-33309559

RESUMEN

Diverse nanoparticulate systems have been engineered as vehicles towards enhancing the bioavailability of orally administrated vaccines. Substantial evidence suggests that targeting microfold cells (M cells) within Peyer's patches (PPs) is a prerequisite for vaccine-loaded nanocarriers to induce an effective antigen-specific immune response. Improved understanding of the contribution of M cells to sampling luminal nanoparticles into the underlying gut associated lymphoid tissues would accelerate the development of oral vaccine formulations. Herein, a novel clearing-based whole tissue mount/imaging technique was developed to enable the specific distribution of nanoparticles within ex vivo murine PPs to be quantitatively determined at the cellular level. This revealed that 200 nm nanoparticles modified with M cell targeting ligands (lectin Ulex europaeus agglutinin-1, UEA-1) were translocated into subepithelial domes 7.6 and 16.3 times greater than the non-targeted ones at 60 min and 120 min, respectively. This approach provides a new methodology to quantitatively investigate the transcytotic activity of M cells for particulate formulations, which may aid in the design of improved oral vaccines.


Asunto(s)
Nanopartículas , Vacunas , Animales , Inmunidad Mucosa , Mucosa Intestinal , Ratones , Ganglios Linfáticos Agregados
18.
Acta Pharm Sin B ; 11(8): 2449-2468, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34522594

RESUMEN

Orally administered drug entities have to survive the harsh gastrointestinal environment, penetrate the enteric epithelia and circumvent hepatic metabolism before reaching the systemic circulation. Whereas the gastrointestinal stability can be well maintained by taking proper measures, hepatic metabolism presents as a formidable barrier to drugs suffering from first-pass metabolism. The pharmaceutical academia and industries are seeking alternative pathways for drug transport to circumvent problems associated with the portal pathway. Intestinal lymphatic transport is emerging as a promising pathway to this end. In this review, we intend to provide an updated overview on the rationale, strategies, factors and applications involved in intestinal lymphatic transport. There are mainly two pathways for peroral lymphatic transport-the chylomicron and the microfold cell pathways. The underlying mechanisms are being unraveled gradually and nowadays witness increasing research input and applications.

19.
Biomed Pharmacother ; 121: 109653, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31810138

RESUMEN

Gut microbiota plays an important role in the gut and have become a hotspot of recent research interests. Commensal microbiota in gut exert a variety of effects on the host, from shaping the structure and function of the gut and the immune system to the modulation of nutrient status of the host and the treatment outcomes of some drugs. Gut microbiota and its enzyme product and subsequent products, such as short-chain fatty acid and bile acid, play important roles in the biotransformation of drugs via directly or indirectly affecting drug absorption, toxicity, metabolism and bioavailability. Drugs, especially antibiotics, also affect the homeostasis of probiotics and the integrity and function of the intestinal mucosa. These interplaying processes produce a variety of important metabolites of the host and drugs and affect the balance of microbiota and the mucosal barrier then modulate the function of drugs. Gut microbiota imbalance is associated with a broad range of disease mechanisms, and this association denotes a new drug-therapeutic avenue. The present review summarizes how gut microbiota acts as an "invisible organ" to directly or indirectly modulate the function of drugs, on the aspects of probiotic homeostasis, drugs and host nutritional metabolism, AJC, mucus layer and microfold cells.


Asunto(s)
Microbioma Gastrointestinal , Preparaciones Farmacéuticas/metabolismo , Microbioma Gastrointestinal/efectos de los fármacos , Homeostasis/efectos de los fármacos , Humanos , Fenómenos Fisiológicos de la Nutrición/efectos de los fármacos , Probióticos/farmacología
20.
Front Immunol ; 10: 1323, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31244859

RESUMEN

Microfold (M) cells residing in the follicle-associated epithelium of mucosa-associated lymphoid tissues are specialized for sampling luminal antigens to initiate mucosal immune responses. In the past decade, glycoprotein 2 (GP2) and Tnfaip2 were identified as reliable markers for M cells in the Peyer's patches of the intestine. Furthermore, RANKL-RANK signaling, as well as the canonical and non-canonical NFκB pathways downstream, is essential for M-cell differentiation from the intestinal stem cells. However, the molecular characterization and differentiation mechanisms of M cells in the lower respiratory tract, where organized lymphoid tissues exist rarely, remain to be fully elucidated. Therefore, this study aimed to explore M cells in the lower respiratory tract in terms of their specific molecular markers, differentiation mechanism, and functions. Immunofluorescence analysis revealed a small number of M cells expressing GP2, Tnfaip2, and RANK is present in the lower respiratory tract of healthy mice. The intraperitoneal administration of RANKL in mice effectively induced M cells, which have a high capacity to take up luminal substrates, in the lower respiratory epithelium. The airway M cells associated with lymphoid follicles were frequently detected in the pathologically induced bronchus-associated lymphoid tissue (iBALT) in the murine models of autoimmune disease as well as pulmonary emphysema. These findings demonstrate that RANKL is a common inducer of M cells in the airway and digestive tracts and that M cells are associated with the respiratory disease. We also established a two-dimensional culture method for airway M cells from the tracheal epithelium in the presence of RANKL successfully. This model may be useful for functional studies of M cells in the sampling of antigens at airway mucosal surfaces.


Asunto(s)
Inmunidad Mucosa , Ligando RANK/inmunología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Enfermedades Respiratorias/inmunología , Enfermedades Respiratorias/patología , Animales , Bronquiolos/inmunología , Bronquiolos/patología , Técnicas de Cultivo de Célula , Fumar Cigarrillos/efectos adversos , Fumar Cigarrillos/inmunología , Fumar Cigarrillos/patología , Modelos Animales de Enfermedad , Enfisema/inmunología , Enfisema/patología , Femenino , Proteínas Ligadas a GPI/inmunología , Tejido Linfoide/inmunología , Tejido Linfoide/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/patología , Receptor Activador del Factor Nuclear kappa-B/inmunología , Transducción de Señal/inmunología , Factores de Necrosis Tumoral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA