Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Intervalo de año de publicación
1.
Sci Rep ; 14(1): 19235, 2024 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-39164530

RESUMEN

This study aimed to determine the effects of Saccharomyces yeast postbiotics on cell turnover, immune responses, and oxidative stress in the jejunal mucosa of pigs. Thirty-two newly weaned pigs at 6.05 ± 0.24 kg were assigned to two dietary treatments based on a randomized complete block design. The treatments were control group receiving a basal diet and a group supplemented with Saccharomyces yeast postbiotics (175 g/ton diet) in the basal diet. After 35 d of the study, pigs were euthanized and jejunal mucosa were collected to assess immune status, oxidative stress, barrier markers, cell proliferation, and apoptosis. Saccharomyces yeast postbiotics reduced (P < 0.05) the fecal score from d 3 to d 7 and tended to increase the gene expression of interferon-γ (IFN-γ) (P = 0.071) and mammalian/mechanistic target of rapamycin (mTOR) (P = 0.080), decrease the gene expression of B-cell lymphoma 2-associated X protein 1 (BAX1) (P < 0.05), tended to decrease the gene expression of serum and glucocorticoid-induced protein kinase 1 (SGK1) (P = 0.066), increased (P < 0.05) cell proliferation in the crypts, and tended to increase the villus height (P = 0.078) and crypt depth (P = 0.052) in the jejunum. In conclusion, the supplementation of Saccharomyces yeast postbiotics in nursery diets reduced diarrhea within the first week after weaning and provided protection to the villi in the jejunum by enhancing the immune responses of nursery pigs, promoting crypt cell proliferation, and reducing the expression of genes associated with apoptosis without affecting inflammatory and oxidative stress status in the jejunum of the nursery pigs.


Asunto(s)
Mucosa Intestinal , Yeyuno , Estrés Oxidativo , Saccharomyces cerevisiae , Animales , Estrés Oxidativo/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Porcinos , Yeyuno/metabolismo , Yeyuno/inmunología , Yeyuno/efectos de los fármacos , Yeyuno/microbiología , Probióticos/administración & dosificación , Probióticos/farmacología , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Destete , Alimentación Animal
2.
J Virol ; 98(8): e0103924, 2024 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-39012142

RESUMEN

In maintaining organismal homeostasis, gut immunity plays a crucial role. The coordination between the microbiota and the immune system through bidirectional interactions regulates the impact of microorganisms on the host. Our research focused on understanding the relationships between substantial changes in jejunal intestinal flora and metabolites and intestinal immunity during porcine epidemic diarrhea virus (PEDV) infection in piglets. We discovered that Lactobacillus rhamnosus GG (LGG) could effectively prevent PEDV infection in piglets. Further investigation revealed that LGG metabolites interact with type 3 innate lymphoid cells (ILC3s) in the jejunum of piglets through the aryl hydrocarbon receptor (AhR). This interaction promotes the activation of ILC3s and the production of interleukin-22 (IL-22). Subsequently, IL-22 facilitates the proliferation of IPEC-J2 cells and activates the STAT3 signaling pathway, thereby preventing PEDV infection. Moreover, the AhR receptor influences various cell types within organoids, including intestinal stem cells (ISCs), Paneth cells, and enterocytes, to promote their growth and development, suggesting that AhR has a broad impact on intestinal health. In conclusion, our study demonstrated the ability of LGG to modulate intestinal immunity and effectively prevent PEDV infection in piglets. These findings highlight the potential application of LGG as a preventive measure against viral infections in livestock.IMPORTANCEWe observed high expression of the AhR receptor on pig and human ILC3s, although its expression was negligible in mouse ILC3s. ILC3s are closely related to the gut microbiota, particularly the secretion of IL-22 stimulated by microbial signals, which plays a crucial regulatory role in intestinal immunity. In our study, we found that metabolites produced by beneficial gut bacteria interact with ILC3s through AhR, thereby maintaining intestinal immune homeostasis in pigs. Moreover, LGG feeding can enhance the activation of ILC3s and promote IL-22 secretion in the intestines of piglets, ultimately preventing PEDV infection.


Asunto(s)
Infecciones por Coronavirus , Inmunidad Innata , Interleucina-22 , Interleucinas , Linfocitos , Virus de la Diarrea Epidémica Porcina , Receptores de Hidrocarburo de Aril , Animales , Receptores de Hidrocarburo de Aril/metabolismo , Porcinos , Interleucinas/metabolismo , Virus de la Diarrea Epidémica Porcina/inmunología , Linfocitos/inmunología , Linfocitos/metabolismo , Infecciones por Coronavirus/inmunología , Infecciones por Coronavirus/prevención & control , Infecciones por Coronavirus/veterinaria , Infecciones por Coronavirus/virología , Infecciones por Coronavirus/metabolismo , Microbioma Gastrointestinal/inmunología , Enfermedades de los Porcinos/inmunología , Enfermedades de los Porcinos/virología , Enfermedades de los Porcinos/prevención & control , Enfermedades de los Porcinos/microbiología , Yeyuno/inmunología , Yeyuno/metabolismo , Transducción de Señal , Ligandos , Intestinos/inmunología , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo
3.
mBio ; 15(8): e0131624, 2024 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-38953637

RESUMEN

Human intestinal enteroids (HIEs) are gaining recognition as physiologically relevant models of the intestinal epithelium. While HIEs from adults are used extensively in biomedical research, few studies have used HIEs from infants. Considering the dramatic developmental changes that occur during infancy, it is important to establish models that represent infant intestinal characteristics and physiological responses. We established jejunal HIEs from infant surgical samples and performed comparisons to jejunal HIEs from adults using RNA sequencing (RNA-Seq) and morphologic analyses. We then validated differences in key pathways through functional studies and determined whether these cultures recapitulate known features of the infant intestinal epithelium. RNA-Seq analysis showed significant differences in the transcriptome of infant and adult HIEs, including differences in genes and pathways associated with cell differentiation and proliferation, tissue development, lipid metabolism, innate immunity, and biological adhesion. Validating these results, we observed a higher abundance of cells expressing specific enterocyte, goblet cell, and enteroendocrine cell markers in differentiated infant HIE monolayers, and greater numbers of proliferative cells in undifferentiated 3D cultures. Compared to adult HIEs, infant HIEs portray characteristics of an immature gastrointestinal epithelium including significantly shorter cell height, lower epithelial barrier integrity, and lower innate immune responses to infection with an oral poliovirus vaccine. HIEs established from infant intestinal tissues reflect characteristics of the infant gut and are distinct from adult cultures. Our data support the use of infant HIEs as an ex vivo model to advance studies of infant-specific diseases and drug discovery for this population. IMPORTANCE: Tissue or biopsy stem cell-derived human intestinal enteroids are increasingly recognized as physiologically relevant models of the human gastrointestinal epithelium. While enteroids from adults and fetal tissues have been extensively used for studying many infectious and non-infectious diseases, there are few reports on enteroids from infants. We show that infant enteroids exhibit both transcriptomic and morphological differences compared to adult cultures. They also differ in functional responses to barrier disruption and innate immune responses to infection, suggesting that infant and adult enteroids are distinct model systems. Considering the dramatic changes in body composition and physiology that begin during infancy, tools that appropriately reflect intestinal development and diseases are critical. Infant enteroids exhibit key features of the infant gastrointestinal epithelium. This study is significant in establishing infant enteroids as age-appropriate models for infant intestinal physiology, infant-specific diseases, and responses to pathogens.


Asunto(s)
Mucosa Intestinal , Humanos , Lactante , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Adulto , Diferenciación Celular , Yeyuno/citología , Yeyuno/inmunología , Transcriptoma , Organoides , Inmunidad Innata , Femenino , Masculino , Recién Nacido , Enterocitos
4.
Microbiol Res ; 286: 127821, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-38941923

RESUMEN

Radiation injury to the intestine is one of the most common complications in patients undergoing abdominal or pelvic cavity radiotherapy. In this study, we investigated the potential protective effect of Lactobacillus rhamnosus GG (LGG) on radiation-induced intestinal injury and its underlying mechanisms. Mice were assigned to a control group, a 10 Gy total abdominal irradiation (TAI) group, or a group pretreated with 108 CFU LGG for three days before TAI. Small intestine and gut microbiota were analyzed 3.5 days post-exposure. LGG intervention improved intestinal structure, reduced jejunal DNA damage, and inhibited the inflammatory cGAS/STING pathway. Furthermore, LGG reduced M1 proinflammatory macrophage and CD8+ T cell infiltration, restoring the balance between Th17 and Treg cells in the inflamed jejunum. LGG also partially restored the gut microbiota. These findings suggest the possible therapeutic radioprotective effect of probiotics LGG in alleviating radiation-induced intestinal injury by maintaining immune homeostasis and reshaping gut microbiota.


Asunto(s)
Microbioma Gastrointestinal , Lacticaseibacillus rhamnosus , Ratones Endogámicos C57BL , Probióticos , Animales , Microbioma Gastrointestinal/efectos de la radiación , Ratones , Probióticos/administración & dosificación , Traumatismos por Radiación/inmunología , Macrófagos/inmunología , Intestinos/microbiología , Intestinos/efectos de la radiación , Intestinos/inmunología , Daño del ADN , Linfocitos T CD8-positivos/inmunología , Proteínas de la Membrana/metabolismo , Linfocitos T Reguladores/inmunología , Masculino , Células Th17/inmunología , Yeyuno/efectos de la radiación , Yeyuno/inmunología , Yeyuno/microbiología , Protectores contra Radiación/farmacología , Protectores contra Radiación/uso terapéutico , Traumatismos Experimentales por Radiación/inmunología , Traumatismos Experimentales por Radiación/prevención & control , Nucleotidiltransferasas
5.
Microb Pathog ; 192: 106691, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38759933

RESUMEN

Necrotic enteritis (NE) is a potentially fatal poultry disease that causes enormous economic losses in the poultry industry worldwide. The study aimed to evaluate the effects of dietary organic yeast-derived selenium (Se) on immune protection against experimental necrotic enteritis (NE) in commercial broilers. Chickens were fed basal diets supplemented with different Se levels (0.25, 0.50, and 1.00 Se mg/kg). To induce NE, Clostridium perfringens (C. perfringens) was orally administered at 14 days of age post hatch. The results showed that birds fed 0.25 Se mg/kg exhibited significantly increased body weight gain compared with the non-supplemented/infected birds. There were no significant differences in gut lesions between the Se-supplemented groups and the non-supplemented group. The antibody levels against α-toxin and NetB toxin increased with the increase between 0.25 Se mg/kg and 0.50 Se mg/kg. In the jejunal scrapings and spleen, the Se-supplementation groups up-regulated the transcripts for pro-inflammatory cytokines IL-1ß, IL-6, IL-8, iNOS, and LITAF and avian ß-defensin 6, 8, and 13 (AvBD6, 8 and 13). In conclusion, supplementation with organic yeast-derived Se alleviates the negative consequences and provides beneficial protection against experimental NE.


Asunto(s)
Alimentación Animal , Pollos , Infecciones por Clostridium , Clostridium perfringens , Citocinas , Suplementos Dietéticos , Enteritis , Enfermedades de las Aves de Corral , Selenio , Animales , Enteritis/prevención & control , Enteritis/veterinaria , Enteritis/inmunología , Enteritis/microbiología , Selenio/farmacología , Selenio/administración & dosificación , Enfermedades de las Aves de Corral/prevención & control , Enfermedades de las Aves de Corral/inmunología , Clostridium perfringens/inmunología , Infecciones por Clostridium/prevención & control , Infecciones por Clostridium/veterinaria , Infecciones por Clostridium/inmunología , Citocinas/metabolismo , Toxinas Bacterianas/inmunología , Necrosis , beta-Defensinas/metabolismo , Yeyuno/efectos de los fármacos , Yeyuno/inmunología , Yeyuno/microbiología , Yeyuno/patología , Bazo/inmunología , Levaduras , Óxido Nítrico Sintasa de Tipo II/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Interleucina-1beta/metabolismo , Anticuerpos Antibacterianos/sangre
6.
Int Immunopharmacol ; 135: 112333, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38805907

RESUMEN

Macrophages are one of the important immune cells, which play important roles in innate and adaptive immune. However, the roles of macrophages in food allergy are not thoroughly understood. To investigate the roles of macrophages during food allergy, we focused on the relationship between macrophage polarization and allergic responses induced by tropomyosin (TM) in the present study. Arg 1 and CD206 expressions in the TM group were significantly higher than those of the PBS group, while iNOS and TNF-α expressions were no obvious difference, moreover, the morphology of macrophages stimulated by TM was similar to that of M2 macrophages. These results indicated macrophages were mainly polarized toward M2 phenotypes in vitro. The antibodies, mMCP-1, histamine and cytokines, revealed that macrophages could participate in food allergy, and macrophage polarization was associated with changes in allergic-related factors. The cytokine levels of M2 phenotypes were significantly higher than those of M1 phenotypes in peripheral blood. The mRNA expressions and protein levels of Arg1 and iNOS in the jejunum and peritoneal cells indicated that M2 phenotypes were the major macrophage in these tissues compared with M1 phenotypes. Hence, macrophage polarization plays an important role in food allergy.


Asunto(s)
Arginasa , Hipersensibilidad a los Alimentos , Macrófagos , Ratones Endogámicos BALB C , Palaemonidae , Tropomiosina , Animales , Tropomiosina/inmunología , Hipersensibilidad a los Alimentos/inmunología , Ratones , Macrófagos/inmunología , Arginasa/metabolismo , Palaemonidae/inmunología , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico Sintasa de Tipo II/genética , Citocinas/metabolismo , Modelos Animales de Enfermedad , Lectinas Tipo C/metabolismo , Lectinas Tipo C/genética , Receptores de Superficie Celular/metabolismo , Receptores de Superficie Celular/genética , Lectinas de Unión a Manosa/metabolismo , Femenino , Receptor de Manosa , Yeyuno/inmunología , Yeyuno/patología , Células Cultivadas , Histamina/metabolismo , Activación de Macrófagos
7.
J Food Sci ; 89(5): 3037-3047, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38563099

RESUMEN

Bovine casein is a major allergen present in cow milk to induce anaphylaxis. In this study, the potential allergenicity of enzymatically hydrolyzed casein (HC) was evaluated based on in vitro and in vivo. The results showed that Alcalase and Protamex treatment (AT, PT) reduced the potential allergenicity of CN, with the greatest reductions of 68.25% and 50.75%, respectively. In addition, in vivo results showed that HC effectively alleviated allergic response symptoms of Balb/c mice; a significant tendency toward decreased serum IgG1 and mast cell tryptase levels was observed, accompanied by a decrease of Th2-associated IL-4, IL-5, and IL-13 and an increase of IFN-γ levels in spleen. Moreover, the inflammation of the lung, jejunum, and ileum was remarkably ameliorated. The findings indicated that HC induced a shift toward Th1 response and maintained the Th1/Th2 immune balance. Importantly, our results provide the basis for the production of hypoallergenic dairy products.


Asunto(s)
Alérgenos , Caseínas , Ratones Endogámicos BALB C , Células Th2 , Animales , Ratones , Caseínas/inmunología , Alérgenos/inmunología , Femenino , Células Th2/inmunología , Hidrólisis , Inmunoglobulina G/sangre , Modelos Animales de Enfermedad , Bovinos , Bazo/inmunología , Hipersensibilidad a la Leche/inmunología , Interferón gamma/metabolismo , Células TH1/inmunología , Interleucina-4/metabolismo , Triptasas/metabolismo , Citocinas/metabolismo , Yeyuno/inmunología , Leche/inmunología , Leche/química , Interleucina-13/inmunología , Interleucina-13/metabolismo , Anafilaxia/inmunología , Anafilaxia/inducido químicamente , Anafilaxia/prevención & control , Interleucina-5/inmunología
8.
Mucosal Immunol ; 17(4): 565-583, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38555027

RESUMEN

Studies have reported the occurrence of gastrointestinal (GI) symptoms, primarily diarrhea, in COVID-19. However, the pathobiology regarding COVID-19 in the GI tract remains limited. This work aimed to evaluate SARS-CoV-2 Spike protein interaction with gut lumen in different experimental approaches. Here, we present a novel experimental model with the inoculation of viral protein in the murine jejunal lumen, in vitro approach with human enterocytes, and molecular docking analysis. Spike protein led to increased intestinal fluid accompanied by Cl- secretion, followed by intestinal edema, leukocyte infiltration, reduced glutathione levels, and increased cytokine levels [interleukin (IL)-6, tumor necrosis factor-α, IL-1ß, IL-10], indicating inflammation. Additionally, the viral epitope caused disruption in the mucosal histoarchitecture with impairment in Paneth and goblet cells, including decreased lysozyme and mucin, respectively. Upregulation of toll-like receptor 2 and toll-like receptor 4 gene expression suggested potential activation of local innate immunity. Moreover, this experimental model exhibited reduced contractile responses in jejunal smooth muscle. In barrier function, there was a decrease in transepithelial electrical resistance and alterations in the expression of tight junction proteins in the murine jejunal epithelium. Additionally, paracellular intestinal permeability increased in human enterocytes. Finally, in silico data revealed that the Spike protein interacts with cystic fibrosis transmembrane conductance regulator (CFTR) and calcium-activated chloride conductance (CaCC), inferring its role in the secretory effect. Taken together, all the events observed point to gut impairment, affecting the mucosal barrier to the innermost layers, establishing a successful experimental model for studying COVID-19 in the GI context.


Asunto(s)
COVID-19 , Mucosa Intestinal , SARS-CoV-2 , Glicoproteína de la Espiga del Coronavirus , Glicoproteína de la Espiga del Coronavirus/metabolismo , Glicoproteína de la Espiga del Coronavirus/inmunología , Animales , SARS-CoV-2/fisiología , SARS-CoV-2/inmunología , Humanos , Ratones , COVID-19/inmunología , COVID-19/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Yeyuno/inmunología , Yeyuno/metabolismo , Yeyuno/patología , Yeyuno/virología , Simulación del Acoplamiento Molecular , Enterocitos/metabolismo , Enterocitos/virología , Inmunidad Innata , Citocinas/metabolismo , Modelos Animales de Enfermedad , Masculino , Relevancia Clínica
9.
Artículo en Inglés | WPRIM (Pacífico Occidental) | ID: wpr-199403

RESUMEN

Plasmablastic lymphoma (PBL) is a recently identified entity that is considered to be a type of diffuse large B-cell lymphoma with a unique immunophenotype and a predilection for the oral cavity of patients with the human immunodeficiency virus (HIV). Although its clinical features may help in the differential diagnosis, an extraoral location in a patient without HIV makes it more difficult to suspect clinically. This case report is the first to describe a patient with PBL originating from the jejunum in a 60-yr-old, HIV-seronegative man. Computed tomography of the face, chest and abdomen showed about a 9.4x9.0 cm mass of the proximal jejunum, multiple masses in the musculoskeletal soft tissue, and multiple lymphadenopathies. The histological examinations demonstrated a large cell lymphoma with plasmablastic differentiation. The neoplastic cells were diffusely positive for MUM1, epithelial membrane antigen and lambda light chains, and focally positive for CD79a; but negative for CD3, CD20, CD30, CD34, CD45RO, CD56, CD99, and CD117. The proliferation index by Ki-67 immunohistochemistry was approximately 70%. These findings were compatible with the diagnosis of PBL. The findings in this case suggest that PBL should be included in the differential diagnosis of a small bowel mass even in a HIV-negative patient.


Asunto(s)
Humanos , Masculino , Persona de Mediana Edad , Diagnóstico Diferencial , Inmunofenotipificación , Neoplasias del Yeyuno/inmunología , Yeyuno/inmunología , Linfoma Inmunoblástico de Células Grandes/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA