Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Nat Immunol ; 9(7): 769-76, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18516037

RESUMEN

The intestinal cell types responsible for defense against pathogenic organisms remain incompletely characterized. Here we identify a subset of CD11c(hi)CD11b(hi) lamina propria dendritic cells (LPDCs) that expressed Toll-like receptor 5 (TLR5) in the small intestine. When stimulated by the TLR5 ligand flagellin, TLR5(+) LPDCs induced the differentiation of naive B cells into immunoglobulin A-producing plasma cells by a mechanism independent of gut-associated lymphoid tissue. In addition, by a mechanism dependent on TLR5 stimulation, these LPDCs promoted the differentiation of antigen-specific interleukin 17-producing T helper cells and type 1 T helper cells. Unlike spleen DCs, the LPDCs specifically produced retinoic acid, which, in a dose-dependent way, supported the generation and retention of immunoglobulin A-producing cells in the lamina propria and positively regulated the differentiation interleukin 17-producing T helper cells. Our findings demonstrate unique properties of LPDCs and the importance of TLR5 for adaptive immunity in the intestine.


Asunto(s)
Formación de Anticuerpos , Células Dendríticas/inmunología , Inmunidad Celular , Inmunidad Mucosa , Receptor Toll-Like 5/biosíntesis , Animales , Linfocitos B/inmunología , Células Cultivadas , Células Dendríticas/metabolismo , Ensayo de Inmunoadsorción Enzimática , Flagelina/inmunología , Citometría de Flujo , Inmunoglobulina A/biosíntesis , Inmunoglobulina A/inmunología , Inmunohistoquímica , Activación de Linfocitos/inmunología , Ratones , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T/inmunología , Tretinoina/inmunología , Tretinoina/metabolismo
2.
Gastroenterology ; 152(8): 1998-2010, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28246016

RESUMEN

BACKGROUND & AIMS: Obesity and metabolic syndrome have been associated with alterations to the intestinal microbiota. However, few studies examined the effects of obesity on the intestinal immune system. We investigated changes in subsets of intestinal CD4+ T-helper (TH) cells with obesity and the effects of gut-tropic TH17 cells in mice on a high-fat diet (HFD). METHODS: We isolated immune cells from small intestine and adipose tissue of C57BL/6 mice fed a normal chow diet or a HFD for 10 weeks and analyzed the cells by flow cytometry. Mice fed a vitamin A-deficient HFD were compared with mice fed a vitamin A-sufficient HFD. Obese RAG1-deficient mice were given injections of only regulatory T cells or a combination of regulatory T cells and TH17 cells (wild type or deficient in integrin ß7 subunit or interleukin 17 [IL17]). Mice were examined for weight gain, fat mass, fatty liver, glucose tolerance, and insulin resistance. Fecal samples were collected before and after T cell transfer and analyzed for microbiota composition by metagenomic DNA sequencing and quantitative polymerase chain reaction. RESULTS: Mice placed on a HFD became obese, which affected the distribution of small intestinal CD4+ TH cells. Intestinal tissues from obese mice had significant reductions in the proportion of TH17 cells but increased proportion of TH1 cells, compared with intestinal tissues from nonobese mice. Depletion of vitamin A in obese mice further reduced the proportion of TH17 cells in small intestine; this reduction correlated with more weight gain and worsening of glucose intolerance and insulin resistance. Adoptive transfer of in vitro-differentiated gut-tropic TH17 cells to obese mice reduced these metabolic defects, which required the integrin ß7 subunit and IL17. Delivery of TH17 cells to intestines of mice led to expansion of commensal microbes associated with leanness. CONCLUSIONS: In mice, intestinal TH17 cells contribute to development of a microbiota that maintains metabolic homeostasis, via IL17. Gut-homing TH17 cells might be used to reduce metabolic disorders in obese individuals.


Asunto(s)
Traslado Adoptivo , Inmunidad Mucosa , Resistencia a la Insulina , Intestino Delgado/inmunología , Síndrome Metabólico/prevención & control , Obesidad/prevención & control , Células Th17/trasplante , Animales , Células Cultivadas , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Heces/microbiología , Microbioma Gastrointestinal/inmunología , Genotipo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Interacciones Huésped-Patógeno , Cadenas beta de Integrinas/genética , Cadenas beta de Integrinas/metabolismo , Interleucina-17/deficiencia , Interleucina-17/genética , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/inmunología , Síndrome Metabólico/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/genética , Obesidad/inmunología , Obesidad/microbiología , Fenotipo , Células Th17/inmunología , Células Th17/microbiología , Factores de Tiempo , Deficiencia de Vitamina A/complicaciones
3.
J Allergy Clin Immunol ; 137(2): 507-516.e8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26433560

RESUMEN

BACKGROUND: The incidence of food allergies has increased dramatically during the last decade. Recently, probiotics have been studied for the prevention and treatment of allergic disease. OBJECTIVE: We examined whether Bifidobacterium longum KACC 91563 and Enterococcus faecalis KACC 91532 have the capacity to suppress food allergies. METHODS: B longum KACC 91563 and E faecalis KACC 91532 were administered to BALB/c wild-type mice, in which food allergy was induced by using ovalbumin and alum. Food allergy symptoms and various immune responses were assessed. RESULTS: B longum KACC 91563, but not E faecalis KACC 91532, alleviated food allergy symptoms. Extracellular vesicles of B longum KACC 91563 bound specifically to mast cells and induced apoptosis without affecting T-cell immune responses. Furthermore, injection of family 5 extracellular solute-binding protein, a main component of extracellular vesicles, into mice markedly reduced the occurrence of diarrhea in a mouse food allergy model. CONCLUSION: B longum KACC 91563 induces apoptosis of mast cells specifically and alleviates food allergy symptoms. Accordingly, B longum KACC 91563 and family 5 extracellular solute-binding protein exhibit potential as therapeutic approaches for food allergies.


Asunto(s)
Proteínas Bacterianas/inmunología , Bifidobacterium/inmunología , Vesículas Extracelulares/inmunología , Hipersensibilidad a los Alimentos/inmunología , Hipersensibilidad a los Alimentos/microbiología , Inmunomodulación , Mastocitos/inmunología , Animales , Apoptosis/inmunología , Proteínas Bacterianas/metabolismo , Bifidobacterium/metabolismo , Recuento de Células , Citocinas/biosíntesis , Modelos Animales de Enfermedad , Endocitosis/inmunología , Vesículas Extracelulares/metabolismo , Hipersensibilidad a los Alimentos/metabolismo , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Intestino Delgado/microbiología , Intestino Delgado/patología , Mastocitos/metabolismo , Mastocitos/microbiología , Ratones , Probióticos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
4.
Gut ; 64(2): 260-71, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24902766

RESUMEN

OBJECTIVE: Programmed death-ligand 1 (PD-L1) has been shown to negatively regulate immune responses via its interaction with PD-1 receptor. In this study, we investigated the effects of PD-L1-Fc treatment on intestinal inflammation using two murine models of inflammatory colitis induced by dextran sulfate sodium (DSS) and T-cell transfer. DESIGN: The anti-colitis effect of adenovirus expressing Fc-conjugated PD-L1 (Ad/PD-L1-Fc) and recombinant PD-L1-Fc protein was evaluated in DSS-treated wild-type and Rag-1 knockout (KO) mice. We examined differentiation of T-helper cells, frequency of innate immune cells, and cytokine production by dendritic cells (DCs) in the colon from DSS-treated mice after PD-L1-Fc administration. In Rag-1 KO mice reconstituted with CD4 CD45RB(high) T cells, we assessed the treatment effect of PD-L1-Fc protein on the development of colitis. RESULTS: Administration of Ad/PD-L1-Fc significantly ameliorated DSS-induced colitis, which was accompanied by diminished frequency of interleukin (IL)-17A-producing CD4 T cells and increased interferon-γ-producing CD4 T cells in the colon of DSS-fed mice. The anti-colitic effect of PD-L1-Fc treatment was also observed in DSS-treated Rag-1 KO mice, indicating lymphoid cell independency. PD-L1-Fc modulated cytokine production by colonic DCs and the effect was dependent on PD-1 expression. Furthermore, PD-L1-Fc protein could significantly reduce the severity of colitis in CD4 CD45RB(high) T-cell-transferred Rag-1 KO mice. CONCLUSIONS: Based on the protective effect of PD-L1-Fc against DSS-induced and T-cell-induced colitis, our results suggest that PD-1-mediated inhibitory signals have a crucial role in limiting the development of colonic inflammation. This implicates that PD-L1-Fc may provide a novel therapeutic approach to treat inflammatory bowel disease.


Asunto(s)
Antígeno B7-H1/uso terapéutico , Colitis Ulcerosa/prevención & control , Factores Inmunológicos/uso terapéutico , Enfermedad Aguda , Adenoviridae/genética , Animales , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Antígeno B7-H1/farmacología , Diferenciación Celular/efectos de los fármacos , Colitis Ulcerosa/etiología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colon/inmunología , Citocinas/biosíntesis , Células Dendríticas/inmunología , Sulfato de Dextran , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos/métodos , Vectores Genéticos , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Inmunidad Innata , Inmunidad Mucosa , Fragmentos Fc de Inmunoglobulinas/genética , Fragmentos Fc de Inmunoglobulinas/metabolismo , Fragmentos Fc de Inmunoglobulinas/farmacología , Fragmentos Fc de Inmunoglobulinas/uso terapéutico , Factores Inmunológicos/genética , Factores Inmunológicos/metabolismo , Factores Inmunológicos/farmacología , Mucosa Intestinal/inmunología , Transfusión de Linfocitos , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Recombinantes de Fusión/farmacología , Proteínas Recombinantes de Fusión/uso terapéutico , Linfocitos T Colaboradores-Inductores/inmunología , Células Th17/inmunología
5.
Plant Biotechnol J ; 13(1): 62-72, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25065685

RESUMEN

Pandemics in poultry caused by the highly pathogenic avian influenza (HPAI) A virus occur too frequently globally, and there is growing concern about the HPAI A virus due to the possibility of a pandemic among humans. Thus, it is important to develop a vaccine against HPAI suitable for both humans and animals. Various approaches are underway to develop such vaccines. In particular, an edible vaccine would be a convenient way to vaccinate poultry because of the behaviour of the animals. However, an edible vaccine is still not available. In this study, we developed a strategy of effective vaccination of mice by the oral administration of transgenic Arabidopsis plants (HA-TG) expressing haemagglutinin (HA) in the endoplasmic reticulum (ER). Expression of HA in the ER resulted in its high-level accumulation, N-glycosylation, protection from proteolytic degradation and long-term stability. Oral administration of HA-TG with saponin elicited high levels of HA-specific systemic IgG and mucosal IgA responses in mice, which resulted in protection against a lethal influenza virus infection with attenuated inflammatory symptoms. Based on these results, we propose that oral administration of freeze-dried leaf powders from transgenic plants expressing HA in the ER together with saponin is an attractive strategy for vaccination against influenza A virus.


Asunto(s)
Adyuvantes Inmunológicos/farmacología , Retículo Endoplásmico/metabolismo , Glicoproteínas Hemaglutininas del Virus de la Influenza/inmunología , Subtipo H5N1 del Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Saponinas/inmunología , Vacunación , Administración Oral , Animales , Formación de Anticuerpos/efectos de los fármacos , Formación de Anticuerpos/inmunología , Especificidad de Anticuerpos/efectos de los fármacos , Especificidad de Anticuerpos/inmunología , Antígenos Virales/inmunología , Arabidopsis/genética , Relación Dosis-Respuesta Inmunológica , Femenino , Inmunidad Humoral/efectos de los fármacos , Inmunidad Mucosa/efectos de los fármacos , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/inmunología , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/virología , Plantas Modificadas Genéticamente , Neumonía/inmunología , Neumonía/patología , Neumonía/prevención & control , Neumonía/virología , Proteínas Recombinantes de Fusión/metabolismo
6.
J Immunol ; 190(10): 5296-305, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23589614

RESUMEN

Small intestinal innate lymphoid cells (ILCs) regulate intestinal epithelial cell homeostasis and help to prevent pathogenic bacterial infections by producing IL-22. In a global gene-expression analysis comparing small intestinal ILCs (Lin(-)c-Kit(+)Sca-1(-) cells) with non-ILCs (Lin(-)c-Kit(-)Sca-1(-) cells), we found that Lin(-)c-Kit(+)Sca-1(-) cells highly expressed the mRNAs for Il22, antimicrobial peptides, Csf2rb2 (Il3r), mast cell proteases, and Rorc. We then subdivided the Lin(-)c-Kit(+)Sca-1(-) cells into three groups--Lin(-)c-Kit(+)NKp46(-)CD4(-), Lin(-)c-Kit(+)NKp46(-)CD4(+) (CD4(+) LTi-like cells), and Lin(-)c-Kit(+)NKp46(+) (NKp46(+) ILC22 cells)--and showed that the Lin(-)c-Kit(+)NKp46(-)CD4(-) cells produced the highest level of IL-22 protein after IL-1ß, IL-23, or IL-1ß and IL-23 stimulation. In addition, we showed that the majority of the Lin(-)c-Kit(+)NKp46(-)CD4(-) population was IL-7Rα(+)CD34(-)ß7(int) cells, and IL-7Rα(-) cells could be divided into three subsets (CD34(+)ß7(int), CD34(-)ß7(int), and CD34(int)ß7(hi) cells). The IL-7Rα(+)CD34(-)ß7(int) cells strongly expressed the transcripts for Il17f and Il22 after costimulation with IL-1ß and IL-23. The IL-7Rα(-)CD34(+)ß7(int) and IL-7Rα(-)CD34(int)ß7(hi) cells predominantly expressed the transcripts for mast cell proteases and differentiated almost entirely into mast cells after 1 wk in culture medium supplemented with a cytokine mixture, whereas the IL-7Rα(-)CD34(-)ß7(int) cells highly expressed α-defensins and showed no differentiation. Taken together, these findings indicate that the IL-7Rα(-)CD34(+)ß7(int) and IL-7Rα(-)CD34(int)ß7(hi) populations are mast cell progenitors, and the IL-7Rα(+)CD34(-)ß7(int) (CD4(-) LTi-like cells) and IL-7Rα(-)CD34(-)ß7(int) populations within Lin(-)c-Kit(+)NKp46(-)CD4(-) cells may control intestinal homeostasis and provide intestinal protection by producing high levels of IL-22 and α-defensins, respectively.


Asunto(s)
Infecciones Bacterianas/inmunología , Interleucina-1beta/metabolismo , Interleucinas/biosíntesis , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Linfocitos/metabolismo , Animales , Antígenos CD34 , Antígenos Ly/metabolismo , Infecciones Bacterianas/prevención & control , Antígenos CD4/metabolismo , Diferenciación Celular , Células Cultivadas , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Interleucina-23/metabolismo , Subunidad alfa del Receptor de Interleucina-7/metabolismo , Interleucinas/inmunología , Mucosa Intestinal/citología , Linfocitos/inmunología , Ratones , Ratones Endogámicos BALB C , Receptor 1 Gatillante de la Citotoxidad Natural/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Receptores de Interleucina-7 , alfa-Defensinas/biosíntesis , alfa-Defensinas/inmunología , Interleucina-22
7.
Nature ; 456(7219): 264-8, 2008 Nov 13.
Artículo en Inglés | MEDLINE | ID: mdl-18849965

RESUMEN

Systems for protein degradation are essential for tight control of the inflammatory immune response. Autophagy, a bulk degradation system that delivers cytoplasmic constituents into autolysosomes, controls degradation of long-lived proteins, insoluble protein aggregates and invading microbes, and is suggested to be involved in the regulation of inflammation. However, the mechanism underlying the regulation of inflammatory response by autophagy is poorly understood. Here we show that Atg16L1 (autophagy-related 16-like 1), which is implicated in Crohn's disease, regulates endotoxin-induced inflammasome activation in mice. Atg16L1-deficiency disrupts the recruitment of the Atg12-Atg5 conjugate to the isolation membrane, resulting in a loss of microtubule-associated protein 1 light chain 3 (LC3) conjugation to phosphatidylethanolamine. Consequently, both autophagosome formation and degradation of long-lived proteins are severely impaired in Atg16L1-deficient cells. Following stimulation with lipopolysaccharide, a ligand for Toll-like receptor 4 (refs 8, 9), Atg16L1-deficient macrophages produce high amounts of the inflammatory cytokines IL-1beta and IL-18. In lipopolysaccharide-stimulated macrophages, Atg16L1-deficiency causes Toll/IL-1 receptor domain-containing adaptor inducing IFN-beta (TRIF)-dependent activation of caspase-1, leading to increased production of IL-1beta. Mice lacking Atg16L1 in haematopoietic cells are highly susceptible to dextran sulphate sodium-induced acute colitis, which is alleviated by injection of anti-IL-1beta and IL-18 antibodies, indicating the importance of Atg16L1 in the suppression of intestinal inflammation. These results demonstrate that Atg16L1 is an essential component of the autophagic machinery responsible for control of the endotoxin-induced inflammatory immune response.


Asunto(s)
Autofagia/genética , Proteínas Portadoras/genética , Regulación de la Expresión Génica/efectos de los fármacos , Interleucina-1beta/biosíntesis , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Adenina/análogos & derivados , Adenina/farmacología , Animales , Proteínas Relacionadas con la Autofagia , Quimera , Colitis/inducido químicamente , Colitis/inmunología , Sulfato de Dextran/farmacología , Femenino , Interleucina-1beta/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación
8.
Microorganisms ; 12(6)2024 May 27.
Artículo en Inglés | MEDLINE | ID: mdl-38930468

RESUMEN

Given the recognized involvement of the gut microbiome in the development of obesity, considerable efforts are being made to discover probiotics capable of preventing and managing obesity. In this study, we report the discovery of Lactiplantibacillus plantarum GBCC_F0227, isolated from fermented food, which exhibited superior triglyceride catabolism efficacy compared to L. plantarum WCSF1. Molecular analysis showed elevated expression levels of α/ß hydrolases with lipase activity (abH04, abH08_1, abH08_2, abH11_1, and abH11_2) in L. plantarum GBCC_F0227 compared to L. plantarum WCFS1, demonstrating its enhanced lipolytic activity. In a high-fat-diet (HFD)-induced mouse obesity model, the administration of L. plantarum GBCC_F0227 mitigated weight gain, reduced blood triglycerides, and diminished fat mass. Furthermore, L. plantarum GBCC_F0227 upregulated adiponectin gene expression in adipose tissue, indicative of favorable metabolic modulation, and showed robust growth and low cytotoxicity, underscoring its industrial viability. Therefore, our findings encourage the further investigation of L. plantarum GBCC_F0227's therapeutic applications for the prevention and treatment of obesity and associated metabolic diseases.

9.
Cells ; 12(11)2023 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-37296626

RESUMEN

Mast cells act as key effector cells of inflammatory responses through degranulation. Mast cell degranulation is induced by the activation of cell surface receptors, such as FcεRI, MRGPRX2/B2, and P2RX7. Each receptor, except FcεRI, varies in its expression pattern depending on the tissue, which contributes to their differing involvement in inflammatory responses depending on the site of occurrence. Focusing on the mechanism of allergic inflammatory responses by mast cells, this review will describe newly identified mast cell receptors in terms of their involvement in degranulation induction and patterns of tissue-specific expression. In addition, new drugs targeting mast cell degranulation for the treatment of allergy-related diseases will be introduced.


Asunto(s)
Hipersensibilidad , Mastocitos , Humanos , Mastocitos/metabolismo , Receptores de IgE/metabolismo , Hipersensibilidad/metabolismo , Desarrollo de Medicamentos , Proteínas del Tejido Nervioso/metabolismo , Receptores de Neuropéptido/metabolismo , Receptores Acoplados a Proteínas G/metabolismo
10.
Clin Immunol ; 144(3): 190-9, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22836084

RESUMEN

IL-12p40 homodimer is a natural antagonist of IL-12 and IL-23, which are potent pro-inflammatory cytokines required for Th1 and Th17 immune responses, respectively. It has been reported that Th17 response is involved in inflammatory bowel disease (IBD), a chronic disorder of the digestive system with steadily increasing incidence. Here, we investigated the effects of IL-12p40 delivered via recombinant adenovirus (rAd/IL-12p40) or mesenchymal stem cells (MSC/IL-12p40) in a dextran sulfate sodium salt (DSS)-induced colitis model. Injection of rAd/IL-12p40 or MSC/IL-12p40 efficiently attenuated colitis symptoms and tissue damage, leading to an increased survival rate. Moreover, IL-12p40 delivery suppressed IL-17A, but enhanced IFN-γ production from mesenteric lymph node cells, supporting the preferential suppression of IL-23 by IL-12p40 homodimer in vitro and the suppression of Th17 responses in vivo. Our results demonstrate that IL-12p40 delivery ameliorates DSS-induced colitis by suppressing IL-17A production and inflammation in the intestinal mucosa, providing an effective new therapeutic strategy for IBDs.


Asunto(s)
Colitis/inmunología , Inflamación/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Subunidad p40 de la Interleucina-12/inmunología , Interleucina-17/inmunología , Mucosa Intestinal/inmunología , Adenoviridae/inmunología , Animales , Colitis/inducido químicamente , Sulfato de Dextran , Modelos Animales de Enfermedad , Femenino , Interferón gamma/inmunología , Interleucina-12/inmunología , Interleucina-23/inmunología , Ganglios Linfáticos/inmunología , Células Madre Mesenquimatosas/inmunología , Ratones , Ratones Endogámicos C57BL , Ratas , Ratas Sprague-Dawley , Células Th17/inmunología
11.
Nat Commun ; 13(1): 5669, 2022 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-36167830

RESUMEN

IgE is central to the development of allergic diseases, and its neutralization alleviates allergic symptoms. However, most of these antibodies are based on IgG1, which is associated with an increased risk of fragment crystallizable-mediated side effects. Moreover, omalizumab, an anti-IgE antibody approved for therapeutic use, has limited benefits for patients with high IgE levels. Here, we assess a fusion protein with extracellular domain of high affinity IgE receptor, FcεRIα, linked to a IgD/IgG4 hybrid Fc domain we term IgETRAP, to reduce the risk of IgG1 Fc-mediated side effects. IgETRAP shows enhanced IgE binding affinity compared to omalizumab. We also see an enhanced therapeutic effect of IgETRAP in food allergy models when combined with Bifidobacterium longum, which results in mast cell number and free IgE levels. The combination of IgETRAP and B. longum may therefore represent a potent treatment for allergic patients with high IgE levels.


Asunto(s)
Bifidobacterium longum , Hipersensibilidad a los Alimentos , Bifidobacterium longum/metabolismo , Suplementos Dietéticos , Hipersensibilidad a los Alimentos/terapia , Humanos , Inmunoglobulina D , Inmunoglobulina E , Inmunoglobulina G , Omalizumab/uso terapéutico , Receptores de IgE/metabolismo
12.
Opt Express ; 19(14): 13089-96, 2011 Jul 04.
Artículo en Inglés | MEDLINE | ID: mdl-21747461

RESUMEN

The combination of two-photon microscopy (TPM) and optical coherence tomography (OCT) is useful in conducting in-vivo tissue studies, because they provide complementary information regarding tissues. In the present study, we developed a new combined system using separate light sources and scanners for individually optimal imaging conditions. TPM used a Ti-Sapphire laser and provided molecular and cellular information in microscopic tissue regions. Meanwhile, OCT used a wavelength-swept source centered at 1300 nm and provided structural information in larger tissue regions than TPM. The system was designed to do simultaneous imaging by combining light from both sources. TPM and OCT had the field of view values of 300 µm and 800 µm on one side respectively with a 20x objective. TPM had resolutions of 0.47 µm and 2.5 µm in the lateral and axial directions respectively, and an imaging speed of 40 frames/s. OCT had resolutions of 5 µm and 8 µm in lateral and axial directions respectively, a sensitivity of 97dB, and an imaging speed of 0.8 volumes per second. This combined system was tested with simple microsphere specimens, and was then applied to image small intestine and ear tissues of mouse models ex-vivo. Molecular, cellular, and structural information of the tissues were visualized using the proposed combined system.


Asunto(s)
Aumento de la Imagen/instrumentación , Iluminación/instrumentación , Microscopía de Fluorescencia por Excitación Multifotónica/instrumentación , Técnica de Sustracción/instrumentación , Tomografía de Coherencia Óptica/instrumentación , Animales , Diseño de Equipo , Análisis de Falla de Equipo , Ratones , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
13.
FEBS Lett ; 582(20): 3018-24, 2008 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-18675811

RESUMEN

Nepmucin/CLM-9 is an Ig domain-containing sialomucin expressed in vascular endothelial cells. Here we show that, like CD31, nepmucin was localized to interendothelial contacts and to vesicle-like structures along the cell border and underwent intracellular recycling. Functional analyses showed that nepmucin mediated homotypic and heterotypic cell adhesion via its Ig domain. Nepmucin-expressing endothelial cells showed enhanced lymphocyte transendothelial migration (TEM), which was abrogated by anti-nepmucin mAbs that block either homophilic or heterophilic binding. Notably, the mAbs that inhibited homophilic binding blocked TEM without affecting lymphocyte adhesion. These results suggest that endothelial nepmucin promotes lymphocyte TEM using multiple adhesion pathways.


Asunto(s)
Movimiento Celular , Endotelio Vascular/inmunología , Linfocitos/inmunología , Sialomucinas/inmunología , Animales , Adhesión Celular/inmunología , Endotelio Vascular/citología , Inmunoglobulinas/inmunología , Ratones , Ratones Endogámicos C57BL , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/inmunología , Estructura Terciaria de Proteína , Sialomucinas/genética , Vesículas Transportadoras/inmunología
14.
Sci Rep ; 8(1): 14174, 2018 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-30242205

RESUMEN

Paneth cells are one of the principal epithelial cell types in the small intestine, located at the base of intestinal crypts. Paneth cells play key roles in intestinal host-microbe homeostasis via granule secretion, and their dysfunction is implicated in pathogenesis of several diseases including Crohn's disease. Despite their physiological importance, study of Paneth cells has been hampered by the limited accessibility and lack of labeling methods. In this study, we developed a simple in vivo imaging method of Paneth cells in the intact mouse small intestine by using moxifloxacin and two-photon microscopy (TPM). Moxifloxacin, an FDA-approved antibiotic, was used for labeling cells and its fluorescence was strongly observed in Paneth cell granules by TPM. Moxifloxacin labeling of Paneth cell granules was confirmed by molecular counterstaining. Comparison of Paneth cells in wild type, genetically obese (ob/ob), and germ-free (GF) mice showed different granule distribution. Furthermore, Paneth cell degranulation was observed in vivo. Our study suggests that TPM with moxifloxacin labeling can serve as a useful tool for studying Paneth cell biology and related diseases.


Asunto(s)
Intestino Delgado/patología , Células de Paneth/patología , Animales , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Gránulos Citoplasmáticos/metabolismo , Modelos Animales de Enfermedad , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Intestino Delgado/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Microscopía/métodos , Moxifloxacino/metabolismo , Células de Paneth/metabolismo , Fotones
15.
Immune Netw ; 17(1): 41-47, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-28261019

RESUMEN

Eosinophils are potent effector cells implicated in allergic responses and helminth infections. Responding to stimuli, they release their granule-derived cytotoxic proteins and are involved in inflammatory processes. However, under homeostatic conditions, eosinophils are abundantly present in the intestine and are constantly in contact with the gut microbiota and maintain the balance of immune responses without inflammation. This situation indicates that intestinal eosinophils have an anti-inflammatory function unlike allergic eosinophils. In support of this notion, some papers have shown that eosinophils have different phenotypes depending on the site of residence and are a heterogeneous cell population. Recently, it was reported that eosinophils in the small intestine and adipose tissue, respectively, contribute to homeostasis of intestinal immune responses and metabolism. Accordingly, in this review, we summarize new functions of eosinophils demonstrated in recent studies and discuss their homeostatic functions.

16.
Yonsei Med J ; 58(6): 1083-1091, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29047231

RESUMEN

Gut microbiota play critical physiological roles in energy extraction from the intestine and in the control of systemic immunity, as well as local intestinal immunity. Disturbance of gut microbiota leads to the development of several diseases, such as colitis, inflammatory bowel diseases, metabolic disorders, cancer, etc. From a metabolic point of view, the gut is a large metabolic organ and one of the first to come into contact with dietary fats. Interestingly, excessive dietary fat has been incriminated as a primary culprit of metabolic syndrome and obesity. After intake of high-fat diet or Western diet, extensive changes in gut microbiota have been observed, which may be an underlying cause of alterations in whole body metabolism and nutrient homeostasis. Here, we summarize recent data on changes in the gut microbiota and immunity associated with dietary fat, as well as their relationships with the pathogenesis of metabolic syndrome. These findings may provide insight into the understanding of the complex pathophysiology related to the development of metabolic diseases and offer an opportunity to develop novel candidates for therapeutic agents.


Asunto(s)
Dieta Alta en Grasa , Microbioma Gastrointestinal , Síndrome Metabólico , Obesidad/fisiopatología , Grasas de la Dieta , Humanos , Enfermedades Inflamatorias del Intestino , Intestinos
17.
Immunol Lett ; 190: 247-256, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28860040

RESUMEN

Oral vaccine responsiveness is often lower in children from less developed countries. Childhood malnutrition may be associated with poor immune response to oral vaccines. The present study was designed to investigate whether protein energy malnutrition (PEM) impairs B cell immunity and ultimately reduces oral vaccine efficacy in a mouse model. Purified isocaloric diets containing low protein (1/10 the protein of the control diet) were used to determine the effect of PEM. PEM increased both nonspecific total IgA and oral antigen-specific IgA in serum without alteration of gut permeability. However, PEM decreased oral antigen-specific IgA in feces, which is consistent with decreased expression of polymeric Immunoglobulin receptor (pIgR) in the small intestine. Of note, polymeric IgA was predominant in serum under PEM. In addition, PEM altered B cell development status in the bone marrow and increased the frequency of IgA-secreting B cells, as well as IgA secretion by long-lived plasma cells in the small intestinal lamina propria. Moreover, PEM reduced the protective efficacy of the mucosally administered cholera vaccine and recombinant attenuated Salmonella enterica serovar Typhimurium vaccine in a mouse model. Our results suggest that PEM can impair mucosal immunity where IgA plays an important role in host protection and may partly explain the reduced efficacy of oral vaccines in malnourished subjects.


Asunto(s)
Linfocitos B/inmunología , Vacunas contra el Cólera/inmunología , Inmunoglobulina A/biosíntesis , Mucosa Intestinal/inmunología , Desnutrición Proteico-Calórica/inmunología , Vacunas contra la Salmonella/inmunología , Administración Oral , Animales , Niño , Países Desarrollados , Modelos Animales de Enfermedad , Femenino , Humanos , Inmunidad Humoral , Ratones , Ratones Endogámicos C57BL , Resultado del Tratamiento , Vacunación
18.
Science ; 351(6275): 858-63, 2016 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-26822607

RESUMEN

Dietary antigens are normally rendered nonimmunogenic through a poorly understood "oral tolerance" mechanism that involves immunosuppressive regulatory T (Treg) cells, especially Treg cells induced from conventional T cells in the periphery (pTreg cells). Although orally introducing nominal protein antigens is known to induce such pTreg cells, whether a typical diet induces a population of pTreg cells under normal conditions thus far has been unknown. By using germ-free mice raised and bred on an elemental diet devoid of dietary antigens, we demonstrated that under normal conditions, the vast majority of the small intestinal pTreg cells are induced by dietary antigens from solid foods. Moreover, these pTreg cells have a limited life span, are distinguishable from microbiota-induced pTreg cells, and repress underlying strong immunity to ingested protein antigens.


Asunto(s)
Proteínas en la Dieta/inmunología , Dispepsia/inmunología , Microbioma Gastrointestinal/inmunología , Intestino Delgado/inmunología , Intestino Delgado/microbiología , Linfocitos T Reguladores/inmunología , Animales , Antígenos/inmunología , Dieta , Vida Libre de Gérmenes , Tolerancia Inmunológica , Inmunidad Mucosa , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL
19.
J Exp Med ; 213(4): 555-67, 2016 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-26951334

RESUMEN

Eosinophils play proinflammatory roles in helminth infections and allergic diseases. Under steady-state conditions, eosinophils are abundantly found in the small intestinal lamina propria, but their physiological function is largely unexplored. In this study, we found that small intestinal eosinophils down-regulate Th17 cells. Th17 cells in the small intestine were markedly increased in the ΔdblGATA-1 mice lacking eosinophils, and an inverse correlation was observed between the number of eosinophils and that of Th17 cells in the small intestine of wild-type mice. In addition, small intestinal eosinophils suppressed the in vitro differentiation of Th17 cells, as well as IL-17 production by small intestinal CD4(+)T cells. Unlike other small intestinal immune cells or circulating eosinophils, we found that small intestinal eosinophils have a unique ability to constitutively secrete high levels of IL-1 receptor antagonist (IL-1Ra), a natural inhibitor of IL-1ß. Moreover, small intestinal eosinophils isolated from IL-1Ra-deficient mice failed to suppress Th17 cells. Collectively, our results demonstrate that small intestinal eosinophils play a pivotal role in the maintenance of intestinal homeostasis by regulating Th17 cells via production of IL-1Ra.


Asunto(s)
Cistinil Aminopeptidasa/inmunología , Eosinófilos/inmunología , Intestino Delgado/inmunología , Células Th17/inmunología , Animales , Cistinil Aminopeptidasa/genética , Eosinófilos/citología , Interleucina-17/genética , Interleucina-17/inmunología , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Intestino Delgado/citología , Ratones , Ratones Transgénicos , Células Th17/citología
20.
J Leukoc Biol ; 95(4): 621-9, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24319287

RESUMEN

PD-1 is a well-established negative regulator of T cell responses by inhibiting proliferation and cytokine production of T cells via interaction with its ligands, B7-H1 (PD-L1) and B7-DC (PD-L2), expressed on non-T cells. Recently, PD-1 was found to be expressed in innate cells, including activated DCs, and plays roles in suppressing production of inflammatory cytokines. In this study, we demonstrate that PD-1 KO DCs exhibited prolonged longevity compared with WT DCs in the dLNs after transfer of DCs into hind footpads. Interestingly, upon LPS stimulation, WT DCs increased the expression of PD-1 and started to undergo apoptosis. DCs, in spleen of LPS-injected PD-1 KO mice, were more resistant to LPS-mediated apoptosis in vivo than WT controls. Moreover, treatment of blocking anti-PD-1 mAb during DC maturation resulted in enhanced DC survival, suggesting that PD-1:PD-L interactions are involved in DC apoptosis. As a result, PD-1-deficient DCs augmented T cell responses in terms of antigen-specific IFN-γ production and proliferation of CD4 and CD8 T cells to a greater degree than WT DCs. Moreover, PD-1 KO DCs exhibited increased MAPK1 and CD40-CD40L signaling, suggesting a possible mechanism for enhanced DC survival in the absence of PD-1 expression. Taken together, our findings further extend the function of PD-1, which plays an important role in apoptosis of activated DCs and provides important implications for PD-1-mediated immune regulation.


Asunto(s)
Células Dendríticas/inmunología , Receptor de Muerte Celular Programada 1/fisiología , Traslado Adoptivo , Animales , Apoptosis , Antígenos CD40/fisiología , Ligando de CD40/fisiología , Supervivencia Celular , Células Cultivadas , Células Dendríticas/citología , Lipopolisacáridos/farmacología , Ratones , Proteína Quinasa 1 Activada por Mitógenos/fisiología , Linfocitos T/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA