Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
2.
Cutan Ocul Toxicol ; 43(2): 124-128, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38284163

RESUMEN

PURPOSE: To study the effects of the anti-IL-23A antibody risankizumab on the IL-36γ/IL-23A/IL-17A signalling cascade we used a newly developed 3D skin model consisting of primary human keratinocytes, fibroblasts and γδ-T-cells. METHODS: In this in vitro study we developed new full-thickness 3D skin models containing normal human epidermal keratinocytes (NHEK), normal human dermal fibroblasts (NHDF) and IL-23A responsive and IL-17A producing γδ-T-cells. The effects of IL-36γ stimulation with and without risankizumab treatment on IL-23A and IL-17A expression were examined at the RNA and protein levels. RESULTS: In preliminary monolayer experiments stimulation of γδ-T-cells with IL-23A promoted the IL-17A expression that was inhibited after risankizumab treatment. Using 3D skin models containing γδ-T-cells, we found that stimulation with IL-36γ significantly increased not only IL-23A but also IL-17A expression. These effects were inhibited by concomitant treatment with risankizumab. CONCLUSIONS: Our results showed that blockade of IL-23A has inhibitory effects on the IL-36γ/IL-23A feedforward loop. Our newly developed 3D skin model containing IL-23A responsive and IL-17A producing γδ-T-cells enables molecular analysis of targeted therapies aimed at the IL-36γ/IL-23A/IL-17A signalling cascade in psoriasis.


Asunto(s)
Anticuerpos Monoclonales , Interleucina-17 , Subunidad p19 de la Interleucina-23 , Queratinocitos , Piel , Humanos , Interleucina-17/metabolismo , Queratinocitos/efectos de los fármacos , Queratinocitos/metabolismo , Anticuerpos Monoclonales/farmacología , Subunidad p19 de la Interleucina-23/metabolismo , Piel/efectos de los fármacos , Piel/metabolismo , Piel/inmunología , Células Cultivadas , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Interleucina-1/metabolismo , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/inmunología , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Psoriasis/tratamiento farmacológico , Psoriasis/inmunología
3.
Biochem Pharmacol ; 213: 115629, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37257721

RESUMEN

Autoimmune uveitis is a non-infectious, inflammatory intraocular disease that affects the uveal and adjacent tissues. It frequently causes varying degrees of visual loss. Evidence for the strong association between activated γδ T cells and the development of autoimmune uveitis is growing. The innate and adaptive immune response are connected in the early phases by the γδ T cells that contain the γ and δ chains. γδ T cells can identify antigens in a manner that is not constrained by the MHC. When activated by various pathways, γδ T cells can not only secrete pro-inflammatory factors early on (such as IL-17), but they can also promote Th17 cells responses, which ultimately exacerbates autoimmune uveitis. Therefore, we review the mechanisms by which γδ T cells affect autoimmune uveitis in different activation and disease states. Moreover, we also prospect for immunotherapies targeting different γδ T cell-related action pathways, providing a reference for exploring new drug for the treatment of autoimmune uveitis.


Asunto(s)
Enfermedades Autoinmunes , Inmunoterapia , Linfocitos Intraepiteliales , Activación de Linfocitos , Uveítis , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/inmunología , Uveítis/tratamiento farmacológico , Uveítis/inmunología , Enfermedades Autoinmunes/tratamiento farmacológico , Enfermedades Autoinmunes/inmunología , Humanos , Animales , Células Th17/inmunología , Interleucina-17/metabolismo , Inmunoterapia/métodos
4.
Front Immunol ; 12: 699478, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34721375

RESUMEN

Angiogenesis is an essential physiological process and hallmark of cancer. Currently, antiangiogenic therapy, mostly targeting the vascular endothelial growth factor (VEGF)/VEGFR2 signaling axis, is commonly used in the clinic for solid tumors. However, antiangiogenic therapies for breast cancer patients have produced limited survival benefits since cancer cells rapidly resistant to anti-VEGFR2 therapy. We applied the low-dose and high-dose VEGFR2 mAb or VEGFR2-tyrosine kinase inhibitor (TKI) agents in multiple breast cancer mouse models and found that low-dose VEGFR2 mAb or VEGFR2-TKI achieved good effects in controlling cancer progression, while high-dose treatment was not effective. To further investigate the mechanism involved in regulating the drug resistance, we found that high-dose anti-VEGFR2 treatment elicited IL17A expression in γδ T cells via VEGFR1-PI3K-AKT pathway activation and then promoted N2-like neutrophil polarization, thus inducing CD8+ T cell exhaustion to shape an immunosuppressive microenvironment. Combining anti-VEGFR2 therapy with immunotherapy such as IL17A, PD-1 or Ly-6G mAb therapy, which targeting the immunomodulatory axis of "γδT17 cells-N2 neutrophils" in vivo, showed promising therapeutic effects in breast cancer treatment. This study illustrates the potential mechanism of antiangiogenic therapy resistance in breast cancer and provides synergy treatment for cancer.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Neoplasias de la Mama/inmunología , Resistencia a Antineoplásicos/inmunología , Linfocitos Intraepiteliales/efectos de los fármacos , Neutrófilos/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Animales , Femenino , Interleucina-17/inmunología , Linfocitos Intraepiteliales/inmunología , Ratones , Neutrófilos/inmunología , Microambiente Tumoral/inmunología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
5.
Front Immunol ; 12: 729085, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34630403

RESUMEN

Recently, many discoveries have elucidated the cellular and molecular diversity in the leukemic microenvironment and improved our knowledge regarding their complex nature. This has allowed the development of new therapeutic strategies against leukemia. Advances in biotechnology and the current understanding of T cell-engineering have led to new approaches in this fight, thus improving cell-mediated immune response against cancer. However, most of the investigations focus only on conventional cytotoxic cells, while ignoring the potential of unconventional T cells that until now have been little studied. γδ T cells are a unique lymphocyte subpopulation that has an extensive repertoire of tumor sensing and may have new immunotherapeutic applications in a wide range of tumors. The ability to respond regardless of human leukocyte antigen (HLA) expression, the secretion of antitumor mediators and high functional plasticity are hallmarks of γδ T cells, and are ones that make them a promising alternative in the field of cell therapy. Despite this situation, in particular cases, the leukemic microenvironment can adopt strategies to circumvent the antitumor response of these lymphocytes, causing their exhaustion or polarization to a tumor-promoting phenotype. Intervening in this crosstalk can improve their capabilities and clinical applications and can make them key components in new therapeutic antileukemic approaches. In this review, we highlight several characteristics of γδ T cells and their interactions in leukemia. Furthermore, we explore strategies for maximizing their antitumor functions, aiming to illustrate the findings destined for a better mobilization of γδ T cells against the tumor. Finally, we outline our perspectives on their therapeutic applicability and indicate outstanding issues for future basic and clinical leukemia research, in the hope of contributing to the advancement of studies on γδ T cells in cancer immunotherapy.


Asunto(s)
Antineoplásicos Inmunológicos/uso terapéutico , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Inmunoterapia Adoptiva , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/trasplante , Leucemia/terapia , Animales , Antineoplásicos Inmunológicos/efectos adversos , Proliferación Celular/efectos de los fármacos , Humanos , Inhibidores de Puntos de Control Inmunológico/efectos adversos , Inmunoterapia Adoptiva/efectos adversos , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Leucemia/genética , Leucemia/inmunología , Leucemia/metabolismo , Activación de Linfocitos/efectos de los fármacos , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Fenotipo , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología , Receptores Quiméricos de Antígenos/metabolismo , Transducción de Señal , Microambiente Tumoral
6.
Biomed Pharmacother ; 144: 112283, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34628169

RESUMEN

Psoriasis is a chronic inflammatory skin disease characterized by epidermal hyperplasia and cellular infiltration. Studies have shown that disease development depends on proinflammatory cytokines, such as interleukin (IL)-23 and IL-17. It has been suggested that IL-23 produced by innate immune cells, such as macrophages, stimulates a subset of helper T cells to release IL-17, promoting neutrophil recruitment and keratinocyte proliferation. However, recent studies have revealed the crucial role of γδT cells in psoriasis pathogenesis as the primary source of dermal IL-17. The nuclear receptors REV-ERBs are ligand-dependent transcription factors recognized as circadian rhythm regulators. REV-ERBs negatively regulate IL-17-producing helper T cells, whereas the involvement of REV-ERBs in regulating IL-17-producing γδT (γδT17) cells remains unclear. Here we revealed the regulatory mechanism involving γδT17 cells through REV-ERBs. γδT17 cell levels were remarkably elevated in the secondary lymphoid organs of mice that lacked an isoform of REV-ERBs. A synthetic REV-ERB agonist, SR9009, suppressed γδT17 cells in vitro and in vivo. Topical application of SR9009 to the skin reduced the inflammatory symptoms of psoriasiform dermatitis in mice. The results of this study provide a novel therapeutic approach for psoriasis targeting REV-ERBs in γδT17 cells.


Asunto(s)
Antiinflamatorios/farmacología , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/efectos de los fármacos , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/agonistas , Psoriasis/tratamiento farmacológico , Pirrolidinas/farmacología , Piel/efectos de los fármacos , Tiofenos/farmacología , Administración Cutánea , Animales , Antiinflamatorios/administración & dosificación , Células Cultivadas , Modelos Animales de Enfermedad , Regulación hacia Abajo , Femenino , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Ratones Noqueados , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo D de la Subfamilia 1 de Receptores Nucleares/metabolismo , Psoriasis/inmunología , Psoriasis/metabolismo , Psoriasis/patología , Pirrolidinas/administración & dosificación , Transducción de Señal , Piel/inmunología , Piel/metabolismo , Tiofenos/administración & dosificación
7.
Cell Rep ; 36(9): 109574, 2021 08 31.
Artículo en Inglés | MEDLINE | ID: mdl-34469732

RESUMEN

Neuroinflammation in patients with Alzheimer's disease (AD) and related mouse models has been recognized for decades, but the contribution of the recently described meningeal immune population to AD pathogenesis remains to be addressed. Here, using the 3xTg-AD model, we report an accumulation of interleukin-17 (IL-17)-producing cells, mostly γδ T cells, in the brain and the meninges of female, but not male, mice, concomitant with the onset of cognitive decline. Critically, IL-17 neutralization into the ventricles is sufficient to prevent short-term memory and synaptic plasticity deficits at early stages of disease. These effects precede blood-brain barrier disruption and amyloid-beta or tau pathology, implying an early involvement of IL-17 in AD pathology. When IL-17 is neutralized at later stages of disease, the onset of short-memory deficits and amyloidosis-related splenomegaly is delayed. Altogether, our data support the idea that cognition relies on a finely regulated balance of "inflammatory" cytokines derived from the meningeal immune system.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Conducta Animal , Encéfalo/metabolismo , Cognición , Mediadores de Inflamación/metabolismo , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/metabolismo , Enfermedades Neuroinflamatorias/metabolismo , Sinapsis/metabolismo , Enfermedad de Alzheimer/patología , Enfermedad de Alzheimer/prevención & control , Enfermedad de Alzheimer/psicología , Animales , Antiinflamatorios/farmacología , Anticuerpos Monoclonales/farmacología , Anticuerpos Neutralizantes/farmacología , Conducta Animal/efectos de los fármacos , Encéfalo/efectos de los fármacos , Encéfalo/patología , Cognición/efectos de los fármacos , Modelos Animales de Enfermedad , Femenino , Mediadores de Inflamación/antagonistas & inhibidores , Interleucina-17/antagonistas & inhibidores , Linfocitos Intraepiteliales/efectos de los fármacos , Masculino , Memoria a Corto Plazo , Ratones de la Cepa 129 , Ratones Transgénicos , Enfermedades Neuroinflamatorias/patología , Enfermedades Neuroinflamatorias/prevención & control , Enfermedades Neuroinflamatorias/psicología , Plasticidad Neuronal , Sinapsis/efectos de los fármacos , Sinapsis/patología
8.
Front Immunol ; 12: 687961, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34335589

RESUMEN

Graft-versus-host disease (GVHD) is a pathology in which chemokines and their receptors play essential roles in directing the migration of alloreactive donor T cells into GVHD organs, thereby leading to further target tissue damage. Currently, acute GVHD (aGVHD) remains a major cause of high morbidity and mortality in patients who underwent allogeneic hematopoietic cell transplantation (alloHCT). The identification of immune cells that correlate with aGVHD is important and intriguing. To date, the involvement of innate-like γδ T cells in the pathogenesis of aGVHD is unclear. Herein, we found that primary human γδ T cells did not directly trigger allogeneic reactions. Instead, we revealed that γδ T cells facilitated the migration of CD4 T cells via the SDF-1-CXCR4 axis. These results indicate indirect regulation of γδ T cells in the development of aGVHD rather than a direct mechanism. Furthermore, we showed that the expression of CXCR4 was significantly elevated in γδ T cells and CD4 and CD8 T cells in recipients who experienced grades II-IV aGVHD after alloHCT. Consistently, CXCR4-expressing γδ T cells and CD4 T cells were induced in the target organs of mice suffering aGVHD. The depletion of γδ T cells in transplant grafts and treatment with AMD3100, an inhibitor of CXCR4 signaling, delayed the onset of aGVHD and prolonged survival in mice. Taken together, these findings suggest a role for γδ T cells in recruiting alloreactive CD4 T cells to target tissues through the expression of CXCR4. Our findings may help in understanding the mechanism of aGVHD and provide novel therapeutic targets.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Quimiocina CXCL12/metabolismo , Quimiotaxis de Leucocito , Enfermedad Injerto contra Huésped/etiología , Trasplante de Células Madre Hematopoyéticas/efectos adversos , Linfocitos Intraepiteliales/metabolismo , Receptores CXCR4/metabolismo , Adolescente , Adulto , Animales , Bencilaminas/farmacología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Ciclamas/farmacología , Modelos Animales de Enfermedad , Femenino , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/metabolismo , Enfermedad Injerto contra Huésped/prevención & control , Humanos , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/inmunología , Masculino , Ratones Endogámicos NOD , Persona de Mediana Edad , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Receptores CXCR4/antagonistas & inhibidores , Transducción de Señal , Trasplante Homólogo , Adulto Joven
9.
Gastroenterology ; 161(4): 1270-1287.e19, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34224738

RESUMEN

BACKGROUND & AIMS: The molecular checkpoints driving T cell activation and cytokine responses in ulcerative colitis (UC) are incompletely understood. Here, we studied the Tec kinase ITK in UC. METHODS: We analyzed patients with inflammatory bowel disease (n = 223) and evaluated ITK activity as well as the functional effects of cyclosporine-A (CsA). In addition, 3 independent murine colitis models were used to investigate the functional role of ITK. Finally, the activity of ITK was blocked via pharmacological inhibitors and genetically engineered mice. Readout parameters were mini-endoscopy, histopathology, mucosal T cell apoptosis, and cytokine production. RESULTS: We found an expansion of pITK-expressing mucosal CD4+ T cells in UC rather than Crohn's disease that correlated with disease severity. CsA suppressed activation of ITK in cultured CD4+ T cells and calcineurin-containing microclusters adjacent to the T cell receptor signaling complex. Functionally, the capacity of CsA to suppress activity of experimental colitis was critically dependent on ITK. Genetic inactivation of Itk via gene targeting or induction of allele-sensitive Itk mutants prevented experimental colitis in 3 colitis models, and treatment with pharmacological ITK blockers suppressed established colitis. In addition, ITK controlled apoptosis and activation of mucosal Th2 and Th17 lymphocytes via NFATc2 signaling pathways. CONCLUSIONS: ITK activation was detected in UC and could be down-regulated in cultured T cells by CsA administration. Selective targeting of ITK emerges as an attractive approach for treatment of chronic intestinal inflammation and potentially UC by driving resolution of mucosal inflammation.


Asunto(s)
Antiinflamatorios/farmacología , Colitis Ulcerosa/prevención & control , Colon/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Linfocitos Intraepiteliales/efectos de los fármacos , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Animales , Apoptosis/efectos de los fármacos , Células Cultivadas , Colitis Ulcerosa/enzimología , Colitis Ulcerosa/inmunología , Colitis Ulcerosa/patología , Colon/enzimología , Colon/inmunología , Colon/patología , Ciclosporina/farmacología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/enzimología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Linfocitos Intraepiteliales/enzimología , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/patología , Ratones Noqueados , Terapia Molecular Dirigida , Fosforilación , Proteínas Tirosina Quinasas/genética , Proteínas Tirosina Quinasas/metabolismo , Transducción de Señal
10.
Front Immunol ; 12: 635076, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34025642

RESUMEN

Hyperforin is a major active constituent of Hypericum perforatum L. extract, which is widely used for the treatment of depressive disorders. Recent studies have reported that hyperforin reduced inflammation in stroke and suppressed proliferation and differentiation in keratinocytes. Psoriasis is a chronic immune-mediated inflammatory skin disease in which the IL-23/IL-17 axis plays an important role. To investigate the underlying inflammatory mechanisms and response of hyperforin in psoriasis, we use imiquimod (IMQ)-induced mice model, in vitro cultured murine splenic γδ T cells, and HaCaT cells in this study. Data showed that hyperforin reduced epidermal thickness and decreased IMQ-induced pathological scores of cutaneous skin lesions in mice. Meanwhile we proved that hyperforin suppressed infiltration of CD3+ T cells and downregulated expression of Il1, Il6, Il23, Il17a, Il22, antimicrobial peptides (AMPs) in the skin lesion. Hyperforin significantly inhibited imiquimod-induced splenomegaly, reduced serum levels of TNF-α and IL-6, and IL-17A in splenocytes and draining lymph nodes. Our study also suggested that hyperforin lessened the infiltration of γδ T cell and CCR6+ γδ T cells in spleen and lymph nodes. Hyperforin also suppressed the typical psoriasis-like inflammatory responses and the infiltration of IL-17A+ cells in dermal γδ T cells of IMQ treated Tcrd-/- mice transferred with γδ T cells. In vitro studies, hyperforin reduced the expression and secretion of IL-17A in γδ T cells, and suppressed the activation of MAPK/STAT3 pathways in human keratinocyte HaCaT cells and γδ T cells. In conclusion, hyperforin alleviates IMQ-induced inflammation in psoriasis through suppressing the immune responses exerted by IL-17 A-producing γδ T cells and related cytokines by modulating MAPK/STAT3 pathways. Our study provided a novel therapeutic tragedy for psoriasis by which hyperforin attenuates psoriasis-related inflammatory responses.


Asunto(s)
Antiinflamatorios/farmacología , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/efectos de los fármacos , Floroglucinol/análogos & derivados , Psoriasis/prevención & control , Piel/efectos de los fármacos , Terpenos/farmacología , Traslado Adoptivo , Animales , Modelos Animales de Enfermedad , Genes Codificadores de los Receptores de Linfocitos T , Células HaCaT , Humanos , Imiquimod , Interleucina-17/genética , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Linfocitos Intraepiteliales/trasplante , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Floroglucinol/farmacología , Fosforilación , Psoriasis/inducido químicamente , Psoriasis/inmunología , Psoriasis/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Piel/inmunología , Piel/metabolismo
11.
J Immunother Cancer ; 9(5)2021 05.
Artículo en Inglés | MEDLINE | ID: mdl-34011536

RESUMEN

BACKGROUND: Gamma-delta (γδ) T lymphocytes are primed to potently respond to pathogens and transformed cells by recognizing a broad range of antigens. However, adoptive immunotherapy with γδT cells has exhibited mixed treatment responses. Better understanding of γδT cell biology and stratifying healthy donors for allogeneic adoptive therapy is clinically needed to fully realize the therapeutic potential of γδT cells. METHODS: We examine 98 blood samples from healthy donors and measure their expansion capacity after zoledronate stimulation, and test the migration and cytotoxic effector function of expanded γδT cells in 2D culture, 3D tumor spheroid and patient-derived melanoma organoid assays. RESULTS: We find that γδT cell expansion capacity is independent of expansion methods, gender, age and HLA type. Basal γδT cell levels in Peripheral blood mononuclear cell (PBMC) correlate well with their expansion, migration and cytotoxic effector capacity in vitro. Circulating γδT cells with lower expression of PD-1, CTLA-4, Eomes, T-bet and CD69, or higher IFN-γ production expand better. γδT cells with central memory and effector memory phenotypes are significantly more abundant in good expanders. A cut-off level of 0.82% γδT cells in PBMC stratifies good versus poor γδT cell expansion with a sensitivity of 97.78%, specificity of 90.48% and area under the curve of 0.968 in a healthy individual. Donors with higher Vδ2 Index Score in PBMC have greater anti-tumor functions including migratory function and cytotoxicity. CONCLUSIONS: Our results demonstrate that the interindividual γδT cell functions correlate with their circulating levels in healthy donors. Examination of circulating γδT cell level may be used to select healthy donors to participate in γδT-based immunotherapies.


Asunto(s)
Proliferación Celular , Linfocitos Intraepiteliales/inmunología , Activación de Linfocitos , Adulto , Biomarcadores/metabolismo , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular/efectos de los fármacos , Técnicas de Cocultivo , Citotoxicidad Inmunológica , Femenino , Voluntarios Sanos , Humanos , Memoria Inmunológica , Inmunofenotipificación , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/metabolismo , Activación de Linfocitos/efectos de los fármacos , Recuento de Linfocitos , Masculino , Melanoma/inmunología , Melanoma/metabolismo , Melanoma/patología , Persona de Mediana Edad , Fenotipo , Neoplasias Cutáneas/inmunología , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Adulto Joven , Ácido Zoledrónico/farmacología
12.
J Immunother Cancer ; 9(4)2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33849925

RESUMEN

BACKGROUND: Anti-programmed death-ligand 1 (αPD-L1) immunotherapy is approved to treat bladder cancer (BC) but is effective in <30% of patients. Interleukin (IL)-2/αIL-2 complexes (IL-2c) that preferentially target IL-2 receptor ß (CD122) augment CD8+ antitumor T cells known to improve αPD-L1 efficacy. We hypothesized that the tumor microenvironment, including local immune cells in primary versus metastatic BC, differentially affects immunotherapy responses and that IL-2c effects could differ from, and thus complement αPD-L1. METHODS: We studied mechanisms of IL-2c and αPD-L1 efficacy using PD-L1+ mouse BC cell lines MB49 and MBT-2 in orthotopic (bladder) and metastatic (lung) sites. RESULTS: IL-2c reduced orthotopic tumor burden and extended survival in MB49 and MBT-2 BC models, similar to αPD-L1. Using antibody-mediated cell depletions and genetically T cell-deficient mice, we unexpectedly found that CD8+ T cells were not necessary for IL-2c efficacy against tumors in bladder, whereas γδ T cells, not reported to contribute to αPD-L1 efficacy, were indispensable for IL-2c efficacy there. αPD-L1 responsiveness in bladder required conventional T cells as expected, but not γδ T cells, altogether defining distinct mechanisms for IL-2c and αPD-L1 efficacy. γδ T cells did not improve IL-2c treatment of subcutaneously challenged BC or orthotopic (peritoneal) ovarian cancer, consistent with tissue-specific and/or tumor-specific γδ T cell contributions to IL-2c efficacy. IL-2c significantly altered bladder intratumoral γδ T cell content, activation status, and specific γδ T cell subsets with antitumor or protumor effector functions. Neither IL-2c nor αPD-L1 alone treated lung metastatic MB49 or MBT-2 BC, but their combination improved survival in both models. Combination treatment efficacy in lungs required CD8+ T cells but not γδ T cells. CONCLUSIONS: Mechanistic insights into differential IL-2c and αPD-L1 treatment and tissue-dependent effects could help develop rational combination treatment strategies to improve treatment efficacy in distinct cancers. These studies also provide insights into γδ T cell contributions to immunotherapy in bladder and engagement of adaptive immunity by IL-2c plus αPD-L1 to treat refractory lung metastases.


Asunto(s)
Antígeno B7-H1/antagonistas & inhibidores , Inhibidores de Puntos de Control Inmunológico/farmacología , Subunidad beta del Receptor de Interleucina-2/agonistas , Interleucina-2/farmacología , Linfocitos Intraepiteliales/efectos de los fármacos , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Antígeno B7-H1/inmunología , Antígeno B7-H1/metabolismo , Línea Celular Tumoral , Subunidad beta del Receptor de Interleucina-2/inmunología , Subunidad beta del Receptor de Interleucina-2/metabolismo , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundario , Masculino , Ratones , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/metabolismo , Neoplasias de la Vejiga Urinaria/patología
13.
J Dermatol Sci ; 102(2): 116-125, 2021 May.
Artículo en Inglés | MEDLINE | ID: mdl-33888401

RESUMEN

BACKGROUND: Psoriasis is a chronic inflammatory skin disease. Interleukin (IL)-17A plays a key role in the pathogenesis of psoriasis. Fingolimod, which is available for the treatment of multiple sclerosis, exerts anti-inflammatory effects by sequestrating inflammatory lymphocytes in secondary lymphoid tissues and the thymus. The effect of fingolimod on psoriasis has not been reported yet. OBJECTIVE: Our objectives were to investigate the effect of fingolimod on psoriasis utilizing mice with imiquimod (IMQ)-induced psoriasiform dermatitis, and explore the possibility of fingolimod as a therapeutic agent for psoriasis. METHODS: Psoriasiform dermatitis was induced by imiquimod application on murine shaved back skin for six days. Fingolimod prepared in phosphate-buffered saline (PBS), or PBS alone as a control, was administered intraperitoneally daily from days 0 to 5. RESULTS: Fingolimod ameliorated IMQ-induced psoriasis dermatitis clinically and histologically. On day 6, the mRNA expression level of IL-17A was lower in the skin of fingolimod-treated mice than in that of PBS-treated mice, whereas it was higher in the inguinal lymph nodes of fingolimod-treated mice than in those of PBS-treated mice. Flow cytometric analyses revealed that fingolimod reduced IL-17A-producing ?d T cells infiltrating into the skin, whereas it increased these cells in the inguinal lymph nodes. Fingolimod inhibited egress of Langerhans cells from the skin to lymph nodes. CONCLUSION: Our results demonstrated that fingolimod showed effectiveness for IMQ-induced psoriasiform dermatitis by hindering the emigration of IL-17A-producing ?d T cells from the lymph nodes to the skin, and suggest that fingolimod is a promising candidate for the treatment of psoriasis.


Asunto(s)
Clorhidrato de Fingolimod/farmacología , Linfocitos Intraepiteliales/efectos de los fármacos , Ganglios Linfáticos/efectos de los fármacos , Psoriasis/tratamiento farmacológico , Piel/efectos de los fármacos , Animales , Modelos Animales de Enfermedad , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/inmunología , Evaluación Preclínica de Medicamentos , Femenino , Clorhidrato de Fingolimod/uso terapéutico , Humanos , Imiquimod/administración & dosificación , Imiquimod/inmunología , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Células de Langerhans/inmunología , Células de Langerhans/metabolismo , Ganglios Linfáticos/citología , Ganglios Linfáticos/inmunología , Ganglios Linfáticos/metabolismo , Ratones , Psoriasis/inmunología , Psoriasis/patología , Piel/citología , Piel/inmunología , Piel/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/inmunología
14.
J Immunother Cancer ; 9(3)2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33653802

RESUMEN

BACKGROUND: Although intravesical BCG is the standard treatment of high-grade non-muscle invasive bladder cancer (NMIBC), response rates remain unsatisfactory. In preclinical models, rapamycin enhances BCG vaccine efficacy against tuberculosis and the killing capacity of γδ T cells, which are critical for BCG's antitumor effects. Here, we monitored immunity, safety, and tolerability of rapamycin combined with BCG in patients with NMIBC. METHODS: A randomized double-blind trial of oral rapamycin (0.5 or 2.0 mg daily) versus placebo for 1 month was conducted in patients with NMIBC concurrently receiving 3 weekly BCG instillations (NCT02753309). The primary outcome was induction of BCG-specific γδ T cells, measured as a percentage change from baseline. Post-BCG urinary cytokines and immune cells were examined as surrogates for local immune response in the bladder. Secondary outcomes measured were adverse events (AEs) and tolerability using validated patient-reported questionnaires. RESULTS: Thirty-one patients were randomized (11 placebo, 8 rapamycin 2.0 mg, and 12 rapamycin 0.5 mg). AEs were similar across groups and most were grade 1-2. One (12.5%) patient randomized to 2.0 mg rapamycin was taken off treatment due to stomatitis. No significant differences in urinary symptoms, bowel function, or bother were observed between groups. The median (IQR) percentage change in BCG-specific γδ T cells from baseline per group was as follows: -26% (-51% to 24%) for placebo, 9.6% (-59% to 117%) for rapamycin 0.5 mg (versus placebo, p=0.18), and 78.8% (-31% to 115%) for rapamycin 2.0 mg (versus placebo, p=0.03). BCG-induced cytokines showed a progressive increase in IL-8 (p=0.02) and TNF-α (p=0.04) over time for patients on rapamycin 2.0 mg, whereas patients receiving placebo had no significant change in urinary cytokines. Compared with placebo, patients receiving 2.0 mg rapamycin had increased urinary γδ T cells at the first week of BCG (p=0.02). CONCLUSIONS: Four weeks of 0.5 and 2.0 mg oral rapamycin daily is safe and tolerable in combination with BCG for patients with NMIBC. Rapamycin enhances BCG-specific γδ T cell immunity and boosts urinary cytokines during BCG treatment. Further study is needed to determine long-term rapamycin safety, tolerability and effects on BCG efficacy.


Asunto(s)
Adyuvantes Inmunológicos/administración & dosificación , Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Vacuna BCG/administración & dosificación , Linfocitos Intraepiteliales/efectos de los fármacos , Sirolimus/administración & dosificación , Neoplasias de la Vejiga Urinaria/tratamiento farmacológico , Adyuvantes Inmunológicos/efectos adversos , Administración Intravesical , Adulto , Anciano , Anciano de 80 o más Años , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos , Vacuna BCG/efectos adversos , Citocinas/orina , Método Doble Ciego , Femenino , Humanos , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias , Fenotipo , Sirolimus/efectos adversos , Factores de Tiempo , Resultado del Tratamiento , Microambiente Tumoral , Neoplasias de la Vejiga Urinaria/inmunología , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Vejiga Urinaria/orina , Orina/química , Orina/citología
15.
J Immunother Cancer ; 9(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33436485

RESUMEN

BACKGROUND: Endogenous retroviruses (ERVs) play a role in a variety of biological processes, including embryogenesis and cancer. DNA methyltransferase inhibitors (DNMTi)-induced ERV expression triggers interferon responses in ovarian cancer cells via the viral sensing machinery. Baseline expression of ERVs also occurs in cancer cells, though this process is poorly understood and previously unexplored in epithelial ovarian cancer (EOC). Here, the prognostic and immunomodulatory consequences of baseline ERV expression was assessed in EOC. METHODS: ERV expression was assessed using EOC transcriptional data from The Cancer Genome Atlas (TCGA) and from an independent cohort (Hammersmith Hospital, HH), as well as from untreated or DNMTi-treated EOC cell lines. Least absolute shrinkage and selection operator (LASSO) logistic regression defined an ERV expression score to predict patient prognosis. Immunohistochemistry (IHC) was conducted on the HH cohort. Combination of DNMTi treatment with γδ T cells was tested in vitro, using EOC cell lines and patient-derived tumor cells. RESULTS: ERV expression was found to define clinically relevant subsets of EOC patients. An ERV prognostic score was successfully generated in TCGA and validated in the independent cohort. In EOC patients from this cohort, a high ERV score was associated with better survival (log-rank p=0.0009) and correlated with infiltration of CD8+PD1+T cells (r=0.46, p=0.0001). In the TCGA dataset, a higher ERV score was found in BRCA1/2 mutant tumors, compared to wild type (p=0.015), while a lower ERV score was found in CCNE1 amplified tumors, compared to wild type (p=0.019). In vitro, baseline ERV expression dictates the level of ERV induction in response to DNMTi. Manipulation of an ERV expression threshold by DNMTi resulted in improved EOC cell killing by cytotoxic immune cells. CONCLUSIONS: These findings uncover the potential for baseline ERV expression to robustly inform EOC patient prognosis, influence tumor immune infiltration and affect antitumor immunity.


Asunto(s)
Proteína BRCA1/genética , Proteína BRCA2/genética , Carcinoma Epitelial de Ovario/genética , Ciclina E/genética , Retrovirus Endógenos , Perfilación de la Expresión Génica/métodos , Proteínas Oncogénicas/genética , Neoplasias Ováricas/genética , Carcinoma Epitelial de Ovario/tratamiento farmacológico , Carcinoma Epitelial de Ovario/inmunología , Línea Celular Tumoral , Decitabina/farmacología , Decitabina/uso terapéutico , Retrovirus Endógenos/efectos de los fármacos , Femenino , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/inmunología , Mutación , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/inmunología , Pronóstico , Análisis de Secuencia de ARN , Análisis de Supervivencia
16.
Cancer Lett ; 496: 156-168, 2021 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-33045304

RESUMEN

Gamma delta T cells (γδTc) have tremendous anti-tumoral activity, thus γδTc immunotherapy is currently under development for various malignancies. We targeted breast cancer stem-like cells (BCSC), a rare cell population responsible for patient mortality. BCSC were mostly susceptible to γδTc immunotherapy, yet some escaped. The BCSC secretome rendered γδTc hypo-responsive, and resistant BCSC expressed more PD-L1 and anti-apoptotic protein MCL-1 than non-stem-like cells (NSC). BCSC resistance was partially overcome by dMCL1-2, an MCL-1 degrader, or more fully by blocking PD-1 on γδTc. Increased MICA shedding was prevented by the ADAM inhibitor GW280264X, rendering BCSC as sensitive to γδTc cytotoxicity as NSC. Our data show promising potential for γδTc immunotherapy against BCSC while unraveling immune evasion mechanisms exploited by BCSC, which likely also enable their resistance to cytotoxic T and NK cells. Overcoming this resistance, as we have done here, will improve cancer immunotherapy, leading to better cancer patient outcomes.


Asunto(s)
Proteínas ADAM/antagonistas & inhibidores , Neoplasias de la Mama/tratamiento farmacológico , Resistencia a Antineoplásicos/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Inmunoterapia , Linfocitos Intraepiteliales/inmunología , Células Madre Neoplásicas/efectos de los fármacos , Inhibidores de Proteasas/farmacología , Animales , Apoptosis , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular , Femenino , Humanos , Linfocitos Intraepiteliales/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Células Asesinas Naturales/inmunología , Ratones , Ratones Endogámicos NOD , Ratones SCID , Terapia Molecular Dirigida , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
17.
Immunology ; 162(3): 306-313, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33131052

RESUMEN

Bisphosphonates distributed to bone exert toxic effects specifically towards osteoclasts. On the other hand, intravenous administration of a nitrogen-containing bisphosphonate (N-BP) such as zoledronate induces acute-phase reactions (APRs), including influenza-like fever 1 day later, indicating an interaction with immunocompetent cells circulating blood. Although it has been reported that activation of γδ T cells is pivotal to induce an APR following treatment with zoledronate, downstream events, including the production of inflammatory cytokines after activation of γδ T cells, remain obscure. We investigated the effects of zoledronate on inflammatory cytokine expression in human peripheral blood mononuclear cells (PBMCs) in vitro. While zoledronate induced mRNA expressions of tumour necrosis factor-α (TNF-α), interleukin (IL)-1ß, IL-6 and interferon-γ (IFN-γ) in PBMC, depletion of γδ T cells abolished that zoledronate-induced expression of those cytokines, indicating the necessity of γδ T cells for expression induction by zoledronate. However, which types of cells were responsible for the production of those cytokines in blood remained unclear. As it is generally accepted that monocytes and macrophages are primary sources of inflammatory cytokines, CD14+ cells from PBMC were exposed to zoledronate in the presence of PBMC, which resulted in induced expression of mRNAs for IL-1ß, IL-6 and IFN-γ, but not for TNF-α. These results indicate that CD14+ cells are responsible, at least in part, for the production of IL-1ß, IL-6 and IFN-γ in blood exposed to zoledronate. This suggests that CD14+ cells play an essential role in the occurrence of APRs following N-BP administration.


Asunto(s)
Reacción de Fase Aguda/inducido químicamente , Conservadores de la Densidad Ósea/toxicidad , Citocinas/metabolismo , Mediadores de Inflamación/metabolismo , Linfocitos Intraepiteliales/efectos de los fármacos , Receptores de Lipopolisacáridos/metabolismo , Activación de Linfocitos/efectos de los fármacos , Monocitos/efectos de los fármacos , Ácido Zoledrónico/toxicidad , Reacción de Fase Aguda/inmunología , Reacción de Fase Aguda/metabolismo , Células Cultivadas , Técnicas de Cocultivo , Citocinas/genética , Humanos , Linfocitos Intraepiteliales/inmunología , Linfocitos Intraepiteliales/metabolismo , Monocitos/inmunología , Monocitos/metabolismo
18.
Biochimie ; 179: 146-156, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-33007409

RESUMEN

Psoriasis is a debilitating chronic skin disease with a worldwide prevalence. Its main features include well-marked silvery scales on the skin of hands and feet and back which arise due to hyperproliferation of keratinocytes and infiltration of immune cells in the skin. Multiple interactions exist between adaptive immune cells such as T cells and innate immune cells such as neutrophils and macrophages which are key players in the pathogenesis of psoriasis. Interleukin-2-inducible T-cell kinase (ITK) plays a key role in Th17 cell development through control of several transcription factors. ITK has been shown to control NFATc1, NFkB and STAT3 in CD4+ T cells. Effect of ITK inhibitor in imiquimod (IMQ)-induced psoriasiform inflammation remains to be explored. In the current examination, role of ITK signaling and its inhibition blockade were evaluated on NFATc1, NFkB and STAT3, IL-17A, TNF-α, IFN-γ, Foxp3, IL-10 in CD4+ T cells in IMQ model. Our data display that ITK signaling is involved in IMQ-induced psoriatic inflammation as paralleled by enhancement of p-ITK, NFATc1, p-NFkB and p-STAT3 in CD4+ T cells. It was associated with enhancement of Th17/Th1 cells and neutrophilic inflammation in the skin. Preventive treatment with ITK inhibitor led to a reduction in Th17/Th1 cells and enhancement of Treg cells. Overall, this study suggests that ITK signaling is an important modulator of transcription factor signaling in CD4+ T cells which is associated with Th17/Th1 cells and psoriasiform inflammation in mice. ITK signaling blockade could be a therapeutic target for the treatment of psoriatic inflammation.


Asunto(s)
Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Tirosina Quinasas/fisiología , Psoriasis/tratamiento farmacológico , Psoriasis/metabolismo , Animales , Modelos Animales de Enfermedad , Imiquimod/toxicidad , Inflamación/inmunología , Inflamación/patología , Interleucina-17/metabolismo , Linfocitos Intraepiteliales/efectos de los fármacos , Masculino , Ratones Endogámicos C57BL , Psoriasis/inmunología , Psoriasis/patología , Transducción de Señal/efectos de los fármacos , Piel/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Células TH1/efectos de los fármacos , Células Th17/efectos de los fármacos , Factor de Necrosis Tumoral alfa/metabolismo
19.
Cell Rep Med ; 1(6): 100095, 2020 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-32984856

RESUMEN

Induction of protective mucosal T cell memory remains a formidable challenge to vaccinologists. Using a combination adjuvant strategy that elicits potent CD8 and CD4 T cell responses, we define the tenets of vaccine-induced pulmonary T cell immunity. An acrylic-acid-based adjuvant (ADJ), in combination with Toll-like receptor (TLR) agonists glucopyranosyl lipid adjuvant (GLA) or CpG, promotes mucosal imprinting but engages distinct transcription programs to drive different degrees of terminal differentiation and disparate polarization of TH1/TC1/TH17/TC17 effector/memory T cells. Combination of ADJ with GLA, but not CpG, dampens T cell receptor (TCR) signaling, mitigates terminal differentiation of effectors, and enhances the development of CD4 and CD8 TRM cells that protect against H1N1 and H5N1 influenza viruses. Mechanistically, vaccine-elicited CD4 T cells play a vital role in optimal programming of CD8 TRM and viral control. Taken together, these findings provide further insights into vaccine-induced multifaceted mucosal T cell immunity with implications in the development of vaccines against respiratorypathogens, including influenza virus and SARS-CoV-2.


Asunto(s)
Adyuvantes de Vacunas/farmacología , Pulmón/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Resinas Acrílicas/administración & dosificación , Resinas Acrílicas/farmacología , Adyuvantes de Vacunas/administración & dosificación , Animales , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/inmunología , Inflamación , Virus de la Influenza A/inmunología , Vacunas contra la Influenza/administración & dosificación , Vacunas contra la Influenza/farmacología , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/inmunología , Pulmón/inmunología , Células T de Memoria/efectos de los fármacos , Células T de Memoria/inmunología , Ratones , Infecciones por Orthomyxoviridae/inmunología , Infecciones por Orthomyxoviridae/prevención & control , Infecciones por Orthomyxoviridae/virología , Receptores de Antígenos de Linfocitos T/metabolismo , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología , Receptores Toll-Like/agonistas
20.
Front Immunol ; 11: 1702, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32849598

RESUMEN

TCRαß+CD8αα+ intraepithelial lymphocytes (IELs) are abundant in gastrointestinal (GI) tract and play an important role in regulation of mucosal immunity and tolerance in the gut. However, it is unknown whether TCRαß+CD8αα+ IELs exist in the oral mucosa and if yes, what controls their development. We here identified and characterized TCRαß+CD8αα+ IELs from the murine oral mucosa. We showed that the number and function of TCRαß+CD8αα+ IELs were regulated by TGF-ß. We further revealed that oral TCRαß+CD8αα+ IELs could be altered under systemic inflammatory conditions and by antibiotic treatment at the neonatal age of the mice. Our findings have revealed a previously unrecognized population of oral IELs that may regulate oral mucosal immune responses.


Asunto(s)
Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa , Linfocitos Intraepiteliales/inmunología , Mucosa Bucal/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Animales , Animales Recién Nacidos , Antibacterianos/farmacología , Antiinflamatorios/farmacología , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Microbioma Gastrointestinal , Inmunidad Mucosa/efectos de los fármacos , Interferón gamma/metabolismo , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Linfocitos Intraepiteliales/efectos de los fármacos , Linfocitos Intraepiteliales/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/metabolismo , Fenotipo , Receptor Tipo I de Factor de Crecimiento Transformador beta/genética , Receptor Tipo I de Factor de Crecimiento Transformador beta/metabolismo , Transducción de Señal , Factor de Crecimiento Transformador beta1/genética , Factor de Crecimiento Transformador beta1/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA