Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 7 de 7
Filtrar
1.
Sci Rep ; 10(1): 10798, 2020 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-32612145

RESUMEN

Celiac disease (CD) is an enteropathy triggered by the ingestion of gluten proteins in genetically predisposed individuals and characterized by excessive activation of effector immune cells and enhanced production of inflammatory cytokines. However, factors/mechanisms that amplify the ongoing mucosal inflammation in CD are not fully understood. In this study, we assessed whether mammalian target of Rapamycin (mTOR), a pathway that combines intra- and extra-cellular signals and acts as a central regulator for the metabolism, growth, and function of immune and non-immune cells, sustains CD-associated immune response. Our findings indicate that expression of phosphorylated (p)/active form of mTOR is increased in protein lysates of duodenal biopsy samples taken from patients with active CD (ACD) as compared to normal controls. In ACD, activation of mTOR occurs mainly in the epithelial compartment and associates with enhanced expression of p-4EBP, a downstream target of mTOR complex (mTORC)1, while expression of p-Rictor, a component of mTORC2, is not increased. Stimulation of mucosal explants of inactive CD patients with pepsin-trypsin-digested (PT)-gliadin or IFN-γ/IL-21, two cytokines produced in CD by gluten-specific T cells, increases p-4EBP expression. Consistently, blockade of such cytokines in cultures of ACD mucosal explants reduces p-4EBP. Finally, we show that inhibition of mTORC1 with rapamycin in ACD mucosal explants reduces p-4EBP and production of IL-15, a master cytokine produced by epithelial cells in this disorder. Our data suggest that ACD inflammation is marked by activation of mTORC1 in the epithelial compartment.


Asunto(s)
Enfermedad Celíaca/inmunología , Duodeno/inmunología , Mucosa Intestinal/inmunología , Serina-Treonina Quinasas TOR/inmunología , Biopsia , Enfermedad Celíaca/patología , Duodeno/patología , Femenino , Gliadina/inmunología , Humanos , Inflamación/inmunología , Inflamación/patología , Interferón gamma/inmunología , Interleucinas/inmunología , Mucosa Intestinal/patología , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina/inmunología , Diana Mecanicista del Complejo 2 de la Rapamicina/inmunología , Fosforilación/inmunología , Linfocitos T/inmunología
2.
Oncogene ; 34(27): 3493-503, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25174402

RESUMEN

Colorectal cancers (CRCs) often show a dense infiltrate of cytokine-producing immune/inflammatory cells. The exact contribution of each immune cell subset and cytokine in the activation of the intracellular pathways sustaining CRC cell growth is not understood. Herein, we isolate tumor-infiltrating leukocytes (TILs) and lamina propria mononuclear cells (LPMCs) from the tumor area and the macroscopically unaffected, adjacent, colonic mucosa of patients who underwent resection for sporadic CRC and show that the culture supernatants of TILs, but not of LPMCs, potently enhance the growth of human CRC cell lines through the activation of the oncogenic transcription factors signal transducer and activator of transcription 3 (STAT3) and nuclear factor-kappa B (NF-kB). Characterization of immune cell complexity of TILs and LPMCs reveals no differences in the percentages of T cells, natural killer T cells, natural killer (NK) cells, macrophages and B cells. However, T cells from TILs show a functional switch compared with those from LPMCs to produce large amounts of T helper type 17 (Th17)-related cytokines (that is, interleukin-17A (IL-17A), IL-17F, IL-21 and IL-22), tumor necrosis factor-α (TNF-α) and IL-6. Individual neutralization of IL-17A, IL-17F, IL-21, IL-22, TNF-α or IL-6 does not change TIL-derived supernatant-driven STAT3 and NF-kB activation, as well as their proproliferative effect in CRC cells. In contrast, simultaneous neutralization of both IL-17A and TNF-α, which abrogates NF-kB signaling, and IL-22 and IL-6, which abrogates STAT3 signaling, reduces the mitogenic effect of supernatants in CRC cells. IL-17A, IL-21, IL-22, TNF-α and IL-6 are also produced in excess in the early colonic lesions in a mouse model of sporadic CRC, associated with enhanced STAT3/NF-kB activation. Mice therapeutically given BP-1-102, an orally bioavailable compound targeting STAT3/NF-kB activation and cross-talk, exhibit reduced colon tumorigenesis and diminished expression of STAT3/NF-kB-activating cytokines in the neoplastic areas. These data suggest that strategies aimed at the cotargeting of STAT3/NF-kB activation and interaction between them might represent an attractive and novel approach to combat CRC.


Asunto(s)
Neoplasias Colorrectales/patología , Interleucina-17/farmacología , Interleucina-6/farmacología , Interleucinas/farmacología , FN-kappa B/genética , Factor de Transcripción STAT3/genética , Factor de Necrosis Tumoral alfa/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Células Cultivadas , Neoplasias Colorrectales/genética , Citocinas/metabolismo , Citocinas/farmacología , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HT29 , Humanos , Interleucina-17/metabolismo , Interleucina-6/metabolismo , Interleucinas/metabolismo , Ratones , Ratones Transgénicos , FN-kappa B/metabolismo , Factor de Transcripción STAT3/metabolismo , Células Th17/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Interleucina-22
3.
Cell Death Dis ; 5: e1073, 2014 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-24556688

RESUMEN

Initially identified as an inhibitor of transforming growth factor (TGF)-ß mainly owing to its ability to bind TGF-ß receptor type I and abrogate TGF-ß-driven signaling, Smad7 can interact with additional intracellular proteins and regulate TGF-ß-independent pathways, thus having a key role in the control of neoplastic processes in various organs. Genome-wide association studies have shown that common alleles of Smad7 influence the risk of colorectal cancer (CRC), even though the contribution of Smad7 in colon carcinogenesis is not fully understood. In this study, we assessed the expression and role of Smad7 in human and mouse models of sporadic CRC. We document a significant increase of Smad7 in human CRC relative to the surrounding nontumor tissues and show that silencing of Smad7 inhibits the growth of CRC cell lines both in vitro and in vivo after transplantation into immunodeficient mice. Knockdown of Smad7 results in enhanced phosphorylation of the cyclin-dependent kinase (CDK)2, accumulation of CRC cells in S phase and enhanced cell death. Smad7-deficient CRC cells have lower levels of CDC25A, a phosphatase that dephosphorylates CDK2, and hyperphosphorylated eukaryotic initiation factor 2 (eIF2)α, a negative regulator of CDC25 protein translation. Consistently, knockdown of Smad7 associates with inactivation of eIF2α, lower CDC25A expression and diminished fraction of proliferating cells in human CRC explants, and reduces the number of intestinal tumors in Apc(min/+) mice. Altogether, these data support a role for Smad7 in sustaining colon tumorigenesis.


Asunto(s)
Proliferación Celular , Neoplasias del Colon/metabolismo , Proteína smad7/metabolismo , Animales , Supervivencia Celular , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias del Colon/prevención & control , Quinasa 2 Dependiente de la Ciclina/metabolismo , Modelos Animales de Enfermedad , Factor 2 Eucariótico de Iniciación/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Genes APC , Genes RAG-1 , Terapia Genética , Células HCT116 , Células HT29 , Células Hep G2 , Humanos , Ratones , Ratones Transgénicos , Oligonucleótidos Antisentido/metabolismo , Fosforilación , Transducción de Señal , Factores de Tiempo , Transfección , Fosfatasas cdc25/metabolismo
4.
Mucosal Immunol ; 7(6): 1467-79, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24850427

RESUMEN

In inflammatory bowel disease (IBD), tissue damage is driven by an excessive immune response, poorly controlled by counter-regulatory mechanisms. SIRT1, a class III NAD+-dependent deacetylase, regulates negatively the expression of various proteins involved in the control of immune-inflammatory pathways, such as Stat3, Smad7, and NF-κB. Here we examined the expression, regulation, and function of SIRT1 in IBD. SIRT1 RNA and protein expression was less pronounced in whole biopsies and lamina propria mononuclear cells (LPMCs) of IBD patients in comparison with normal controls. SIRT1 expression was downregulated in control LPMC by tumor necrosis factor (TNF)-α and interleukin (IL)-21, and upregulated in IBD LPMC by neutralizing TNF-α and IL-21antibodies. Consistently, SIRT1 expression was increased in mucosal samples taken from IBD patients successfully treated with Infliximab. Treatment of IBD LPMC with Cay10591, a specific SIRT1 activator, reduced NF-κB activation and inhibited inflammatory cytokine synthesis, whereas Ex527, an inhibitor of SIRT1, increased interferon (IFN)-γ in control LPMC. SIRT1 was also reduced in mice with colitis induced by 2,4,6-trinitrobenzenesulphonic acid or oxazolone. Cay10591 prevented and cured experimental colitis whereas Ex527 exacerbated disease by modulating T cell-derived cytokine response. Data indicate that SIRT1 is downregulated in IBD patients and colitic mice and suggest that SIRT1 activation can help attenuate inflammatory signals in the gut.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/inmunología , Enfermedades Inflamatorias del Intestino/inmunología , Intestinos/inmunología , Sirtuina 1/inmunología , Adyuvantes Inmunológicos/efectos adversos , Adyuvantes Inmunológicos/farmacología , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Femenino , Regulación Enzimológica de la Expresión Génica/genética , Humanos , Enfermedades Inflamatorias del Intestino/inducido químicamente , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/genética , Enfermedades Inflamatorias del Intestino/patología , Infliximab , Interferón gamma/genética , Interferón gamma/inmunología , Interleucinas/genética , Interleucinas/inmunología , Intestinos/patología , Masculino , Ratones , Persona de Mediana Edad , Oxazolona/efectos adversos , Oxazolona/farmacología , Sirtuina 1/genética , Ácido Trinitrobencenosulfónico/toxicidad , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
5.
Mucosal Immunol ; 6(2): 244-55, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22785229

RESUMEN

Celiac disease (CD)-associated inflammation is characterized by high interleukin- 21 (IL-21), but the mechanisms that control IL-21 production are not fully understood. Here we analyzed IL-21 cell sources and examined how IL-21 production is regulated in CD. Intraepithelial lymphocytes (IELs) and lamina propria lymphocytes (LPLs), isolated from CD patients and non-CD controls, were analyzed for cell markers, cytokines, and transcription factors by flow cytometry. IL-21 was highly produced by CD4+ and CD4+/CD8+ IELs and LPLs in active CD. IL-21-producing cells coexpressed interferon-γ (IFN-γ) and to a lesser extent T helper type 17 (Th17) cytokines. Treatment of control LPLs with IL-15, a cytokine overproduced in CD, activated Akt and STAT3 (signal transducer and activator of transcription 3), thus enhancing IL-21 synthesis. Active CD biopsies contained elevated levels of Akt, and blockade of IL-15 in those samples reduced IL-21. Similarly, neutralization of IL-15 in biopsies of inactive CD patients inhibited peptic-tryptic digest of gliadin-induced IL-21 expression. These findings indicate that in CD, IL-15 positively regulates IL-21 production.


Asunto(s)
Enfermedad Celíaca/metabolismo , Interleucina-15/metabolismo , Interleucinas/biosíntesis , Mucosa Intestinal/metabolismo , Enfermedad Celíaca/genética , Enfermedad Celíaca/patología , Células Cultivadas , Expresión Génica , Humanos , Interferón gamma/metabolismo , Interleucinas/genética , Mucosa Intestinal/patología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Receptores CXCR5/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
6.
Mucosal Immunol ; 4(2): 239-44, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20944558

RESUMEN

An altered balance between effector and regulatory factors is supposed to sustain the tissue-damaging immune response in inflammatory bowel disease (IBD). We have recently shown that in IBD, there is a defective synthesis of the counter-regulatory cytokine, interleukin (IL)-25. In this study we investigated factors that control IL-25 production in the gut. IBD patients produced less IL-25 when compared with normal controls. Stimulation of normal intestinal explants with tumor necrosis factor-α (TNF-α), but not interferon-γ (IFN-γ) or IL-21, reduced IL-25 synthesis. Consistently, IL-25 production was enhanced by anti-TNF-α both in vitro and in vivo. Upregulation of IL-25 was also seen in normal colonic explants stimulated with transforming growth factor-ß1 (TGF-ß1). As in IBD, TGF-ß1 activity is abrogated by Smad7, we next assessed whether inhibition of Smad7 with an antisense oligonucleotide enhanced IL-25 expression. Knockdown of Smad7 was accompanied by an increase in IL-25 production. Data show that IL-25 production is differently regulated by TNF-α and TGF-ß1 in the human gut.


Asunto(s)
Regulación de la Expresión Génica , Interleucina-17/metabolismo , Mucosa Intestinal/inmunología , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Enfermedad Celíaca/inmunología , Regulación de la Expresión Génica/inmunología , Técnicas de Silenciamiento del Gen , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Interleucina-17/genética , Proteína smad7/genética , Proteína smad7/metabolismo
7.
Artículo en Inglés | WPRIM | ID: wpr-29718

RESUMEN

Many oral and maxillofacial bone defects are not self-healing. Guided bone regeneration (GBR), which uses a barrier membrane to prevent the soft tissues from invading the defect to enable slower-growing bone cells to penetrate the area, was developed as a therapy in the 1980s. Although there has been some success with GBR in some clinical situations, better treatments are needed. This review discusses the concept of GBR focusing on bioactive membranes that incorporate osteoconductive materials, growth factors and cells for improved oral and maxillofacial bone regeneration.


Asunto(s)
Regeneración Ósea , Sustitutos de Huesos , Péptidos y Proteínas de Señalización Intercelular , Membranas , Células Madre
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA