Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Oncoimmunology ; 11(1): 2096535, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35832043

RESUMEN

The microbiome is now regarded as one of the hallmarks of cancer and several strategies to modify the gut microbiota to improve immune checkpoint inhibitor (ICI) activity are being evaluated in clinical trials. Preliminary data regarding the upper gastro-intestinal microbiota indicated that Helicobacter pylori seropositivity was associated with a negative prognosis in patients amenable to ICI. In 97 patients with advanced melanoma treated with ICI, we assessed the impact of H. pylori on outcomes and microbiome composition. We performed H. pylori serology and profiled the fecal microbiome with metagenomics sequencing. Among the 97 patients, 22% were H. pylori positive (Pos). H. pylori Pos patients had a significantly shorter overall survival (p = .02) compared to H. pylori negative (Neg) patients. In addition, objective response rate and progression-free survival were decreased in H. pylori Pos patients. Metagenomics sequencing did not reveal any difference in diversity indexes between the H. pylori groups. At the taxa level, Eubacterium ventriosum, Mediterraneibacter (Ruminococcus) torques, and Dorea formicigenerans were increased in the H. pylori Pos group, while Alistipes finegoldii, Hungatella hathewayi and Blautia producta were over-represented in the H. pylori Neg group. In a second independent cohort of patients with NSCLC, diversity indexes were similar in both groups and Bacteroides xylanisolvens was increased in H. pylori Neg patients. Our results demonstrated that the negative impact of H. pylori on outcomes seem to be independent from the fecal microbiome composition. These findings warrant further validation and development of therapeutic strategies to eradicate H. pylori in immuno-oncology arena.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Infecciones por Helicobacter , Helicobacter pylori , Neoplasias Pulmonares , Melanoma , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Infecciones por Helicobacter/tratamiento farmacológico , Infecciones por Helicobacter/microbiología , Helicobacter pylori/fisiología , Humanos , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Melanoma/tratamiento farmacológico , Síndrome
2.
Front Immunol ; 13: 899161, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35677057

RESUMEN

Helicobacter pylori infects the gastric mucosa of a large number of humans. Although asymptomatic in the vast majority of cases, H pylori infection can lead to the development of peptic ulcers gastric adenocarcinoma and mucosa-associated lymphoid tissue (MALT) lymphoma. Using a variety of mechanisms, H pylori locally suppresses the function of the host immune system to establish chronic infection. Systemic immunomodulation has been observed in both clinical and pre-clinical studies, which have demonstrated that H pylori infection is associated with reduced incidence of inflammatory diseases, such as asthma and Crohn's disease. The introduction of immunotherapies in the arsenal of anti-cancer drugs has revealed a new facet of H pylori-induced immune suppression. In this review, we will describe the intimate interactions between H pylori and its host, and formulate hypothtyeses describing the detrimental impact of H pylori infection on the efficacy of cancer immunotherapies.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Linfoma de Células B de la Zona Marginal , Mucosa Gástrica , Infecciones por Helicobacter/complicaciones , Humanos , Inmunoterapia/efectos adversos , Linfoma de Células B de la Zona Marginal/patología
3.
Helicobacter ; 27(2): e12875, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35092634

RESUMEN

BACKGROUND: Helicobacter pylori (H. pylori) colonizes the human gastric mucosa with a high worldwide prevalence. Currently, H. pylori is eradicated by the use of antibiotics. However, elevated antibiotic resistance suggests new therapeutic strategies need to be envisioned: one approach being prophylactic vaccination. Pre-clinical and clinical data show that a urease-based vaccine is efficient in decreasing H. pylori infection through the mobilization of T helper (Th) cells, especially Th17 cells. Th17 cells produce interleukins such as IL-22 and IL-17, among others, and are key players in vaccine efficacy. Recently, granulocyte-macrophage colony-stimulating factor (GM-CSF)-producing Th17 cells have been identified. AIM: This study explores the possibility that GM-CSF plays a role in the reduction of H. pylori infection following vaccination. RESULTS: We demonstrate that GM-CSF+ IL-17+ Th17 cells accumulate in the stomach mucosa of H. pylori infected mice during the vaccine-induced reduction of H. pylori infection. Secondly, we provide evidence that vaccinated GM-CSF deficient mice only modestly reduce H. pylori infection. Conversely, we observe that an increase in GM-CSF availability reduces H. pylori burden in chronically infected mice. Thirdly, we show that GM-CSF, by acting on gastric epithelial cells, promotes the production of ßdefensin3, which exhibits H. pylori bactericidal activities. CONCLUSION: Taken together, we demonstrate a key role of GM-CSF, most probably originating from Th17 cells, in the vaccine-induced reduction of H. pylori infection.


Asunto(s)
Vacunas Bacterianas , Factor Estimulante de Colonias de Granulocitos y Macrófagos , Infecciones por Helicobacter , Helicobacter pylori , Animales , Vacunas Bacterianas/inmunología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/inmunología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/prevención & control , Ratones , Células Th17 , Vacunación
4.
Cancer Discov ; 12(4): 1070-1087, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-35031549

RESUMEN

Several approaches to manipulate the gut microbiome for improving the activity of cancer immune-checkpoint inhibitors (ICI) are currently under evaluation. Here, we show that oral supplementation with the polyphenol-rich berry camu-camu (CC; Myrciaria dubia) in mice shifted gut microbial composition, which translated into antitumor activity and a stronger anti-PD-1 response. We identified castalagin, an ellagitannin, as the active compound in CC. Oral administration of castalagin enriched for bacteria associated with efficient immunotherapeutic responses (Ruminococcaceae and Alistipes) and improved the CD8+/FOXP3+CD4+ ratio within the tumor microenvironment. Moreover, castalagin induced metabolic changes, resulting in an increase in taurine-conjugated bile acids. Oral supplementation of castalagin following fecal microbiota transplantation from ICI-refractory patients into mice supported anti-PD-1 activity. Finally, we found that castalagin binds to Ruminococcus bromii and promoted an anticancer response. Altogether, our results identify castalagin as a polyphenol that acts as a prebiotic to circumvent anti-PD-1 resistance. SIGNIFICANCE: The polyphenol castalagin isolated from a berry has an antitumor effect through direct interactions with commensal bacteria, thus reprogramming the tumor microenvironment. In addition, in preclinical ICI-resistant models, castalagin reestablishes the efficacy of anti-PD-1. Together, these results provide a strong biological rationale to test castalagin as part of a clinical trial. This article is highlighted in the In This Issue feature, p. 873.


Asunto(s)
Microbioma Gastrointestinal , Animales , Bacterias , Trasplante de Microbiota Fecal , Humanos , Ratones , Polifenoles/farmacología , Polifenoles/uso terapéutico
5.
Gut ; 71(3): 457-466, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-34253574

RESUMEN

OBJECTIVE: In this study, we determined whether Helicobacter pylori (H. pylori) infection dampens the efficacy of cancer immunotherapies. DESIGN: Using mouse models, we evaluated whether immune checkpoint inhibitors or vaccine-based immunotherapies are effective in reducing tumour volumes of H. pylori-infected mice. In humans, we evaluated the correlation between H. pylori seropositivity and the efficacy of the programmed cell death protein 1 (PD-1) blockade therapy in patients with non-small-cell lung cancer (NSCLC). RESULTS: In mice engrafted with MC38 colon adenocarcinoma or B16-OVA melanoma cells, the tumour volumes of non-infected mice undergoing anticytotoxic T-lymphocyte-associated protein 4 and/or programmed death ligand 1 or anti-cancer vaccine treatments were significantly smaller than those of infected mice. We observed a decreased number and activation status of tumour-specific CD8+ T cells in the tumours of infected mice treated with cancer immunotherapies independent of the gut microbiome composition. Additionally, by performing an in vitro co-culture assay, we observed that dendritic cells of infected mice promote lower tumour-specific CD8+ T cell proliferation. We performed retrospective human clinical studies in two independent cohorts. In the Dijon cohort, H. pylori seropositivity was found to be associated with a decreased NSCLC patient survival on anti-PD-1 therapy. The survival median for H. pylori seropositive patients was 6.7 months compared with 15.4 months for seronegative patients (p=0.001). Additionally, in the Montreal cohort, H. pylori seropositivity was found to be associated with an apparent decrease of NSCLC patient progression-free survival on anti-PD-1 therapy. CONCLUSION: Our study unveils for the first time that the stomach microbiota affects the response to cancer immunotherapies and that H. pylori serology would be a powerful tool to personalize cancer immunotherapy treatment.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Infecciones por Helicobacter/complicaciones , Inhibidores de Puntos de Control Inmunológico/uso terapéutico , Neoplasias Pulmonares/tratamiento farmacológico , Adenocarcinoma/microbiología , Adenocarcinoma/patología , Animales , Vacunas contra el Cáncer/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/microbiología , Carcinoma de Pulmón de Células no Pequeñas/patología , Neoplasias del Colon/microbiología , Neoplasias del Colon/patología , Modelos Animales de Enfermedad , Femenino , Helicobacter pylori , Humanos , Neoplasias Pulmonares/microbiología , Neoplasias Pulmonares/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Estudios Retrospectivos
6.
Sci Adv ; 7(29)2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34272244

RESUMEN

The mechanisms maintaining adult lymphatic vascular specialization throughout life and their role in coordinating inter-organ communication to sustain homeostasis remain elusive. We report that inactivation of the mechanosensitive transcription factor Foxc2 in adult lymphatic endothelium leads to a stepwise intestine-to-lung systemic failure. Foxc2 loss compromised the gut epithelial barrier, promoted dysbiosis and bacterial translocation to peripheral lymph nodes, and increased circulating levels of purine metabolites and angiopoietin-2. Commensal microbiota depletion dampened systemic pro-inflammatory cytokine levels, corrected intestinal lymphatic dysfunction, and improved survival. Foxc2 loss skewed the specialization of lymphatic endothelial subsets, leading to populations with mixed, pro-fibrotic identities and to emergence of lymph node-like endothelial cells. Our study uncovers a cross-talk between lymphatic vascular function and commensal microbiota, provides single-cell atlas of lymphatic endothelial subtypes, and reveals organ-specific and systemic effects of dysfunctional lymphatics. These effects potentially contribute to the pathogenesis of diseases, such as inflammatory bowel disease, cancer, or lymphedema.


Asunto(s)
Vasos Linfáticos , Linfedema , Células Endoteliales/metabolismo , Endotelio Linfático/metabolismo , Endotelio Linfático/patología , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Humanos , Vasos Linfáticos/metabolismo , Linfedema/metabolismo , Linfedema/patología
7.
Vaccine ; 39(27): 3590-3601, 2021 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-34049736

RESUMEN

Helicobacter pylori (Hp) colonizes the human gastric mucosa with a high worldwide prevalence. Currently, Hp can be eradicated by the use of antibiotics. Due to the increase of antibiotic resistance, new therapeutic strategies need to be devised: one such approach being prophylactic vaccination. Pre-clinical and clinical data showed that a urease-based vaccine is efficient in decreasing Hp infection through the mobilization of T helper (Th)-dependent immune effectors, including eosinophils. Preliminary data have shown that upon vaccination and subsequent Hp infection, eosinophils accumulate in the gastric mucosa, suggesting a possible implication of this granulocyte subset in the vaccine-induced reduction of Hp infection. In our study, we confirm that activated eosinophils, expressing CD63, CD40, MHCII and PD-L1 at their cell surface, infiltrate the gastric mucosa during vaccine-induced reduction of Hp infection. Strikingly, we provide evidence that bone marrow derived eosinophils efficiently kill Hp in vitro, suggesting that eosinophils may participate to the vaccine-induced reduction of Hp infection. However, conversely to our expectations, the absence of eosinophils does not decrease the efficacy of this Hp vaccine in vivo. Indeed, vaccinated mice that have been genetically ablated of the eosinophil lineage or that have received anti-Sialic acid-binding immunoglobulin-like lectin F eosinophil-depleting antibodies, display a lower Hp colonization when compared to their eosinophil sufficient counterparts. Although the vaccine induces similar urease-specific humoral and Th responses in both eosinophil sufficient and deficient mice, a decreased production of anti-inflammatory cytokines, such as IL-10, TGFß, and calgranulin B, was specifically observed in eosinophil depleted mice. Taken together, our results suggest that gastric eosinophils maintain an anti-inflammatory environment, thus sustaining chronic Hp infection. Because eosinophils are one of the main immune effectors mobilized by Th2 responses, our study strongly suggests that the formulation of an Hp vaccine needs to include an adjuvant that preferentially primes Hp-specific Th1/Th17 responses.


Asunto(s)
Infecciones por Helicobacter , Helicobacter pylori , Animales , Eosinófilos , Mucosa Gástrica , Infecciones por Helicobacter/prevención & control , Ratones , Estómago
8.
iScience ; 23(3): 100891, 2020 Mar 27.
Artículo en Inglés | MEDLINE | ID: mdl-32114379

RESUMEN

Expression of Toll-interacting protein (Tollip), a potent TLR modulator, decreases in patients with inflammatory bowel diseases (IBD), whereas Tollip-/- mice are susceptible to colitis. Tollip expression was shown to be reduced in sporadic adenoma. In contrast, we found variable Tollip expression in patients with colitis-associated adenomas. In Tollip-/- mice challenged to develop colitis-associated cancer (CAC), tumor formation was significantly reduced owing to decreased mucosal proliferative and apoptotic indexes. This protection was associated with blunt inflammatory responses without significant changes in microbial composition. mRNA expression of Cd62l and Ccr5 homing receptors was reduced in colons of untreated Tollip-/- mice, whereas CD62L+ CD8+ T cells accumulated in the periphery. In Tollip-deficient adenomas Ctla-4 mRNA expression and tumor-infiltrating CD4+ Foxp3+ regulatory T cell (Treg) were decreased. Our data show that protection from CAC in Tollip-deficient mice is associated with defects in lymphocyte accumulation and composition in colitis-associated adenomas.

9.
Sci Rep ; 9(1): 6135, 2019 04 16.
Artículo en Inglés | MEDLINE | ID: mdl-30992496

RESUMEN

The maintenance of homeostasis in the gut is a major challenge for the immune system. Here we demonstrate that the transcription factor MAF plays a central role in T cells for the prevention of gastro-intestinal inflammation. Conditional knock out mice lacking Maf in all T cells developed spontaneous late-onset colitis, correlating with a decrease of FOXP3+RORγt+ T cells proportion, dampened IL-10 production in the colon and an increase of inflammatory TH17 cells. Strikingly, FOXP3+ specific conditional knock out mice for MAF did not develop colitis and demonstrated normal levels of IL-10 in their colon, despite the incapacity of regulatory T cells lacking MAF to suppress colon inflammation in Rag1-/- mice transferred with naïve CD4+ T cells. We showed that one of the cellular sources of IL-10 in the colon of these mice are TH17 cells. Thus, MAF is critically involved in the maintenance of the gut homeostasis by regulating the balance between Treg and TH17 cells either at the level of their differentiation or through the modulation of their functions.


Asunto(s)
Colitis/genética , Proteínas Proto-Oncogénicas c-maf/genética , Linfocitos T Reguladores/patología , Células Th17/patología , Animales , Células Cultivadas , Colitis/inmunología , Colitis/patología , Femenino , Factores de Transcripción Forkhead/análisis , Factores de Transcripción Forkhead/inmunología , Eliminación de Gen , Interleucina-10/análisis , Interleucina-10/inmunología , Masculino , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/análisis , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Proteínas Proto-Oncogénicas c-maf/inmunología , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
10.
Rev Med Suisse ; 13(572): 1480-1486, 2017 Aug 30.
Artículo en Francés | MEDLINE | ID: mdl-28853805

RESUMEN

Ulcerative colitis (UC) has a prevalence of 1 in 1000 inhabitants in Switzerland. The diagnosis of UC is based on a typical clinical presentation that involves bloody diarrhea, characteristic endoscopic features with continuous inflammation involving the rectum, and compatible histology. UC develops in genetically susceptible individuals with a dysregulated mucosal immune system. This article highlights latest insights into the pathogenesis, diagnosis, and therapy of UC.


La rectocolite ulcéro-hémorragique (RCUH) représente une maladie fréquente en Suisse avec une prévalence de 1 sur 1000 habitants. Le diagnostic se base sur la présentation clinique typique avec des diarrhées sanglantes, une image endoscopique caractéristique avec une inflammation continue qui touche pratiquement toujours le rectum, et une image histologique compatible. La RCUH se développe chez des individus avec prédisposition génétique et une dérégulation du système immunitaire colique. Cet article fait le point sur la pathogenèse, le diagnostic et les approches thérapeutiques de cette maladie.


Asunto(s)
Colitis Ulcerosa , Colitis Ulcerosa/diagnóstico , Colitis Ulcerosa/terapia , Diarrea , Hemorragia Gastrointestinal/etiología , Humanos , Inflamación
11.
Helicobacter ; 21 Suppl 1: 26-9, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27531535

RESUMEN

The tight control of the innate and adaptive immune responses in the stomach mucosa during chronic Helicobacter pylori infection is of prime importance for the bacteria to persist and for the host to prevent inflammation-driven diseases. This review summarizes recent data on the roles of innate and adaptive immune responses during H. pylori/host interactions. In addition, the latest preclinical developments of H. pylori vaccines are discussed with a special focus on the clinical trial reported by Zeng et al., who provided evidence that oral vaccination significantly reduces the acquisition of natural H. pylori infection in children.


Asunto(s)
Vacunas Bacterianas/inmunología , Infecciones por Helicobacter/inmunología , Infecciones por Helicobacter/prevención & control , Helicobacter pylori/inmunología , Inflamación/prevención & control , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/aislamiento & purificación , Infecciones por Helicobacter/patología , Interacciones Huésped-Patógeno , Humanos , Inflamación/patología , Ensayos Clínicos Controlados Aleatorios como Asunto
12.
Toxins (Basel) ; 8(6)2016 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-27338474

RESUMEN

One of the major toxins secreted by H. pylori is the Vacuolating cytotoxin A (VacA) named after its ability to induce the formation of "vacuole"-like membrane vesicles in the cytoplasm of gastric cells. VacA has been associated with the disruption of mitochondrial functions, stimulation of apoptosis, blockade of T cell proliferation and promotion of regulatory T cells, thereby making it a promising vaccine target. Immunity to bacterial virulence factors is well known to protect humans against bacterial infections; hence, detoxified VacA has been evaluated as a vaccine antigen. Our short review summarizes the pre-clinical and clinical data that have been published on the use of VacA in the development of the H. pylori vaccine.


Asunto(s)
Antígenos Bacterianos/inmunología , Proteínas Bacterianas/inmunología , Toxinas Bacterianas/inmunología , Vacunas , Animales , Antígenos Bacterianos/fisiología , Proteínas Bacterianas/fisiología , Humanos
13.
Inflamm Bowel Dis ; 22(7): 1723-36, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27271495

RESUMEN

Innate immune responses are key to maintain adequate host-microbial interactions. However, those signals are needed to efficiently trigger rapid and targeted antimicrobial responses in case of pathogen encounter. Several molecules have evolved to regulate intensity and coordinate signaling to avoid detrimental consequences to the host. Regulation can occur at the cell surface, within the cytoplasm, and at the transcriptional level. Innate immune regulation seems to be equally important than stimulation, as disruption of immunoregulatory molecules modulates the risk for several diseases. This is the case for colitis and inflammatory bowel disease but also colorectal cancer and intestinal infections. In this review, we recapitulate the molecular mechanisms underlying regulation of innate immune signals and mention their implications in several disease states including inflammatory bowel disease.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/inmunología , Inmunidad Innata , Inmunidad Mucosa , Enfermedades Inflamatorias del Intestino/inmunología , Mucosa Intestinal/inmunología , Neoplasias/inmunología , Receptores de Superficie Celular/inmunología , Animales , Membrana Celular/inmunología , Citoplasma/inmunología , Humanos , Infecciones/inmunología , Transducción de Señal/inmunología
14.
Infect Immun ; 83(11): 4217-28, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26283332

RESUMEN

Despite the proven ability of immunization to reduce Helicobacter infection in mouse models, the precise mechanism of protection has remained elusive. In this study, we evaluated the role of inflammatory monocytes in the vaccine-induced reduction of Helicobacter felis infection. We first showed by using flow cytometric analysis that Ly6C(low) major histocompatibility complex class II-positive chemokine receptor type 2 (CCR2)-positive CD64(+) inflammatory monocytes accumulate in the stomach mucosa during the vaccine-induced reduction of H. felis infection. To determine whether inflammatory monocytes played a role in the protection, these cells were depleted with anti-CCR2 depleting antibodies. Indeed, depletion of inflammatory monocytes was associated with an impaired vaccine-induced reduction of H. felis infection on day 5 postinfection. To determine whether inflammatory monocytes had a direct or indirect role, we studied their antimicrobial activities. We observed that inflammatory monocytes produced tumor necrosis factor alpha and inducible nitric oxide synthase (iNOS), two major antimicrobial factors. Lastly, by using a Helicobacter in vitro killing assay, we showed that mouse inflammatory monocytes and activated human monocytes killed H. pylori in an iNOS-dependent manner. Collectively, these data show that inflammatory monocytes play a direct role in the immunization-induced reduction of H. felis infection from the gastric mucosa.


Asunto(s)
Vacunas Bacterianas/inmunología , Infecciones por Helicobacter/inmunología , Helicobacter felis/inmunología , Monocitos/inmunología , Animales , Vacunas Bacterianas/administración & dosificación , Vacunas Bacterianas/genética , Femenino , Infecciones por Helicobacter/genética , Infecciones por Helicobacter/microbiología , Infecciones por Helicobacter/prevención & control , Helicobacter felis/genética , Humanos , Inmunización , Ratones , Ratones Endogámicos BALB C , Monocitos/microbiología , Óxido Nítrico Sintasa de Tipo II/genética , Óxido Nítrico Sintasa de Tipo II/inmunología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/inmunología
15.
J Exp Med ; 212(3): 369-83, 2015 Mar 09.
Artículo en Inglés | MEDLINE | ID: mdl-25732303

RESUMEN

NLR family apoptosis inhibitory proteins (NAIPs) belong to both the Nod-like receptor (NLR) and the inhibitor of apoptosis (IAP) families. NAIPs are known to form an inflammasome with NLRC4, but other in vivo functions remain unexplored. Using mice deficient for all NAIP paralogs (Naip1-6(Δ/Δ)), we show that NAIPs are key regulators of colorectal tumorigenesis. Naip1-6(Δ/Δ) mice developed increased colorectal tumors, in an epithelial-intrinsic manner, in a model of colitis-associated cancer. Increased tumorigenesis, however, was not driven by an exacerbated inflammatory response. Instead, Naip1-6(Δ/Δ) mice were protected from severe colitis and displayed increased antiapoptotic and proliferation-related gene expression. Naip1-6(Δ/Δ) mice also displayed increased tumorigenesis in an inflammation-independent model of colorectal cancer. Moreover, Naip1-6(Δ/Δ) mice, but not Nlrc4-null mice, displayed hyper-activation of STAT3 and failed to activate p53 18 h after carcinogen exposure. This suggests that NAIPs protect against tumor initiation in the colon by promoting the removal of carcinogen-elicited epithelium, likely in a NLRC4 inflammasome-independent manner. Collectively, we demonstrate a novel epithelial-intrinsic function of NAIPs in protecting the colonic epithelium against tumorigenesis.


Asunto(s)
Colitis/patología , Neoplasias del Colon/patología , Proteína Inhibidora de la Apoptosis Neuronal/metabolismo , Animales , Colitis/genética , Colitis/metabolismo , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Células Epiteliales/patología , Inflamasomas/genética , Inflamasomas/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína Inhibidora de la Apoptosis Neuronal/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo
16.
Pathobiology ; 81(4): 183-9, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25170537

RESUMEN

OBJECTIVE: Macrophages play a critical role in intestinal wound repair. However, the molecular pathways that regulate macrophage wound repair activities remain poorly understood. The aim of this study was to evaluate the role of GM-CSF receptor signaling in the wound repair activities of macrophages. METHODS: Murine macrophages were differentiated from bone marrow cells and human macrophages from monocytes isolated from peripheral blood mononuclear cells of Crohn's disease (CD) patients. In vitro models were used to study the repair activities of macrophages. RESULTS: We provide evidence that GM-CSF receptor signaling is required for murine macrophages to promote epithelial repair. In addition, we demonstrate that the deficient repair properties of macrophages from CD patients with active disease can be recovered via GM-CSF therapy. CONCLUSION: Our data support a critical role of the GM-CSF signaling pathway in the pro-repair activities of mouse and human macrophages.


Asunto(s)
Células Epiteliales/fisiología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Macrófagos/fisiología , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Transducción de Señal , Animales , Células de la Médula Ósea/inmunología , Células CACO-2 , Enfermedad de Crohn/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Monocitos/inmunología , Cicatrización de Heridas
17.
World J Gastroenterol ; 20(19): 5583-93, 2014 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-24914318

RESUMEN

Helicobacter pylori (H. pylori) infection is one of the most common infections in human beings worldwide. H. pylori express lipopolysaccharides and flagellin that do not activate efficiently Toll-like receptors and express dedicated effectors, such as γ-glutamyl transpeptidase, vacuolating cytotoxin (vacA), arginase, that actively induce tolerogenic signals. In this perspective, H. pylori can be considered as a commensal bacteria belonging to the stomach microbiota. However, when present in the stomach, H. pylori reduce the overall diversity of the gastric microbiota and promote gastric inflammation by inducing Nod1-dependent pro-inflammatory program and by activating neutrophils through the production of a neutrophil activating protein. The maintenance of a chronic inflammation in the gastric mucosa and the direct action of virulence factors (vacA and cytotoxin-associated gene A) confer pro-carcinogenic activities to H. pylori. Hence, H. pylori cannot be considered as symbiotic bacteria but rather as part of the pathobiont. The development of a H. pylori vaccine will bring health benefits for individuals infected with antibiotic resistant H. pylori strains and population of underdeveloped countries.


Asunto(s)
Infecciones por Helicobacter/inmunología , Helicobacter pylori , Antibacterianos/uso terapéutico , Proteínas Bacterianas/genética , Linfocitos T CD4-Positivos/inmunología , Gastritis/inmunología , Gastritis/microbiología , Tracto Gastrointestinal/inmunología , Tracto Gastrointestinal/microbiología , Humanos , Inmunidad Innata , Inmunoglobulina A/inmunología , Inmunoglobulina G/inmunología , Inflamación/inmunología , Inflamación/microbiología , Úlcera Péptica/inmunología , Úlcera Péptica/microbiología , Estómago/inmunología , Estómago/microbiología
18.
Inflamm Bowel Dis ; 20(4): 660-70, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24572204

RESUMEN

BACKGROUND: The intestinal epithelium accommodates with a myriad of commensals to maintain immunological homeostasis, but the underlying mechanisms regulating epithelial responsiveness to flora-derived signals remain poorly understood. Herein, we sought to determine the role of the Toll/interleukin (IL)-1 receptor regulator Toll-interacting protein (Tollip) in intestinal homeostasis. METHODS: Colitis susceptibility was determined after oral dextran sulfate sodium (DSS) administration or by breeding Tollip on an IL-10 background. The intestinal flora was depleted with 4 antibiotics before DSS exposure to assess its contribution in colitis onset. Bone marrow chimeras were generated to identify the cellular compartment, whereby Tollip may negatively regulate intestinal inflammation in response to DSS. Tollip-dependent epithelial barrier functions were studied in vitro by using Tollip-knockdown in Caco-2 cells and in vivo by immunohistochemistry and fluorescein isothiocyanate-labeled dextran gavage. RESULTS: Genetic ablation of Tollip did not lead to spontaneous intestinal inflammatory disorders. However, Tollip deficiency aggravated spontaneous disease onset in IL-10 mice and increased susceptibility to DSS colitis. Increased colitis severity in Tollip-deficient mice was not improved by bacterial flora depletion using broad-spectrum antibiotics. In addition, DSS exposure of bone marrow chimeric mice revealed a protective role for Tollip in nonhematopoietic cells. Knockdown of Tollip in epithelial cells led to exaggerated NFκ-B activity and proinflammatory cytokine secretion. Finally, DSS-treated Tollip mice showed enhanced intestinal permeability and increased epithelial apoptosis when compared with wild-type controls, a finding that coincided with tight junction alterations on injury. CONCLUSION: Overall, our data show an essential role for Tollip on colitis susceptibility in mice.


Asunto(s)
Colitis/genética , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Péptidos y Proteínas de Señalización Intracelular/genética , ARN Mensajero/metabolismo , Uniones Estrechas/metabolismo , Animales , Antibacterianos/farmacología , Apoptosis , Células de la Médula Ósea , Células CACO-2 , Quimera , Colitis/inducido químicamente , Colitis/patología , Sulfato de Dextran , Heces/microbiología , Femenino , Técnicas de Silenciamiento del Gen , Hemostasis , Humanos , Interleucina-10/genética , Interleucina-1beta/genética , Interleucina-6/genética , Interleucina-8/metabolismo , Mucosa Intestinal/patología , Péptidos y Proteínas de Señalización Intracelular/deficiencia , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Microbiota/efectos de los fármacos , FN-kappa B/metabolismo , Peptidoglicano/farmacología , Permeabilidad
19.
Pathobiology ; 81(2): 86-93, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24457836

RESUMEN

OBJECTIVE: Macrophages play a critical role in wound repair. However, the specific role of the different macrophage subtypes in wound repair remains incompletely understood. The aim of this study was to compare the wound repair activities of undifferentiated macrophages (M0), classically activated macrophages (M1) and alternatively activated (M2) macrophages. METHODS: The macrophage repair activities of intestinal wounds were evaluated using in vitro and in vivo models. RESULTS: All three macrophage subtypes enhanced wound closure in vitro, with the M2 macrophages demonstrating greater repair activities than the M0 and M1 macrophages. Injection of M0 and M2 macrophages into mice with experimental dextran sodium sulfate-induced colitis significantly enhanced ulcer repair when compared to control mice. In contrast, injection of M1 macrophages did not affect ulcer repair. CONCLUSIONS: These results underscore the wound repair capacity of different macrophage subsets. Notably, wound repair activity is not restricted to M2 macrophages, as the current literature suggests.


Asunto(s)
Activación de Macrófagos/fisiología , Macrófagos/citología , Cicatrización de Heridas/fisiología , Animales , Células 3T3 BALB , Diferenciación Celular , Colitis/patología , Modelos Animales de Enfermedad , Femenino , Ratones , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
20.
Am J Pathol ; 182(6): 2155-67, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23583421

RESUMEN

Inflammatory bowel diseases are commonly complicated by weight and bone loss. We hypothesized that IL-15, a pro-inflammatory cytokine expressed in colitis and an osteoclastogenic factor, could play a central role in systemic and skeletal complications of inflammatory bowel diseases. We evaluated the effects of an IL-15 antagonist, CRB-15, in mice with chronic colitis induced by oral 2% dextran sulfate sodium for 1 week, followed by another 1% for 2 weeks. During the last 2 weeks, mice were treated daily with CRB-15 or an IgG2a control antibody. Intestinal inflammation, disease severity, and bone parameters were evaluated at days 14 and 21. CRB-15 improved survival, early weight loss, and colitis clinical score, although colon damage and inflammation were prevented in only half the survivors. CRB-15 also delayed loss of femur bone mineral density and trabecular microarchitecture. Bone loss was characterized by decreased bone formation, but increased bone marrow osteoclast progenitors and osteoclast numbers on bone surfaces. CRB-15 prevented the suppression of osteoblastic markers of bone formation, and reduced osteoclast progenitors at day 14, but not later. However, by day 21, CRB-15 decreased tumor necrosis factor α and increased IL-10 expression in bone, paralleling a reduction of osteoclasts. These results delineate the role of IL-15 on the systemic and skeletal manifestations of chronic colitis and provide a proof-of-concept for future therapeutic developments.


Asunto(s)
Colitis/prevención & control , Interleucina-15/antagonistas & inhibidores , Osteoporosis/prevención & control , Proteínas Recombinantes de Fusión/uso terapéutico , Animales , Peso Corporal/efectos de los fármacos , Densidad Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Enfermedad Crónica , Colitis/inducido químicamente , Colitis/complicaciones , Colitis/fisiopatología , Citocinas/metabolismo , Sulfato de Dextran , Evaluación Preclínica de Medicamentos/métodos , Femenino , Fémur/patología , Fémur/fisiopatología , Mediadores de Inflamación/metabolismo , Interleucina-15/farmacología , Interleucina-15/uso terapéutico , Ratones , Ratones Endogámicos C57BL , Osteoblastos/efectos de los fármacos , Osteoblastos/patología , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteoporosis/etiología , Osteoporosis/patología , Osteoporosis/fisiopatología , Proteínas Recombinantes de Fusión/farmacología , Índice de Severidad de la Enfermedad , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA