Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Clin Invest ; 132(18)2022 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-36106640

RESUMEN

Forkhead box O transcriptional factors, especially FoxO1 and FoxO3a, play critical roles in physiologic and pathologic immune responses. However, the function of FoxO4, another main member of the FoxO family, in lymphoid cells is still poorly understood. Here, we showed that loss of FoxO4 in T cells augmented IFN-γ production of Th1 cells in vitro. Correspondingly, conditional deletion of FoxO4 in CD4+ T cells enhanced T cell-specific responses to Listeria monocytogenes infection in vivo. Genome-wide occupancy and transcriptomic analyses identified Dkk3 (encoding the Dickkopf-3 protein) as a direct transcriptional target of FoxO4. Consistent with the FoxO4-DKK3 relationship, recombinant DKK3 protein restored normal levels of IFN-γ production in FoxO4-deficient Th1 cells through the downregulation of lymphoid enhancer-binding factor 1 (Lef1) expression. Together, our data suggest a potential FoxO4/DKK3 axis in Th1 cell differentiation, providing what we believe to be an important insight and supplement for FoxO family proteins in T lymphocyte biology and revealing a promising target for the treatment of immune-related diseases.


Asunto(s)
Antiinfecciosos , Células TH1 , Antibacterianos , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Perfilación de la Expresión Génica , Células TH1/metabolismo
2.
JCI Insight ; 7(11)2022 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-35471938

RESUMEN

K-ras-mutant lung adenocarcinoma (KM-LUAD) is associated with abysmal prognosis and is tightly linked to tumor-promoting inflammation. A human mAb, canakinumab, targeting the proinflammatory cytokine IL-1ß, significantly decreased the risk of lung cancer in the Canakinumab Anti-inflammatory Thrombosis Outcomes Study. Interestingly, we found high levels of IL-1ß in the lungs of mice with K-rasG12D-mutant tumors (CC-LR mice). Here, we blocked IL-1ß using an anti-IL-1ß mAb in cohorts of 6- or 14-week-old CC-LR mice to explore its preventive and therapeutic effect, respectively. IL-1ß blockade significantly reduced lung tumor burden, which was associated with reprogramming of the lung microenvironment toward an antitumor phenotype characterized by increased infiltration of cytotoxic CD8+ T cells (with high IFN-γ and granzyme B expression but low programmed cell death 1 [PD-1] expression) while suppressing neutrophils and polymorphonuclear (PMN) myeloid-derived suppressor cells. When querying the Cancer Genome Atlas data set, we found positive correlations between IL1B expression and infiltration of immunosuppressive PMNs and expression of their chemoattractant, CXCL1, and PDCD1 expressions in patients with KM-LUAD. Our data provide evidence that IL-1ß blockade may be a preventive strategy for high-risk individuals and an alternative therapeutic approach in combination with currently available treatments for KM-LUAD.


Asunto(s)
Adenocarcinoma del Pulmón , Anticuerpos Monoclonales Humanizados , Interleucina-1beta , Neoplasias Pulmonares , Adenocarcinoma del Pulmón/tratamiento farmacológico , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/inmunología , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Citocinas/biosíntesis , Citocinas/inmunología , Genes ras , Humanos , Interleucina-1beta/antagonistas & inhibidores , Interleucina-1beta/inmunología , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Ratones , Terapia Molecular Dirigida , Mutación , Neutrófilos/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Microambiente Tumoral
3.
J Immunother Cancer ; 9(6)2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34083422

RESUMEN

BACKGROUND: Multiple types of immune cells producing IL-17 are found in the tumor microenvironment. However, their roles in tumor progression and exhaustion of CD8+ tumor-infiltrating lymphocytes (TILs) remain unclear. METHODS: To determine the role of type 17 immunity in tumor, we investigated the growth of B16F10 melanoma and the exhaustion of CD8+ TILs in Il17a-/- mice, Il17aCreR26DTA mice, RORγt inhibitor-treated mice, or their respective control mice. Adoptive transfer of tumor-specific IL-17-producing T cells was performed in B16F10-bearing congenic mice. Anti-CD4 or anti-Ly6G antibodies were used to deplete CD4+ T cells or CD11b+Gr-1hi myeloid cells in vivo, respectively. Correlation between type 17 immunity and T cell exhaustion in human cancer was evaluated by interrogating TCGA dataset. RESULTS: Depletion of CD4+ T cells promotes the exhaustion of CD8+ T cells with a concomitant increase in IL-17-producing CD8+ T (Tc17) cells in the tumor. Unlike IFN-γ-producing CD8+ T (Tc1) cells, tumor-infiltrating Tc17 cells exhibit CD103+KLRG1-IL-7Rαhi tissue resident memory-like phenotypes and are poorly cytolytic. Adoptive transfer of IL-17-producing tumor-specific T cells increases, while depletion of IL-17-producing cells decreases, the frequency of PD-1hiTim3+TOX+ terminally exhausted CD8+ T cells in the tumor. Blockade of IL-17 or RORγt pathway inhibits exhaustion of CD8+ T cells and also delays tumor growth in vivo. Consistent with these results, human TCGA analyses reveal a strong positive correlation between type 17 and CD8+ T cell exhaustion signature gene sets in multiple cancers. CONCLUSION: IL-17-producing cells promote terminal exhaustion of CD8+ T cells and tumor progression in vivo, which can be reversed by blockade of IL-17 or RORγt pathway. These findings unveil a novel role for IL-17-producing cells as tumor-promoting cells facilitating CD8+ T cell exhaustion, and propose type 17 immunity as a promising target for cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Eliminación de Gen , Interleucina-17/genética , Melanoma Experimental/terapia , Traslado Adoptivo , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/trasplante , Línea Celular Tumoral , Femenino , Humanos , Masculino , Melanoma Experimental/genética , Melanoma Experimental/inmunología , Ratones , Microambiente Tumoral
4.
Nat Immunol ; 21(11): 1467, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32884131

RESUMEN

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

5.
Oncoimmunology ; 9(1): 1682380, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32002289

RESUMEN

Oncogenic KRAS-mutant lung cancers remain treatment refractory. A better understanding of the immune response of KRAS-mutant lung cancers is required to facilitate the development of potential therapeutic strategies. Regulatory T cells (Tregs) are a subset of immune cells that promote tumor progression through suppressing anti-tumor immune response. Here, we used KrasG12D lung cancer mice to examine the characteristics of tumor-infiltrating Tregs. In tumor-bearing animals, Tregs are increased during tumor progression. Of note, a majority of Tregs that localized in lung tumors of Kras-mutant mice expressed ST2, a receptor for IL-33, which are different from Tregs in secondary lymphoid organs. To investigate the function of local Tregs influencing immune response in primary lung tumor development, we used anti-ST2 antibody to deplete Tregs in lung tumors of Kras-mutant mice. Treatment of Kras-mutant mice with anti-ST2 antibody resulted in depletion of activated Tregs in lung tumor while leaving Tregs in secondary lymphoid organs intact. Also, localized Tregs depletion led to a significant reduction in lung tumor burden. Immune response after Tregs depletion in tumors showed restoration of NK cell activity and enhanced Th1 activity, with increased CD8 cytotoxic T cell response. In addition, we found that the M2 macrophage signature in lung tumors was suppressed upon Tregs depletion, accompanied by upregulation of surface expression of MHC-II molecules and reduced expression of Arg1, Mmp12, Cxcl2, and Chi3l3. These data suggest that therapeutic strategies targeting activated Tregs in lung cancer have the potential to restrain tumor progression by enhancing anti-tumor immunity.


Asunto(s)
Neoplasias Pulmonares , Linfocitos T Reguladores , Animales , Linfocitos T CD8-positivos , Células Asesinas Naturales , Neoplasias Pulmonares/tratamiento farmacológico , Ratones , Proteínas Proto-Oncogénicas p21(ras)/genética
6.
Cardiovasc Intervent Radiol ; 43(2): 311-321, 2020 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-31591689

RESUMEN

PURPOSE: To determine the tumor immune cell landscape after transcatheter arterial bland embolization (TAE) in a clinically relevant rat hepatocellular carcinoma (HCC) model. MATERIALS AND METHODS: Buffalo rats (n = 21) bearing syngeneic McArdle RH-7777 rat hepatoma cells implanted into the left hepatic lobe underwent TAE using 70-150 µm beads (n = 9) or hepatic artery saline infusion (n = 12). HCC nodules, peritumoral margin, adjacent non-cancerous liver, and splenic parenchyma were collected and disaggregated to generate single-cell suspensions for immunological characterization 14 d after treatment. Changes in tumor-infiltrating immune subsets including CD4 T cells (Th17 and Treg), CD8 cytotoxic T cells (IFNγ), and neutrophils were evaluated by multiparameter flow cytometry. Migration and colony formation assays were performed to examine the effect of IL-17, a signature cytokine of Th17 cells, on McArdle RH-7777 hepatoma cells under conditions simulating post-embolization environment (i.e., hypoxia and nutrient privation). Statistical significance was determined by the Student unpaired t test or one-way ANOVA. RESULTS: TAE induces increased infiltration of Th17 cells in liver tumors when compared with controls 14 d after treatment (0.29 ± 0.01 vs. 0.19 ± 0.02; p = 0.02). A similar pattern was observed in the spleen (1.41 ± 0.13 vs. 0.57 ± 0.08; p < 0.001), indicating both local and systemic effect. No significant differences in the percentage of FoxP3 + Tregs, IFNγ-producing CD4 T cells, and CD8 T cells were observed between groups (p > 0.05). In vitro post-embolization assays demonstrated that IL-17 reduces McA-RH7777 cell migration at 24-48 h (p = 0.003 and p = 0.002, respectively). CONCLUSION: Transcatheter hepatic arterial bland embolization induces local and systemic increased infiltration of Th17 cells and expression of their signature cytokine IL-17. In a simulated post-embolization environment, IL-17 significantly reduced McA-RH7777 cell migration.


Asunto(s)
Carcinoma Hepatocelular/terapia , Embolización Terapéutica/métodos , Neoplasias Hepáticas/terapia , Células Th17/metabolismo , Animales , Carcinoma Hepatocelular/diagnóstico por imagen , Carcinoma Hepatocelular/inmunología , Modelos Animales de Enfermedad , Humanos , Hígado/diagnóstico por imagen , Hígado/inmunología , Neoplasias Hepáticas/diagnóstico por imagen , Neoplasias Hepáticas/inmunología , Imagen por Resonancia Magnética/métodos , Masculino , Radiología Intervencionista/métodos , Ratas , Ratas Endogámicas BUF , Células Th17/inmunología
7.
Front Immunol ; 10: 1172, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31244826

RESUMEN

Interleukin-17D (IL-17D) belongs to the IL-17 family of cytokines. While the members of the IL-17 family have been implicated in inflammation and host defense, the function of IL-17D remains unclear. Here, we showed that the lack of IL-17D expression confers protection against Listeria infection. A deficiency in IL-17D also resulted in less weight loss with reduced pathogen burden during influenza A virus infection. During infection, the loss of IL-17D resulted in compromised CD8 T cell activity. CD8 T cell depletion in IL-17D-deficient mice restored the bacterial burden to a level similar to that found in WT mice. Similarly, IL-17D-deficient mice in a RAG-deficient background had no difference in bacterial and viral burden compared to WT mice. IL-17D controlled CD8 T cell activity in part by suppressing the function of dendritic cells. We found that IL-17D from the non-hematopoietic compartment regulates protective immunity during infection. Together, our data led to the identification of IL-17D as a critical cytokine during intracellular bacteria and virus infection that suppresses the activity of CD8 T cells by regulating dendritic cells.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Inmunomodulación , Infecciones/etiología , Infecciones/metabolismo , Interleucina-17/metabolismo , Activación de Linfocitos/inmunología , Animales , Biomarcadores , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Expresión Génica , Inmunofenotipificación , Terapia de Inmunosupresión , Interleucina-17/genética , Ratones , Ratones Transgénicos , Índice de Severidad de la Enfermedad , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/metabolismo
8.
Arch Pharm Res ; 42(7): 549-559, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30941641

RESUMEN

Th17 cells are a specialized subset of CD4+ T cells that are essential in driving inflammation during autoimmune disease and infection through a signature cytokine IL-17. Th17 cells have been found in various human cancers. The function of these cells in cancers is highly context-dependent; both tumor-promoting and tumor-suppressing activity have been reported. IL-17 and IL-22, Th17-derived cytokines, influence the tumor microenvironment by directly promoting transformed cell properties and neighboring stromal cell activity. These cytokines are also involved in regulation of the immune system by modulating the activities of myeloid cells and T cells. These findings suggest that Th17 cells and their cytokines are a key mediator of cancer development, representing a potential target for cancer therapy. Herein, I review recent preclinical studies on the function of Th17 cells and IL-17 in cancer and discuss possible therapeutic approaches to harness Th17 cells for cancer immunotherapy.


Asunto(s)
Inmunoterapia , Interleucina-17/inmunología , Neoplasias/inmunología , Neoplasias/terapia , Células Th17/inmunología , Animales , Humanos
9.
Cancer Immunol Res ; 6(7): 788-797, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29764837

RESUMEN

Somatic KRAS mutations are the most common oncogenic variants in lung cancer and are associated with poor prognosis. Using a Kras-induced lung cancer mouse model, CC-LR, we previously showed a role for inflammation in lung tumorigenesis through activation of the NF-κB pathway, along with induction of interleukin 6 (IL6) and an IL17-producing CD4+ T-helper cell response. IL22 is an effector molecule secreted by CD4+ and γδ T cells that we previously found to be expressed in CC-LR mice. IL22 mostly signals through the STAT3 pathway and is thought to act exclusively on nonhematopoietic cells with basal IL22 receptor (IL22R) expression on epithelial cells. Here, we found that higher expression of IL22R1 in patients with KRAS-mutant lung adenocarcinoma was an independent indicator of poor recurrence-free survival. We then showed that genetic ablation of Il22 in CC-LR mice (CC-LR/IL22KO mice) caused a significant reduction in tumor number and size. This was accompanied by significantly lower tumor cell proliferation, angiogenesis, and STAT3 activation. Il22 ablation was also associated with significant reduction in lung-infiltrating inflammatory cells and expression of protumor inflammatory cytokines. Conversely, this was accompanied with increased antitumor Th1 and cytotoxic CD8+ T-cell responses, while suppressing the protumor immunosuppressive T regulatory cell response. In CC-LR/IL22KO mice, we found significantly reduced expression of core stemness genes and the number of prototypical SPC+CCSP+ stem cells. Thus, we conclude that IL22 promotes Kras-mutant lung tumorigenesis by driving a protumor inflammatory microenvironment with proliferative, angiogenic, and stemness contextual cues in epithelial/tumor cells. Cancer Immunol Res; 6(7); 788-97. ©2018 AACR.


Asunto(s)
Interleucinas/metabolismo , Neoplasias Pulmonares/etiología , Neoplasias Pulmonares/metabolismo , Mutación , Células Madre Neoplásicas/inmunología , Células Madre Neoplásicas/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Animales Modificados Genéticamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/inmunología , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Inmunohistoquímica , Interleucinas/genética , Neoplasias Pulmonares/patología , Ratones , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Microambiente Tumoral
10.
J Allergy Clin Immunol ; 141(6): 2061-2073.e5, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-28943467

RESUMEN

BACKGROUND: Allergic asthma is a heterogeneous chronic inflammatory disease of the airways with a massive infiltration of eosinophils or neutrophils mediated by allergen-specific TH2 and TH17 cells, respectively. Therefore successful treatment of allergic asthma will require suppression of both TH2 and TH17 cells. OBJECTIVE: We sought to investigate the role of the TH17 cell pathway in regulating TH2 cell responses in allergic asthma. METHODS: Allergic asthma was induced by intranasal challenge with proteinase allergens in C57BL/6, Il17a-/-Il17f-/-, and retinoic acid receptor-related orphan receptor γt (RORγt)gfp/gfp mice. A pharmacologic RORγt inhibitor was used to evaluate its preventive and therapeutic effects in allergic asthma. Characteristics of allergic airway inflammation were analyzed by using flow cytometry, histology, quantitative real-time PCR, and ELISA. Mixed bone marrow chimeric mice, fate mapping analysis, short hairpin RNA transduction, and in vitro T-cell differentiation were used for mechanistic studies. RESULTS: Mice deficient in IL-17A and IL-17F, as well as RORγt, exhibited a significant reduction not only in TH17 cell responses but also in TH2 cell responses in an animal model of allergic asthma. Similarly, mice treated with an RORγt inhibitor had significantly diminished TH17 and TH2 cell responses, leading to reduced neutrophil and eosinophil numbers in the airway. RORγt-deficient T cells were intrinsically defective in differentiating into TH2 cells and expressed increased levels of B-cell lymphoma 6 (Bcl6). Bcl6 knockdown resulted in a remarkable restoration of TH2 cell differentiation in RORγt-deficient T cells. Blockade of RORγt also significantly hampered the differentiation of human TH2 and TH17 cells from naive CD4+ T cells. CONCLUSION: RORγt in T cells is required for optimal TH2 cell differentiation by suppressing Bcl6 expression; this finding suggests that targeting RORγt might be a promising approach for the treatment of allergic asthma by concomitantly suppressing TH17 and TH2 cell responses in the airway.


Asunto(s)
Asma/inmunología , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Células Th17/inmunología , Células Th2/inmunología , Animales , Diferenciación Celular/inmunología , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores
11.
Cell Rep ; 21(1): 195-207, 2017 Oct 03.
Artículo en Inglés | MEDLINE | ID: mdl-28978473

RESUMEN

Th17 cells are potent mediators in autoimmune diseases, and RORγt is required for their development. Recent studies have shown that RORγt+ Treg cells in the gut regulate intestinal inflammation by inhibiting effector T cell function. In the current study, we report that RORγt+ Treg cells were also found in lymph nodes following immunization. Not only distinct from intestinal RORγt+ Treg cells in their transcriptomes, peripheral RORγt+ Treg cells were derived from Foxp3+ thymic Treg cells in an antigen-specific manner. Development of these RORγt+ Treg cells, coined T regulatory 17 (Tr17) cells, depended on IL-6/Stat3 signaling. Tr17 cells showed suppressive activity against antigen-specific effector T cells in vitro. In addition, Tr17 cells efficiently inhibited myelin-specific Th17-cell-mediated CNS auto-inflammation in a passive EAE model. Collectively, our study demonstrates that Tr17 cells are effector Treg cells that potentially restrict autoimmunity.


Asunto(s)
Autoinmunidad/genética , Encefalomielitis Autoinmune Experimental/genética , Factores de Transcripción Forkhead/genética , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Linfocitos T Reguladores/inmunología , Células Th17/inmunología , Traslado Adoptivo , Animales , Diferenciación Celular , Encefalomielitis Autoinmune Experimental/inducido químicamente , Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , Factores de Transcripción Forkhead/inmunología , Regulación de la Expresión Génica , Genes Reporteros , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Proteína Coestimuladora de Linfocitos T Inducibles/genética , Proteína Coestimuladora de Linfocitos T Inducibles/inmunología , Interleucina-6/genética , Interleucina-6/inmunología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Glicoproteína Mielina-Oligodendrócito/administración & dosificación , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/inmunología , Fragmentos de Péptidos/administración & dosificación , Receptores CCR6/genética , Receptores CCR6/inmunología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Transducción de Señal , Linfocitos T Reguladores/patología , Linfocitos T Reguladores/trasplante , Células Th17/patología
12.
Cell Res ; 27(8): 1034-1045, 2017 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-28685773

RESUMEN

The interaction between tumor and the immune system is still poorly understood. Significant clinical responses have been achieved in cancer patients treated with antibodies against the CTLA4 and PD-1/PD-L1 checkpoints; however, only a small portion of patients responded to the therapies, indicating a need to explore additional co-inhibitory molecules for cancer treatment. B7-H3, a member of the B7 superfamily, was previously shown by us to inhibit T-cell activation and autoimmunity. In this study, we have analyzed the function of B7-H3 in tumor immunity. Expression of B7-H3 was found in multiple tumor lines, tumor-infiltrating dendritic cells, and macrophages. B7-H3-deficient mice or mice treated with an antagonistic antibody to B7-H3 showed reduced growth of multiple tumors, which depended on NK and CD8+ T cells. With a putative receptor expressed by cytotoxic lymphocytes, B7-H3 inhibited their activation, and its deficiency resulted in increased cytotoxic lymphocyte function in tumor-bearing mice. Combining blockades of B7-H3 and PD-1 resulted in further enhanced therapeutic control of late-stage tumors. Taken together, our results indicate that the B7-H3 checkpoint may serve as a novel target for immunotherapy against cancer.


Asunto(s)
Antígenos B7/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Celular , Células Asesinas Naturales/inmunología , Proteínas de Neoplasias/inmunología , Neoplasias/inmunología , Animales , Antígenos B7/genética , Linfocitos T CD8-positivos/patología , Línea Celular Tumoral , Femenino , Humanos , Células Asesinas Naturales/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas de Neoplasias/genética , Neoplasias/genética , Neoplasias/patología , Neoplasias/terapia
13.
Oncoimmunology ; 5(10): e1229724, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27853654

RESUMEN

Tumor necrosis factor (TNF) is known as an important regulator of tumor microenvironment and inflammation. TNF levels are markedly elevated in the bronchoalveolar lavage fluid (BALF) of patients with chronic obstructive pulmonary disease (COPD), which is an independent risk factor for lung cancer. We have previously shown that COPD-like airway inflammation promotes lung cancer in a K-ras mutant mouse model (CC-LR mouse). This was associated with a significant increase of neutrophils in BALF, accompanied by a marked increase in TNF level, suggesting a link between COPD, TNF, and lung cancer promotion. Therefore, we first overexpressed TNF in the airway epithelium of CC-LR mice, which promoted lung cancer by ∼2-fold. This was associated with increased numbers of Ki67 and CD31 positive cells in lung tumors of CC-LR/TNF-Tg mice. We also found a robust increase in NF-κB activation, and numbers of neutrophils and myeloid-derived suppressor cells (MDSCs) in lung. Accordingly, we depleted MDSCs in CC-LR/TNF-Tg mice, which lead to significant tumor suppression emphasizing on the role of TNF-induced MDSCs in K-ras induced lung tumorigenesis. Finally, we targeted TNF expression by crossing CC-LR mice with TNF knock-out mice (CC-LR/TNF-KO), which resulted in a significant decrease in lung tumor burden in the absence or presence of COPD-like airway inflammation. Interestingly, there were less MDSCs and lower Ki67 and CD31 expression in the lung of the CC-LR/TNF-KO mice. We conclude that TNF links COPD to lung cancer promotion by induction of an immunosuppressive MDSC response, and subsequent amplification of proliferation and angiogenesis in tumors.

14.
Cancer Res ; 76(11): 3189-99, 2016 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-27197187

RESUMEN

Activating mutations of K-ras are the most common oncogenic alterations found in lung cancer. Unfortunately, attempts to target K-ras-mutant lung tumors have thus far failed, clearly indicating the need for new approaches in patients with this molecular profile. We have previously shown NF-κB activation, release of IL6, and activation of its responsive transcription factor STAT3 in K-ras-mutant lung tumors, which was further amplified by the tumor-enhancing effect of chronic obstructive pulmonary disease (COPD)-type airway inflammation. These findings suggest an essential role for this inflammatory pathway in K-ras-mutant lung tumorigenesis and its enhancement by COPD. Therefore, here we blocked IL6 using a monoclonal anti-IL6 antibody in a K-ras-mutant mouse model of lung cancer in the absence or presence of COPD-type airway inflammation. IL6 blockade significantly inhibited lung cancer promotion, tumor cell-intrinsic STAT3 activation, tumor cell proliferation, and angiogenesis markers. Moreover, IL6 inhibition reduced expression of protumor type 2 molecules (arginase 1, Fizz 1, Mgl, and IDO), number of M2-type macrophages and granulocytic myeloid-derived suppressor cells, and protumor T-regulatory/Th17 cell responses. This was accompanied by increased expression of antitumor type 1 molecule (Nos2), and antitumor Th1/CD8 T-cell responses. Our study demonstrates that IL6 blockade not only has direct intrinsic inhibitory effect on tumor cells, but also reeducates the lung microenvironment toward an antitumor phenotype by altering the relative proportion between protumor and antitumor immune cells. This information introduces IL6 as a potential druggable target for prevention and treatment of K-ras-mutant lung tumors. Cancer Res; 76(11); 3189-99. ©2016 AACR.


Asunto(s)
Anticuerpos Monoclonales/farmacología , Carcinoma de Pulmón de Células no Pequeñas/patología , Interleucina-6/antagonistas & inhibidores , Neoplasias Pulmonares/patología , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Microambiente Tumoral/efectos de los fármacos , Adenocarcinoma/tratamiento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/patología , Animales , Apoptosis , Western Blotting , Carcinogénesis , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Carcinoma de Pulmón de Células no Pequeñas/genética , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Técnicas para Inmunoenzimas , Interleucina-6/genética , Interleucina-6/inmunología , Interleucina-6/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/genética , Ratones , FN-kappa B/genética , FN-kappa B/metabolismo , Estadificación de Neoplasias , Pronóstico , Enfermedad Pulmonar Obstructiva Crónica/tratamiento farmacológico , Enfermedad Pulmonar Obstructiva Crónica/genética , Enfermedad Pulmonar Obstructiva Crónica/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Tasa de Supervivencia , Células Tumorales Cultivadas
15.
Oncoimmunology ; 4(1): e955704, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25949859

RESUMEN

Although inflammation has been linked to lung cancer pathogenesis, very little is known about the critical players during lung cancer development. We found that Kras mutation in lung epithelial cells preferentially leads to recruitment of Th17 cells, which produce IL-17, a signature cytokine that promotes inflammation. We demonstrated IL-17 is critical for tumor growth in part by recruiting tumorigenic GR1+ CD11b+ cells.

16.
Immunity ; 42(4): 692-703, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25888259

RESUMEN

The interleukin-17 (IL-17) family of cytokines has emerged as a critical player in inflammatory diseases. Among them, IL-25 has been shown to be important in allergic inflammation and protection against parasitic infection. Here we have demonstrated that IL-17B, a poorly understood cytokine, functions to inhibit IL-25-driven inflammation. IL-17B and IL-25, both binding to the interleukin-17 receptor B (IL-17RB), were upregulated in their expression after acute colonic inflammation. Individual inhibition of these cytokines revealed opposing functions in colon inflammation: IL-25 was pathogenic but IL-17B was protective. Similarly opposing phenotypes were observed in Citrobacter rodentium infection and allergic asthma. Moreover, IL-25 was found to promote IL-6 production from colon epithelial cells, which was inhibited by IL-17B. Therefore, our data demonstrate that IL-17B is an anti-inflammatory cytokine in the IL-17 family.


Asunto(s)
Asma/inmunología , Colitis/inmunología , Disbiosis/inmunología , Infecciones por Enterobacteriaceae/inmunología , Interleucina-17/inmunología , Interleucinas/inmunología , Mucosa Intestinal/inmunología , Animales , Antibacterianos , Asma/inducido químicamente , Asma/genética , Asma/patología , Línea Celular , Citrobacter rodentium/inmunología , Colitis/inducido químicamente , Colitis/genética , Colitis/patología , Disbiosis/inducido químicamente , Disbiosis/genética , Disbiosis/patología , Infecciones por Enterobacteriaceae/genética , Infecciones por Enterobacteriaceae/microbiología , Infecciones por Enterobacteriaceae/patología , Células Epiteliales/inmunología , Células Epiteliales/patología , Regulación de la Expresión Génica , Interleucina-17/deficiencia , Interleucina-17/genética , Interleucina-6/genética , Interleucina-6/inmunología , Interleucinas/deficiencia , Interleucinas/genética , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ovalbúmina , Unión Proteica , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/inmunología , Transducción de Señal , Dodecil Sulfato de Sodio
17.
Proc Natl Acad Sci U S A ; 111(15): 5664-9, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24706787

RESUMEN

Lung cancer development is associated with extensive pulmonary inflammation. In addition, the linkage between chronic obstructive pulmonary disease (COPD) and lung cancer has been demonstrated in population-based studies. IL-17-producing CD4 helper T cells (Th17 cells) play a critical role in promoting chronic tissue inflammation. Although Th17 cells are found in human COPD and lung cancer, their role is not understood. We have thus used a mouse model of lung cancer, in which an oncogenic form of K-ras (K-ras(G12D)), frequently found in human lung cancer, is restrictedly expressed in lung epithelial cells [via Clara cell secretory protein (CCSP(cre))]. In this model, Th17 and Treg but not Th1 cells were found enriched at the tumor tissues. When CCSP(cre)/K-ras(G12D) mice were weekly challenged with a lysate of nontypeable Haemophilus influenza (NTHi), which induces COPD-type inflammation and accelerates the tumor growth, they showed greatly enhanced Th17 cell infiltration in the lung tissues. Lack of IL-17, but not IL-17F, resulted in a significant reduction in lung tumor numbers in CCSP(cre)/K-ras(G12D) mice and also those treated with NTHi. Absence of IL-17 not only resulted in reduction of tumor cell proliferation and angiogenesis, but also decreased the expression of proinflammatory mediators and reduced recruitment of myeloid cells. Depletion of Gr-1(+)CD11b(+) myeloid cells in CCSP(cre)/K-ras(G12D) mice suppressed tumor growth in lung, indicating Gr-1(+)CD11b(+) myeloid cells recruited by IL-17 play a protumor role. Taken together, our data demonstrate a critical role for Th17 cell-mediated inflammation in lung tumorigenesis and suggest a novel way for prevention and treatment of this disease.


Asunto(s)
Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/fisiopatología , Neumonía/inmunología , Enfermedad Pulmonar Obstructiva Crónica/inmunología , Células Th17/inmunología , Animales , Cartilla de ADN/genética , Citometría de Flujo , Genes ras/genética , Haemophilus influenzae/inmunología , Inmunohistoquímica , Neoplasias Pulmonares/complicaciones , Ratones , Ratones Mutantes , Mutación/genética , Células Mieloides/inmunología , Células Mieloides/metabolismo , Neumonía/complicaciones , Enfermedad Pulmonar Obstructiva Crónica/complicaciones , Reacción en Cadena en Tiempo Real de la Polimerasa , Uteroglobina/metabolismo
18.
PLoS Pathog ; 9(9): e1003628, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-24068935

RESUMEN

Although the protective functions by T helper 17 (Th17) cytokines against extracellular bacterial and fungal infection have been well documented, their importance against intracellular bacterial infection remains unclear. Here, we investigated the contribution of Th17 responses to host defense against intracellular bacteria Listeria monocytogenes and found that Th17 cell generation was suppressed in this model. Unexpectedly, mice lacking both p35 and EBI3 cleared L. monocytogenes as efficiently as wild-type mice, whereas p35-deficient mice failed to do so. Furthermore, both innate cells and pathogen-specific T cells from double-deficient mice produced significantly higher IL-17 and IL-22 compared to wild-type mice. The bacterial burden in the liver of double-deficient mice treated with anti-IL-17 was significantly increased compared to those receiving a control Ab. Transfer of Th17 cells specific for listeriolysin O as well as administration of IL-17 and IL-22 significantly suppressed bacterial growth in p35-deficient mice, indicating the critical contribution of Th17 responses to host defense against the intracellular pathogen in the absence of IL-12 and proper Th1 responses. Our findings unveil a novel immune evasion mechanism whereby the intracellular bacteria exploit IL-27EBI3 to suppress Th17-mediated protective immunity.


Asunto(s)
Regulación hacia Abajo , Inmunidad Celular , Subunidad p35 de la Interleucina-12/metabolismo , Listeria monocytogenes/inmunología , Listeriosis/inmunología , Receptores de Citocinas/metabolismo , Células Th17/inmunología , Animales , Carga Bacteriana , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/microbiología , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/trasplante , Células Cultivadas , Regulación de la Expresión Génica , Evasión Inmune , Inmunidad Innata , Subunidad p35 de la Interleucina-12/genética , Interleucina-17/genética , Interleucina-17/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Listeria monocytogenes/crecimiento & desarrollo , Listeria monocytogenes/aislamiento & purificación , Listeriosis/microbiología , Listeriosis/patología , Listeriosis/terapia , Hígado/inmunología , Hígado/microbiología , Ratones Endogámicos C57BL , Ratones Noqueados , Antígenos de Histocompatibilidad Menor , Receptores de Citocinas/genética , Bazo/inmunología , Bazo/metabolismo , Bazo/microbiología , Bazo/patología , Células Th17/metabolismo , Células Th17/microbiología , Células Th17/patología
19.
Nat Immunol ; 14(7): 732-40, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23727894

RESUMEN

Transcription factors of the STAT family are critical in the cytokine-mediated functional differentiation of CD4(+) helper T cells. Signaling inhibitors of the SOCS family negatively regulate the activation of STAT proteins; however, their roles in the differentiation and function of helper T cells are not well understood. Here we found that the SOCS protein CIS, which was substantially induced by interleukin 4 (IL-4), negatively regulated the activation of STAT3, STAT5 and STAT6 in T cells. CIS-deficient mice spontaneously developed airway inflammation, and CIS deficiency in T cells led to greater susceptibility to experimental allergic asthma. CIS-deficient T cells showed enhanced differentiation into the TH2 and TH9 subsets of helper T cells. STAT5 and STAT6 regulated IL-9 expression by directly binding to the Il9 promoter. Our data thus demonstrate a critical role for CIS in controlling the proallergic generation of helper T cells.


Asunto(s)
Asma/inmunología , Inflamación/inmunología , Factores de Transcripción STAT/inmunología , Proteínas Supresoras de la Señalización de Citocinas/inmunología , Linfocitos T Reguladores/inmunología , Animales , Diferenciación Celular/inmunología , Histocitoquímica , Immunoblotting , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosforilación , ARN/química , ARN/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal , Proteínas Supresoras de la Señalización de Citocinas/genética
20.
J Immunol ; 189(9): 4226-30, 2012 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-23024280

RESUMEN

In the IL-17 family of cytokines, much is known about the sources and functions of IL-17, IL-17F, and IL-25 in the host defense against infection and in inflammatory diseases; however, the physiological function of IL-17C remains poorly understood. Using mice deficient in IL-17C, we demonstrate that this cytokine is crucial for the regulation of an acute experimental colitis elicited by dextran sulfate sodium. In this model, mice lacking IL-17C exhibited exacerbated disease that was associated with increased IL-17 expression by γδ T cells and Th17 cells. Moreover, IL-17C directly regulated the expression of the tight junction molecule occludin by colonic epithelial cells. Thus, our data suggest that IL-17C plays a critical role in maintaining mucosal barrier integrity.


Asunto(s)
Colitis/inmunología , Colitis/patología , Mediadores de Inflamación/fisiología , Interleucina-17/fisiología , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Animales , Línea Celular , Colitis/genética , Colon/citología , Colon/inmunología , Colon/patología , Sulfato de Dextran/toxicidad , Modelos Animales de Enfermedad , Epitelio/inmunología , Epitelio/metabolismo , Epitelio/patología , Predisposición Genética a la Enfermedad , Mediadores de Inflamación/metabolismo , Interleucina-17/biosíntesis , Interleucina-17/deficiencia , Mucosa Intestinal/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...