Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Exp Med ; 220(2)2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36367776

RESUMEN

Immune checkpoint blockade (ICB) has revolutionized cancer treatment, yet quality of life and continuation of therapy can be constrained by immune-related adverse events (irAEs). Limited understanding of irAE mechanisms hampers development of approaches to mitigate their damage. To address this, we examined whether mice gained sensitivity to anti-CTLA-4 (αCTLA-4)-mediated toxicity upon disruption of gut homeostatic immunity. We found αCTLA-4 drove increased inflammation and colonic tissue damage in mice with genetic predisposition to intestinal inflammation, acute gastrointestinal infection, transplantation with a dysbiotic fecal microbiome, or dextran sodium sulfate administration. We identified an immune signature of αCTLA-4-mediated irAEs, including colonic neutrophil accumulation and systemic interleukin-6 (IL-6) release. IL-6 blockade combined with antibiotic treatment reduced intestinal damage and improved αCTLA-4 therapeutic efficacy in inflammation-prone mice. Intestinal immune signatures were validated in biopsies from patients with ICB colitis. Our work provides new preclinical models of αCTLA-4 intestinal irAEs, mechanistic insights into irAE development, and potential approaches to enhance ICB efficacy while mitigating irAEs.


Asunto(s)
Colitis , Interleucina-6 , Ratones , Animales , Calidad de Vida , Colitis/patología , Inmunoterapia , Inflamación
2.
Elife ; 102021 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-34378531

RESUMEN

Maintenance of immune homeostasis involves a synergistic relationship between the host and the microbiome. Canonical interferon (IFN) signaling controls responses to acute microbial infection, through engagement of the STAT1 transcription factor. However, the contribution of tonic levels of IFN to immune homeostasis in the absence of acute infection remains largely unexplored. We report that STAT1 KO mice spontaneously developed an inflammatory disease marked by myeloid hyperplasia and splenic accumulation of hematopoietic stem cells. Moreover, these animals developed inflammatory bowel disease. Profiling gut bacteria revealed a profound dysbiosis in the absence of tonic IFN signaling, which triggered expansion of TH17 cells and loss of splenic Treg cells. Reduction of bacterial load by antibiotic treatment averted the TH17 bias and blocking IL17 signaling prevented myeloid expansion and splenic stem cell accumulation. Thus, tonic IFNs regulate gut microbial ecology, which is crucial for maintaining physiologic immune homeostasis and preventing inflammation.


Asunto(s)
Disbiosis/inmunología , Microbioma Gastrointestinal , Inflamación/genética , Interferones/administración & dosificación , Interleucina-17/genética , Factor de Transcripción STAT1/genética , Animales , Femenino , Interleucina-17/metabolismo , Ratones , Ratones Noqueados , Factor de Transcripción STAT1/metabolismo
3.
J Immunother Cancer ; 8(1)2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32273347

RESUMEN

BACKGROUND: Type 1 conventional dendritic cells (cDC1s) possess efficient antigen presentation and cross-presentation activity, as well as potent T cell priming ability. Tissue-resident cDC1s (CD103+ cDC1s in mice, CD141+ cDC1s in humans) are linked with improved tumor control, yet the efficacy of immunotherapy using this population is understudied. METHODS: We generated murine CD103+ cDC1s in vitro and examined their expression of cDC1-related factors, antigen cross-presentation activity, and accumulation in tumor-draining lymph nodes (TdLNs). The antitumor efficacy of the in vitro-generated CD103+ cDC1s was studied in murine melanoma and osteosarcoma models. We evaluated tumor responses on vaccination with CD103+ cDC1s, compared these to vaccination with monocyte-derived DCs (MoDCs), tested CD103+ cDC1 vaccination with checkpoint blockade, and examined the antimetastatic activity of CD103+ cDC1s. RESULTS: In vitro-generated CD103+ cDC1s produced cDC1-associated factors such as interleukin-12p70 and CXCL10, and demonstrated antigen cross-presentation activity on stimulation with the toll-like receptor 3 agonist polyinosinic:polycytidylic acid (poly I:C). In vitro-generated CD103+ cDC1s also migrated to TdLNs following poly I:C treatment and intratumoral delivery. Vaccination with poly I:C-activated and tumor antigen-loaded CD103+ cDC1s enhanced tumor infiltration of tumor antigen-specific and interferon-γ+ CD8+ T cells, and suppressed melanoma and osteosarcoma growth. CD103+ cDC1s showed superior antitumor efficacy compared with MoDC vaccination, and led to complete regression of 100% of osteosarcoma tumors in combination with CTLA-4 antibody-mediated checkpoint blockade. In vitro-generated CD103+ cDC1s effectively protected mice from pulmonary melanoma and osteosarcoma metastases. CONCLUSIONS: Our data indicate an in vitro-generated CD103+ cDC1 vaccine elicits systemic and long-lasting tumor-specific T cell-mediated cytotoxicity, which restrains primary and metastatic tumor growth. The CD103+ cDC1 vaccine was superior to MoDCs and enhanced response to immune checkpoint blockade. These results indicate the potential for new immunotherapies based on use of cDC1s alone or in combination with checkpoint blockade.


Asunto(s)
Antígenos CD/inmunología , Células Dendríticas/inmunología , Cadenas alfa de Integrinas/inmunología , Neoplasias Pulmonares/inmunología , Melanoma Experimental/inmunología , Osteosarcoma/inmunología , Sarcoma Experimental/inmunología , Vacunas/administración & dosificación , Animales , Presentación de Antígeno/inmunología , Antígenos CD/metabolismo , Antígenos de Neoplasias/inmunología , Neoplasias Óseas/inmunología , Neoplasias Óseas/patología , Neoplasias Óseas/terapia , Reactividad Cruzada , Células Dendríticas/trasplante , Inmunoterapia , Técnicas In Vitro , Cadenas alfa de Integrinas/metabolismo , Neoplasias Pulmonares/secundario , Neoplasias Pulmonares/terapia , Melanoma Experimental/patología , Melanoma Experimental/terapia , Ratones , Ratones Endogámicos C57BL , Osteosarcoma/patología , Osteosarcoma/terapia , Sarcoma Experimental/patología , Sarcoma Experimental/terapia , Células Tumorales Cultivadas
4.
Cancer Immunol Res ; 8(6): 794-805, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32213626

RESUMEN

Clinical response rates after adoptive cell therapy (ACT) are highly correlated with in vivo persistence of the infused T cells. However, antigen-specific T cells found in tumor sites are often well-differentiated effector cells with limited persistence. Central memory CD8+ T cells, capable of self-renewal, represent desirable ACT products. We report here that exposure to a histone deacetylase inhibitor (HDACi) and IL21 could reprogram differentiated human CD8+ T cells into central memory-like T cells. Dedifferentiation of CD8+ T cells was initiated by increased H3 acetylation and chromatin accessibility at the CD28 promoter region. This led to IL21-mediated pSTAT3 binding to the CD28 region, and subsequent upregulation of surface CD28 and CD62L (markers of central memory T cells). The reprogrammed cells exhibited enhanced proliferation in response to both IL2 and IL15, and a stable memory-associated transcriptional signature (increased Lef1 and Tcf7). Our findings support the application of IL21 and HDACi for the in vitro generation of highly persistent T-cell populations that can augment the efficacy of adoptively transferred T cells.


Asunto(s)
Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/inmunología , Diferenciación Celular , Inhibidores de Histona Desacetilasas/farmacología , Memoria Inmunológica/inmunología , Interleucinas/metabolismo , Linfocitos T/inmunología , Adolescente , Antígenos CD28/genética , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/metabolismo , Proliferación Celular , Células Cultivadas , Humanos , Memoria Inmunológica/efectos de los fármacos , Inmunoterapia Adoptiva , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo
5.
Cancers (Basel) ; 12(1)2020 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-31947933

RESUMEN

Conventional dendritic cells (cDCs) are a critical immune population, composed of multiple subsets, and responsible for controlling adaptive immunity and tolerance. Although migratory type 1 cDCs (CD103+ cDC1s in mice) are necessary to mount CD8+ T cell-mediated anti-tumor immunity, whether and how tumors modulate CD103+ cDC1 function remain understudied. Signal Transducer and Activator of Transcription 3 (STAT3) mediates the intracellular signaling of tumor-associated immunosuppressive cytokines, such as interleukin (IL)-10; thus, we hypothesized that STAT3 restrained anti-tumor immune responses elicited by CD103+ cDC1s. Herein, we show that in vitro-derived STAT3-deficient (Stat3∆/∆) CD103+ cDC1s are refractory to the inhibitory effects of IL-10 on Toll-like receptor 3 (TLR3) agonist-induced maturation responses. In a tumor vaccination approach, we found Stat3∆/∆ CD103+ cDC1s restrained mammary gland tumor growth and increased mouse survival more effectively than STAT3-sufficient CD103+ cDC1s. In addition, vaccination with Stat3∆/∆ CD103+ cDC1s elicited increased amounts of tumor antigen-specific CD8+ T cells and IFN-γ+ CD4+ T cells in tumors and tumor-draining lymph nodes versus phosphate-buffered saline (PBS)-treated animals. Furthermore, IL-10 receptor-deficient CD103+ cDC1s controlled tumor growth to a similar degree as Stat3∆/∆ CD103+ cDC1s. Taken together, our data reveal an inhibitory role for STAT3 in CD103+ cDC1 maturation and regulation of anti-tumor immunity. Our results also suggest IL-10 is a key factor eliciting immunosuppressive STAT3 signaling in CD103+ cDC1s in breast cancer. Thus, inhibition of STAT3 in cDC1s may provide an important strategy to improve their efficacy in tumor vaccination approaches and cDC1-mediated control of anti-tumor immunity.

6.
Cell Res ; 29(6): 474-485, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-31086255

RESUMEN

NF-κB, a family of transcription factors regulating diverse biological processes including immune responses, is activated by canonical and noncanonical pathways based on degradation of IκBα and processing of the IκB-like protein p100, respectively. Although p100 responds to noncanonical NF-κB stimuli for processing, it does not undergo degradation, but rather becomes accumulated, along with canonical NF-κB activation. We show here that the stability of p100 is tightly controlled by a deubiquitinase, Otub1. Otub1 deficiency not only promotes signal-induced p100 processing and noncanonical NF-κB activation but also causes steady-state p100 degradation, leading to aberrant NF-κB activation in the canonical pathway. B-cell-conditional deletion of Otub1 results in B-cell hyperplasia, antibody hyper-production, and lupus-like autoimmunity. Otub1-deficient B cells display aberrantly activated phenotypes and overproduce the cytokine IL-6, contributing to autoimmunity induction. Thus, maintenance of p100 stability by Otub1 serves as an unusual mechanism of NF-κB regulation that prevents autoimmunity.


Asunto(s)
Autoinmunidad , Cisteína Endopeptidasas/metabolismo , Proteínas I-kappa B/metabolismo , FN-kappa B/metabolismo , Animales , Células Cultivadas , Cisteína Endopeptidasas/deficiencia , Enzimas Desubicuitinizantes , Células HEK293 , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Estabilidad Proteica
7.
Leukemia ; 33(11): 2640-2653, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31028278

RESUMEN

CXCR5 mediates homing of both B and follicular helper T (TFH) cells into follicles of secondary lymphoid organs. We found that CXCR5+CD8+ T cells are present in human tonsils and follicular lymphoma, inhibit TFH-mediated B cell differentiation, and exhibit strong cytotoxic activity. Consistent with these findings, adoptive transfer of CXCR5+CD8+ T cells into an animal model of lymphoma resulted in significantly greater antitumor activity than CXCR5-CD8+ T cells. Furthermore, RNA-Seq-based transcriptional profiling revealed 77 differentially expressed genes unique to CXCR5+CD8+ T cells. Among these, a signature comprised of 33 upregulated genes correlated with improved survival in follicular lymphoma patients. We also showed that CXCR5+CD8+ T cells could be induced and expanded ex vivo using IL-23 plus TGF-ß, suggesting a possible strategy to generate these cells for clinical application. In summary, our study identified CXCR5+CD8+ T cells as a distinct T cell subset with ability to suppress TFH-mediated B cell differentiation, exert strong antitumor activity, and confer favorable prognosis in follicular lymphoma patients.


Asunto(s)
Linfocitos T CD8-positivos/citología , Receptores CXCR5/metabolismo , Subgrupos de Linfocitos T/citología , Traslado Adoptivo , Animales , Linfocitos B/citología , Diferenciación Celular , Técnicas de Cocultivo , Femenino , Centro Germinal/inmunología , Humanos , Leucocitos Mononucleares/citología , Activación de Linfocitos , Linfoma Folicular/inmunología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Tonsila Palatina/citología , Receptores de Antígenos de Linfocitos T/genética , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo
8.
Gastroenterology ; 157(1): 163-178, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-30885780

RESUMEN

BACKGROUND & AIMS: The peroxisome proliferator-activated receptor delta (PPARD) regulates cell metabolism, proliferation, and inflammation and has been associated with gastric and other cancers. Villin-positive epithelial cells are a small population of quiescent gastric progenitor cells. We expressed PPARD from a villin promoter to investigate the role of these cells and PPARD in development of gastric cancer. METHODS: We analyzed gastric tissues from mice that express the Ppard (PPARD1 and PPARD2 mice) from a villin promoter, and mice that did not carry this transgene (controls), by histology and immunohistochemistry. We performed cell lineage-tracing experiments and analyzed the microbiomes, chemokine and cytokine production, and immune cells and transcriptomes of stomachs of these mice. We also performed immunohistochemical analysis of PPARD levels in 2 sets of human gastric tissue microarrays. RESULTS: Thirty-eight percent of PPARD mice developed spontaneous, invasive gastric adenocarcinomas, with severe chronic inflammation. Levels of PPARD were increased in human gastric cancer tissues, compared with nontumor tissues, and associated with gastric cancer stage and grade. We found an inverse correlation between level of PPARD in tumor tissue and patient survival time. Gastric microbiomes from PPARD and control mice did not differ significantly. Lineage-tracing experiments identified villin-expressing gastric progenitor cells (VGPCs) as the origin of gastric tumors in PPARD mice. In these mice, PPARD up-regulated CCL20 and CXCL1, which increased infiltration of the gastric mucosa by immune cells. Immune cell production of inflammatory cytokines promoted chronic gastric inflammation and expansion and transformation of VGPCs, leading to tumorigenesis. We identified a positive-feedback loop between PPARD and interferon gamma signaling that sustained gastric inflammation to induce VGPC transformation and gastric carcinogenesis. CONCLUSIONS: We found PPARD overexpression in VPGCs to result in inflammation, dysplasia, and tumor formation. PPARD and VGPCs might be therapeutic targets for stomach cancer.


Asunto(s)
Carcinogénesis/genética , Transformación Celular Neoplásica/genética , Citocinas/inmunología , Mucosa Gástrica/metabolismo , Interferón gamma/inmunología , Receptores Citoplasmáticos y Nucleares/genética , Células Madre/metabolismo , Estómago/inmunología , Adenocarcinoma/genética , Adenocarcinoma/inmunología , Animales , Carcinogénesis/inmunología , Linaje de la Célula , Transformación Celular Neoplásica/inmunología , Quimiocina CCL20/metabolismo , Quimiocina CXCL1/metabolismo , Quimiocinas , Retroalimentación Fisiológica , Perfilación de la Expresión Génica , Inflamación , Ratones , Microbiota/inmunología , Proteínas de Microfilamentos/genética , Células Madre/inmunología , Estómago/microbiología , Neoplasias Gástricas/genética , Neoplasias Gástricas/inmunología
9.
Mol Immunol ; 110: 24-39, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-29549977

RESUMEN

Dendritic cells (DCs) are the principal antigen-presenting cells of the immune system and play key roles in controlling immune tolerance and activation. As such, DCs are chief mediators of tumor immunity. DCs can regulate tolerogenic immune responses that facilitate unchecked tumor growth. Importantly, however, DCs also mediate immune-stimulatory activity that restrains tumor progression. For instance, emerging evidence indicates the cDC1 subset has important functions in delivering tumor antigens to lymph nodes and inducing antigen-specific lymphocyte responses to tumors. Moreover, DCs control specific therapeutic responses in cancer including those resulting from immune checkpoint blockade. DC generation and function is influenced profoundly by cytokines, as well as their intracellular signaling proteins including STAT transcription factors. Regardless, our understanding of DC regulation in the cytokine-rich tumor microenvironment is still developing and must be better defined to advance cancer treatment. Here, we review literature focused on the molecular control of DCs, with a particular emphasis on cytokine- and STAT-mediated DC regulation. In addition, we highlight recent studies that delineate the importance of DCs in anti-tumor immunity and immune therapy, with the overall goal of improving knowledge of tumor-associated factors and intrinsic DC signaling cascades that influence DC function in cancer.


Asunto(s)
Diferenciación Celular/genética , Células Dendríticas/fisiología , Homeostasis , Inflamación , Neoplasias , Animales , Homeostasis/genética , Homeostasis/inmunología , Humanos , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología
10.
Proc Natl Acad Sci U S A ; 115(10): E2311-E2319, 2018 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-29463696

RESUMEN

Blood cell formation must be appropriately maintained throughout life to provide robust immune function, hemostasis, and oxygen delivery to tissues, and to prevent disorders that result from over- or underproduction of critical lineages. Persistent inflammation deregulates hematopoiesis by damaging hematopoietic stem and progenitor cells (HSPCs), leading to elevated myeloid cell output and eventual bone marrow failure. Nonetheless, antiinflammatory mechanisms that protect the hematopoietic system are understudied. The transcriptional regulator STAT3 has myriad roles in HSPC-derived populations and nonhematopoietic tissues, including a potent antiinflammatory function in differentiated myeloid cells. STAT3 antiinflammatory activity is facilitated by STAT3-mediated transcriptional repression of Ube2n, which encodes the E2 ubiquitin-conjugating enzyme Ubc13 involved in proinflammatory signaling. Here we demonstrate a crucial role for STAT3 antiinflammatory activity in preservation of HSPCs and lineage-balanced hematopoiesis. Conditional Stat3 removal from the hematopoietic system led to depletion of the bone marrow lineage- Sca-1+ c-Kit+ CD150+ CD48- HSPC subset (LSK CD150+ CD48- cells), myeloid-skewed hematopoiesis, and accrual of DNA damage in HSPCs. These responses were accompanied by intrinsic transcriptional alterations in HSPCs, including deregulation of inflammatory, survival and developmental pathways. Concomitant Ube2n/Ubc13 deletion from Stat3-deficient hematopoietic cells enabled lineage-balanced hematopoiesis, mitigated depletion of bone marrow LSK CD150+ CD48- cells, alleviated HSPC DNA damage, and corrected a majority of aberrant transcriptional responses. These results indicate an intrinsic protective role for STAT3 in the hematopoietic system, and suggest that this is mediated by STAT3-dependent restraint of excessive proinflammatory signaling via Ubc13 modulation.


Asunto(s)
Células Sanguíneas/inmunología , Hematopoyesis , Factor de Transcripción STAT3/inmunología , Animales , Células Sanguíneas/citología , Linaje de la Célula , Femenino , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Células Mieloides/inmunología , Factor de Transcripción STAT3/genética , Enzimas Ubiquitina-Conjugadoras/genética , Enzimas Ubiquitina-Conjugadoras/inmunología
11.
Exp Hematol ; 56: 7-15, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28911907

RESUMEN

MicroRNA-22 (miR-22) is a highly conserved microRNA that can regulate cell proliferation, oncogenesis, and cell maturation, especially during stress. In hematopoietic stem cells (HSCs), miR-22 has been reported to be involved in the regulation of key self-renewal factors, including Tet2. Recent work demonstrates that miR-22 also participates in regulation of the interferon (IFN) response, and expression profiling studies suggest that it is variably expressed at different stages in erythroid differentiation. We thus hypothesized that miR-22 regulates maturation of erythroid progenitors during stress hematopoiesis through its interaction with IFN. We compared the blood and bone marrow of wild-type (WT) and miR-22-deficient mice at baseline and upon infectious challenge with systemic lymphochoriomeningitis (LCMV) virus. miR-22-deficient mice maintained platelet counts better than WT mice during infection, but they showed significantly reduced red blood cells and hemoglobin. Analysis of bone marrow progenitors demonstrated better overall survival and improved HSC homeostasis in infected miR-22-null mice compared with WT, which was attributable to a blunted IFN response to LCMV challenge in the miR-22-null mice. We found that miR-22 was expressed exclusively in stage II erythroid precursors and downregulated upon infection in WT mice. Our results indicate that miR-22 promotes the IFN response to viral infection and that it functions at baseline as a brake to slow erythroid differentiation and maintain adequate erythroid potential. Impaired regulation of erythrogenesis in the absence of miR-22 can lead to anemia during infection.


Asunto(s)
Anemia/metabolismo , Células Precursoras Eritroides/metabolismo , Eritropoyesis , Interferón-alfa/metabolismo , Coriomeningitis Linfocítica/metabolismo , Virus de la Coriomeningitis Linfocítica/metabolismo , MicroARNs/metabolismo , Estrés Fisiológico , Anemia/genética , Anemia/virología , Animales , Células Precursoras Eritroides/patología , Perfilación de la Expresión Génica , Interferón-alfa/genética , Coriomeningitis Linfocítica/genética , Virus de la Coriomeningitis Linfocítica/genética , Ratones , Ratones Noqueados , MicroARNs/genética
12.
Oncotarget ; 8(23): 38008-38021, 2017 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-28402942

RESUMEN

Mesenchymal stem or stromal cells (MSCs) are non-hematopoietic stem cells that facilitate tissue regeneration through mechanisms involving self-renewal and differentiation, supporting angiogenesis and tissue cell survival, and limiting inflammation. MSCs were originally identified and expanded in long-term cultures of cells from bone marrow and other organs; and their native identity was recently confined into pericytes and adventitial cells in vascularized tissue. The multipotency, as well as the trophic and immunosuppressive effects, of MSCs have prompted the rapid development of clinical applications for many diseases involving tissue inflammation and immune disorders, including inflammatory bowel disease. Although standard criteria have been established to define MSCs, their therapeutic efficacy has varied significantly among studies due to their natural heterogenicity. Thus, understanding the biological and immunological features of MSCs is critical to standardize and optimize MSCs-based therapy. In this review, we highlight the cellular and molecular mechanisms involved in MSCs-mediated tissue repair and immunosuppression. We also provide an update on the current development of MSCs-based clinical trials, with a detailed discussion of MSC-based cell therapy in inflammatory bowel disease.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Enfermedades Inflamatorias del Intestino/terapia , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Humanos , Terapia de Inmunosupresión , Células Madre Mesenquimatosas/citología
13.
J Exp Med ; 214(5): 1493-1507, 2017 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-28356390

RESUMEN

Dendritic cells (DCs) are crucial for mediating immune responses but, when deregulated, also contribute to immunological disorders, such as autoimmunity. The molecular mechanism underlying the function of DCs is incompletely understood. In this study, we have identified TANK-binding kinase 1 (TBK1), a master innate immune kinase, as an important regulator of DC function. DC-specific deletion of Tbk1 causes T cell activation and autoimmune symptoms and also enhances antitumor immunity in animal models of cancer immunotherapy. The TBK1-deficient DCs have up-regulated expression of co-stimulatory molecules and increased T cell-priming activity. We further demonstrate that TBK1 negatively regulates the induction of a subset of genes by type I interferon receptor (IFNAR). Deletion of IFNAR1 could largely prevent aberrant T cell activation and autoimmunity in DC-conditional Tbk1 knockout mice. These findings identify a DC-specific function of TBK1 in the maintenance of immune homeostasis and tolerance.


Asunto(s)
Autoinmunidad/fisiología , Células Dendríticas/fisiología , Neoplasias/inmunología , Proteínas Serina-Treonina Quinasas/fisiología , Linfocitos T/fisiología , Animales , Homeostasis , Activación de Linfocitos/fisiología , Ratones , Ratones Noqueados , Neoplasias/fisiopatología , Factor de Transcripción STAT3/fisiología
14.
Sci Signal ; 9(447): ra94, 2016 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-27678219

RESUMEN

Despite the potent ability of dendritic cells (DCs) to stimulate lymphocyte responses and host immunity, granulocyte-macrophage colony-stimulating factor-derived DCs (GM-DCs) used as antitumor vaccines have demonstrated relatively modest success in cancer immunotherapy. We found that injecting GM-DCs into melanoma tumors in mice, or culturing GM-DCs with melanoma-secreted cytokines or melanoma-conditioned medium, rapidly suppressed DC-intrinsic expression of the gene encoding inhibitor of differentiation 2 (ID2), a transcriptional regulator. Melanoma-associated cytokines repressed Id2 transcription in murine DCs through the activation of signal transducer and activator of transcription 3 (STAT3). Enforced expression of ID2 in GM-DCs (ID2-GM-DCs) suppressed their production of the proinflammatory cytokine tumor necrosis factor-α (TNF-α). Vaccination with ID2-GM-DCs slowed the progression of melanoma tumors and enhanced animal survival, which was associated with an increased abundance of tumor-infiltrating interferon-γ-positive CD4(+) effector and CD8(+) cytotoxic T cells and a decreased number of tumor-infiltrating regulatory CD4(+) T cells. The efficacy of the ID2-GM-DC vaccine was improved by combinatorial treatment with a blocking antibody to programmed cell death protein-1 (PD-1), a current immunotherapy that overcomes suppressive immune checkpoint signaling. Collectively, our data reveal a previously unrecognized STAT3-mediated immunosuppressive mechanism in DCs and indicate that DC-intrinsic ID2 promotes tumor immunity by modulating tumor-associated CD4(+) T cell responses. Thus, inhibiting STAT3 or overexpressing ID2 selectively in DCs may improve the efficiency of DC vaccines in cancer therapy.


Asunto(s)
Células Dendríticas/inmunología , Inmunidad Celular , Proteína 2 Inhibidora de la Diferenciación/inmunología , Melanoma/inmunología , Factor de Transcripción STAT3/inmunología , Transducción de Señal/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/patología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Vacunas contra el Cáncer/genética , Vacunas contra el Cáncer/inmunología , Línea Celular Tumoral , Células Dendríticas/patología , Proteína 2 Inhibidora de la Diferenciación/genética , Melanoma/genética , Melanoma/patología , Melanoma/terapia , Ratones , Ratones Noqueados , Receptor de Muerte Celular Programada 1/genética , Receptor de Muerte Celular Programada 1/inmunología , Factor de Transcripción STAT3/genética , Transducción de Señal/genética
15.
Cytokine Growth Factor Rev ; 31: 1-15, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27185365

RESUMEN

The transcriptional regulator STAT3 has key roles in vertebrate development and mature tissue function including control of inflammation and immunity. Mutations in human STAT3 associate with diseases such as immunodeficiency, autoimmunity and cancer. Strikingly, however, either hyperactivation or inactivation of STAT3 results in human disease, indicating tightly regulated STAT3 function is central to health. Here, we attempt to summarize information on the numerous and distinct biological actions of STAT3, and highlight recent discoveries, with a specific focus on STAT3 function in the immune and hematopoietic systems. Our goal is to spur investigation on mechanisms by which aberrant STAT3 function drives human disease and novel approaches that might be used to modulate disease outcome.


Asunto(s)
Factor de Transcripción STAT3/inmunología , Inmunidad Adaptativa , Animales , Humanos , Enfermedades del Sistema Inmune/inmunología , Inmunidad Innata , Mutación , Factor de Transcripción STAT3/genética
16.
Eur J Haematol ; 97(3): 261-70, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26660446

RESUMEN

Heterozygous mutations in the transcriptional regulator GATA-2 associate with multilineage immunodeficiency, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML). The majority of these mutations localize in the zinc finger (ZnF) domains, which mediate GATA-2 DNA binding. Deregulated hematopoiesis with GATA-2 mutation frequently develops in adulthood, yet GATA-2 function in the bone marrow remains unresolved. To investigate this, we conditionally deleted the GATA-2 C-terminal ZnF (C-ZnF) coding sequences in adult mice. Upon Gata2 C-ZnF deletion, we observed rapid peripheral cytopenia, bone marrow failure, and decreased c-Kit expression on hematopoietic progenitors. Transplant studies indicated GATA-2 has a cell-autonomous role in bone marrow hematopoiesis. Moreover, myeloid lineage populations were particularly sensitive to Gata2 hemizygosity, while molecular assays indicated GATA-2 regulates c-Kit expression in multilineage progenitor cells. Enforced c-Kit expression in Gata2 C-ZnF-deficient hematopoietic progenitors enhanced myeloid colony activity, suggesting GATA-2 sustains myelopoiesis via a cell intrinsic role involving maintenance of c-Kit expression. Our results provide insight into mechanisms regulating hematopoiesis in bone marrow and may contribute to a better understanding of immunodeficiency and bone marrow failure associated with GATA-2 mutation.


Asunto(s)
Anemia Aplásica/genética , Enfermedades de la Médula Ósea/genética , Médula Ósea/patología , Factor de Transcripción GATA2/genética , Hemoglobinuria Paroxística/genética , Dominios y Motivos de Interacción de Proteínas/genética , Proteínas Proto-Oncogénicas c-kit/deficiencia , Eliminación de Secuencia , Dedos de Zinc/genética , Anemia Aplásica/diagnóstico , Anemia Aplásica/metabolismo , Anemia Aplásica/mortalidad , Animales , Biomarcadores , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Enfermedades de la Médula Ósea/diagnóstico , Enfermedades de la Médula Ósea/metabolismo , Enfermedades de la Médula Ósea/mortalidad , Trastornos de Fallo de la Médula Ósea , Huesos/patología , Inmunoprecipitación de Cromatina , Descalcificación Patológica/genética , Modelos Animales de Enfermedad , Factor de Transcripción GATA2/química , Factor de Transcripción GATA2/metabolismo , Expresión Génica , Regulación de la Expresión Génica , Frecuencia de los Genes , Genes Reporteros , Genotipo , Hematopoyesis/genética , Células Madre Hematopoyéticas/metabolismo , Hemoglobinuria Paroxística/diagnóstico , Hemoglobinuria Paroxística/metabolismo , Hemoglobinuria Paroxística/mortalidad , Secuenciación de Nucleótidos de Alto Rendimiento , Inmunofenotipificación , Ratones , Ratones Noqueados , Pronóstico , Células de Población Lateral
17.
Cancer Discov ; 6(2): 202-16, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26645196

RESUMEN

UNLABELLED: T cell-mediated immunotherapies are promising cancer treatments. However, most patients still fail to respond to these therapies. The molecular determinants of immune resistance are poorly understood. We show that loss of PTEN in tumor cells in preclinical models of melanoma inhibits T cell-mediated tumor killing and decreases T-cell trafficking into tumors. In patients, PTEN loss correlates with decreased T-cell infiltration at tumor sites, reduced likelihood of successful T-cell expansion from resected tumors, and inferior outcomes with PD-1 inhibitor therapy. PTEN loss in tumor cells increased the expression of immunosuppressive cytokines, resulting in decreased T-cell infiltration in tumors, and inhibited autophagy, which decreased T cell-mediated cell death. Treatment with a selective PI3Kß inhibitor improved the efficacy of both anti-PD-1 and anti-CTLA-4 antibodies in murine models. Together, these findings demonstrate that PTEN loss promotes immune resistance and support the rationale to explore combinations of immunotherapies and PI3K-AKT pathway inhibitors. SIGNIFICANCE: This study adds to the growing evidence that oncogenic pathways in tumors can promote resistance to the antitumor immune response. As PTEN loss and PI3K-AKT pathway activation occur in multiple tumor types, the results support the rationale to further evaluate combinatorial strategies targeting the PI3K-AKT pathway to increase the efficacy of immunotherapy.


Asunto(s)
Anticuerpos/administración & dosificación , Melanoma/tratamiento farmacológico , Melanoma/genética , Fosfohidrolasa PTEN/deficiencia , Linfocitos T/inmunología , Aminopiridinas/administración & dosificación , Aminopiridinas/uso terapéutico , Animales , Anticuerpos/uso terapéutico , Antígeno CTLA-4/inmunología , Línea Celular Tumoral , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Inmunoterapia/métodos , Melanoma/inmunología , Ratones , Morfolinas/administración & dosificación , Morfolinas/uso terapéutico , Receptor de Muerte Celular Programada 1/inmunología
18.
FASEB J ; 29(6): 2359-70, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25713055

RESUMEN

The IL-6/signal transducer and activator of transcription 3 (STAT3) pathway is a critical signaling pathway for colitis-associated colorectal cancer (CAC). Peroxisome proliferator-activated receptor (PPAR)-δ, a lipid nuclear receptor, up-regulates IL-6. 15-Lipoxygenase-1 (15-LOX-1), which is crucial to production of lipid signaling mediators to terminate inflammation, down-regulates PPAR-δ. 15-LOX-1 effects on IL-6/STAT3 signaling and CAC tumorigenesis have not been determined. We report that intestinally targeted transgenic 15-LOX-1 expression in mice inhibited azoxymethane- and dextran sodium sulfate-induced CAC, IL-6 expression, STAT3 phosphorylation, and IL-6/STAT3 downstream target (Notch3 and MUC1) expression. 15-LOX-1 down-regulation was associated with IL-6 up-regulation in human colon cancer mucosa. Reexpression of 15-LOX-1 in human colon cancer cells suppressed IL-6 mRNA expression, STAT3 phosphorylation, IL-6 promoter activity, and PPAR-δ mRNA and protein expression. PPAR-δ overexpression in colonic epithelial cells promoted CAC tumorigenesis in mice and increased IL-6 expression and STAT3 phosphorylation, whereas concomitant 15-LOX-1 expression in colonic epithelial cells (15-LOX-1-PPAR-δ-Gut mice) suppressed these effects: the number of tumors per mouse (mean ± sem) was 4.22 ± 0.68 in wild-type littermates, 6.67 ± 0.83 in PPAR-δ-Gut mice (P = 0.026), and 2.25 ± 0.25 in 15-LOX-1-PPAR-δ-Gut mice (P = 0.0006). Identification of 15-LOX-1 suppression of PPAR-δ to inhibit IL-6/STAT3 signaling-driven CAC tumorigenesis provides mechanistic insights that can be used to molecularly target CAC.


Asunto(s)
Araquidonato 15-Lipooxigenasa/metabolismo , Colitis/metabolismo , Neoplasias del Colon/metabolismo , Interleucina-6/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Araquidonato 15-Lipooxigenasa/genética , Azoximetano , Western Blotting , Células CACO-2 , Línea Celular Tumoral , Proliferación Celular/genética , Colitis/inducido químicamente , Colitis/genética , Neoplasias del Colon/inducido químicamente , Neoplasias del Colon/genética , Sulfato de Dextran , Expresión Génica , Células HCT116 , Humanos , Inmunohistoquímica , Interleucina-6/genética , Ratones Transgénicos , PPAR delta/genética , PPAR delta/metabolismo , Fosforilación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética
19.
Nat Commun ; 5: 5798, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25503582

RESUMEN

The transcriptional regulator STAT3 curbs pro-inflammatory cytokine production mediated by NF-κB signalling in innate immune cells, yet the mechanism by which this occurs has been unclear. Here we identify STAT3 as a pivotal negative regulator of Ubc13, an E2 ubiquitin-conjugating enzyme that facilitates TRAF6 K63-linked ubiquitination and NF-κB activation. Ubc13 accumulates intracellularly in the absence of STAT3. Depletion of Ubc13 in Stat3-deficient macrophages subdues excessive RANKL- or LPS-dependent gene expression, indicating that Ubc13 overexpression mediates enhanced transcriptional responses in the absence of STAT3. In RANKL-activated macrophages, STAT3 is stimulated by autocrine IL-6 and inhibits accrual of Ets-1, Set1 methyltransferase and trimethylation of histone H3 lysine 4 (H3K4me3) at the Ube2n (Ubc13) promoter. These results delineate a mechanism by which STAT3 operates as a transcriptional repressor on Ube2n, thus modulating NF-κB activity by regulation of Ubc13 abundance. Our data suggest that this pathway plays important roles in bone homeostasis and restraint of inflammation.


Asunto(s)
Fibroblastos/metabolismo , Macrófagos/metabolismo , Ligando RANK/farmacología , Factor de Transcripción STAT3/genética , Receptor Toll-Like 4/genética , Enzimas Ubiquitina-Conjugadoras/genética , Animales , Femenino , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Regulación de la Expresión Génica , Células HEK293 , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/genética , Histonas/metabolismo , Humanos , Interleucina-6/genética , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/citología , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Noqueados , FN-kappa B/genética , FN-kappa B/metabolismo , Cultivo Primario de Células , Proteína Proto-Oncogénica c-ets-1 , Ligando RANK/genética , Ligando RANK/metabolismo , Factor de Transcripción STAT3/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/genética , Factor 6 Asociado a Receptor de TNF/metabolismo , Receptor Toll-Like 4/metabolismo , Transcripción Genética , Enzimas Ubiquitina-Conjugadoras/antagonistas & inhibidores , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitinación
20.
Immunol Rev ; 261(1): 84-101, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25123278

RESUMEN

The term innate immunity typically refers to a quick but non-specific host defense response against invading pathogens. The innate immune system comprises particular immune cell populations, epithelial barriers, and numerous secretory mediators including cytokines, chemokines, and defense peptides. Innate immune cells are also now recognized to play important contributing roles in cancer and pathological inflammatory conditions. Innate immunity relies on rapid signal transduction elicited upon pathogen recognition via pattern recognition receptors (PRRs) and cell:cell communication conducted by soluble mediators, including cytokines. A majority of cytokines involved in innate immune signaling use a molecular cascade encompassing receptor-associated Jak protein tyrosine kinases and STAT (signal transducer and activator of transcription) transcriptional regulators. Here, we focus on roles for STAT proteins in three major innate immune subsets: neutrophils, macrophages, and dendritic cells (DCs). While knowledge in this area is only now emerging, understanding the molecular regulation of these cell types is necessary for developing new approaches to treat human disorders such as inflammatory conditions, autoimmunity, and cancer.


Asunto(s)
Células Dendríticas/inmunología , Enfermedades del Sistema Inmune/inmunología , Macrófagos/inmunología , Neoplasias/inmunología , Neutrófilos/inmunología , Factores de Transcripción STAT/metabolismo , Animales , Comunicación Celular/inmunología , Citocinas/metabolismo , Humanos , Inmunidad Innata , Quinasas Janus/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Factores de Transcripción STAT/genética , Transducción de Señal , Activación Transcripcional
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...