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











Base de dados
Intervalo de ano de publicação
1.
Clin Immunol ; 263: 110223, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38636890

RESUMO

Idiopathic severe aplastic anemia (SAA) is a disease of bone marrow failure caused by T-cell-induced destruction of hematopoietic stem and progenitor cells (HSPCs), however the mechanism remains unclear. We performed single-cell RNA sequencing of PBMCs and BMMCs from SAA patients and healthy donors and identified a CD8+ T cell subset with a tissue residency phenotype (Trm) in bone marrow that exhibit high IFN-γ and FasL expression and have a higher ability to induce apoptosis in HSPCs in vitro through FasL expression. CD8+ Trm cells were induced by IL-15 presented by IL-15Rα on monocytes, especially CD16+ monocytes, which were increased in SAA patients. CD16+ monocytes contributed to IL-15-induced CD38+CXCR6+ pre-Trm differentiation into CD8+ Trm cells, which can be inhibited by the CD38 inhibitor 78c. Our results demonstrate that IL-15-induced CD8+ Trm cells are pathogenic cells that mediate HSPC destruction in SAA patients and are therapeutic targets for future treatments.


Assuntos
Anemia Aplástica , Linfócitos T CD8-Positivos , Proteínas Ligadas por GPI , Células-Tronco Hematopoéticas , Interleucina-15 , Monócitos , Receptores de IgG , Humanos , Anemia Aplástica/imunologia , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/efeitos dos fármacos , Interleucina-15/farmacologia , Interleucina-15/imunologia , Receptores de IgG/metabolismo , Receptores de IgG/imunologia , Monócitos/imunologia , Monócitos/efeitos dos fármacos , Feminino , Masculino , Adulto , Células-Tronco Hematopoéticas/imunologia , Proteínas Ligadas por GPI/metabolismo , Proteínas Ligadas por GPI/imunologia , Pessoa de Meia-Idade , Proteína Ligante Fas/metabolismo , Proteína Ligante Fas/imunologia , Adulto Jovem , Adolescente , Interferon gama/imunologia , Interferon gama/metabolismo , Receptores de Interleucina-15/metabolismo , Receptores de Interleucina-15/imunologia , Apoptose/efeitos dos fármacos , Diferenciação Celular/imunologia
2.
Cancer Commun (Lond) ; 42(6): 536-557, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35615815

RESUMO

BACKGROUND: Interleukin-15 (IL-15) is a promising immunotherapeutic agent owing to its powerful immune-activating effects. However, the clinical benefits of these treatments are limited. Crosstalk between tumor cells and immune cells plays an important role in immune escape and immunotherapy drug resistance. Herein, this study aimed to obtain in-depth understanding of crosstalk in the tumor microenvironment for providing potential therapeutic strategies to prevent tumor progression. METHODS: T-cell killing assays and co-culture models were developed to determine the role of crosstalk between macrophages and tumor cells in breast cancer resistant to IL-15. Western blotting, histological analysis, CRISPR-Cas9 knockout, multi-parameter flow cytometry, and tumor cell-macrophage co-injection mouse models were developed to examine the mechanism by which IL-15Rα+ tumor-associated macrophages (TAMs) regulate breast cancer cell resistance to IL-15. RESULTS: We found that macrophages contributed to the resistance of tumor cells to IL-15, and tumor cells induced macrophages to express high levels of the α subunit of the IL-15 receptor (IL-15Rα). Further investigation showed that IL-15Rα+ TAMs reduced the protein levels of chemokine CX3C chemokine ligand 1 (CX3CL1) in tumor cells to inhibit the recruitment of CD8+ T cells by releasing the IL-15/IL-15Rα complex (IL-15Rc). Administration of an IL-15Rc blocking peptide markedly suppressed breast tumor growth and overcame the resistance of cancer cells to anti- programmed cell death protein 1 (PD-1) antibody immunotherapy. Interestingly, Granulocyte-macrophage colony-stimulating factor (GMCSF) induced γ chain (γc) expression to promote tumor cell-macrophage crosstalk, which facilitated tumor resistance to IL-15. Additionally, we observed that the non-transcriptional regulatory function of hypoxia inducible factor-1alpha (HIF-1α) was essential for IL-15Rc to regulate CX3CL1 expression in tumor cells. CONCLUSIONS: The IL-15Rc-HIF-1α-CX3CL1 signaling pathway serves as a crosstalk between macrophages and tumor cells in the tumor microenvironment of breast cancer. Targeting this pathway may provide a potential therapeutic strategy for enhancing the efficacy of cancer immunotherapy.


Assuntos
Neoplasias da Mama , Interleucina-15 , Animais , Neoplasias da Mama/terapia , Linfócitos T CD8-Positivos/metabolismo , Feminino , Humanos , Interleucina-15/metabolismo , Interleucina-15/farmacologia , Camundongos , Receptores de Interleucina-15/imunologia , Microambiente Tumoral , Macrófagos Associados a Tumor
4.
Clin Cancer Res ; 26(9): 2216-2230, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32019860

RESUMO

PURPOSE: We hypothesized that the combination of a local stimulus for activating tumor-specific T cells and an anti-immunosuppressant would improve treatment of gliomas. Virally encoded IL15Rα-IL15 as the T-cell activating stimulus and a prostaglandin synthesis inhibitor as the anti-immunosuppressant were combined with adoptive transfer of tumor-specific T cells. EXPERIMENTAL DESIGN: Two oncolytic poxviruses, vvDD vaccinia virus and myxoma virus, were each engineered to express the fusion protein IL15Rα-IL15 and a fluorescent protein. Viral gene expression (YFP or tdTomato Red) was confirmed in the murine glioma GL261 in vitro and in vivo. GL261 tumors in immunocompetent C57BL/6J mice were treated with vvDD-IL15Rα-YFP vaccinia virus or vMyx-IL15Rα-tdTr combined with other treatments, including vaccination with GARC-1 peptide (a neoantigen for GL261), rapamycin, celecoxib, and adoptive T-cell therapy. RESULTS: vvDD-IL15Rα-YFP and vMyx-IL15Rα-tdTr each infected and killed GL261 cells in vitro. In vivo, NK cells and CD8+ T cells were increased in the tumor due to the expression of IL15Rα-IL15. Each component of a combination treatment contributed to prolonging survival: an oncolytic virus, the IL15Rα-IL15 expressed by the virus, a source of T cells (whether by prevaccination or adoptive transfer), and prostaglandin inhibition all synergized to produce elimination of gliomas in a majority of mice. vvDD-IL15Rα-YFP occasionally caused ventriculitis-meningitis, but vMyx-IL15Rα-tdTr was safe and effective, causing a strong infiltration of tumor-specific T cells and eliminating gliomas in 83% of treated mice. CONCLUSIONS: IL15Rα-IL15-armed oncolytic poxviruses provide potent antitumor effects against brain tumors when combined with adoptive T-cell therapy, rapamycin, and celecoxib.


Assuntos
Neoplasias Encefálicas/terapia , Celecoxib/farmacologia , Sinergismo Farmacológico , Glioma/terapia , Imunoterapia/métodos , Terapia Viral Oncolítica/métodos , Sirolimo/farmacologia , Animais , Neoplasias Encefálicas/imunologia , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Terapia Combinada , Inibidores de Ciclo-Oxigenase 2/farmacologia , Modelos Animais de Doenças , Feminino , Glioma/imunologia , Glioma/metabolismo , Imunossupressores/farmacologia , Imunoterapia Adotiva , Interleucina-15/imunologia , Masculino , Camundongos Endogâmicos C57BL , Myxoma virus/genética , Myxoma virus/isolamento & purificação , Receptores de Interleucina-15/imunologia , Vaccinia virus/genética
5.
Immunology ; 159(2): 221-230, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31663113

RESUMO

Oxidized low-density lipoprotein (LDL) contributes to cardiovascular disease in part by mediating activation and maturation of monocytes and macrophages. Furthermore, co-localization studies using histochemical approaches have implicated a potential role for oxidized LDL as a mediator of interleukin-15 (IL-15) expression in myeloid cells of atherosclerotic plaque. The latter activity could be an important pro-inflammatory mechanism that mediates myeloid cell/T-cell crosstalk. Here, we examined the responses of primary human monocytes to highly oxidized LDL molecules. Oxidized LDL readily induced secretion of chemokines MCP-1 (CCL2) and GRO-α (CXCL1) but unlike lipopolysaccharide (LPS), has limited capacity to induce a variety of other cytokines including tumor necrosis factor-α, IL-6, IL-1ß and interferon-γ-induced protein-10 and also displayed a poor capacity to induce p-Akt or P-S6 signaling. Failure of oxidized LDL to induce IL-1ß secretion was associated with limited induction of caspase-1 activation. Furthermore, despite finding evidence that oxidized LDL could enhance the expression of IL-15 and IL-15 receptor expression in monocytes, we found no evidence that it could confer IL-15 transpresentation capability to these cells. This observation contrasted with induction of IL-15 transpresentation in lipopolysaccharide-stimulated monocytes. Overall, our data suggest that highly oxidized LDL is a selective inducer of monocyte activation. Sterile inflammatory mediators, particularly those implicated in Toll-like receptor 4 signaling, may play a role in vascular pathology but the activities of these agents are not uniform.


Assuntos
Interleucina-15/metabolismo , Interleucina-1beta/metabolismo , Lipoproteínas LDL/farmacologia , Monócitos/efeitos dos fármacos , Caspase 1/metabolismo , Células Cultivadas , Citocinas/imunologia , Citocinas/metabolismo , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interleucina-15/imunologia , Interleucina-1beta/imunologia , Lipopolissacarídeos/farmacologia , Monócitos/imunologia , Monócitos/metabolismo , Fosforilação , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores de Interleucina-15/imunologia , Receptores de Interleucina-15/metabolismo , Proteínas Quinases S6 Ribossômicas/metabolismo , Via Secretória , Transdução de Sinais , Linfócitos T/efeitos dos fármacos , Linfócitos T/imunologia , Linfócitos T/metabolismo
6.
Immunity ; 51(3): 479-490.e6, 2019 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-31402259

RESUMO

Natural killer (NK) cells are cytotoxic type 1 innate lymphoid cells (ILCs) that defend against viruses and mediate anti-tumor responses, yet mechanisms controlling their development and function remain incompletely understood. We hypothesized that the abundantly expressed microRNA-142 (miR-142) is a critical regulator of type 1 ILC biology. Interleukin-15 (IL-15) signaling induced miR-142 expression, whereas global and ILC-specific miR-142-deficient mice exhibited a cell-intrinsic loss of NK cells. Death of NK cells resulted from diminished IL-15 receptor signaling within miR-142-deficient mice, likely via reduced suppressor of cytokine signaling-1 (Socs1) regulation by miR-142-5p. ILCs persisting in Mir142-/- mice demonstrated increased expression of the miR-142-3p target αV integrin, which supported their survival. Global miR-142-deficient mice exhibited an expansion of ILC1-like cells concurrent with increased transforming growth factor-ß (TGF-ß) signaling. Further, miR-142-deficient mice had reduced NK-cell-dependent function and increased susceptibility to murine cytomegalovirus (MCMV) infection. Thus, miR-142 critically integrates environmental cues for proper type 1 ILC homeostasis and defense against viral infection.


Assuntos
Homeostase/imunologia , Imunidade Inata/imunologia , Linfócitos/imunologia , MicroRNAs/imunologia , Animais , Linhagem Celular , Feminino , Células HEK293 , Humanos , Células Matadoras Naturais/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Muromegalovirus/imunologia , Células NIH 3T3 , Receptores de Interleucina-15/imunologia , Transdução de Sinais/imunologia , Proteínas Supressoras da Sinalização de Citocina/imunologia , Fator de Crescimento Transformador beta/imunologia
7.
Arthritis Rheumatol ; 71(4): 529-541, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30407753

RESUMO

OBJECTIVE: To obtain the comprehensive transcriptome profile of human citrulline-specific B cells from patients with rheumatoid arthritis (RA). METHODS: Citrulline- and hemagglutinin-specific B cells were sorted by flow cytometry using peptide-streptavidin conjugates from the peripheral blood of RA patients and healthy individuals. The transcriptome profile of the sorted cells was obtained by RNA-sequencing, and expression of key protein molecules was evaluated by aptamer-based SOMAscan assay and flow cytometry. The ability of these proteins to effect differentiation of osteoclasts and proliferation and migration of synoviocytes was examined by in vitro functional assays. RESULTS: Citrulline-specific B cells, in comparison to citrulline-negative B cells, from patients with RA differentially expressed the interleukin-15 receptor α (IL-15Rα) gene as well as genes related to protein citrullination and cyclic AMP signaling. In analyses of an independent cohort of cyclic citrullinated peptide-seropositive RA patients, the expression of IL-15Rα protein was enriched in citrulline-specific B cells from the patients' peripheral blood, and surprisingly, all B cells from RA patients were capable of producing the epidermal growth factor ligand amphiregulin (AREG). Production of AREG directly led to increased migration and proliferation of fibroblast-like synoviocytes, and, in combination with anti-citrullinated protein antibodies, led to the increased differentiation of osteoclasts. CONCLUSION: To the best of our knowledge, this is the first study to document the whole transcriptome profile of autoreactive B cells in any autoimmune disease. These data identify several genes and pathways that may be targeted by repurposing several US Food and Drug Administration-approved drugs, and could serve as the foundation for the comparative assessment of B cell profiles in other autoimmune diseases.


Assuntos
Artrite Reumatoide/genética , Artrite Reumatoide/imunologia , Autoanticorpos/genética , Linfócitos B/imunologia , Transcriptoma/imunologia , Artrite Reumatoide/sangue , Autoanticorpos/imunologia , Citocinas/sangue , Citocinas/imunologia , Receptores ErbB/sangue , Receptores ErbB/imunologia , Humanos , Leucócitos Mononucleares/imunologia , Receptores de Interleucina-15/sangue , Receptores de Interleucina-15/imunologia , Análise de Sequência de RNA , Transdução de Sinais/imunologia
8.
Proc Natl Acad Sci U S A ; 116(2): 599-608, 2019 01 08.
Artigo em Inglês | MEDLINE | ID: mdl-30587590

RESUMO

Previous studies have provided evidence that IL-15 expression within human tumors is crucial for optimal antitumor responses; however, the regulation of IL-15 within the tumor microenvironment (TME) is unclear. We report herein, in analyses of mice implanted with various tumor cell lines, soluble IL-15/IL-15Rα complexes (sIL-15 complexes) are abundant in the interstitial fluid of tumors with expression preceding the infiltration of tumor-infiltrating lymphocytes. Moreover, IL-15 as well as type I IFN, which regulates IL-15, was required for establishing normal numbers of CD8 T cells and natural killer cells in tumors. Depending on tumor type, both the tumor and the stroma are sources of sIL-15 complexes. In analyses of IL-15 reporter mice, most myeloid cells in the TME express IL-15 with CD11b+Ly6Chi cells being the most abundant, indicating there is a large source of IL-15 protein in tumors that lies sequestered within the tumor stroma. Despite the abundance of IL-15-expressing cells, the relative levels of sIL-15 complexes are low in advanced tumors but can be up-regulated by local stimulator of IFN genes (STING) activation. Furthermore, while treatment of tumors with STING agonists leads to tumor regression, optimal STING-mediated immunity and regression of distant secondary tumors required IL-15 expression. Overall, our study reveals the dynamic regulation of IL-15 in the TME and its importance in antitumor immunity. These findings provide insight into an unappreciated attribute of the tumor landscape that contributes to antitumor immunity, which can be manipulated therapeutically to enhance antitumor responses.


Assuntos
Regulação Neoplásica da Expressão Gênica/imunologia , Interleucina-15/imunologia , Melanoma/imunologia , Proteínas de Neoplasias/imunologia , Microambiente Tumoral/imunologia , Animais , Linhagem Celular Tumoral , Inflamação/genética , Inflamação/imunologia , Inflamação/patologia , Interleucina-15/genética , Melanoma/genética , Melanoma/patologia , Proteínas de Membrana/genética , Proteínas de Membrana/imunologia , Camundongos , Camundongos Knockout , Proteínas de Neoplasias/genética , Receptores de Interleucina-15/genética , Receptores de Interleucina-15/imunologia , Microambiente Tumoral/genética
9.
Cancer Immunol Immunother ; 67(4): 675-689, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29392336

RESUMO

Natural killer (NK) cells are innate cytotoxic lymphocytes that play a fundamental role in the immunosurveillance of cancers. NK cells of cancer patients exhibit impaired function mediated by immunosuppressive factors released from the tumor microenvironment (TME), such as transforming growth factor (TGF)-ß1. An interleukin (IL)-15 superagonist/IL-15 receptor α fusion complex (IL-15SA/IL-15RA; ALT-803) activates the IL-15 receptor on CD8 T cells and NK cells, and has shown significant anti-tumor activity in several in vivo studies. This in vitro study investigated the efficacy of IL-15SA/IL-15RA on TGF-ß1-induced suppression of NK cell-cytotoxic function. IL-15SA/IL-15RA inhibited TGF-ß1 from decreasing NK cell lysis of four of four tumor cell lines (H460, LNCap, MCF7, MDA-MB-231). IL-15SA/IL-15RA rescued healthy donor and cancer patient NK cell-cytotoxicity, which had previously been suppressed by culture with TGF-ß1. TGF-ß1 downregulated expression of NK cell-activating markers and cytotoxic granules, such as CD226, NKG2D, NKp30, granzyme B, and perforin. Smad2/3 signaling was responsible for this TGF-ß1-induced downregulation of NK cell-activating markers and cytotoxic granules. IL-15SA/IL-15RA blocked Smad2/3-induced transcription, resulting in the rescue of NK cell-cytotoxic function from TGF-ß1-induced suppression. These findings suggest that in addition to increasing NK cell function via promoting the IL-15 signaling pathway, IL-15SA/IL-15RA can function as an inhibitor of TGF-ß1 signaling, providing a potential remedy for NK cell dysfunction in the immunosuppressive tumor microenvironment.


Assuntos
Anticorpos Monoclonais/administração & dosagem , Citotoxicidade Imunológica/imunologia , Interleucina-15/imunologia , Células Matadoras Naturais/imunologia , Neoplasias/imunologia , Receptores de Interleucina-15/imunologia , Proteínas Recombinantes de Fusão/imunologia , Fator de Crescimento Transformador beta1/farmacologia , Citotoxicidade Celular Dependente de Anticorpos , Humanos , Terapia de Imunossupressão , Células Matadoras Naturais/efeitos dos fármacos , Ativação Linfocitária , Neoplasias/terapia , Células Tumorais Cultivadas
10.
Cytokine ; 92: 68-74, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28110119

RESUMO

Astrocytes play a pivotal role in neuronal survival under the condition of post-ischemic brain inflammation, but the relevant astrocyte-derived mediators of ischemic brain injury remain to be defined. IL-15 supports survival of multiple lymphocyte lineages in the peripheral immune system, but the role of IL-15 in inflammatory disease of the central nervous system is not well defined. Recent research has shown an increase of IL-15-expressing astrocytes in the ischemic brain. Since astrocytes promote neuron survival under cerebral ischemia by buffering excess extracellular glutamate and producing growth factors, recovery of astrocyte function could be of benefit for stroke therapy. Here, we report that IL-15 is the pro-survival cytokine that prevents astrocyte death from oxygen glucose deprivation (OGD)-induced damage. Astrocytes up-regulate expression of the IL-15/IL-15Rα complex under OGD, whereas OGD down-regulates the levels of pSTAT5 and pAkt in astrocytes. IL-15 treatment ameliorates the decline of pAkt, decreases the percentage of annexin V+ cells, inhibits the activation of caspase-3, and activates the Akt pathway to promote astrocyte survival in response to OGD. We further identified that activation of Akt, but not PKCα/ßI, is essential for astrocyte survival under OGD. Taken together, this study reveals the function of IL-15 in astrocyte survival via Akt phosphorylation in response to OGD-induced damage.


Assuntos
Astrócitos/imunologia , Interleucina-15/farmacologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Animais , Anexina A5/imunologia , Morte Celular/imunologia , Hipóxia Celular/imunologia , Ativação Enzimática/efeitos dos fármacos , Ativação Enzimática/imunologia , Glucose/imunologia , Interleucina-15/imunologia , Camundongos , Oxigênio/imunologia , Receptores de Interleucina-15/imunologia , Fator de Transcrição STAT5/imunologia
11.
Clin Cancer Res ; 23(11): 2817-2830, 2017 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-27986749

RESUMO

Purpose: Adoptive cell transfer (ACT) is a promising immunotherapeutic approach for cancer. Host lymphodepletion is associated with favorable ACT therapy outcomes, but it may cause detrimental effects in humans. We tested the hypothesis that IL15 administration enhances ACT in the absence of lymphodepletion. We previously showed that bioactive IL15 in vivo comprises a stable complex of the IL15 chain with the IL15 receptor alpha chain (IL15Rα), termed heterodimeric IL15 (hetIL15).Experimental Design: We evaluated the effects of the combination regimen ACT + hetIL15 in the absence of lymphodepletion by transferring melanoma-specific Pmel-1 T cells into B16 melanoma-bearing mice.Results: hetIL15 treatment delayed tumor growth by promoting infiltration and persistence of both adoptively transferred Pmel-1 cells and endogenous CD8+ T cells into the tumor. In contrast, persistence of Pmel-1 cells was severely reduced following irradiation in comparison with mice treated with hetIL15. Importantly, we found that hetIL15 treatment led to the preferential enrichment of Pmel-1 cells in B16 tumor sites in an antigen-dependent manner. Upon hetIL15 administration, tumor-infiltrating Pmel-1 cells showed a "nonexhausted" effector phenotype, characterized by increased IFNγ secretion, proliferation, and cytotoxic potential and low level of PD-1. hetIL15 treatment also resulted in an improved ratio of Pmel-1 to Treg in the tumor.Conclusions: hetIL15 administration improves the outcome of ACT in lymphoreplete hosts, a finding with significant implications for improving cell-based cancer immunotherapy strategies. Clin Cancer Res; 23(11); 2817-30. ©2016 AACR.


Assuntos
Transferência Adotiva/métodos , Imunoterapia Adotiva , Melanoma Experimental/terapia , Animais , Linfócitos T CD8-Positivos/imunologia , Linhagem Celular Tumoral , Humanos , Ativação Linfocitária/efeitos dos fármacos , Ativação Linfocitária/imunologia , Melanoma Experimental/imunologia , Camundongos , Receptores de Interleucina-15/genética , Receptores de Interleucina-15/imunologia , Linfócitos T Reguladores/efeitos dos fármacos , Linfócitos T Reguladores/imunologia
12.
Cytokine ; 82: 95-101, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26778709

RESUMO

IL-15 is a member of the gamma chain family of cytokines (γc - CD132). The IL-15 receptor (IL-15R) complex consists of 3 subunits: the ligand-binding IL-15Rα chain (CD215), the ß chain (CD122; also used by IL-2), and the common γ chain. The biological activities of IL-15 are mostly mediated by the IL-15:IL-15Rα complex, produced by the same cell and 'trans-presented' to responder cells expressing the IL-15Rßγc. The peculiar and almost unique requirement for IL-15 to be trans-presented by IL-15Rα suggests that the biological effects of IL-15 signaling are tightly regulated even at the level of availability of IL-15. Tissue-specific deletion of IL-15Rα has shown macrophage-and dendritic cell-derived IL-15Rα mediate the homeostasis of different CD8(+) T cell subsets. Here we show that hepatocyte and macrophage- specific expression of IL-15Rα is required to maintain the homeostasis of NK and NKT cells in the liver. Thus, homeostasis of IL-15-dependent lymphocyte subsets is also regulated by trans-presentation of IL-15 by non-hematopoietic cells in the tissue environment.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Homeostase/imunologia , Interleucina-15/imunologia , Fígado/imunologia , Transdução de Sinais/imunologia , Animais , Células Dendríticas/imunologia , Homeostase/genética , Interleucina-15/genética , Macrófagos/imunologia , Camundongos , Camundongos Knockout , Receptores de Interleucina-15/genética , Receptores de Interleucina-15/imunologia , Transdução de Sinais/genética
13.
J Biol Chem ; 291(17): 8931-50, 2016 Apr 22.
Artigo em Inglês | MEDLINE | ID: mdl-26719339

RESUMO

Here we report a "configuration-dependent" mechanism of action for IL-15:IL-15Rα (heterodimeric IL-15 or hetIL-15) where the manner by which IL-15:IL-15Rα molecules are presented to target cells significantly affects its function as a vaccine adjuvant. Although the cellular mechanism of IL-15 trans-presentation via IL-15Rα and its importance for IL-15 function have been described, the full effect of the IL-15:IL-15Rα configuration on responding cells is not yet known. We found that trans-presenting IL-15:IL-15Rα in a multivalent fashion on the surface of antigen-encapsulating nanoparticles enhanced the ability of nanoparticle-treated dendritic cells (DCs) to stimulate antigen-specific CD8(+) T cell responses. Localization of multivalent IL-15:IL-15Rα and encapsulated antigen to the same DC led to maximal T cell responses. Strikingly, DCs incubated with IL-15:IL-15Rα-coated nanoparticles displayed higher levels of functional IL-15 on the cell surface, implicating a mechanism for nanoparticle-mediated transfer of IL-15 to the DC surface. Using artificial antigen-presenting cells to highlight the effect of IL-15 configuration on DCs, we showed that artificial antigen-presenting cells presenting IL-15:IL-15Rα increased the sensitivity and magnitude of the T cell response, whereas IL-2 enhanced the T cell response only when delivered in a paracrine fashion. Therefore, the mode of cytokine presentation (configuration) is important for optimal immune responses. We tested the effect of configuration dependence in an aggressive model of murine melanoma and demonstrated significantly delayed tumor progression induced by IL-15:IL-15Rα-coated nanoparticles in comparison with monovalent IL-15:IL-15Rα. The novel mechanism of IL-15 transfer to the surface of antigen-processing DCs may explain the enhanced potency of IL-15:IL-15Rα-coated nanoparticles for antigen delivery.


Assuntos
Apresentação de Antígeno/efeitos dos fármacos , Antígenos de Neoplasias , Linfócitos T CD8-Positivos/imunologia , Materiais Revestidos Biocompatíveis/farmacologia , Células Dendríticas/imunologia , Imunidade Celular/efeitos dos fármacos , Interleucina-15 , Nanopartículas , Neoplasias Experimentais , Receptores de Interleucina-15/imunologia , Animais , Antígenos de Neoplasias/imunologia , Antígenos de Neoplasias/farmacologia , Humanos , Interleucina-15/imunologia , Interleucina-15/farmacologia , Camundongos , Neoplasias Experimentais/tratamento farmacológico , Neoplasias Experimentais/imunologia , Neoplasias Experimentais/patologia
14.
J Immunol ; 195(10): 4810-21, 2015 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-26453750

RESUMO

IL-15 bound to the IL-15Rα-chain (IL-15Rα) is presented in trans to cells bearing the IL-2Rß-chain and common γ-chain. As IL-15 transpresentation occurs in the context of cell-to-cell contacts, it has the potential for regulation by and of other receptor-ligand interactions. In this study, human NK cells were tested for the sensitivity of IL-15 transpresentation to inhibitory receptors. Human cells expressing HLA class I ligands for inhibitory receptors KIR2DL1, KIR2DL2/3, or CD94-NKG2A were transfected with IL-15Rα. Proliferation of primary NK cells in response to transpresented IL-15 was reduced by engagement of either KIR2DL1 or KIR2DL2/3 by cognate HLA-C ligands. Inhibitory KIR-HLA-C interactions did not reduce the proliferation induced by soluble IL-15. Therefore, transpresentation of IL-15 is subject to downregulation by MHC class I-specific inhibitory receptors. Similarly, proliferation of the NKG2A(+) cell line NKL induced by IL-15 transpresentation was inhibited by HLA-E. Coengagement of inhibitory receptors, either KIR2DL1 or CD94-NKG2A, did not inhibit phosphorylation of Stat5 but inhibited selectively phosphorylation of Akt and S6 ribosomal protein. IL-15Rα was not excluded from, but was evenly distributed across, inhibitory synapses. These findings demonstrate a novel mechanism to attenuate IL-15-dependent NK cell proliferation and suggest that inhibitory NK cell receptors contribute to NK cell homeostasis.


Assuntos
Proliferação de Células/fisiologia , Interleucina-15/imunologia , Células Matadoras Naturais/imunologia , Subfamília C de Receptores Semelhantes a Lectina de Células NK/imunologia , Subfamília D de Receptores Semelhantes a Lectina de Células NK/imunologia , Receptores KIR2DL1/imunologia , Feminino , Antígenos HLA-C/imunologia , Humanos , Subunidade beta de Receptor de Interleucina-2/imunologia , Células Matadoras Naturais/citologia , Masculino , Fosforilação/imunologia , Proteínas Proto-Oncogênicas c-akt/imunologia , Receptores de Interleucina-15/imunologia , Receptores KIR2DL2/imunologia , Receptores KIR2DL3/imunologia
15.
Cancer Immunol Res ; 3(3): 219-27, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25736261

RESUMO

IL2 and IL15, members of the 4α-helix bundle family of cytokines, play pivotal roles in the control of the life and death of lymphocytes. Although their heterotrimeric receptors have two receptor subunits in common, these two cytokines have contrasting roles in adaptive immune responses. The unique role of IL2 through maintenance of fitness of regulatory T cells and activation-induced cell death is the elimination of self-reactive T cells to prevent autoimmunity. In contrast with IL2, IL15 is dedicated to the prolonged maintenance of memory T-cell responses to invading pathogens. Blockade of IL2 and IL15 using monoclonal antibodies has been reported to be of value in the treatment of patients with leukemia, autoimmune disorders, and in the prevention of allograft rejection. IL2 has been approved by the FDA for the treatment of patients with malignant renal cell cancer and metastatic malignant melanoma. Clinical trials involving recombinant human IL15 given by bolus infusions have been completed, and studies assessing subcutaneous and continuous intravenous infusions are under way in patients with metastatic malignancy. Furthermore, clinical trials are being initiated that employ the combination of IL15 with IL15Rα(+/-) IgFc.


Assuntos
Carcinoma de Células Renais/tratamento farmacológico , Interleucina-15/imunologia , Interleucina-2/imunologia , Linfócitos/fisiologia , Melanoma/tratamento farmacológico , Animais , Anticorpos Monoclonais/uso terapêutico , Ensaios Clínicos como Assunto , Humanos , Imunoterapia , Interleucina-15/genética , Interleucina-15/uso terapêutico , Interleucina-2/genética , Interleucina-2/uso terapêutico , Neoplasias/tratamento farmacológico , Receptores de Interleucina-15/imunologia , Receptores de Interleucina-2/imunologia , Transdução de Sinais , Linfócitos T Reguladores/imunologia
16.
Immunity ; 41(2): 230-43, 2014 Aug 21.
Artigo em Inglês | MEDLINE | ID: mdl-25148024

RESUMO

CD8αα(+) intraepithelial lymphocytes (IELs) are instrumental in maintaining the epithelial barrier in the intestine. Similar to natural killer cells and other innate lymphoid cells, CD8αα(+) IELs constitutively express the T-box transcription factor T-bet. However, the precise role of T-bet for the differentiation or function of IELs is unknown. Here we show that mice genetically deficient for T-bet lacked both TCRαß(+) and TCRγδ(+) CD8αα(+) IELs and thus are more susceptible to chemically induced colitis. Although T-bet was induced in thymic IEL precursors (IELPs) as a result of agonist selection and interleukin-15 (IL-15) receptor signaling, it was dispensable for the generation of IELPs. Subsequently, T-bet was required for the IL-15-dependent activation, differentiation, and expansion of IELPs in the periphery. Our study reveals a function of T-bet as a central transcriptional regulator linking agonist selection and IL-15 signaling with the emergence of CD8αα(+) IELs.


Assuntos
Antígenos CD8/biossíntese , Interleucina-15/genética , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Receptores de Antígenos de Linfócitos T gama-delta/genética , Proteínas com Domínio T/genética , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/imunologia , Diferenciação Celular/imunologia , Colite/induzido quimicamente , Colite/imunologia , Células Epiteliais/imunologia , Interleucina-15/imunologia , Mucosa Intestinal/citologia , Mucosa Intestinal/imunologia , Intestinos/citologia , Intestinos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interleucina-15/imunologia , Transdução de Sinais/imunologia , Proteínas com Domínio T/biossíntese
17.
Cancer Res ; 74(17): 4908-21, 2014 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-24980552

RESUMO

Despite its aggressive nature, triple-negative breast cancer (TNBC) often exhibits leucocyte infiltrations that correlate with favorable prognosis. In this study, we offer an explanation for this apparent conundrum by defining TNBC cell subsets that overexpress the IL15 immune receptor IL15RA. This receptor usually forms a heterotrimer with the IL2 receptors IL2RB and IL2RG, which regulates the proliferation and differentiation of cytotoxic T cells and NK cells. However, unlike IL15RA, the IL2RB and IL2RG receptors are not upregulated in basal-like TNBC breast cancer cells that express IL15RA. Mechanistic investigations indicated that IL15RA signaling activated JAK1, STAT1, STAT2, AKT, PRAS40, and ERK1/2 in the absence of IL2RB and IL2RG, whereas neither STAT5 nor JAK2 were activated. RNAi-mediated attenuation of IL15RA established its role in cell growth, apoptosis, and migration, whereas expression of the IL15 cytokine in IL15RA-expressing cells stimulated an autocrine signaling cascade that promoted cell proliferation and migration and blocked apoptosis. Notably, coexpression of IL15RA and IL15 was also sufficient to activate peripheral blood mononuclear cells upon coculture in a paracrine signaling manner. Overall, our findings offer a mechanistic explanation for the paradoxical association of some high-grade breast tumors with better survival outcomes, due to engagement of the immune stroma.


Assuntos
Linfócitos/imunologia , Receptores de Interleucina-15/genética , Receptores de Interleucina-15/imunologia , Neoplasias de Mama Triplo Negativas/genética , Neoplasias de Mama Triplo Negativas/imunologia , Animais , Apoptose/genética , Apoptose/imunologia , Linhagem Celular Tumoral , Movimento Celular/genética , Movimento Celular/imunologia , Proliferação de Células/genética , Feminino , Humanos , Interleucina-15/genética , Interleucina-15/imunologia , Leucócitos Mononucleares/imunologia , Camundongos , Comunicação Parácrina/genética , Comunicação Parácrina/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia
18.
Clin Cancer Res ; 20(8): 2044-50, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24737791

RESUMO

Interleukin-15 (IL-15) is a proinflammatory cytokine involved in the development, survival, proliferation, and activation of multiple lymphocyte lineages utilizing a variety of signaling pathways. IL-15 utilizes three distinct receptor chains in at least two different combinations to signal and exert its effects on the immune system. The binding of IL-15 to its receptor complex activates an "immune-enhancing" signaling cascade in natural killer cells and subsets of T cells, as well as the induction of a number of proto-oncogenes. Additional studies have explored the role of IL-15 in the development and progression of cancer, notably leukemia of large granular lymphocytes, cutaneous T-cell lymphoma, and multiple myeloma. This review provides an overview of the molecular events in the IL-15 signaling pathway and the aberrancies in its regulation that are associated with chronic inflammation and cancer. We briefly explore the potential therapeutic opportunities that have arisen as a result of these studies to further the treatment of cancer. These involve both targeting the disruption of IL-15 signaling as well as IL-15-mediated enhancement of innate and antigen-specific immunity.


Assuntos
Imunidade Inata/imunologia , Interleucina-15/imunologia , Linfócitos/imunologia , Neoplasias/imunologia , Transdução de Sinais/imunologia , Células Apresentadoras de Antígenos/imunologia , Células Apresentadoras de Antígenos/metabolismo , Humanos , Inflamação/imunologia , Inflamação/metabolismo , Interleucina-15/metabolismo , Linfócitos/metabolismo , Modelos Imunológicos , Neoplasias/metabolismo , Receptores de Interleucina-15/imunologia , Receptores de Interleucina-15/metabolismo
19.
J Immunol ; 192(6): 2659-66, 2014 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-24510964

RESUMO

In the thymus, interactions with both cortical and medullary microenvironments regulate the development of self-tolerant conventional CD4(+) and CD8(+) αßT cells expressing a wide range of αßTCR specificities. Additionally, the cortex is also required for the development of invariant NKT (iNKT) cells, a specialized subset of T cells that expresses a restricted αßTCR repertoire and is linked to the regulation of innate and adaptive immune responses. Although the role of the cortex in this process is to enable recognition of CD1d molecules expressed by CD4(+)CD8(+) thymocyte precursors, the requirements for additional thymus microenvironments during iNKT cell development are unknown. In this study, we reveal a role for medullary thymic epithelial cells (mTECs) during iNKT cell development in the mouse thymus. This requirement for mTECs correlates with their expression of genes required for IL-15 trans-presentation, and we show that soluble IL-15/IL-15Rα complexes restore iNKT cell development in the absence of mTECs. Furthermore, mTEC development is abnormal in iNKT cell-deficient mice, and early stages in iNKT cell development trigger receptor activator for NF-κB ligand-mediated mTEC development. Collectively, our findings demonstrate that intrathymic iNKT cell development requires stepwise interactions with both the cortex and the medulla, emphasizing the importance of thymus compartmentalization in the generation of both diverse and invariant αßT cells. Moreover, the identification of a novel requirement for iNKT cells in thymus medulla development further highlights the role of both innate and adaptive immune cells in thymus medulla formation.


Assuntos
Diferenciação Celular/imunologia , Microambiente Celular/imunologia , Células Epiteliais/imunologia , Células T Matadoras Naturais/imunologia , Animais , Antígenos CD1d/genética , Antígenos CD1d/imunologia , Antígenos CD1d/metabolismo , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Microambiente Celular/efeitos dos fármacos , Microambiente Celular/genética , Células Epiteliais/metabolismo , Células Epiteliais/transplante , Citometria de Fluxo , Interleucina-15/administração & dosagem , Interleucina-15/genética , Interleucina-15/imunologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células T Matadoras Naturais/metabolismo , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/genética , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/imunologia , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Ligante RANK/imunologia , Ligante RANK/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/imunologia , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Receptores de Interleucina-15/administração & dosagem , Receptores de Interleucina-15/genética , Receptores de Interleucina-15/imunologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Timócitos/citologia , Timócitos/imunologia , Timócitos/metabolismo , Timo/citologia , Timo/imunologia , Timo/metabolismo , Fator de Transcrição RelB/genética , Fator de Transcrição RelB/imunologia , Fator de Transcrição RelB/metabolismo
20.
Cytokine Growth Factor Rev ; 24(5): 455-66, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23791986

RESUMO

IL-15 is a member of the IL-2 family of cytokines whose signaling pathways are a bridge between innate and adaptive immune response. IL-15 is part of the intestinal mucosal barrier, and functions to modulate gut homeostasis. IL-15 has pivotal roles in the control of development, proliferation and survival of both innate and adaptive immune cells. IL-15 becomes up-regulated in the inflamed tissue of intestinal inflammatory disease, such as IBD, Celiac Disease and related complications. Indeed, several studies have reported that IL-15 may participate to the pathogenesis of these diseases. Furthermore, although IL-15 seems to be responsible for inflammation and autoimmunity, it also may increase the immune response against cancer. For these reasons, we decided to study the intestinal mucosa as an 'immunological niche', in which immune response, inflammation and local homeostasis are modulated. Understanding the role of the IL-15/IL-15R system will provide a scientific basis for the development of new approaches that use IL-15 for immunotherapy of autoimmune diseases and malignancies. Indeed, a better understanding of the complexity of the mucosal immune system will contribute to the general understanding of immuno-pathology, which could lead to new therapeutical tools for widespread immuno-mediated diseases.


Assuntos
Imunidade Adaptativa , Doença Celíaca/imunologia , Neoplasias Gastrointestinais/imunologia , Imunidade Inata , Doenças Inflamatórias Intestinais/imunologia , Interleucina-15/imunologia , Animais , Doença Celíaca/patologia , Neoplasias Gastrointestinais/patologia , Humanos , Imunidade nas Mucosas , Doenças Inflamatórias Intestinais/patologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/patologia , Receptores de Interleucina-15/imunologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA