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








Base de dados
Intervalo de ano de publicação
1.
Immune Netw ; 24(1): e2, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38455465

RESUMO

Studies over the last 2 decades have identified IL-17 and IL-21 as key cytokines in the modulation of a wide range of immune responses. IL-17 serves as a critical defender against bacterial and fungal pathogens, while maintaining symbiotic relationships with commensal microbiota. However, alterations in its levels can lead to chronic inflammation and autoimmunity. IL-21, on the other hand, bridges the adaptive and innate immune responses, and its imbalance is implicated in autoimmune diseases and cancer, highlighting its important role in both health and disease. Delving into the intricacies of these cytokines not only opens new avenues for understanding the immune system, but also promises innovative advances in the development of therapeutic strategies for numerous diseases. In this review, we will discuss an updated view of the immunobiology and therapeutic potential of IL-17 and IL-21.

2.
Int J Antimicrob Agents ; 63(3): 107082, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38163552

RESUMO

Global spread of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2) Omicron subvariants, such as BA.4, BA.5 and XBB.1.5, has been leading the recent wave of coronavirus disease 2019 (COVID-19). Unique mutations in the spike proteins of these emerging Omicron subvariants caused immune evasion from the pre-existing protective immunity induced by vaccination or natural infection. Previously, we developed AdCLD-CoV19-1, a non-replicating recombinant adenoviral vector that encodes the receptor binding domain of the spike protein of the ancestral SARS-CoV-2 strain. Based on the same recombinant adenoviral vector platform, updated vaccines that cover unique mutations found in each Omicron subvariant, including BA.1, BA.2, BA.4.1 and BA.5, were constructed. Preclinical studies revealed that each updated vaccine as a booster shot following primary vaccination targeting the ancestral strain improved neutralizing antibody responses against the pseudovirus of its respective strain most effectively. Of note, boosting with a vaccine targeting the BA.1 or BA.2 Omicron subvariant was most effective in neutralization against the pseudovirus of the BA.2.75 strain, whereas BA.4.1/5-adapted booster shots were most effective in neutralization against the BQ.1, BQ1.1 and BF.7 strains. Therefore, it is imperative to develop a vaccination strategy that can cover the unique spike mutations of currently circulating Omicron subvariants in order to prevent the next wave of COVID-19.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Camundongos , SARS-CoV-2/genética , COVID-19/prevenção & controle , Anticorpos Neutralizantes , Vetores Genéticos , Adenoviridae/genética
3.
Exp Mol Med ; 55(11): 2287-2299, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37907738

RESUMO

CD8 T cells play crucial roles in immune surveillance and defense against infections and cancer. After encountering antigenic stimulation, naïve CD8 T cells differentiate and acquire effector functions, enabling them to eliminate infected or malignant cells. Traditionally, cytotoxic T cells, characterized by their ability to produce effector cytokines and release cytotoxic granules to directly kill target cells, have been recognized as the constituents of the predominant effector T-cell subset. However, emerging evidence suggests distinct subsets of effector CD8 T cells that each exhibit unique effector functions and therapeutic potential. This review highlights recent advancements in our understanding of CD8 T-cell subsets and the contributions of these cells to various disease pathologies. Understanding the diverse roles and functions of effector CD8 T-cell subsets is crucial to discern the complex dynamics of immune responses in different disease settings. Furthermore, the development of immunotherapeutic approaches that specifically target and regulate the function of distinct CD8 T-cell subsets holds great promise for precision medicine.


Assuntos
Linfócitos T CD8-Positivos , Subpopulações de Linfócitos T , Antígenos , Citocinas
4.
Adv Mater ; 35(36): e2303080, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37249019

RESUMO

To demonstrate potent efficacy, a cancer vaccine needs to activate both innate and adaptive immune cells. Personalized cancer vaccine strategies often require the identification of patient-specific neoantigens; however, the clonal and mutational heterogeneity of cancer cells presents inherent challenges. Here, extracellular nanovesicles derived from alpha-galactosylceramide-conjugated autologous acute myeloid leukemia (AML) cells (ECNV-αGC) are presented as a personalized therapeutic vaccine that activates both innate and adaptive immune responses, bypassing the need to identify patient-specific neoantigens. ECNV-αGC vaccination directly engages with and activates both invariant natural killer T (iNKT) cells and leukemia-specific CD8+ T cells in mice with AML, thereby promoting long-term anti-leukemic immune memory. ECNV-αGC sufficiently serves as an antigen-presenting platform that can directly activate antigen-specific CD8+ T cells even in the absence of dendritic cells, thereby demonstrating a multifaceted cellular mechanism of immune activation. Moreover, ECNV-αGC vaccination results in a significantly lower AML burden and higher percentage of leukemia-free survivors among cytarabine-treated hosts with AML. Human AML-derived ECNV-αGCs activate iNKT cells in both healthy individuals and patients with AML regardless of responsiveness to conventional therapies. Together, autologous AML-derived ECNV-αGCs may be a promising personalized therapeutic vaccine that efficiently establishes AML-specific long-term immunity without requiring the identification of neoantigens.


Assuntos
Vacinas Anticâncer , Leucemia Mieloide Aguda , Células T Matadoras Naturais , Humanos , Animais , Camundongos , Linfócitos T CD8-Positivos , Ativação Linfocitária , Leucemia Mieloide Aguda/terapia
5.
Phytochemistry ; 212: 113688, 2023 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-37121294

RESUMO

Camellia sinensis L. (Theaceae) leaves have been used as a beverage in both Eastern and Western cultures for a long time, while its root has not been intensively studied. In this study, seven undescribed triterpenoid saponins (1-7) and twelve known saponins (8-19) with different combinations of substituents, such as oxygenated isoprenyl substituents and sugar moieties, and lengths of sugar chains, were isolated from the C. sinensis roots. Their structures were unequivocally determined using one- and two-dimensional nuclear magnetic resonance data and acid hydrolysis analysis. Investigation of the biological activities of isolated compounds revealed that only those without functional acetyl groups exhibited cytotoxic activities against mouse and human cancer cells (B16F10) and human cervical cancer cell line (HeLa) at 50 µM. Compounds with an aldehyde group at C-23 of aglycone showed immunomodulatory activity against Th1 and Th17 cells at 10 µM. Ten compounds with biological activities from C. sinensis roots extracts, including three previously undescribed ones (3, 6, and 7), were identified.


Assuntos
Antineoplásicos Fitogênicos , Antineoplásicos , Camellia sinensis , Camellia , Saponinas , Triterpenos , Humanos , Animais , Camundongos , Triterpenos/farmacologia , Triterpenos/química , Antineoplásicos Fitogênicos/química , Saponinas/farmacologia , Saponinas/química , Açúcares , Camellia/química
6.
J Clin Invest ; 132(18)2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36106640

RESUMO

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.


Assuntos
Anti-Infecciosos , Células Th1 , Antibacterianos , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Perfilação da Expressão Gênica , Células Th1/metabolismo
7.
J Clin Med ; 11(9)2022 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-35566588

RESUMO

Background: The platelet-to-white blood cell ratio (PWR) is a hematologic marker of the systemic inflammatory response. Recently, the PWR was revealed to have a role as an independent prognostic factor for mortality in patients with hepatitis B virus (HBV)-related acute-on-chronic failure (ACLF) and HBV-related liver cirrhosis (LC) with acute decompensation (AD). However, the prognostic role of the PWR still needs to be investigated in LC patients with AD. In this study, we analyzed whether the PWR could stratify the risk of adverse outcomes (death or liver transplantation (LT)) in these patients. Methods: A prospective cohort of 1670 patients with AD of liver cirrhosis ((age: 55.2 ± 7.8, male = 1226 (73.4%)) was enrolled and evaluated for 28-day and overall adverse outcomes. Results: During a median follow-up of 8.0 months (range, 1.9−15.5 months), 424 (25.4%) patients had adverse outcomes (death = 377, LT = 47). The most common etiology of LC was alcohol use (69.7%). The adverse outcome rate was higher for patients with a PWR ≤ 12.1 than for those with a PWR > 12.1. A lower PWR level was a prognostic factor for 28-day adverse outcomes (PWR: hazard ratio 1.707, p = 0.034) when adjusted for the etiology of cirrhosis, infection, ACLF, and the MELD score. In the subgroup analysis, the PWR level stratified the risk of 28-day adverse outcomes regardless of the presence of ACLF or the main form of AD but not for those with bacterial infection. Conclusions: A lower PWR level was associated with 28-day adverse outcomes, indicating that the PWR level can be a useful and simple tool for stratifying the risk of 28-day adverse outcomes in LC patients with AD.

8.
Adv Mater ; 34(9): e2106516, 2022 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-34962660

RESUMO

Despite the clinically proven efficacies of immune checkpoint blockades, including anti-cytotoxic T lymphocyte-associated protein 4 antibody (αCTLA-4), the low response rate and immune-related adverse events (irAEs) in cancer patients represent major drawbacks of the therapy. These drawbacks of αCTLA-4 therapy are mainly due to the suboptimal activation of tumor-specific cytotoxic T lymphocytes (CTLs) and the systemic nonspecific activation of T cells. To overcome such drawbacks, αCTLA-4 is delivered by dendritic cell-derived nanovesicles presenting tumor antigens (DCNV-TAs) that exclusively interact with tumor-specific T cells, leading to selective activation of tumor-specific CTLs. Compared to conventional αCTLA-4 therapy, treatment with αCTLA-4-conjugated DCNV-TAs significantly inhibits tumor growth and reduces irAEs in syngeneic tumor-bearing mice. This study demonstrates that the spatiotemporal presentation of both αCTLA-4 and tumor antigens enables selective activation of tumor-specific T cells and potentiates the antitumor efficacy of αCTLA-4 without inducing systemic irAEs.


Assuntos
Inibidores de Checkpoint Imunológico , Neoplasias , Animais , Antígenos de Neoplasias , Humanos , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico , Linfócitos T Citotóxicos
9.
Cancer Lett ; 520: 38-47, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34224797

RESUMO

Understanding the rationale of combining immunotherapy and other anticancer treatment modalities is of great interest because of interpatient variability in single-agent immunotherapy. Here, we demonstrated that topoisomerase I inhibitors, a class of chemotherapeutic drugs, can alter the tumor immune landscape, corroborating their antitumor effects combined with immunotherapy. We observed that topotecan-conditioned TC-1 tumors were occupied by a vast number of monocytic cells that highly express CD11c, CD64, and costimulatory molecules responsible for the favorable changes in the tumor microenvironment. Ly6C+MHC-II+CD11chiCD64hi cells, referred to as topotecan-induced monocyte-derived dendritic cells (moDCs), proliferate and activate antigen-specific CD8+ T cells to levels equivalent to those of conventional DCs. Phenotypic changes in Ly6C+ cells towards moDCs were similarly induced by exposure to topotecan in vitro, which was more profoundly facilitated in the presence of tumor cells. Notably, anti-M-CSFR reversed the acquisition of DC-like properties of topotecan-induced moDCs, leading to the abolition of the antitumor effect of topotecan combined with a cancer vaccine. In short, topoisomerase I inhibitors generate monocyte-derived antigen-presenting cells in tumors, which could be mediated by M-CSF-M-CSFR signaling.


Assuntos
Células Apresentadoras de Antígenos/imunologia , Imunoterapia , Neoplasias/terapia , Inibidores da Topoisomerase I/farmacologia , Animais , Antígenos Ly/imunologia , Antígeno CD11c/imunologia , Vacinas Anticâncer/imunologia , Vacinas Anticâncer/farmacologia , Proliferação de Células/genética , Técnicas de Cocultura , Terapia Combinada , Células Dendríticas/efeitos dos fármacos , Células Dendríticas/imunologia , Humanos , Imunidade Inata/efeitos dos fármacos , Imunidade Inata/imunologia , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Linfócitos do Interstício Tumoral/imunologia , Camundongos , Neoplasias/imunologia , Neoplasias/patologia , Receptores de IgG/imunologia , Linfócitos T/imunologia , Inibidores da Topoisomerase I/imunologia , Topotecan/farmacologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/imunologia
10.
J Immunother Cancer ; 9(6)2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-34083422

RESUMO

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.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Deleção de Genes , Interleucina-17/genética , Melanoma Experimental/terapia , Transferência Adotiva , Animais , Linfócitos T CD4-Positivos/imunologia , Linfócitos T CD8-Positivos/transplante , Linhagem Celular Tumoral , Feminino , Humanos , Masculino , Melanoma Experimental/genética , Melanoma Experimental/imunologia , Camundongos , Microambiente Tumoral
11.
Exp Mol Med ; 53(5): 993-1004, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-34050263

RESUMO

Pathogenic conversion of Th17 cells into multifunctional helper T cells or Th1 cells contributes to the pathogenesis of autoimmune diseases; however, the mechanism regulating the plasticity of Th17 cells remains unclear. Here, we found that Th17 cells expressed latent TGF-ß1 in a manner dependent on autocrine TGF-ß1. By employing IL-17-producing cell-specific Tgfb1 conditional knockout and fate-mapping systems, we demonstrated that TGF-ß1-deficient Th17 cells are relatively susceptible to becoming IFN-γ producers through IL-12Rß2 and IL-27Rα upregulation. TGF-ß1-deficient Th17 cells exacerbated tissue inflammation compared to TGF-ß1-sufficient Th17 cells in adoptive transfer models of experimental autoimmune encephalomyelitis and colitis. Thus, TGF-ß1 production by Th17 cells provides an essential autocrine signal for maintaining the stability and regulating the pathogenicity of Th17 cells in vivo.


Assuntos
Autoimunidade , Imunomodulação , Células Th17/imunologia , Células Th17/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Comunicação Autócrina , Biomarcadores , Colite/etiologia , Colite/metabolismo , Colite/patologia , Suscetibilidade a Doenças , Encefalomielite Autoimune Experimental/etiologia , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Regulação da Expressão Gênica , Imunofenotipagem , Contagem de Linfócitos , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Receptores de Interleucina-12/genética , Receptores de Interleucina-12/metabolismo , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Fator de Crescimento Transformador beta1/genética
13.
PLoS One ; 16(1): e0246108, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33513159

RESUMO

This study provides a review of methods used in the determination of organochlorine pesticides (OCPs) in ginseng and compares the effectiveness of three extraction methods (Quick, Easy, Cheap, Effective, Rugged, and Safe (QuEChERS), a modified QuEChERS and a Fast Pesticide Extraction (FaPEx)) in the analyses of 20 OCPs in ginseng root samples. For each method, sample mass, solvent volume and sorbent mass were varied to identify the optimum combination to effectively isolate analytes of interest from the complex sample matrix. Extracts were analyzed using the gas chromatography-µ-electron capture detector (GC-µ-ECD), and confirmatory analyses performed by gas chromatography-tandem-mass spectrometry (GC-MS/MS). Eighteen out of 20 OCPs spiked onto in-house prepared ginseng samples produced acceptable recoveries (51-156%) when extracted using QuEChERS and FaPEx. All 20 analytes, including dichlorodiphenyldichloroethane (p, p'- DDD) and dichlorodiphenyltrichloroethane (o, p'-DDT), produced acceptable recoveries (51-129%) with the use of a modified QuEChERS method. The applicability of the modified QuEChERS method was demonstrated through the analysis of ginseng samples grown in endosulfan-treated soil. The samples were analyzed by both GC-µ-ECD and GC-MS/MS with no significant difference identified in the results of each analytical method. This study highlights the applicability of the modified QuEChERS method, in combination with GC- µ-ECD, to determine organochlorine pesticides in ginseng. This may be especially useful for laboratories in developing countries and less advanced institutions without access to MS/MS instrumentation.


Assuntos
Hidrocarbonetos Clorados/análise , Panax/química , Resíduos de Praguicidas/análise , Endossulfano/análise , Cromatografia Gasosa-Espectrometria de Massas , Limite de Detecção
15.
Vaccines (Basel) ; 8(3)2020 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-32707803

RESUMO

For cancer vaccines, the selection of optimal tumor-associated antigens (TAAs) that can maximize the immunogenicity of the vaccine without causing unwanted adverse effects is challenging. In this study, we developed two engineered Human epidermal growth factor receptor 2 (HER2) antigens, K965 and K1117, and compared their immunogenicity to a previously reported truncated HER2 antigen, K684, within a B cell and monocyte-based vaccine (BVAC). We found that BVAC-K965 and BVAC-K1117 induced comparable antigen-specific antibody responses and antigen-specific T cell responses to BVAC-K684. Interestingly, BVAC-K1117 induced more potent antitumor activity than the other vaccines in murine CT26-HER2 tumor models. In addition, BVAC-K1117 showed enhanced antitumor effects against truncated p95HER2-expressing CT26 tumors compared to BVAC-K965 and BVAC-K684 based on the survival analysis by inducing T cell responses against intracellular domain (ICD) epitopes. The increased ICD epitope-specific T cell responses induced by BVAC-K1117 compared to BVAC-K965 and BVAC-K684 were recapitulated in human leukocyte antigen (HLA)-untyped human PBMCs and HLA-A*0201 PBMCs. Furthermore, we also observed synergistic antitumor effects between BVAC-K1117 and anti-PD-L1 antibody treatment against CT26-HER2 tumors. Collectively, our findings demonstrate that inclusion of a sufficient number of ICD epitopes of HER2 in cellular vaccines can improve the antitumor activity of the vaccine and provide a way to optimize the efficacy of anticancer cellular vaccines targeting HER2.

16.
Biomolecules ; 10(6)2020 05 27.
Artigo em Inglês | MEDLINE | ID: mdl-32471185

RESUMO

Transforming growth factor beta 1 (TGF-ß1) is an immunosuppresive cytokine that plays an essential role in immune homeostasis. It is well known that regulatory T (Treg) cells express TGF-ß1; however, the role of autocrine TGF-ß1 in the development, function, and stability of Treg cells remains poorly understood. We found that Treg cell-derived TGF-ß1 was not required for the development of thymic Treg cells in mice, but played a role in the expression of latency-associated peptide and optimal suppression of naïve T cell proliferation in vitro. Moreover, the frequency of Treg cells was significantly reduced in the mesenteric lymph nodes of the Treg cell-specific TGF-ß1-deficient mice, which was associated with increased frequency of IFN-γ-producers among Treg cells. TGF-ß1-deficient Treg cells were more prone to express IFN-γ than TGF-ß1-sufficient Treg cells in a dendritic cell-mediated stimulation in vitro as well as in an adoptive transfer study in vivo. Mechanistically, TGF-ß1-deficient Treg cells expressed higher levels of Il12rb2 and were more sensitive to IL-12-induced conversion into IFN-γ-producing Treg cells or IFN-γ-producing exTreg cells than TGF-ß1-sufficient Treg cells. Our findings demonstrate that autocrine TGF-ß1 plays a critical role in the optimal suppressive activity and stability of Treg cells by downregulating IL-12R on Treg cells.


Assuntos
Regulação para Baixo , Fatores de Transcrição Forkhead/metabolismo , Receptores de Interleucina-12/metabolismo , Linfócitos T Reguladores/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Animais , Comunicação Autócrina , Fatores de Transcrição Forkhead/genética , Camundongos , Camundongos Endogâmicos , Camundongos Knockout , Camundongos Transgênicos , Receptores de Interleucina-12/genética
17.
Immune Netw ; 20(1): e6, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-32158594

RESUMO

IL-17 is produced by RAR-related orphan receptor gamma t (RORγt)-expressing cells including Th17 cells, subsets of γδT cells and innate lymphoid cells (ILCs). The biological significance of IL-17-producing cells is well-studied in contexts of inflammation, autoimmunity and host defense against infection. While most of available studies in tumor immunity mainly focused on the role of T-bet-expressing cells, including cytotoxic CD8+ T cells and NK cells, and their exhaustion status, the role of IL-17-producing cells remains poorly understood. While IL-17-producing T-cells were shown to be anti-tumorigenic in adoptive T-cell therapy settings, mice deficient in type 17 genes suggest a protumorigenic potential of IL-17-producing cells. This review discusses the features of IL-17-producing cells, of both lymphocytic and myeloid origins, as well as their suggested pro- and/or anti-tumorigenic functions in an organ-dependent context. Potential therapeutic approaches targeting these cells in the tumor microenvironment will also be discussed.

18.
Cancer Immunol Res ; 8(5): 698-709, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32122993

RESUMO

Although treatment with the glucocorticoid-induced tumor necrosis factor receptor-related protein (GITR) agonistic antibody (DTA-1) has shown antitumor activity in various tumor models, the underlying mechanism is not fully understood. Here, we demonstrate that interleukin (IL)-21-producing follicular helper T (Tfh) cells play a crucial role in DTA-1-induced tumor inhibition. The administration of DTA-1 increased IL21 expression by Tfh cells in an antigen-specific manner, and this activation led to enhanced antitumor cytotoxic T lymphocyte (CTL) activity. Mice treated with an antibody that neutralizes the IL21 receptor exhibited decreased antitumor activity when treated with DTA-1. Tumor growth inhibition by DTA-1 was abrogated in Bcl6 fl/fl Cd4 Cre mice, which are genetically deficient in Tfh cells. IL4 was required for optimal induction of IL21-expressing Tfh cells by GITR costimulation, and c-Maf mediated this pathway. Thus, our findings identify GITR costimulation as an inducer of IL21-expressing Tfh cells and provide a mechanism for the antitumor activity of GITR agonism.


Assuntos
Anticorpos Monoclonais/farmacologia , Citocinas/metabolismo , Proteína Relacionada a TNFR Induzida por Glucocorticoide/agonistas , Interleucinas/imunologia , Neoplasias/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Proteína Relacionada a TNFR Induzida por Glucocorticoide/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
19.
Oncoimmunology ; 9(1): 1682380, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32002289

RESUMO

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.


Assuntos
Neoplasias Pulmonares , Linfócitos T Reguladores , Animais , Linfócitos T CD8-Positivos , Células Matadoras Naturais , Neoplasias Pulmonares/tratamento farmacológico , Camundongos , Proteínas Proto-Oncogênicas p21(ras)/genética
20.
FASEB J ; 34(3): 4462-4481, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-31989715

RESUMO

Myeloid progenitor cells have generally been considered the predominant source of myeloid cells under steady-state conditions. Here we show that NK cells contributed to a myeloid cell lineage pool in naïve and tumor-bearing mice. Using fate tracing of NKp46+ cells, we found that myeloid cells could be derived from NK cells. Notably, among mature CD11b+ CD27+ NK cells, c-Kit+ CD24+ NK cells were capable of differentiating into a range of myeloid lineages in vitro and produced neutrophils and monocytes in vivo. The differentiation was completely inhibited by NK-stimulating cytokines. In addition to the potential for differentiation into myeloid cells, c-Kit+ CD24+ NK cells retained NK cell phenotypes and effector functions. Mechanistically, GATA-2 was necessary for the differentiation of c-Kit+ CD24+ NK cells. Therefore, we discovered that GATA-2-dependent differentiation of c-Kit+ CD24+ NK cells contributes to myeloid cell development and identified a novel pathway for myeloid lineage commitment under physiological conditions.


Assuntos
Proliferação de Células/fisiologia , Células Mieloides/citologia , Células Mieloides/metabolismo , Animais , Antígenos Ly/genética , Antígenos Ly/metabolismo , Antígeno CD11b/genética , Antígeno CD11b/metabolismo , Antígeno CD24/genética , Antígeno CD24/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Ensaio de Imunoadsorção Enzimática , Feminino , Citometria de Fluxo , Lentivirus/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Monócitos/metabolismo , Receptor 1 Desencadeador da Citotoxicidade Natural/genética , Receptor 1 Desencadeador da Citotoxicidade Natural/metabolismo , Neutrófilos/metabolismo , Fagocitose/genética , Fagocitose/fisiologia , Reação em Cadeia da Polimerase em Tempo Real , Análise de Sequência de RNA , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/genética , Membro 7 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA