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

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cancer Sci ; 111(11): 4000-4011, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32798273

RESUMO

Heterotypic interactions between tumor cells and macrophages can enable tumor progression and hold potential for the development of therapeutic interventions. However, the communication between tumors and macrophages and its mechanism are poorly understood. Here, we find that tumor-associated macrophages (TAM) from tumor-bearing mice have high amounts of lipid as compared to macrophages from tumor-free mice. TAM also present high lipid content in clinical human gastric cancer patients. Functionally, TAM with high lipid levels are characterized by polarized M2-like profiling, and exhibit decreased phagocytic potency and upregulated programmed death ligand 1 (PD-L1) expression, blocking anti-tumor T cell responses to support their immunosuppressive function. Mechanistically, Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway analysis identifies the specific PI3K pathway enriched within lipid-laid TAM. Lipid accumulation in TAM is mainly caused by increased uptake of extracellular lipids from tumor cells, which leads to the upregulated expression of gamma isoform of phosphoinositide 3-kinase (PI3K-γ) polarizing TAM to M2-like profiling. Correspondingly, a preclinical gastric cancer model is used to show pharmacological targeting of PI3K-γ in high-lipid TAM with a selective inhibitor, IPI549. IPI549 restores the functional activity of macrophages and substantially enhances the phagocytosis activity and promotes cytotoxic-T-cell-mediated tumor regression. Collectively, this symbiotic tumor-macrophage interplay provides a potential therapeutic target for gastric cancer patients through targeting PI3K-γ in lipid-laden TAM.


Assuntos
Metabolismo dos Lipídeos , Ativação de Macrófagos/imunologia , Macrófagos/imunologia , Macrófagos/metabolismo , Neoplasias Gástricas/etiologia , Neoplasias Gástricas/metabolismo , Animais , Linhagem Celular Tumoral , Transformação Celular Neoplásica , Modelos Animais de Doenças , Suscetibilidade a Doenças , Feminino , Imunofluorescência , Humanos , Imunofenotipagem , Lipidômica , Ativação de Macrófagos/genética , Camundongos , Modelos Biológicos , Fagocitose , Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias Gástricas/patologia , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo , Subpopulações de Linfócitos T/patologia , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia , Macrófagos Associados a Tumor/imunologia , Macrófagos Associados a Tumor/metabolismo , Macrófagos Associados a Tumor/patologia
2.
Proc Natl Acad Sci U S A ; 112(5): 1535-40, 2015 Feb 03.
Artigo em Inglês | MEDLINE | ID: mdl-25605927

RESUMO

Inflammation is critical for host defense, but without appropriate control, it can cause chronic disease or even provoke fatal responses. Here we identify a mechanism that limits the inflammatory response. Probing the responses of macrophages to the key sensory Toll-like receptors, we identify that the Broad-complex, Tramtrack and Bric-a-brac/poxvirus and zinc finger (BTB/POZ), transcriptional regulator promyelocytic leukemia zinc finger (PLZF) limits the expression of inflammatory gene products. In accord with this finding, PLZF-deficient animals express higher levels of potent inflammatory cytokines and mount exaggerated inflammatory responses to infectious stimuli. Temporal quantitation of inflammatory gene transcripts shows increased gene induction in the absence of PLZF. Genome-wide analysis of histone modifications distinguish that PLZF establishes basal activity states of early response genes to maintain immune homeostasis and limit damaging inflammation. We show that PLZF stabilizes a corepressor complex that encompasses histone deacetylase activity to control chromatin. Together with our previous demonstration that PLZF promotes the antiviral response, these results suggest a strategy that could realize one of the major goals of immune therapy to retain immune resistance to pathogens while curbing damaging inflammation.


Assuntos
Cromatina/metabolismo , Inflamação/metabolismo , Fatores de Transcrição Kruppel-Like/metabolismo , Transdução de Sinais , Animais , Infecções Bacterianas/metabolismo , Imunoprecipitação da Cromatina , Transferência Ressonante de Energia de Fluorescência , Histona Desacetilases/metabolismo , Macrófagos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Proteína com Dedos de Zinco da Leucemia Promielocítica , Reação em Cadeia da Polimerase em Tempo Real
3.
J Transl Med ; 15(1): 206, 2017 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-29025424

RESUMO

BACKGROUND: Understanding immune phenotypes and human gastric disease in situ requires an approach that leverages multiplexed immunohistochemistry (mIHC) with multispectral imaging to facilitate precise image analyses. METHODS: We developed a novel 4-color mIHC assay based on tyramide signal amplification that allowed us to reliably interrogate immunologic checkpoints, including programmed death-ligand 1 (PD-L1), cytotoxic T cells (CD8+T) and regulatory T cells (Foxp3), in formalin-fixed, paraffin-embedded tissues of various human gastric diseases. By observing cell phenotypes within the disease tissue microenvironment, we were able to determine specific co-localized staining combinations and various measures of cell density. RESULTS: We found that PD-L1 was expressed in gastric ulcer and in tumor cells (TCs), as well as in tumor-infiltrating immune cells (TIICs), but not in normal gastric mucosa or other gastric intraepithelial neoplastic tissues. Furthermore, we found no significant reduction in CD8+T cells, whereas the ratio of CD8+T:Foxp3 cells and CD8+T:PD-L1 cells was suppressed in tumor tissues and elevated in adjacent normal tissues. An unsupervised hierarchical analysis also identified correlations between CD8+T and Foxp3+ tumor-infiltrating lymphocyte (TIL) densities and average PD-L1 levels. Three main groups were identified based on the results of CD8+T:PD-L1 ratios in gastric tumor tissues. Furthermore, integrating CD8+T:Foxp3 ratios, which increased the complexity for immune phenotype status, revealed 6-7 clusters that enabled the separation of gastric cancer patients at the same clinical stage into different risk-group subsets. CONCLUSIONS: Characterizing immune phenotypes in human gastric disease tissues via multiplexed immunohistochemistry may help guide PD-L1 clinical therapy. Observing unique disease tissue microenvironments can improve our understanding of immune phenotypes and cell interactions within these microenvironments, providing the ability to predict safe responses to immunotherapies.


Assuntos
Imuno-Histoquímica/métodos , Gastropatias/imunologia , Gastropatias/patologia , Antígeno B7-H1/metabolismo , Linfócitos T CD8-Positivos/imunologia , Fatores de Transcrição Forkhead/metabolismo , Humanos , Fenótipo
4.
Clin Immunol ; 157(1): 65-77, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25572535

RESUMO

The signal transducer and activator of transcription (STAT) activity plays an important role in the differentiation and imbalance of Th17 and Treg cells in acute coronary syndrome (ACS) patients. We determined that the basal STAT3 phosphorylation level was significantly increased and exhibited a positive relationship with Th17 cells but was negatively correlated with Treg cells in ACS patients. Opposite effects were observed for STAT5 activity. Using the pharmaceutical inhibitor TG101348 or knockdown of STAT3 reduced the number of Th17 cells while promoting the number and function of Treg cells via the Janus kinase2 (JAK2)/STAT3 pathway in ACS patients. Significantly more STAT5 bound to the Foxp3 locus when STAT3 was knocked down, and overexpression of STAT5 led to an increased number of Treg cells but a decreased number of Th17 cells in ACS patients. Our findings demonstrate that modulation of STAT3/STAT5 activity rectifies the imbalance of Th17/Treg cells in ACS patients.


Assuntos
Síndrome Coronariana Aguda/metabolismo , Fator de Transcrição STAT3/metabolismo , Fator de Transcrição STAT5/metabolismo , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Síndrome Coronariana Aguda/imunologia , Angina Estável/imunologia , Diferenciação Celular , Citocinas/sangue , Citometria de Fluxo , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Fator de Transcrição STAT3/genética , Fator de Transcrição STAT5/genética , Linfócitos T Reguladores/citologia , Células Th17/citologia , Regulação para Cima
5.
Immunopharmacol Immunotoxicol ; 36(1): 33-42, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24308297

RESUMO

CONTEXT: Intravenous immunoglobulin (IVIG) has been successfully applied in immune-related diseases of adults and neonates, such as human immunodeficiency virus (HIV) infection and systemic lupus erythematosus (SLE). OBJECTIVE: This study aims to investigate the distinct impacts of IVIG on cultured dendritic cells (DCs) from newborn and healthy adult. MATERIALS AND METHODS: Blood samples were collected from eight full-term newborns and eight healthy adult volunteers. DCs from cord blood and peripheral blood were both cultured in the RPMI 1640 medium containing 10% fetal calf serum, 50 ng/ml granulocyte/macrophage colony-stimulating factor (GM-CSF) and 10 ng/ml recombinant human interleukin-4 (rhIL-4) for 5 d with therapeutic IVIG (20 mg/ml) or physiological IVIG (10 mg/ml). Lipopolysaccharides (LPSs, 1 µg/ml) were added on the fifth day to induce the maturation of immature DCs. The phagocytosis of monocytes, expression of MR (mannose receptor), CD14, CD1a, CD80, CD83, CD86 and MHC II were examined by flow cytometry. The expression of IL-4 mRNA was detected by RT-PCR, while IFN-γ, IL-12 and IL-10 were analyzed by enzyme-linked immunosorbent assay (ELISA) commercial kits. RESULTS: IVIG of therapeutic dose inhibited the phagocytosis, differentiation and maturation of DCs, whereas physiological dose exhibited an accelerated role in vitro, especially on DCs from neonates, but aroused different effects on cytokine secretion. DISCUSSION AND CONCLUSION: The different responses are generally due to immature immune system of neonate, which has a limit capacity to maintain immunity homeostasis. Modulation of DCs phagocytosis, differentiation, maturation and cytokine secretion by IVIG is of potential relevance to its dosage and immune status of patients.


Assuntos
Diferenciação Celular/efeitos dos fármacos , Citocinas/imunologia , Células Dendríticas/imunologia , Imunoglobulinas Intravenosas/farmacologia , Fatores Imunológicos/farmacologia , Fagocitose/efeitos dos fármacos , Adulto , Diferenciação Celular/imunologia , Células Dendríticas/patologia , Relação Dose-Resposta a Droga , Relação Dose-Resposta Imunológica , Feminino , Infecções por HIV/tratamento farmacológico , Infecções por HIV/imunologia , Humanos , Recém-Nascido , Lúpus Eritematoso Sistêmico/tratamento farmacológico , Lúpus Eritematoso Sistêmico/imunologia , Masculino , Fagocitose/imunologia
6.
Int J Mol Sci ; 15(7): 11539-54, 2014 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-24979135

RESUMO

Fatty acid synthase (FASN), responsible for the de novo synthesis of fatty acids, has been shown to act as an oncogene in various human cancers. However, the mechanisms by which FASN favors the progression of ovarian carcinoma remain unknown. In this study, we evaluated FASN expression in ovarian cancer and investigated how FASN regulates the aggressiveness of ovarian cancer cells. Our results show that increased FASN is associated with the peritoneal metastasis of ovarian cancers. Over-expression of FASN results in a significant increase of tumor burden in peritoneal dissemination, accompanied by augment in cellular colony formation and metastatic ability. Correspondingly, FASN knockdown using RNA interference in ovarian cancer cells inhibits the migration in vitro and experimental peritoneal dissemination in vivo. Mechanistic studies reveal that FASN promotes Epithelial-mesenchymal Transition (EMT) via a transcriptional regulation of E-cadherin and N-cadherin, which is also confirmed by luciferase promoter activity analysis. Taken together, our work demonstrates that FASN promotes the peritoneal dissemination of ovarian cancer cells, at least in part through the induction of EMT. These findings suggest that FASN plays a critical role in the peritoneal metastasis of ovarian cancer. Targeting de novo lipogenesis may have a therapeutic potential for advanced ovarian cancer.


Assuntos
Carcinoma/diagnóstico , Transição Epitelial-Mesenquimal , Ácido Graxo Sintase Tipo I/metabolismo , Neoplasias Ovarianas/diagnóstico , Regulação para Cima , Animais , Caderinas/metabolismo , Linhagem Celular Tumoral , Movimento Celular , Ácido Graxo Sintase Tipo I/genética , Feminino , Humanos , Camundongos , Metástase Neoplásica
7.
Eur J Immunol ; 42(10): 2803-14, 2012 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-22711477

RESUMO

Atherosclerosis is a progressive disease with a strong inflammatory component. Here we confirm the existence of a critical imbalance in the ratio of Th17 to Treg-cell populations in peripheral CD4(+) T cells from patients with acute coronary syndrome (ACS), which favors inflammation. This was concurrent with increased IL-17 production from the CD4(+) CD45RA(-) FOXP3(lo) Treg-cell subset, and elevated osteopontin (OPN) levels in serum from ACS patients. We demonstrate a direct effect of OPN in serum from ACS patients on increased IL-17 production by CD4(+) CD45RA(-) FOXP3(lo) T cells, mediated through recruitment of the OPN receptors CD29 and CD44, and dependent on STAT3 and the nuclear hormone receptor retinoic-acid-related orphan receptor-γt (RORγt) pathway, but not IL-6 production. To our knowledge and beyond the disease context of ACS, this study constitutes the first demonstration of a critical role for OPN in the positive regulation of inflammation through increased IL-17 production by CD4(+) CD45RA(-) FOXP3(lo) cells.


Assuntos
Síndrome Coronariana Aguda/imunologia , Linfócitos T CD4-Positivos/imunologia , Inflamação/imunologia , Interleucina-17/metabolismo , Osteopontina/biossíntese , Linfócitos T Reguladores/imunologia , Células Th17/imunologia , Idoso , Células Cultivadas , Feminino , Fatores de Transcrição Forkhead/metabolismo , Humanos , Interleucina-17/genética , Antígenos Comuns de Leucócito/metabolismo , Masculino , Pessoa de Meia-Idade , Membro 3 do Grupo F da Subfamília 1 de Receptores Nucleares/metabolismo , Osteopontina/sangue , Osteopontina/genética , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Regulação para Cima
8.
Small ; 9(19): 3327-35, 2013 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-23463727

RESUMO

NIR-emitting CdSeTe/CdS/ZnS core/shell/shell QD-encoded microbeads are combined with common flow cytometry with one laser for multiplexed detection of hepatitis B virus (HBV). A facile one-pot synthetic route is developed to prepare CdSeTe/CdS/ZnS core/shell/shell QDs with high photoluminescence quantum yield and excellent stability in liquid paraffin, and a Shirasu porous glass (SPG) membrane emulsification technique is applied to incorporate the QDs into polystyrene-maleic anhydride (PSMA) microbeads to obtain highly fluorescent QD-encoded microbeads. The relatively wide NIR photoluminescence full width half maximum of the CdSeTe/CdS/ZnS QDs is used to develop a 'single wavelength' encoding method to obtain different optical codes by changing the wavelengh and emission intensity of the QDs incorporated into the microbeads. Moreover, a detection platform combining NIR-emitting CdSeTe/CdS/ZnS QD-encoded microbeads and Beckman Coulter FC 500 flow cytometry with one laser of 488 nm is successfully used to conduct a 2-plex hybridization assay for hepatitis B surface antigen (HBsAg), hepatitis B e antigen (HBeAg), and a 3-plex hybridization assay for hepatitis B surface antibody (HBsAb), hepatitis B e antibody (HBeAb), and hepatitis B core antibody (HBcAb), which suggests the promising application of NIR QD-encoded microbeads for multiplex immunoassays.


Assuntos
Imunoensaio/métodos , Microesferas , Pontos Quânticos
9.
Oncoimmunology ; 12(1): 2210959, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37197441

RESUMO

T-cell-based immune checkpoint blockade therapy (ICB) can be undermined by local immunosuppressive M2-like tumor-associated macrophages (TAMs). However, modulating macrophages has proved difficult as the molecular and functional features of M2-TAMs on tumor growth are still uncertain. Here we reported that immunosuppressive M2 macrophages render cancer cells resistant to CD8+ T-cell-dependent tumor-killing refractory ICB efficacy by secreting exosomes. Proteomics and functional studies revealed that M2 macrophage-derived exosome (M2-exo) transmitted apolipoprotein E (ApoE) to cancer cells conferring ICB resistance by downregulated MHC-I expression curbing tumor intrinsic immunogenicity. Mechanistically, M2 exosomal ApoE diminished the tumor-intrinsic ATPase activity of binding immunoglobulin protein (BiP) to decrease tumor MHC-I expression. Sensitizing ICB efficacy can be achieved by the administration of ApoE ligand, EZ-482, enhancing ATPase activity of BiP to boost tumor-intrinsic immunogenicity. Therefore, ApoE may serve as a predictor and a potential therapeutic target for ICB resistance in M2-TAMs-enriched cancer patients. Collectively, our findings signify that the exosome-mediated transfer of functional ApoE from M2 macrophages to the tumor cells confers ICB resistance. Our findings also provide a preclinical rationale for treating M2-enriched tumors with ApoE ligand, EZ-482, to restore sensitivity to ICB immunotherapy.


Assuntos
Exossomos , Humanos , Ligantes , Linhagem Celular Tumoral , Macrófagos/metabolismo , Imunoterapia , Antígenos de Neoplasias , Apolipoproteínas E/metabolismo , Adenosina Trifosfatases/metabolismo
10.
Nat Commun ; 14(1): 8465, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-38123560

RESUMO

Inflammasome activity is important for the immune response and is instrumental in numerous clinical conditions. Here we identify a mechanism that modulates the central Caspase-1 and NLR (Nod-like receptor) adaptor protein ASC (apoptosis-associated speck-like protein containing a CARD). We show that the function of ASC in assembling the inflammasome is controlled by its modification with SUMO (small ubiquitin-like modifier) and identify that the nuclear ZBTB16 (zinc-finger and BTB domain-containing protein 16) promotes this SUMOylation. The physiological significance of this activity is demonstrated through the reduction of acute inflammatory pathogenesis caused by a constitutive hyperactive inflammasome by ablating ZBTB16 in a mouse model of Muckle-Wells syndrome. Together our findings identify an further mechanism by which ZBTB16-dependent control of ASC SUMOylation assembles the inflammasome to promote this pro-inflammatory response.


Assuntos
Inflamassomos , Proteína 3 que Contém Domínio de Pirina da Família NLR , Animais , Camundongos , Proteínas Adaptadoras de Sinalização CARD/genética , Proteínas Adaptadoras de Sinalização CARD/metabolismo , Caspase 1/metabolismo , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/genética , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Ligação Proteica , Sumoilação
11.
J Hematol Oncol ; 15(1): 47, 2022 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-35488243

RESUMO

The gut microbiota have long been recognized to play a key role in human health and disease. Currently, several lines of evidence from preclinical to clinical research have gradually established that the gut microbiota can modulate antitumor immunity and affect the efficacy of cancer immunotherapies, especially immune checkpoint inhibitors (ICIs). Deciphering the underlying mechanisms reveals that the gut microbiota reprogram the immunity of the tumor microenvironment (TME) by engaging innate and/or adaptive immune cells. Notably, one of the primary modes by which the gut microbiota modulate antitumor immunity is by means of metabolites, which are small molecules that could spread from their initial location of the gut and impact local and systemic antitumor immune response to promote ICI efficiency. Mechanistic exploration provides novel insights for developing rational microbiota-based therapeutic strategies by manipulating gut microbiota, such as fecal microbiota transplantation (FMT), probiotics, engineered microbiomes, and specific microbial metabolites, to augment the efficacy of ICI and advance the age utilization of microbiota precision medicine.


Assuntos
Microbioma Gastrointestinal , Microbiota , Transplante de Microbiota Fecal , Humanos , Imunidade , Imunoterapia
12.
Front Immunol ; 13: 935552, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35874784

RESUMO

The profiling of the tumor immune microenvironment (TIME) is critical for guiding immunotherapy strategies. However, how the composition of the immune landscape affects the tumor progression of gastric cancer (GC) is ill-defined. Here, we used mass cytometry to perform simultaneous in-depth immune profiling of the tumor, adjacent tissues, and blood cells from GC patients and revealed a unique GC tumor-immune signature, where CD8+ T cells were present at a lower frequency in tumor tissues compared to adjacent tissues, whereas regulatory T cells and tumor-associated macrophages (TAMs) were significantly increased, indicating strong suppressive TIME in GC. Incorporated with oncogenic genomic traits, we found that the unique immunophenotype was interactively shaped by a specific GC gene signature across tumor progression. Earlier-stage GC lesions with IFN signaling enrichment harbored significantly altered T-cell compartments while advanced GC featured by metabolism signaling activation was accumulated by TAMs. Interestingly, PD-1 expression on CD8+ T cells was relatively higher in earlier-stage GC patients, indicating that these patients may derive more benefits from PD-1 inhibitors. The dynamic properties of diverse immune cell types revealed by our study provide new dimensions to the immune landscape of GC and facilitate the development of novel immunotherapy strategies for GC patients.


Assuntos
Neoplasias Gástricas , Linfócitos T CD8-Positivos , Humanos , Imunofenotipagem , Neoplasias Gástricas/patologia , Linfócitos T Reguladores , Microambiente Tumoral
13.
Cancer Cell ; 40(1): 36-52.e9, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-34822775

RESUMO

Reinvigoration of antitumor immunity remains an unmet challenge. Our retrospective analyses revealed that cancer patients who took antihistamines during immunotherapy treatment had significantly improved survival. We uncovered that histamine and histamine receptor H1 (HRH1) are frequently increased in the tumor microenvironment and induce T cell dysfunction. Mechanistically, HRH1-activated macrophages polarize toward an M2-like immunosuppressive phenotype with increased expression of the immune checkpoint VISTA, rendering T cells dysfunctional. HRH1 knockout or antihistamine treatment reverted macrophage immunosuppression, revitalized T cell cytotoxic function, and restored immunotherapy response. Allergy, via the histamine-HRH1 axis, facilitated tumor growth and induced immunotherapy resistance in mice and humans. Importantly, cancer patients with low plasma histamine levels had a more than tripled objective response rate to anti-PD-1 treatment compared with patients with high plasma histamine. Altogether, pre-existing allergy or high histamine levels in cancer patients can dampen immunotherapy responses and warrant prospectively exploring antihistamines as adjuvant agents for combinatorial immunotherapy.


Assuntos
Histamina/metabolismo , Imunoterapia , Neoplasias/tratamento farmacológico , Microambiente Tumoral/efeitos dos fármacos , Linhagem Celular Tumoral/efeitos dos fármacos , Humanos , Tolerância Imunológica/imunologia , Imunoterapia/métodos , Macrófagos/imunologia , Neoplasias/imunologia , Receptores Histamínicos/imunologia , Receptores Histamínicos/metabolismo , Microambiente Tumoral/imunologia
14.
Nat Commun ; 13(1): 5413, 2022 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-36109526

RESUMO

Anti-cancer immunity and response to immune therapy is influenced by the metabolic states of the tumours. Immune checkpoint blockade therapy (ICB) is known to involve metabolic adaptation, however, the mechanism is not fully known. Here we show, by metabolic profiling of plasma samples from melanoma-bearing mice undergoing anti-PD1 and anti-CTLA4 combination therapy, that higher levels of purine metabolites, including inosine, mark ICB sensitivity. Metabolic profiles of ICB-treated human cancers confirm the association between inosine levels and ICB sensitivity. In mouse models, inosine supplementation sensitizes tumours to ICB, even if they are intrinsically ICB resistant, by enhancing T cell-mediated cytotoxicity and hence generating an immunologically hotter microenvironment. We find that inosine directly inhibits UBA6 in tumour cells, and lower level of UBA6 makes the tumour more immunogenic and this is reflected in favourable outcome following ICB therapy in human melanomas. Transplanted mouse melanoma and breast cancer cells with genetic ablation of Uba6 show higher sensitivity to ICB than wild type tumours. Thus, we provide evidence of an inosine-regulated UBA6-dependent pathway governing tumour-intrinsic immunogenicity and hence sensitivity to immune checkpoint inhibition, which might provide targets to overcome ICB resistance.


Assuntos
Inibidores de Checkpoint Imunológico , Melanoma , Animais , Terapia Combinada , Humanos , Inosina/farmacologia , Melanoma/patologia , Camundongos , Radioimunoterapia , Microambiente Tumoral , Enzimas Ativadoras de Ubiquitina
15.
Cancer Discov ; 12(7): 1742-1759, 2022 07 06.
Artigo em Inglês | MEDLINE | ID: mdl-35420681

RESUMO

Despite the popular use of dietary supplements during conventional cancer treatments, their impacts on the efficacies of prevalent immunotherapies, including immune-checkpoint therapy (ICT), are unknown. Surprisingly, our analyses of electronic health records revealed that ICT-treated patients with cancer who took vitamin E (VitE) had significantly improved survival. In mouse models, VitE increased ICT antitumor efficacy, which depended on dendritic cells (DC). VitE entered DCs via the SCARB1 receptor and restored tumor-associated DC functionality by directly binding to and inhibiting protein tyrosine phosphatase SHP1, a DC-intrinsic checkpoint. SHP1 inhibition, genetically or by VitE treatment, enhanced tumor antigen cross-presentation by DCs and DC-derived extracellular vesicles (DC-EV), triggering systemic antigen-specific T-cell antitumor immunity. Combining VitE with DC-recruiting cancer vaccines or immunogenic chemotherapies greatly boosted ICT efficacy in animals. Therefore, combining VitE supplement or SHP1-inhibited DCs/DC-EVs with DC-enrichment therapies could substantially augment T-cell antitumor immunity and enhance the efficacy of cancer immunotherapies. SIGNIFICANCE: The impacts of nutritional supplements on responses to immunotherapies remain unexplored. Our study revealed that dietary vitamin E binds to and inhibits DC checkpoint SHP1 to increase antigen presentation, prime antitumor T-cell immunity, and enhance immunotherapy efficacy. VitE-treated or SHP1-silenced DCs/DC-EVs could be developed as potent immunotherapies. This article is highlighted in the In This Issue feature, p. 1599.


Assuntos
Vacinas Anticâncer , Neoplasias , Animais , Vacinas Anticâncer/uso terapêutico , Células Dendríticas , Imunoterapia , Camundongos , Neoplasias/tratamento farmacológico , Proteína Tirosina Fosfatase não Receptora Tipo 6 , Vitamina E/metabolismo
16.
Am J Cancer Res ; 11(5): 2005-2024, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34094666

RESUMO

Prevention of estrogen receptor-negative (ER-) breast cancer is an unmet challenge, although tamoxifen and aromatase inhibitors can successfully decrease the incidence of ER-positive (ER+) breast cancer. PI3K pathway activation has been detected in tamoxifen-resistant ER- breast lesions of patients. Here, we further ratified that the PI3K pathway is significantly activated in premalignant ER- breast lesions compared with paired normal tissues of patients, which prompted our assessment of targeting PI3K on inhibition of ER- mammary tumor initiation and progression. Both genetic knockdown of PIK3CA or intervention with low-doses of a PI3K inhibitor (GDC-0941) prevented the dysplasia phenotype of semi-transformed human ER- mammary epithelial cells in 3-dimensional culture in vitro. Importantly, low-dose GDC-0941 treatment significantly delayed mammary tumor initiation in the MMTV-neu mouse model without exhibiting discernable adverse effects. Interestingly, increased CD8+/GZMB+ T-cells were detected in mammary tissue after GDC-0941 treatment, suggesting enhanced immune surveillance. Mechanistically, elevated expression of potent T-cell chemo-attractants, including CCL5 and CXCL10, were detected both in vitro and in vivo after GDC-0941 treatment. Furthermore, inhibition of PI3K significantly increased T-cell recruitment in a CCL5/CXCL10-dependent manner. In human ER- breast cancer, PI3K activation is correlated with significantly reduced CCL5, CXCL10 and CD8A expression, suggesting that the decreased CD8+ T-cell recruitment and escape of immune surveillance may contribute to ER- breast cancer development. In summary, our study indicates that low-dose PI3K inhibitor treatment may intervene early stage ER- breast cancer development by enhancing immune surveillance via CCL5/CXCL10.

17.
Clin Immunol ; 134(3): 277-88, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-19900843

RESUMO

The transcription factor Foxp3 plays a key role in CD4(+)CD25(+) regulatory T (Treg) cell function. A correlation has been shown between survival and the frequency of tumor-infiltrating Foxp3-positive Treg cells in cancer patients. However, few studies have characterized the regulation of Foxp3 expression and function in Treg cells, which are known to comprise distinct subsets, with different roles in the complex tumor microenvironment. Here, we show that significantly more Foxp3-positive Treg cells accumulated in gastric tumors. In addition, we found increased expression of Foxp3 protein per cell in tumor-infiltrating Treg cells. Moreover, elevated Foxp3 expression in tumor-infiltrating Treg cells was associated with the TNM stage in gastric cancer patients. Importantly, further investigation within the tumor microenvironment showed that expression of Foxp3 in Treg cells correlated with expression of cyclooxygenase-2 (COX-2) and prostaglandin E(2) (PGE(2)). Furthermore, Treg cells with higher levels of Foxp3 were able to suppress the proliferation of autologous CD4(+)CD25(-) T cells. The suppression of the effector T-cell response was reversed by COX inhibitors and PGE(2) receptor-specific antagonists. Our data demonstrate a mechanism by which tumor-infiltrating Treg cells with increased Foxp3 expression can mediate immune suppression via COX-2/PGE(2) production in the gastric cancer microenvironment. Thus, we provide new insights into overcoming regulatory T-cell activity, which may be beneficial for the treatment of human gastric cancer.


Assuntos
Ciclo-Oxigenase 2/imunologia , Fatores de Transcrição Forkhead/biossíntese , Neoplasias Gástricas/enzimologia , Neoplasias Gástricas/imunologia , Linfócitos T Reguladores/imunologia , Ciclo-Oxigenase 2/genética , Dinoprostona/genética , Dinoprostona/imunologia , Feminino , Citometria de Fluxo , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/imunologia , Humanos , Imuno-Histoquímica , Ativação Linfocitária , Masculino , Pessoa de Meia-Idade , Estadiamento de Neoplasias , RNA Neoplásico/química , RNA Neoplásico/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia
18.
Clin Dev Immunol ; 2010: 357541, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20379374

RESUMO

Lactadherin, as one of the immune components in the breast milk, might play a role in the intestinal immune system of newborn. Therefore, we investigated the effect of lactadherin-feeding in early time on the development of intestinal immune system compared with naturally rearing and artificially rearing (non-lactadherin). In the present study, we observed that the Peyer's Patches (PP) from the pups of artificially reared group with lactadherin added were characterized by an excess of OX62(+)CD4(+)SIRP(+) DC cells and a higher expression of CD3(+)CD4(+)CD25(+)T cells. Additionally, this study also demonstrated that IL-10 production was dramatically increased when lactadherin was present in culture medium compared with lactadherin-absent culture. These results suggested that lactadherin could adjust intestinal DCs activity, induce CD3(+)CD4(+)CD25(+)T cell differentiation, and enhance IL-10 production.


Assuntos
Células Dendríticas/imunologia , Mucosa Intestinal/imunologia , Proteínas do Leite/imunologia , Leite/imunologia , Animais , Antígenos de Diferenciação/imunologia , Antígenos de Superfície , Aleitamento Materno , Complexo CD3/imunologia , Antígenos CD4/imunologia , Humanos , Interleucina-10/imunologia , Subunidade alfa de Receptor de Interleucina-2/imunologia , Mucosa Intestinal/crescimento & desenvolvimento , Nódulos Linfáticos Agregados/imunologia , Ratos , Ratos Sprague-Dawley , Linfócitos T Reguladores/imunologia
19.
Front Immunol ; 11: 1731, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32849616

RESUMO

The immunosuppressive status of the tumor microenvironment (TME) remains poorly defined due to a lack of understanding regarding the function of tumor-associated macrophages (TAMs), which are abundant in the TME. TAMs are crucial drivers of tumor progression, metastasis, and resistance to therapy. Intra- and inter-tumoral spatial heterogeneities are potential keys to understanding the relationships between subpopulations of TAMs and their functions. Antitumor M1-like and pro-tumor M2-like TAMs coexist within tumors, and the opposing effects of these M1/M2 subpopulations on tumors directly impact current strategies to improve antitumor immune responses. Recent studies have found significant differences among monocytes or macrophages from distinct tumors, and other investigations have explored the existence of diverse TAM subsets at the molecular level. In this review, we discuss emerging evidence highlighting the redefinition of TAM subpopulations and functions in the TME and the possibility of separating macrophage subsets with distinct functions into antitumor M1-like and pro-tumor M2-like TAMs during the development of tumors. Such redefinition may relate to the differential cellular origin and monocyte and macrophage plasticity or heterogeneity of TAMs, which all potentially impact macrophage biomarkers and our understanding of how the phenotypes of TAMs are dictated by their ontogeny, activation status, and localization. Therefore, the detailed landscape of TAMs must be deciphered with the integration of new technologies, such as multiplexed immunohistochemistry (mIHC), mass cytometry by time-of-flight (CyTOF), single-cell RNA-seq (scRNA-seq), spatial transcriptomics, and systems biology approaches, for analyses of the TME.


Assuntos
Plasticidade Celular , Macrófagos/imunologia , Neoplasias/imunologia , Animais , Regulação Neoplásica da Expressão Gênica , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patologia , Fenótipo , Transdução de Sinais , Transcriptoma , Microambiente Tumoral
20.
Aging (Albany NY) ; 12(9): 8372-8396, 2020 05 04.
Artigo em Inglês | MEDLINE | ID: mdl-32365332

RESUMO

Increased expression of the kinesin family member 23 (KIF23) has been verified in gastric cancer (GC) and its upregulation contributes to cell proliferation. Even though, the role of KIF23 has not been fully elucidated in GC, and the mechanisms of KIF23 as an oncogene remain unknown. To further identify its potential role in GC, we analyzed gene expression data from GC patients in GEO and TCGA datasets. KIF23 was upregulated in GC, and increased expression of KIF23 correlated with poor prognosis. Importantly, KIF23 inhibition not only suppressed GC cell proliferation, tumorigenesis, but also migration and invasion, and arrested the cell cycle in the G2/M phase. Mechanistic investigations confirmed that KIF23 activated the Wnt/ß-catenin signaling pathway by directly interacting with APC membrane recruitment 1 (Amer1). Furthermore, KIF23 exhibited competitive binding with Amer1 to block the association of Amer1 with adenomatous polyposis coli (APC), thus relocating Amer1 from the membrane and cytoplasm to the nucleus and attenuating the ability of Amer1 to negatively regulate Wnt/ß-catenin signaling, resulting in activation of this signaling pathway. Collectively, our findings demonstrated that KIF23 promoted GC cell proliferation by directly interacting with Amer1 and activating the Wnt/ß-catenin signaling pathway.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Regulação Neoplásica da Expressão Gênica , Proteínas Associadas aos Microtúbulos/genética , Neoplasias Gástricas/patologia , Proteínas Supressoras de Tumor/genética , Via de Sinalização Wnt , Animais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Nus , Neoplasias Gástricas/genética , Regulação para Cima , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa