Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Int J Mol Sci ; 22(1)2020 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-33396463

RESUMO

Tumor-infiltrating immune cells phenotype is associated with tumor progression. However, little is known about the phenotype of the peripheral blood mononuclear cells (PBMC) from breast cancer patients. We investigated MMP1 and MMP11 expression in PBMC from breast cancer patients and we analyzed gene expression changes upon their interaction with cancer cells and cancer-associated fibroblasts (CAF). We measured the impact of PBMC on proinflammatory gene expression in breast cancer cells, normal fibroblast (NF), and CAF and the impact on proliferation and invasiveness capacity of breast cancer cells. Gene expression of MMP1 and MMP11 in PBMC from breast cancer patients (n = 54) and control (n = 28); expression of IL1A, IL6, IL17, IFNß, and NFĸB in breast cancer cell lines (MCF-7 and MDA-MB-231); and, additionally, IL10 and MMP11 in CAF and NF were analyzed by qRT-PCR before and after co-culture. Our results show the existence of a subpopulation of breast cancer patients (25.9%) with very high levels of MMP11 gene expression in PBMC. Also, gene expression of MMP1 and MMP11 increases in PBMC after co-culture with breast cancer cell lines, NF or CAF. PBMC from healthy or breast cancer patients induce an increased proliferation rate on MCF-7 and an increased invasiveness capacity of MDA-MB-231. Finally, we show a differential expression profile of inflammatory genes in NF and CAF when co-cultured with control or breast cancer PBMC. We have observed that MMPs' expression in PBMC is regulated by the microenvironment, while the expression of inflammatory genes in NF or CAF is differentially regulated by PBMC. These findings confirm the importance of the crosstalk between stromal cells and suggest that PBMC would play a role in promoting aggressive tumor behavior.


Assuntos
Neoplasias da Mama/patologia , Fibroblastos Associados a Câncer/patologia , Fibroblastos/patologia , Regulação Enzimológica da Expressão Gênica , Leucócitos Mononucleares/patologia , Metaloproteinase 11 da Matriz/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Fibroblastos Associados a Câncer/metabolismo , Estudos de Casos e Controles , Técnicas de Cocultura , Feminino , Fibroblastos/metabolismo , Seguimentos , Regulação Neoplásica da Expressão Gênica , Humanos , Leucócitos Mononucleares/metabolismo , Metaloproteinase 1 da Matriz/genética , Metaloproteinase 11 da Matriz/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Prognóstico , Receptor ErbB-2/metabolismo , Receptores de Estrogênio/metabolismo , Receptores de Progesterona/metabolismo , Células Estromais/metabolismo , Células Estromais/patologia , Microambiente Tumoral
2.
Cell Immunol ; 337: 48-53, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30832981

RESUMO

The Bromodomain and Extra-terminal (BET) family of proteins were first recognized as important epigenetic regulators in inflammatory processes; however, there is increasing evidence to support the notion that BET proteins also play a critical role in 'reading' chromatin and recruiting chromatin-regulating enzymes to control gene expression in a number of pathologic processes, including cancer. To this end, the mechanisms by which BET proteins regulate chromatin remodeling and promote tumor-associated inflammation have been heavily studied over the past decade. This article to review the biology of BET protein dysfunction in promoting tumor-associated inflammation and cancer progression and the application of small molecule inhibitors that target specific BET proteins, alone or in combination with immunomodulatory agents as a novel therapeutic strategy for cancer patients.


Assuntos
Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Neoplasias/terapia , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Animais , Proteínas de Ciclo Celular/antagonistas & inibidores , Transformação Celular Neoplásica/imunologia , Montagem e Desmontagem da Cromatina/fisiologia , Epigênese Genética/genética , Regulação da Expressão Gênica/genética , Humanos , Inflamação/metabolismo , Neoplasias/patologia , Proteínas Nucleares/genética , Proteínas/antagonistas & inibidores , Proteínas/metabolismo , Proteínas/fisiologia , Fatores de Transcrição/antagonistas & inibidores
3.
Exp Cell Res ; 336(2): 211-22, 2015 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-26172215

RESUMO

The human CD99 protein is a 32-kDa type I transmembrane glycoprotein, while CD98 is a disulfide-linked 125-kDa heterodimeric type II transmembrane glycoprotein. It has been previously shown that CD99 and CD98 oppositely regulate ß1 integrin signaling, though the mechanisms by which this regulation occurs are not known. Our results revealed that antibody-mediated crosslinking of CD98 induced FAK phosphorylation at Y397 and facilitated the formation of the protein kinase Cα (PKCα)-syntenin-focal adhesion kinase (FAK), focal adhesions (FAs), and IPP-Akt1-syntenin complex, which mediates ß1 integrin signaling. In contrast, crosslinking of CD99 disrupted the formation of the PKCα-syntenin-FAK complex as well as FA via FAK dephosphorylation. The CD99-induced dephosphorylation of FAK was apparently mediated by the recruitment of Src homology region 2 domain-containing phosphatase-2 (SHP2) to the plasma membrane and subsequent activation of its phosphatase activity. Further consequences of the activation of SHP2 included the disruption of FAK-talin and talin-ß1 integrin interactions and attenuation in the formation of the IPP-Akt1-syntenin complex at the plasma membrane, which resulted in reduced cell-ECM adhesion. This report uncovers the molecular mechanisms underlying the inverse regulation of ß1 integrin signaling by CD99 and CD98 and may provide a novel therapeutic approach to treat inflammation and cancer.


Assuntos
Antígenos CD/metabolismo , Moléculas de Adesão Celular/metabolismo , Quinase 1 de Adesão Focal/metabolismo , Proteína-1 Reguladora de Fusão/metabolismo , Integrina beta1/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Antígeno 12E7 , Adesão Celular , Linhagem Celular Tumoral , Adesões Focais/metabolismo , Humanos , Fosforilação , Proteína Quinase C-alfa/metabolismo , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Interferência de RNA , RNA Interferente Pequeno , Transdução de Sinais , Sinteninas/metabolismo
4.
Semin Cancer Biol ; 23(4): 243-51, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23712403

RESUMO

Inflammation, the first line of defense against pathogens can contribute to all phases of tumorigenesis, including tumor initiation, promotion and metastasis. Within this framework, the Toll-like receptor (TLR) pathway plays a central role in inflammation and cancer. Although extremely useful, the classical representation of this, and other pathways in the cellular network in terms of nodes (proteins) and edges (interactions) is incomplete. Structural pathways can help complete missing parts of such diagrams: they demonstrate in detail how signals coming from different upstream pathways merge and propagate downstream, how parallel pathways compensate each other in drug resistant mutants, how multi-subunit signaling complexes form and in particular why they are needed and how they work, how allosteric events can control these proteins and their pathways, and intricate details of feedback loops and how kick in. They can also explain the mechanisms of some oncogenic SNP mutations. Constructing structural pathways is a challenging task. Here, our goal is to provide an overview of inflammation and cancer from the structural standpoint, focusing on the TLR pathway. We use the powerful PRISM (PRotein Interactions by Structural Matching) tool to reveal important structural information of interactions in and within key orchestrators of the TLR pathway, such as MyD88.


Assuntos
Inflamação/metabolismo , Neoplasias/metabolismo , Transdução de Sinais , Receptores Toll-Like/metabolismo , Inflamação/genética , Modelos Moleculares , Mutação , Fator 88 de Diferenciação Mieloide/química , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Neoplasias/genética , Mapeamento de Interação de Proteínas/métodos , Mapas de Interação de Proteínas , Estrutura Terciária de Proteína , Receptores Toll-Like/química , Receptores Toll-Like/genética
5.
Cancers (Basel) ; 14(3)2022 Jan 28.
Artigo em Inglês | MEDLINE | ID: mdl-35158948

RESUMO

Neutrophils are an important component of the tumor microenvironment, and their infiltration has been associated with a poor prognosis for most human tumors. However, neutrophils have been shown to be endowed with both protumor and antitumor activities, reflecting their heterogeneity and plasticity in cancer. A growing body of studies has demonstrated that chemokines and chemokine receptors, which are fundamental regulators of neutrophils trafficking, can affect neutrophil maturation and effector functions. Here, we review human and mouse data suggesting that targeting chemokines or chemokine receptors can modulate neutrophil activity and improve their antitumor properties and the efficiency of immunotherapy.

6.
Eur J Surg Oncol ; 47(5): 931-934, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33139129

RESUMO

BACKGROUND: The aim of the study was to analyze the clinical characteristics and outcomes of patients with de novo secondary adenocarcinoma arising in the operatively transposed colon not exposed to the fecal stream. METHODS: Two investigators collected and reviewed papers from June 1938 to June 2019, reporting patients with adenocarcinoma arising in the transposed colon, not in contact with the fecal stream. RESULTS: Overall, we identified 98 patients with a transposed colonic autograft, positioned as conduit but not in contact with the fecal stream, in whom a secondary de novo adenocarcinoma was diagnosed. In 50% of the patients, the secondary adenocarcinoma was diagnosed at an advanced stage, with a subsequent poor clinical outcome. Earlier diagnosis allowed local resection with long term success. The occurrence of the adenocarcinoma appeared to be closely related to aging, and to clinical evidence of chronic inflammation. CONCLUSIONS: Patients in whom the colon has been surgically transposed to different anatomic positions, away from the fecal stream, can develop a secondary colonic adenocarcinoma with. Aging and chronic inflammation seem to be risk factors for a secondary adenocarcinoma more than time from implant. Screening for polyps and adenocarcinomas in these patients should be considered.


Assuntos
Adenocarcinoma/etiologia , Colo/transplante , Neoplasias do Colo/etiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Fezes , Feminino , Gastrectomia , Humanos , Inflamação/complicações , Masculino , Pessoa de Meia-Idade , Fatores de Risco , Vagina/cirurgia
7.
ACS Appl Mater Interfaces ; 13(22): 25727-25737, 2021 Jun 09.
Artigo em Inglês | MEDLINE | ID: mdl-34048646

RESUMO

Accurate discrimination of inflammations and cancers as well as differential inhibition of cancers are significant for early diagnoses and timely treatments. Nanoparticles have become new modalities for diagnosis and therapy. However, they are still challenged by the efficient delivery of multiple reagents into living cells, discriminating multisignals without any interference, and differential treatments of different diseases. Here, multifunctional spiky topological nanocapsules (STNs) are prepared for the discrimination and differential inhibition of inflammation and cancer. With unique spiky hollow architectures, STNs' advantages including excellent loading capacity, enhanced cellular uptake, DNAs' protection against degradation, target-controlled drug release, and efficient endo-/lysosome escape are demonstrated. Therefore, sequential detection of inflammation-related miR-155 (by external modified hairpin DNAs) and the cancer target of monocarboxylate transporter 1 (MCT1) (by internal loaded pH-sensitive carbon dots and MCT1 inhibitor-AZD3965) are achieved. Furthermore, the release of AZD3965 from the cavities of STNs is controlled by the miR-155 amount (first target). Therefore, the released drug of AZD3965 realizes the stage-dependent differential treatment of diseases via cellular acidosis induced by MCT1 inhibition. Via in vivo evaluations of normal, inflammatory, and liver cancer cells/mice, as well as the efficient inhibition of tumor growth, the possibility of STN-based discrimination and differential treatment is confirmed. This would encourage new strategies for multidiagnosis and differential treatment of early-stage cancer.


Assuntos
Carcinoma Hepatocelular/tratamento farmacológico , Liberação Controlada de Fármacos , Inflamação/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Transportadores de Ácidos Monocarboxílicos/antagonistas & inibidores , Nanocápsulas/administração & dosagem , Pirimidinonas/farmacologia , Simportadores/antagonistas & inibidores , Tiofenos/farmacologia , Animais , Apoptose , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Movimento Celular , Proliferação de Células , Feminino , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/genética , Nanocápsulas/química , Pirimidinonas/química , Tiofenos/química , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Front Genet ; 12: 654517, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34539726

RESUMO

BACKGROUND: Hepatocellular carcinoma (HCC) has become the main cause of cancer death worldwide. More than half of hepatocellular carcinoma developed from hepatitis B virus infection (HBV). The purpose of this study is to find the key genes in the transformation process of liver inflammation and cancer and to inhibit the development of chronic inflammation and the transformation from disease to cancer. METHODS: Two groups of GEO data (including normal/HBV and HBV/HBV-HCC) were selected for differential expression analysis. The differential expression genes of HBV-HCC in TCGA were verified to coincide with the above genes to obtain overlapping genes. Then, functional enrichment analysis, modular analysis, and survival analysis were carried out on the key genes. RESULTS: We identified nine central genes (CDK1, MAD2L1, CCNA2, PTTG1, NEK2) that may be closely related to the transformation of hepatitis B. The survival and prognosis gene markers composed of PTTG1, MAD2L1, RRM2, TPX2, CDK1, NEK2, DEPDC1, and ZWINT were constructed, which performed well in predicting the overall survival rate. CONCLUSION: The findings of this study have certain guiding significance for further research on the transformation of hepatitis B inflammatory cancer, inhibition of chronic inflammation, and molecular targeted therapy of cancer.

9.
Oncoimmunology ; 8(1): e1518628, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30546959

RESUMO

Hepatocellular carcinoma (HCC) is one type of cancers whose carcinogenesis and progression are closely related to chronic inflammation. Identifying the molecular mechanisms for inflammation-related HCC progression will contribute to improve the efficacy of current therapeutics for HCC patients. Many kinds of epigenetic factors, including long non-coding RNAs (lncRNAs), have been discovered to be important in HCC growth and metastasis. However, how the lncRNAs promote HCC progression and what's the application of lncRNA silencing in vivo in suppressing HCC remain to be further investigated. Here, we found that lncRNA metastasis associated lung adenocarcinoma transcript1 (MALAT1) was upregulated in HCC tumor tissues, and knockdown of MALAT1 suppressed proliferation, cell cycle and invasion of HCC cells in response to lipopolysaccharide (LPS) stimulation. Knockdown of MALAT1 significantly inhibited LPS-induced pro-inflammatory mediators IL-6 and CXCL8 expression in HCC cells, which could be restored by overexpressing MALAT1. Mechanistically, MALAT1 recruited Brahma-related gene 1 (BRG1), a catalytic subunit of chromatin remodeling complex switching/sucrose non-fermentable (SWI/SNF), to the promoter region of IL-6 and CXCL8, and thus facilitated NF-κB to induce the expression of these inflammatory factors. Importantly, in vivo silencing of MALAT1 in HCC tissues inhibited growth of HCC xenografts, and also suppressed the expression of pro-inflammatory factors in HCC tissues accordingly. Our results demonstrate that MALAT1 promotes HCC progression by binding BRG1 to epigenetically enhance inflammatory response in HCC tissues, and silencing of MALAT1 may be a potential approach to the treatment of HCC.

10.
Future Med Chem ; 11(16): 2151-2169, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31538525

RESUMO

Recently, PI3Kγ, a vital kinase, which involved in numerous intracellular signaling pathways, has been considered as a promising drug target for the treatment of immune diseases and certain cancers. Before the 21st century, few selective PI3Kγ inhibitors were discovered because no non-conserved structure in the ATP binding sites of PI3Kγ had been found. Since the discovery of the non-ATP binding pocket, the reported structures of potent and selective PI3Kγ inhibitors have become more diverse, and one compound (IPI549) has entered Phase I clinical trial. This review centers on a general overview of PI3Kγ inhibitors in clinical and preclinical as well as further therapeutic applications in human diseases.


Assuntos
Classe Ib de Fosfatidilinositol 3-Quinase/imunologia , Inflamação/tratamento farmacológico , Neoplasias/tratamento farmacológico , Inibidores de Fosfoinositídeo-3 Quinase/química , Inibidores de Fosfoinositídeo-3 Quinase/uso terapêutico , Animais , Classe Ib de Fosfatidilinositol 3-Quinase/química , Ensaios Clínicos como Assunto , Desenvolvimento de Medicamentos , Descoberta de Drogas , Humanos , Inflamação/imunologia , Modelos Moleculares , Neoplasias/imunologia , Inibidores de Fosfoinositídeo-3 Quinase/farmacologia
11.
Front Immunol ; 9: 1930, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30245686

RESUMO

Macrophages represent a heterogeneous cell population and are known to display a remarkable plasticity. In response to distinct micro-environmental stimuli, e.g., tumor stroma vs. infected tissue, they polarize into different cell subtypes. Originally, two subpopulations were defined: classically activated macrophages or M1, and alternatively activated macrophages or M2. Nowadays, the M1/M2 classification is considered as an oversimplified approach that does not adequately cover the total spectrum of macrophage phenotypes observed in vivo. Especially in pathological circumstances, macrophages behave as plastic cells modifying their expression and transcription profile along a continuous spectrum with M1 and M2 phenotypes as extremes. Here, we focus on the effect of chemokines on macrophage differentiation and polarization in physiological and pathological conditions. In particular, we discuss chemokine-induced macrophage polarization in inflammatory diseases, including obesity, cancer, and atherosclerosis.


Assuntos
Aterosclerose/imunologia , Diferenciação Celular/imunologia , Quimiocinas/imunologia , Macrófagos/imunologia , Transdução de Sinais/imunologia , Animais , Aterosclerose/patologia , Humanos , Inflamação/imunologia , Inflamação/patologia , Macrófagos/patologia
12.
Oncoimmunology ; 7(9): e1475874, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30228943

RESUMO

Ocular adnexal sebaceous carcinoma (OASC) is an aggressive malignancy that frequently recurs locally and metastasizes. Surgical extirpation may produce significant aesthetic morbidity, and effective systemic therapies for locally advanced or metastatic disease are largely ineffective. Immune checkpoint inhibitors have shown efficacy in the management of several solid tumors where tumor cell PD-L1 expression correlates with improved response. To determine whether OASC might be amenable to immune checkpoint blockade, we performed comprehensive immune profiling for CD3, CD8, PD-1, FOXP3, and PD-L1 in 24 patients with primary OASC. The composition, distribution and density of the tumor associated immune infiltrate were quantified by automated image analysis and correlated with measures of clinical outcome. Tumor cells in 12 OASCs (50%) expressed PD-L1. Higher densities of CD3+ (p = 0.01), CD8+ (p = 0.006), and PD-1+ (p = 0.024) tumor-associated T cells were associated with higher T category (≥T3a per the 7th edition of the American Joint Committee on Cancer staging manual). Higher tumor cell expression of PD-L1 correlated with higher density of PD-1+ tumor-associated T cells (p = 0.021). Since a CD3+ CD8+ PD-1 + T-cell infiltrate represents a "suppressed T-cell phenotype" apparently permissive toward OASC progression, our findings provide a mechanistic rationale for the effective application of immune checkpoint blockade in OASC to abrogate PD-1/PD-L1 interaction and effectively unleash the immune infiltrate to treat higher-stage tumors.

13.
Front Microbiol ; 9: 663, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29696001

RESUMO

Pathogenic and commensal microbes induce various levels of inflammation and metabolic disease in the host. Inflammation caused by infection leads to increased production of reactive oxygen species (ROS) and subsequent oxidative DNA damage. These in turn cause further inflammation and exacerbation of DNA damage, and pose a risk for cancer development. Helicobacter pylori-mediated inflammation has been implicated in gastric cancer in many previously established studies, and Fusobacterium nucleatum presence has been observed with greater intensity in colorectal cancer patients. Despite ambiguity in the exact mechanism, infection-mediated inflammation may have a link to cancer development through an accumulation of potentially mutagenic DNA damage in surrounding cells. The multiple DNA repair pathways such as base excision, nucleotide excision, and mismatch repair that are employed by cells are vital in the abatement of accumulated mutations that can lead to carcinogenesis. For this reason, understanding the role of DNA repair as an important cellular mechanism in combatting the development of cancer will be essential to characterizing the effect of infection on DNA repair proteins and to identifying early cancer biomarkers that may be targeted for cancer therapies and treatments.

14.
Front Immunol ; 9: 2732, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30532756

RESUMO

HuR is an abundant RNA-binding protein acting as a post-transcriptional regulator of many RNAs including mRNAs encoding inflammatory mediators, cytokines, death signalers and cell cycle regulators. In the context of intestinal pathologies, elevated HuR is considered to enhance the stability and the translation of pro-tumorigenic mRNAs providing the rationale for its pharmacological targeting. However, HuR also possesses specific regulatory functions for innate immunity and cytokine mRNA control which can oppose intestinal inflammation and tumor promotion. Here, we aim to identify contexts of intestinal inflammation where the innate immune and the epithelial functions of HuR converge or diverge. To address this, we use a disease-oriented phenotypic approach using mice lacking HuR either in intestinal epithelia or myeloid-derived immune compartments. These mice were compared for their responses to (a) Chemically induced Colitis; (b) Colitis- associated Cancer (CAC); (c) T-cell mediated enterotoxicity; (d) Citrobacter rodentium-induced colitis; and (e) TNF-driven inflammatory bowel disease. Convergent functions of epithelial and myeloid HuR included their requirement for suppressing inflammation in chemically induced colitis and their redundancies in chronic TNF-driven IBD and microbiota control. In the other contexts however, their functions diversified. Epithelial HuR was required to protect the epithelial barrier from acute inflammatory or infectious degeneration but also to promote tumor growth. In contrast, myeloid HuR was required to suppress the beneficial inflammation for pathogen clearance and tumor suppression. This cellular dichotomy in HuR's functions was validated further in mice engineered to express ubiquitously higher levels of HuR which displayed diminished pathologic and beneficial inflammatory responses, resistance to epithelial damage yet a heightened susceptibility to CAC. Our study demonstrates that epithelial and myeloid HuR affect different cellular dynamics in the intestine that need to be carefully considered for its pharmacological exploitation and points toward potential windows for harnessing HuR functions in intestinal inflammation.


Assuntos
Citrobacter rodentium/imunologia , Colite/imunologia , Proteína Semelhante a ELAV 1/imunologia , Infecções por Enterobacteriaceae/imunologia , Mucosa Intestinal/imunologia , Animais , Colite/genética , Colite/microbiologia , Colite/patologia , Proteína Semelhante a ELAV 1/genética , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Imunidade Inata , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Inflamação/patologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/patologia , Camundongos , Camundongos Transgênicos
15.
Oncoimmunology ; 7(3): e1404215, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29399404

RESUMO

The enzyme arginase-1 reduces the availability of arginine to tumor-infiltrating immune cells, thus reducing T-cell functionality in the tumor milieu. Arginase-1 is expressed by some cancer cells and by immune inhibitory cells, such as myeloid-derived suppressor cells (MDSCs) and tumor-associated macrophages (TAMs), and its expression is associated with poor prognosis. In the present study, we divided the arginase-1 protein sequence into overlapping 20-amino-acid-long peptides, generating a library of 31 peptides covering the whole arginase-1 sequence. Reactivity towards this peptide library was examined in PBMCs from cancer patients and healthy individuals. IFNγ ELISPOT revealed frequent immune responses against multiple arginase-1-derived peptides. We further identified a hot-spot region within the arginase-1 protein sequence containing multiple epitopes recognized by T cells. Next, we examined in vitro-expanded tumor-infiltrating lymphocytes (TILs) isolated from melanoma patients, and detected arginase-1-specific T cells that reacted against epitopes from the hot-spot region. Arginase-1-specific CD4+T cells could be isolated and expanded from peripheral T cell pool of a patient with melanoma, and further demonstrated the specificity and reactivity of these T cells. Overall, we showed that arginase-1-specific T cells were capable of recognizing arginase-1-expressing cells. The activation of arginase-1-specific T cells by vaccination is an attractive approach to target arginase-1-expressing malignant cells and inhibitory immune cells. In the clinical setting, the induction of arginase-1-specific immune responses could induce or increase Th1 inflammation at the sites of tumors that are otherwise excluded due to infiltration with MDSCs and TAMs.

16.
Front Immunol ; 9: 1111, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29875772

RESUMO

Cells undergoing apoptosis produce heterogeneous populations of membrane delimited extracellular vesicles (Apo-EVs) which vary not only in size-from tens of nanometers to several microns-but also in molecular composition and cargo. Apo-EVs carry a variety of potentially biologically active components, including small molecules, proteins, and nucleic acids. Larger forms of Apo-EVs, commonly termed "apoptotic bodies," can carry organelles, such as mitochondria and nuclear fragments. Molecules displayed on the surface of extracellular vesicles (EVs) can contribute substantially to their size, as well as their functions. Thus far, relatively little is known of the functional significance of Apo-EVs apart from their roles in fragmentation of dying cells and indicated immunomodulatory activities. Here, we discuss EV production by dying tumor cells and consider the possible roles of Apo-EVs in a cell death-driven sector of the tumor microenvironment known as the onco-regenerative niche (ORN). We propose that tumor-derived Apo-EVs are significant vehicles of the ORN, functioning as critical intercellular communicators that activate oncogenic tissue repair and regeneration pathways. We highlight important outstanding questions and suggest that Apo-EVs may harbor novel therapeutic targets.


Assuntos
Apoptose , Micropartículas Derivadas de Células/metabolismo , Vesículas Extracelulares/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Microambiente Tumoral , Animais , Transporte Biológico , Biomarcadores , Humanos , Neoplasias/genética , Transdução de Sinais
17.
Oncoimmunology ; 7(6): e1434468, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29872574

RESUMO

Cancer immunotherapists have been searching for biomarkers predicting patient responses to PD-1/PD-L1 blockade in neoplastic cells as well as in the immune system. Now, accumulating evidence indicates that the composition of the intestinal microflora has a major impact on patient prognosis. Here, we enumerate the bacterial species that are associated with favorable outcome of immunotherapy.

18.
Oncoimmunology ; 7(7): e1445454, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29900057

RESUMO

D-2-hydroxyglutarate (D-2HG) is released by various types of malignant cells including acute myeloid leukemia (AML) blasts carrying isocitrate dehydrogenase (IDH) gain-of-function mutations. D-2HG acting as an oncometabolite promotes proliferation, anoikis, and differentiation block of hematopoietic cells in an autocrine fashion. However, prognostic impact of IDH mutations and high D-2HG levels remains controversial and might depend on the overall mutational context. An increasing number of studies focus on the permissive environment created by AML blasts to promote immune evasion. Impact of D-2HG on immune cells remains incompletely understood. Here, we sought out to investigate the effects of D-2HG on T-cells as key mediators of anti-AML immunity. D-2HG was efficiently taken up by T-cells in vitro, which is in line with high 2-HG levels measured in T-cells isolated from AML patients carrying IDH mutations. T-cell activation was slightly impacted by D-2HG. However, D-2HG triggered HIF-1a protein destabilization resulting in metabolic skewing towards oxidative phosphorylation, increased regulatory T-cell (Treg) frequency, and reduced T helper 17 (Th17) polarization. Our data suggest for the first time that D-2HG might contribute to fine tuning of immune responses.

19.
Oncoimmunology ; 7(8): e1458810, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30221055

RESUMO

mTOR inhibition can promote or inhibit immune responses in a context dependent manner, but whether this will represent a net benefit or be contraindicated in the context of immunooncology therapies is less understood. Here, we report that the mTORC1/2 dual kinase inhibitor vistusertib (AZD2014) potentiates anti-tumour immunity in combination with anti-CTLA-4 (αCTLA-4), αPD-1 or αPD-L1 immune checkpoint blockade. Combination of vistusertib and immune checkpoint blocking antibodies led to tumour growth inhibition and improved survival of MC-38 or CT-26 pre-clinical syngeneic tumour models, whereas monotherapies were less effective. Underlying these combinatorial effects, vistusertib/immune checkpoint combinations reduced the occurrence of exhausted phenotype tumour infiltrating lymphocytes (TILs), whilst increasing frequencies of activated Th1 polarized T-cells in tumours. Vistusertib alone was shown to promote a Th1 polarizing proinflammatory cytokine profile by innate primary immune cells. Moreover, vistusertib directly enhanced activation of effector T-cell and survival, an effect that was critically dependent on inhibitor dose. Therefore, these data highlight direct, tumour-relevant immune potentiating benefits of mTOR inhibition that complement immune checkpoint blockade. Together, these data provide a clear rationale to investigate such combinations in the clinic.

20.
Oncoimmunology ; 7(12): e1486353, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30524887

RESUMO

Tryptophan (Trp) metabolism is an important target in immuno-oncology as it represents a powerful immunosuppressive mechanism hijacked by tumors for protection against immune destruction. However, it remains unclear how tumor cells can proliferate while degrading the essential amino acid Trp. Trp is incorporated into proteins after it is attached to its tRNA by tryptophanyl-tRNA synthestases. As the tryptophanyl-tRNA synthestases compete for Trp with the Trp-catabolizing enzymes, the balance between these enzymes will determine whether Trp is used for protein synthesis or is degraded. In human cancers expression of the Trp-degrading enzymes indoleamine-2,3-dioxygenase-1 (IDO1) and tryptophan-2,3-dioxygenase (TDO2) was positively associated with the expression of the tryptophanyl-tRNA synthestase WARS. One mechanism underlying the association between IDO1 and WARS identified in this study is their joint induction by IFNγ released from tumor-infiltrating T cells. Moreover, we show here that IDO1- and TDO2-mediated Trp deprivation upregulates WARS expression by activating the general control non-derepressible-2 (GCN2) kinase, leading to phosphorylation of the eukaryotic translation initiation factor 2α (eIF2α) and induction of activating transcription factor 4 (ATF4). Trp deprivation induced cytoplasmic WARS expression but did not increase nuclear or extracellular WARS levels. GCN2 protected the cells against the effects of Trp starvation and enabled them to quickly make use of Trp for proliferation once it was replenished. Computational modeling of Trp metabolism revealed that Trp deficiency shifted Trp flux towards WARS and protein synthesis. Our data therefore suggest that the upregulation of WARS via IFNγ and/or GCN2-peIF2α-ATF4 signaling protects Trp-degrading cancer cells from excessive intracellular Trp depletion.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA