Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 69
Filtrar
1.
J Leukoc Biol ; 2024 Apr 16.
Artigo em Inglês | MEDLINE | ID: mdl-38626292

RESUMO

Conventional treatments have shown a limited efficacy for pancreatic cancer, and immunotherapy is an emerging option for treatment of this highly fatal malignancy. Neoantigen is critical to improving the efficacy of tumor-specific immunotherapy. The cancer and peripheral blood specimens from human leukocyte antigen (HLA)-A0201 positive pancreatic cancer patient were subjected to next-generation sequencing and bioinformatics analyses were performed to screen high-affinity and highly stable neoepitopes. The activation of cytotoxic T lymphocytes (CTLs) by the mutBCL2A111-20 neoepitope targeting B-cell lymphoma 2-related protein A1 (BCL2A1) mutant epitope was investigated, and the cytotoxicity of mutBCL2A111-20 neoepitope-specific CTLs to pancreatic cancer cells was evaluated. The mutBCL2A111-20 neoepitope was found to present a high immunogenicity and induce CTLs activation and proliferation, and was cytotoxic to mutBCL2A111-20 neoepitope-loaded T2 cells and pancreatic cancer PANC-1-Neo and A2-BxPC-3-Neo cells that overexpressed mutBCL2A111-20 neoepitopes, appearing a targeting neoepitope specificity. In addition, high BCL2A1 expression correlated with a low 5-year progress free interval (PFI) among pancreatic cancer patients. Our findings provide experimental supports to individualized T-cell therapy targeting mutBCL2A111-20 neoepitopes, and provide an option of immunotherapy for pancreatic cancer.

2.
Int Immunopharmacol ; 128: 111477, 2024 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-38183910

RESUMO

Hepatocellular carcinoma (HCC) exhibits high incidence and mortality rates in China. Most cases are often diagnosed at late stages and require multi-strategy therapies. In recent years, immune checkpoint inhibitors (ICIs), particularly programmed cell death protein 1 (PD-1) antibodies, have demonstrated effectiveness in comprehensive HCC treatment. However, the efficacy and prognosis vary greatly among patients. Screening suitable patients and predicting outcomes are crucial for improving the efficacy of ICIs. Although PD-L1 expression levels in tumor cells have been used as predictors of PD-1/PD-L1 antibody therapy, they may not consistently correlate with clinical response in some studies; thus, exploring new biomarkers is necessary. The neutrophil-to-lymphocyte ratio (NLR) emerged as a new predictor of ICI immunotherapy efficacy, and its application in HCC is worth exploring. This study utilizes the Cancer Genome Atlas Liver Hepatocellular Carcinoma Collection (TCGA-LIHC) project in the Genomic Data Commons (GDC) database for methylation and transcriptome data analysis. The correlation between NLR and ICI immunotherapy efficacy for HCC was evaluated, identifying differentially expressed genes. Analysis revealed 74 up-regulated and 445 down-regulated genes in the high-NLR group compared to the low-NLR group. NLR-related differential methylation analysis identified 68 hypermethylated and 65 hypomethylated probes in the NLR high group. Furthermore, a machine learning model using 27 intersecting genes predicted PD-1 antibody therapy efficacy, achieving an AUC value of 0.813. In summary, we established a predictive model for HCC immunotherapy based on 27 genes related to differential expressions and NLR-associated methylation, showing significant potential for clinical research potential in this field.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Carcinoma Hepatocelular/patologia , Antígeno B7-H1 , Receptor de Morte Celular Programada 1 , Neoplasias Hepáticas/patologia , Neutrófilos/patologia , Linfócitos/patologia
3.
Artigo em Inglês | MEDLINE | ID: mdl-37807645

RESUMO

BACKGROUND: Cellular senescence (CS) is thought to be the primary cause of cancer development and progression. This study aimed to investigate the prognostic role and molecular subtypes of CS-associated genes in gastric cancer (GC). METHODS: The CellAge database was utilized to acquire CS-related genes. Expression data and clinical information of GC patients were obtained from The Cancer Genome Atlas (TCGA) database. Patients were then grouped into distinct subtypes using the "ConsesusClusterPlus" R package based on CS-related genes. An in-depth analysis was conducted to assess the gene expression, molecular function, prognosis, gene mutation, immune infiltration, and drug resistance of each subtype. In addition, a CS-associated risk model was developed based on Cox regression analysis. The nomogram, constructed on the basis of the risk score and clinical factors, was formulated to improve the clinical application of GC patients. Finally, several candidate drugs were screened based on the Cancer Therapeutics Response Portal (CTRP) and PRISM Repurposing dataset. RESULTS: According to the cluster result, patients were categorized into two molecular subtypes (C1 and C2). The two subtypes revealed distinct expression levels, overall survival (OS) and clinical presentations, mutation profiles, tumor microenvironment (TME), and drug resistance. A risk model was developed by selecting eight genes from the differential expression genes (DEGs) between two molecular subtypes. Patients with GC were categorized into two risk groups, with the high-risk group exhibiting a poor prognosis, a higher TME level, and increased expression of immune checkpoints. Function enrichment results suggested that genes were enriched in DNA repaired pathway in the low-risk group. Moreover, the Tumor Immune Dysfunction and Exclusion (TIDE) analysis indicated that immunotherapy is likely to be more beneficial for patients in the low-risk group. Drug analysis results revealed that several drugs, including ML210, ML162, dasatinib, idronoxil, and temsirolimus, may contribute to the treatment of GC patients in the high-risk group. Moreover, the risk model genes presented a distinct expression in single-cell levels in the GSE150290 dataset. CONCLUSION: The two molecular subtypes, with their own individual OS rate, expression patterns, and immune infiltration, lay the foundation for further exploration into the GC molecular mechanism. The eight gene signatures could effectively predict the GC prognosis and can serve as reliable markers for GC patients.

4.
BMC Cancer ; 23(1): 429, 2023 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-37170184

RESUMO

OBJECTIVE: To study the effect of inhibitor of differentiation 3 (ID3) on radiotherapy in patients with rectal cancer and to explore its primary mechanism. METHODS: Cell proliferation and clonogenic assays were used to study the relationship between ID3 and radiosensitivity. Co-immunoprecipitation and immunofluorescence were performed to analyze the possible mechanism of ID3 in the radiosensitivity of colorectal cancer. At the same time, a xenograft tumor model of HCT116 cells in nude mice was established to study the effect of irradiation on the tumorigenesis of ID3 knockdown colorectal cancer cells in vivo. Immunohistochemistry was performed to analyze the relationship between ID3 expression and the efficacy of radiotherapy in 46 patients with rectal cancer. RESULTS: Proliferation and clonogenic assays revealed that the radiosensitivity of colorectal cancer cells decreased with ID3 depletion through p53-independent pathway. With the decrease in ID3 expression, MDC1 was downregulated. Furthermore, the expression of ID3, MDC1, and γH2AX increased and formed foci after irradiation. ID3 interacted with PPARγ and form a positive feedback loop to enhance the effect of ID3 on the radiosensitivity of colorectal cancer. Irradiation tests in nude mice also confirmed that HCT116 cells with ID3 knockdown were more affected by irradiation. Immunohistochemical study showed that rectal cancer patients with low expression of ID3 had better radiotherapy efficacy. CONCLUSIONS: ID3 and PPARγ influence the radiosensitivity of colorectal cancer cells by interacting with MDC1 to form a positive feedback loop that promotes DNA damage repair. Patients with low expression of ID3 who received neoadjuvant chemoradiotherapy can obtain a better curative effect.


Assuntos
Reparo do DNA , PPAR gama , Neoplasias Retais , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Dano ao DNA , Reparo do DNA/genética , Retroalimentação , Proteínas Inibidoras de Diferenciação/genética , Camundongos Nus , Proteínas de Neoplasias/genética , PPAR gama/genética , Tolerância a Radiação/genética , Neoplasias Retais/genética , Neoplasias Retais/radioterapia
5.
Eur J Immunol ; 53(5): e2250054, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36794567

RESUMO

High-frequency mutation of the TP53 tumor suppressor gene is observed in multiple human cancers, which promotes cancer progression. However, the mutated gene-encoded protein may serve as a tumor antigen to elicit tumor-specific immune responses. In this study, we detected widespread expression of shared TP53-Y220C neoantigen in hepatocellular carcinoma with low affinity and low stability of binding to HLA-A0201 molecules. We substituted the amino acid sequences VVPCEPPEV with VLPCEPPEV in the TP53-Y220C neoantigen to yield a TP53-Y220C (L2) neoantigen. This altered neoantigen was found to increase affinity and stability and induce more cytotoxic T lymphocytes (CTLs), indicating improvements in immunogenicity. In vitro assays showed the cytotoxicity of CTLs stimulated by both TP53-Y220C and TP53-Y220C (L2) neoantigens against multiple HLA-A0201-positive cancer cells expressing TP53-Y220C neoantigens; however, the TP53-Y220C (L2) neoantigen showed higher cytotoxicity than the TP53-Y220C neoantigen against cancer cells. More importantly, in vivo assays demonstrated greater inhibition of hepatocellular carcinoma cell proliferation by TP53-Y220C (L2) neoantigen-specific CTLs relative to TP53-Y220C neoantigen in zebrafish and nonobese diabetic/severe combined immune deficiency mouse models. The results of this study demonstrate enhanced immunogenicity of the shared TP53-Y220C (L2) neoantigen, which has the potential as dendritic cells or peptide vaccines for multiple cancers.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animais , Camundongos , Humanos , Linfócitos T Citotóxicos , Antígeno HLA-A2/genética , Epitopos , Peixe-Zebra , Antígenos de Neoplasias , Citotoxicidade Imunológica , Proteína Supressora de Tumor p53/genética
6.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-977722

RESUMO

@#[摘 要] 目的:探究含硬化蛋白域蛋白1(SOSTDC1)对宫颈癌细胞恶性生物学行为的调控及其分子机制。方法:收集2020年8月至2022年5月间在福建省肿瘤医院活检或手术切除的53例宫颈癌组织和相应的癌旁组织标本,免疫组化法检测SOSTDC1蛋白在宫颈癌组织及相应癌旁组织中的表达,qPCR法检测正常宫颈细胞、宫颈癌细胞中SOSTDC1 mRNA表达;将SOSTDC1过表达慢病毒(OE-sostdc1)和对照空病毒(NC)感染宫颈癌细胞SiHa及CaSki,将其分为SiHa-OE-sostdc1、SiHa-NC、CaSki-OE-sostdc1、CaSki-NC组,采用WST-1法、细胞集落形成实验、Transwell实验和WB法检测转染各组SiHa及CaSki细胞的增殖、集落形成、迁移和侵袭能力和BMP、Wnt/β-catenin信号途径相关蛋白及上皮-间充质转化(EMT)相关蛋白的表达。用DNA甲基化酶抑制剂5-氮杂2'-脱氧胞苷(5'-Aza-CdR)处理宫颈癌细胞后采用qPCR和WB法检测SOSTDC1 mRNA及蛋白的表达变化,用甲基化特异性PCR(MSP)检测5例配对宫颈癌组织与癌旁组织中SOSTDC1基因启动子区甲基化水平,同时qPCR检测其SOSTDC1 mRNA水平。结果:与癌旁组织比较,SOSTDC1蛋白在宫颈癌组织中呈低表达(P<0.01),且与淋巴结转移与FIGO分期有关联(均P<0.05);与正常宫颈HUCEC细胞比较,SOSTDC1 mRNA在宫颈癌C33A、HeLa、SiHa、CaSki细胞中均呈低表达(均P<0.01)。过表达SOSTDC1显著抑制SiHa及CaSki细胞的增殖、迁移和侵袭能力(均P<0.05)。WB法结果检测显示,过表达SOSTDC1显著抑制SiHa及CaSki细胞中磷酸化Smad、Dvl2/3、β-catenin、VIM、N-cadherin、Snail蛋白的表达(均P<0.05),5'-Aza-CdR处理后的SiHa及CaSki细胞中SOSTDC1 mRNA和蛋白水平均显著增加(均P<0.05),MSP检测结果显示,相较于癌旁组织,宫颈癌组织中SOSTDC1基因启动子区呈高度甲基化,且SOSTDC1 mRNA水平降低(P<0.01)。结论:SOSTDC1在宫颈癌组织中呈低表达且与肿瘤的恶性进展关联,其表达下调与其基因启动子区高度甲基化有关,过表达SOSTDC1可能通过阻断BMP及Wnt/β-catenin信号通路从而抑制SiHa、CaSki细胞的增殖、侵袭和迁移能力。

7.
J Leukoc Biol ; 112(6): 1577-1590, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36222123

RESUMO

The efficacy of conventional treatments for pancreatic cancer remains unsatisfactory, and immunotherapy is an emerging option for adjuvant treatment of this highly deadly disorder. The tumor-associated antigen (TAA) MUC1 is expressed in a variety of human cancers and is overexpressed in more than 90% of pancreatic cancer, which makes it an attractive target for cancer immunotherapy. As a self-protein, MUC1 shows a low immunogenicity because of immune tolerance, and the most effective approach to breaking immune tolerance is alteration of the antigen structure. In this study, the altered MUC11068-1076Y1 epitope (YLQRDISEM) by modification of amino acid residues in sequences presented a higher immunogenicity and elicited more CTLs relative to the wild-type (WT) MUC11068-1076 epitope (ELQRDISEM). In addition, the altered MUC11068-1076Y1 epitope was found to cross-recognize pancreatic cancer cells expressing WT MUC1 peptides in an HLA-A0201-restricted manner and trigger stronger immune responses against pancreatic cancer via the perforin/granzyme apoptosis pathway. As a potential HLA-A0201-restricted CTL epitope, the altered MUC11068-1076Y1 epitope is considered as a promising target for immunotherapy of pancreatic cancer. Alteration of epitope residues may be feasible to solve the problem of the low immunogenicity of TAA and break immune tolerance to induce immune responses against human cancers.


Assuntos
Neoplasias Pancreáticas , Linfócitos T Citotóxicos , Humanos , Antígenos de Neoplasias , Epitopos , Imunoterapia , Mucina-1/genética , Neoplasias Pancreáticas/terapia , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas
8.
BMC Cancer ; 22(1): 549, 2022 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-35568845

RESUMO

BACKGROUND: Accumulated evidence suggests that RING finger proteins (RNFs) are involved in the carcinogenesis of cancers. However, RNF38, a member of the RNF protein family, has not been studied in nasopharyngeal carcinoma (NPC). METHODS: RNF38 expression was analyzed by RT-PCR, Western blotting and Immunohistochemistry. Biological functions of RNF38 were evaluated by cell growth, colony formation, apoptosis, migration and invasion assays in vitro. Xenograft growth and lung metastasis models were conducted to investigate the effect of RNF38 in vivo. Liquid chromatography coupled with tandem mass spectrometry, co-immunoprecipitation, and CHX assay were implemented to detect the interaction among RNF38 and ACTN4. RESULTS: RNF38 was significantly downregulated in NPC cells and tissues. Immunohistochemistry implied that loss of RNF38 was an independent prognostic factor for poor outcomes of NPC patients. Gain- and loss-of-function experiments showed that RNF38 inhibited proliferation and metastasis in NPC in vitro and in vivo. Upregulation of RNF38 promoted apoptosis of NPC cells to etoposide but not cisplatin. ACTN4 was upregulated in NPC and negatively correlated with RNF38. Mechanistic investigations suggested that RNF38 inactivates the NF-𝛋B and ERK1/2 signaling pathways by inducing ubiquitination and degradation of ACTN4. RNF38 suppress the development of NPC by interacting with ACTN4. CONCLUSIONS: RNF38 plays a potential cancer suppressor gene role in NPC tumorigenesis and is a prognostic biomarker in NPC.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias Nasofaríngeas , Actinina/genética , Actinina/metabolismo , Carcinogênese/genética , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Humanos , Carcinoma Nasofaríngeo/patologia , Neoplasias Nasofaríngeas/patologia , Ubiquitinação
9.
J Gastroenterol ; 57(5): 372-386, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35244769

RESUMO

BACKGROUND: Upregulated Kindlin-2 expression in hepatocellular carcinoma (HCC) correlates with metastasis and poor prognosis. In this study, we investigated the molecular mechanism of Kindlin-2 in HCC. METHODS: Kindlin-2 downstream pathways were explored through microRNA sequencing. The Kindlin-2-miR-1258-TCF4 axis was verified using bisulfite sequencing, a luciferase reporter assay, quantitative real-time PCR, and rescue assays. Binding of TCF4 to the Kindlin-2 promoter was confirmed by promoter activity analysis and chromatin immunoprecipitation. RESULTS: MiRNA sequencing identified miR-1258 as a downstream effector of Kindlin-2. MiR-1258 expression was increased following Kindlin-2 knockdown and decreased after Kindlin-2 overexpression. Next, we identified transcription factor 7 like 2 (TCF7L2 or TCF4) as a target of miR-1258 and found that Kindlin-2 upregulated TCF4 expression by epigenetically suppressing miR-1258 in HCC. Furthermore, our results suggest that TCF4 binds to the Kindlin-2 promotor to enhance its transcription. Therefore, Kindlin-2-miR-1258-TCF4 interaction creates a positive feedback loop. Functional assays and animal experiments demonstrated critical roles of miR-1258 and TCF4 in HCC cell migration in vitro and HCC metastasis in vivo. In HCC tissues, Kindlin-2 expression correlated negatively with miR-1258 expression and positively with TCF4 expression. Meanwhile, miR-1258 expression correlated negatively with TCF4 expression. CONCLUSIONS: This study illustrates a novel integrin-independent signaling pathway, Kindlin-2-miR-1258-TCF4, that regulates HCC invasion and metastasis and identifies Kindlin-2 as a promising therapeutic target in HCC.


Assuntos
Carcinoma Hepatocelular , Neoplasias Hepáticas , Proteínas de Membrana , MicroRNAs , Proteínas de Neoplasias , Fator de Transcrição 4 , Animais , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Retroalimentação , Retroalimentação Sensorial , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Proteínas de Membrana/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Metástase Neoplásica , Proteínas de Neoplasias/metabolismo , Fator de Transcrição 4/genética , Fator de Transcrição 4/metabolismo
11.
Gene ; 819: 146240, 2022 Apr 20.
Artigo em Inglês | MEDLINE | ID: mdl-35114275

RESUMO

BACKGROUND: Liver metastasis of colorectal cancer (CRC) remains high mortality and the mechanism is still unknown. Here we investigated the effects of inhibitor of DNA binding 2 (Id2) on growth and liver metastasis of CRC. METHODS: qPCR and western blotting were used to demonstrate mRNA and protein expressions in Id2-knockdown HCT116 cells. Cell growth was observed by cell proliferation assay, colony formation assay and flow cytometry. Cell migration and invasion were observed with wound healing assay and transwell migration and invasion assay. The effects of Id2 knockdown on tumor growth and liver metastasis in vivo were evaluated respectively with subcutaneous tumor model and colorectal liver metastasis model by injecting HCT116 cells into the mesentery triangle of cecum in mice. RESULTS: Id2 overexpression was found in CRC cell lines. Id2 knockdown resulted in a reduction in the proliferation, colony formation, migration and invasion of HCT116 cells. The suppression of cell proliferation was accompanied by the cell cycle arrest in the G0/G1 phase with down-regulation of Cyclin D1, Cyclin E, p-Cdk2/3, Cdk6, p-p27 and up-regulation of p21 and p27. Id2 knockdown reversed epithelial-mesenchymal transition (EMT) through increasing E-Cadherin and inhibiting N-Cadherin, Vimentin, ß-catenin, Snail and Slug. Id2 was also found to inhibit CRC metastasis via MMP2, MMP9 and TIMP-1. Furthermore, Id2 knockdown suppressed CRC liver metastasis in vivo. CONCLUSION: Id2 promotes CRC growth through activation of the PI3K/AKT signaling pathway, and triggers EMT to enhance CRC migration and invasion.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Transição Epitelial-Mesenquimal , Proteína 2 Inibidora de Diferenciação/genética , Proteína 2 Inibidora de Diferenciação/metabolismo , Fígado/metabolismo , Metástase Neoplásica , Animais , Ciclo Celular , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Modelos Animais de Doenças , Feminino , Regulação Neoplásica da Expressão Gênica , Técnicas de Silenciamento de Genes/métodos , Células HCT116 , Humanos , Neoplasias Hepáticas/secundário , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais , Organismos Livres de Patógenos Específicos
12.
Front Immunol ; 13: 1002361, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36618357

RESUMO

Chimeric antigen receptor (CAR) T cells have been proven effective for the treatment of B-cell-mediated malignancies. Currently, the development of efficient tools that supply CAR T cells for the treatment of other malignancies would have great impact. In this study, interleukin (IL)-15 and C-C motif chemokine ligand 19 (CCL19) were introduced into natural killer group 2D (NKG2D)-based CARs to generate 15×19 CAR T cells, which remarkably increased T-cell expansion and promoted the production of central memory T (Tcm) cells. 15×19 CAR T cells showed greater cytotoxicity to gastric cell lines than conventional CAR T cells and produced higher levels of IL-15 and CCL-19, which resulted in increased responder T cell chemotaxis and reduced expression of T cell exhaustion markers. A live zebrafish model was used for single-cell visualization of local cytotoxicity and metastatic cancers. Administration of 15×19 CAR T cells resulted in significant shrinking of gastric cancer xenograft tumors and expansion of 15×19 CAR T cells in zebrafish models. Taken together, these findings demonstrate that 15×19 CAR T cells are highly efficient in killing gastric cancer cells, are effective to avoid off-target effects, and migrate to local and metastatic sites for long-term surveillance of cancers.


Assuntos
Antineoplásicos , Imunoterapia , Receptores de Antígenos Quiméricos , Neoplasias Gástricas , Animais , Humanos , Linhagem Celular Tumoral , Xenoenxertos , Interleucina-15/metabolismo , Ligantes , Neoplasias Gástricas/terapia , Neoplasias Gástricas/metabolismo , Linfócitos T , Peixe-Zebra/metabolismo , Quimiocina CCL19/metabolismo
13.
Xi Bao Yu Fen Zi Mian Yi Xue Za Zhi ; 37(10): 872-880, 2021 Oct.
Artigo em Chinês | MEDLINE | ID: mdl-34670663

RESUMO

Objective To compare the effect of different cytokine combinations combined with anti-CD3/CD28 beads in vitro inducing the generation of central memory T cell (Tcm). Methods Peripheral blood mononuclear cells (PBMCs) were isolated from healthy donors. Naive CD8+ T cells were purified using immunomagnetic beads and stimulated with CD3/CD28 antibody for 48 hours. Cells were treated with different cytokine combinations as follows: Interleukin-2(IL-2), IL-7/IL-15, IL-7/IL-15/IL-21, and IL-7/IL-15/IL-21/IL-23. The automatic blood cell counting instrument was used for cell counting. 5, 6-carboxyfluorescein diacetate succinimidyl ester (CFSE)was employed to test the cell proliferation and flow cytometry was adopted to measure the immune memory phenotype, apoptosis and intracellular factor expression of CD8+ T cells induced by different cytokine combinations. The expression of Bcl-2 was determined by Western blot analysis. Results Unlike other cytokine combinations, IL-7/IL-15/IL-21/IL-23 promoted the proliferation of CD8+ T cells and significantly increased the expression of CD8+CD62L+CD45RA- central memory T cell subsets. At the same time, IL-7/IL-15/IL-21/IL-23 treatment significantly reduced the secretion levels of IFN-γ, perforin, and granzyme B. The level of cell apoptosis was also significantly decreased. Conclusion The cytokines combination of IL-7/IL-15/IL-21/IL-23 can effectively induce the differentiation of naive CD8+T cells to CD8+ Tcm cells in vitro, which provides a new strategy for the generation of human CD8+ Tcm in immunotherapy.


Assuntos
Memória Imunológica , Interleucina-7 , Linfócitos T CD8-Positivos , Humanos , Interleucina-15 , Interleucina-23 , Interleucinas , Leucócitos Mononucleares
14.
J Oncol ; 2021: 6660486, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33936204

RESUMO

Id4 is one of the inhibitors of DNA-binding proteins (Id) and involved in the pathogenesis of numerous cancers. The specific mechanism underlying the Id4-mediated regulation of proliferation, invasion, and metastasis of colorectal cancer (CRC) cells is still largely unclear. In the present study, results showed CRC cells had a lower baseline Id4 expression than normal intestinal epithelial NCM460 cells. In order to explore the role of Id4 in the tumorigenicity, CRC HCT116 cells with stable Id4 expression were used, and results showed Id4 overexpression arrested the cell cycle at the G0/G1 phase, inhibited the cell proliferation and the colony formation, as well as suppressed the migration and invasion. In the in vivo model, Id4 overexpression inhibited the tumor growth and metastasis in the nude mice. Furthermore, Id4 overexpression upregulated the expression of proteins associated with cell proliferation, inhibited the PI3K/AKT pathway, and suppressed epithelial-mesenchymal transition (EMT) of HCT116 cells. Moreover, Id4 significantly decreased cytokeratin 18 (CK18) expression, but CK18 overexpression in Id4 expressing HCT116-Id4 cells rescued the activation of AKT, p-AKT, MMP2, MMP7, and E-cadherin. Collectively, our study indicated Id4 may inhibit CRC growth and metastasis through inhibiting the PI3K/AKT pathway in a CK18-dependent manner and suppressing EMT. Id4 may become a target for the treatment of CRC.

15.
Cancer Biol Med ; 18(4)2021 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-35959968

RESUMO

OBJECTIVE: Neoantigens arising from gene mutations in tumors can induce specific immune responses, and neoantigen-based immunotherapies have been tested in clinical trials. Here, we characterized the efficacy of altered neoepitopes in improving immunogenicity against gastric cancer. METHODS: Raw data of whole-exome sequencing derived from a patient with gastric cancer were analyzed using bioinformatics methods to identify neoepitopes. Neoepitopes were modified by P1Y (the first amino acid was replaced by tyrosine) and P2L (the second amino acid was replaced by leucine). T2 binding and stability assays were used to detect the affinities between the neoepitopes and the HLA molecules, as well as the stabilities of complexes. Dendritic cells (DCs) presented with neoepitopes stimulated naïve CD8+ T cells to induce specific cytotoxic T lymphocytes. ELISA and carboxyfluorescein succinimidyl ester were used to detect IFN-γ and TNF-α levels, and T cell proliferation. Perforin was detected by flow cytometry. The cytotoxicity of T cells was determined using the lactate dehydrogenase assay. RESULTS: Bioinformatics analysis, T2 binding, and stability assays indicated that residue substitution increased the affinity between neoepitopes and HLA molecules, as well as the stabilities of complexes. DCs presented with altered neoepitopes stimulated CD8+T cells to release more IFN-γ and had a greater effect on promoting proliferation than wild-type neoepitopes. CD8+T cells stimulated with altered neoepitopes killed more wild-type neoepitope-pulsed T2 cells than those stimulated with wild-type neoepitopes, by secreting more IFN-γ, TNF-α, and perforin. CONCLUSIONS: Altered neoepitopes exhibited greater immunogenicity than wild-type neoepitopes. Residue substitution could be used as a new strategy for immunotherapy to target neoantigens.


Assuntos
Linfócitos T CD8-Positivos , Neoplasias Gástricas , Aminoácidos/metabolismo , Humanos , Perforina/metabolismo , Neoplasias Gástricas/genética , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/terapia , Fator de Necrose Tumoral alfa/metabolismo
17.
Pathol Res Pract ; 216(11): 153144, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32911346

RESUMO

Non-small cell lung cancer (NSCLC) accounting for 85 % of all lung cancer was one of the main causes of death worldwide. In this study, we investigated the role of circRNA_102179 in NSCLC development. The levels of circRNA_102179 in NSCLC tissues and cell lines were determined by quantitative real-time PCR assay (qRT-PCR). CCK8 and colony formation assays were applied to explore the effect of circRNA_102179 on the growth of NSCLC cells in vitro. Transwell assay was utilized to analyze the impact of circRNA_102179 on the migration and invasion of NSCLC cells. Target prediction and luciferase reporter assay were used to identify the interacting miRNA of circRNA_102179. The interaction among circRNA_102179/ miR-330-5p/HMGB3 was further validated by colony formation and Transwell invasion assays. Finally, the mouse xenograft NSCLC model was used to explore the role of circRNA_102179 in the tumor growth of NSCLC cells in vivo. CircRNA_102179 was overexpressed in NSCLC tissues and cells compared with normal lung tissues and human bronchial epithelial cells (HBEs). The down-regulation of circRNA_102179 markedly reduced the proliferation, migration, and invasion of NSCLC cells. Moreover, down-expression of circRNA_102179 significantly increased the level of miR-330-5p/HMGB3 in NSCLC cells. Further functional experiments indicated that over-expression of miR-330-5p reversed the inhibitory effect of circRNA_102179 on NSCLC cells growth, migration, and invasion. Our results reveal that circRNA_102179 facilitates the proliferation, migration, and invasion of NSCLC cell via modulating miR-330-5p/ HMGB3 axis in NSCLC cells.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/patologia , Proteína HMGB3/genética , Neoplasias Pulmonares/patologia , MicroRNAs/genética , RNA Circular/genética , Animais , Biomarcadores Tumorais/análise , Biomarcadores Tumorais/genética , Carcinoma Pulmonar de Células não Pequenas/genética , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Proteína HMGB3/metabolismo , Xenoenxertos , Humanos , Neoplasias Pulmonares/genética , Camundongos , Invasividade Neoplásica/genética
18.
J Clin Lab Anal ; 34(8): e23350, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32672362

RESUMO

OBJECTIVE: This study aimed to explore the association of A kinase-interacting protein 1 (AKIP1) expression with clinicopathological characteristics and prognosis in gastric cancer patients. METHODS: Data of 260 gastric cancer patients were retrospectively reviewed. AKIP1 expression in tumor tissue and non-cancerous tissue specimens was detected by immunohistochemistry and semi-quantitatively scored according to the staining intensity and density. Moreover, the clinicopathological features were retrieved, and disease-free survival (DFS) and overall survival (OS) were calculated. RESULTS: A kinase-interacting protein 1 expression was increased in tumor tissues compared with non-cancerous tissues (P < .001). In terms of tumor features, tumor AKIP1 high expression correlated with elevated T stage (P < .001) and raised TNM stage (P = .042), while did not correlate with pathological grade (P > .999), tumor size (P = .060), N stage (P = .180), or tumor location (P > .999). Meanwhile, tumor AKIP1 was not associated with the non-tumor features either. Kaplan-Meier curves disclosed that AKIP1 high expression patients had shorter DFS (P = .004) and OS (P = .043) compared with AKIP1 low expression patients. Univariate Cox's regression showed that AKIP1 high expression correlated with shorter DFS (P = .005, hazard ratio [HR] = 1.635) and OS (P = .046, HR = 1.519), whereas multivariate Cox's regression displayed that AKIP1 did not independently predict worse DFS (P = .172, HR = 1.276) or shorter OS (P = .433, HR = 1.183). CONCLUSION: A kinase-interacting protein 1 may serve as a potential biomarker for deteriorative tumor features and poor prognosis in gastric cancer patients.

19.
Cancer Sci ; 111(5): 1711-1723, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32155300

RESUMO

Epstein-Barr virus (EBV) BamHI A rightward transcripts (BART) encoded microRNAs (EBV-miR-BARTs) are abnormally highly expressed in nasopharyngeal carcinoma (NPC). This study aims to investigate the diagnostic and prognostic performance of miR-BART7-3p and miR-BART13-3p. Plasma levels of EBV DNA, miR-BART7-3p, and miR-BART13-3p were examined by quantitative PCR in 483 treatment-naïve NPC patients and 243 controls without NPC. The prognostic performance was examined by comparing plasma levels with rates of distant metastasis during follow-up. The area under the receiver operating characteristic curve for diagnosing NPC was 0.926 for EBV DNA, 0.964 for plasma miR-BART7-3p, 0.973 for miR-BART13-3p, and 0.997 for all three indices. Among 465 NPC patients without distant metastasis, the above-median miR-BART7-3p and EBV DNA were independent risk for shorter distant metastasis-free survival (DMFS) (hazard ratio [HR] = 2.94, 95% confidence interval [CI], 1.44-5.97, P = .003; HR = 2.27, 95% CI, 1.26-4.10, P = .006) in multivariate Cox regression. Epstein-Barr virus DNA, miR-BART7-3p, and miR-BART13-3p after radiotherapy were detectable in 28.6%, 17.6%, and 54.7% of patients, respectively. In multivariate Cox regression, detectable miR-BART7-3p and EBV DNA were independent risks for shorter DMFS (HR = 4.13, 95% CI, 1.89-9.01, P < .001; HR = 2.14, 95% CI, 1.04-4.42, P = .039). The 4-year DMFS rate was 92.0% in subjects (n = 156) with neither detectable miR-BART7-3p nor EBV DNA, 80.0% in subjects (n = 65) with either detectable miR-BART7-3p or EBV DNA, and 52.9% in subjects (n = 24) with both detectable miR-BART7-3p and EBV DNA after radiotherapy (P < .001). Circulating levels of miR-BART7-3p and miR-BART13-3p show excellent diagnostic performance for NPC. The combination of plasma levels of miR-BART7-3p and EBV DNA at diagnosis and after radiotherapy could help stratify patients by risk of poor DMFS.


Assuntos
MicroRNA Circulante/sangue , Herpesvirus Humano 4/genética , Carcinoma Nasofaríngeo/diagnóstico , Neoplasias Nasofaríngeas/diagnóstico , RNA Viral/sangue , Adulto , Idoso , Biomarcadores Tumorais/sangue , DNA Viral/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Carcinoma Nasofaríngeo/sangue , Carcinoma Nasofaríngeo/patologia , Carcinoma Nasofaríngeo/terapia , Neoplasias Nasofaríngeas/sangue , Neoplasias Nasofaríngeas/patologia , Neoplasias Nasofaríngeas/terapia , Prognóstico , Análise de Sobrevida , Adulto Jovem
20.
Onco Targets Ther ; 13: 1757-1765, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32161471

RESUMO

PURPOSE: Immune checkpoint proteins in the tumor microenvironment can enter the blood circulation and are potential markers for liquid biopsy. The aims of this study were to explore differences in immune checkpoint protein expression between patients with nasopharyngeal carcinoma (NPC) and healthy controls and to investigate the prognostic value of the soluble form of programmed death-ligand 1 (sPD-L1) in NPC. METHODS: In total, 242 patients were included in the disease group. Plasma samples from 23 NPC patients and 15 healthy control were used for immune checkpoint protein panel assays. Samples from 219 patients with NPC including 30 paired pre-treatment and post-radiotherapy samples were evaluated by enzyme-linked immunosorbent assay to determine sPD-L1 levels. RESULTS: A total of 14 immune checkpoint proteins, including sPD-L1were upregulated in 23 patients with NPC (all p<0.001) compared with 15 healthy controls. Among 219 patients, the median follow-up time was 50 months (7-82 months). Based on the optimal cutoff value of 93.7 pg/mL, patients with high expression of sPD-L1 had worse distant metastasis-free survival (87.5% vs 74.0%, p=0.006) than those of patients with low expression. Multivariate analysis showed that sPD-L1 (HR=1.99, p=0.048) and EBV-DNA (HR=2.51, p=0.030) were poor prognostic factors for DMFS. In the group with high EBV-DNA expression, DMFS was worse for patients with high sPD-L1 expression than those with low sPD-L1 expression (56.4% vs 82.6%, p=0.002). CONCLUSION: Plasma immune checkpoint protein expression differed significantly between patients with NPC and healthy donors. Plasma sPD-L1 levels are a candidate prognostic biomarker, especially when combined with EBV-DNA.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...