Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
1.
Exp Cell Res ; 439(1): 114073, 2024 May 03.
Artigo em Inglês | MEDLINE | ID: mdl-38704079

RESUMO

Determining the appropriate source of antigens for optimal antigen presentation to T cells is a major challenge in designing dendritic cell (DC) -based therapeutic strategies against hepatocellular carcinoma (HCC). Tumor-derived exosomes (Tex) express a wide range of tumor antigens, making them a promising source of antigens for DC vaccines. As reported, the exosomes secreted by tumor cells can inhibit the antitumor function of immune cells. In this study, we transfected hepatocellular carcinoma cells with Rab27a to enhance the yield of exosomes, which were characterized using transmission electron microscopy and Western blot analysis. We found that Tex secreted by overexpressing Rab27a Hepatocellular carcinoma cell lines pulsed DC is beneficial for the differentiation and maturation of DCs but inhibits the secretion of the IL-12 cytokine. Consequently, we developed a complementary immunotherapy approach by using Tex as an antigen loaded onto DCs, in combination with the cytokine IL-12 to induce antigen-specific cytotoxic T lymphocytes (CTLs). The results indicated that the combination of DC-Tex and IL-12 was more effective in stimulating T lymphocyte proliferation, releasing IFN-γ, and enhancing cytotoxicity compared to using exosomes or IL-12 alone. Additionally, the inclusion of IL-12 also compensated for the reduced IL-2 secretion by DCs caused by Tex. Moreover, in a BALB/c nude mice model of hepatocellular carcinoma, CTLs induced by DC-Tex combined with IL-12 maximized the tumor-specific T-cell immune effect and suppressed tumor growth. Thus, Tex provides a novel and promising source of antigens, with cytokines compensating for the shortcomings of Tex as a tumor antigen. This work helps to clarify the role of exosomes in tumor immunotherapy and may offer a safe and effective prospective strategy for the clinical application of exosome-based cellular immunotherapy.

2.
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.

3.
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
4.
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
5.
Exp Cell Res ; 417(2): 113232, 2022 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-35659970

RESUMO

To date, most studies of exosomes related to hepatocellular carcinoma (HCC) have used commercial cancer cell lines or patient plasma as source material. In this study, we isolated exosomes directly from HCC tissues to investigate the potential of exosomal contents as biomarkers for HCC. Exosomes were identified and verified using transmission electron microscopy, nano-flow cytometry analysis, and western blotting. Tissue-derived exosomal miRNA expression was profiled by high-throughput sequencing, and differential expression of miRNAs was validated by quantitative real-time polymerase chain reaction analysis. The diagnostic performance of differentially expressed exosomal miRNAs for HCC was evaluated by receiver operating characteristic curve analysis. Target genes of these miRNAs were verified using luciferase reporter assays, and their functions were studied through in vitro and rescue assays. In total, 225 differentially expressed exosomal miRNAs were identified in HCC samples compared with adjacent liver tissues, and some were associated with HCC tumorigenesis and progression. Comparison of the expression profiles of tissue-derived and plasma-derived exosomal miRNAs identified hsa-miR-483-5p as the only differentially expressed miRNA detected in both HCC tissue and plasma, and this was in a validation group of HCC patients. Analysis of the diagnostic performance of plasma exosomal hsa-miR-483-5p or plasma hsa-miR-483-5p found that both could differentiate HCC and non-HCC cases. In vitro ectopic miR-483-5p expression promoted HCC cell proliferation. CDK15 was confirmed to bind with miR-483-5p directly, and thus, miR-483-5p may function by downregulating CDK15. Hsa-miR-483-5p represents a potential specific and sensitive biomarker for HCC diagnosis.


Assuntos
Carcinoma Hepatocelular , Exossomos , Neoplasias Hepáticas , MicroRNAs , Biomarcadores/metabolismo , Carcinoma Hepatocelular/diagnóstico , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Exossomos/metabolismo , Humanos , Neoplasias Hepáticas/diagnóstico , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , MicroRNAs/metabolismo
6.
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
7.
Theranostics ; 12(5): 2063-2079, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35265199

RESUMO

Background: Dietary tyrosine regulating melanoma progression has been well-recognized. However, whether tyrosine-based melanin anabolism contributes to pulmonary and cerebral organotropic colonization of melanoma remains elusive. Furthermore, approaches based on targeting tyrosinase activity to inhibiting multi-organ metastasis of melanoma cells need to be designed and validated. Methods: Patients derived melanoma cells and mouse B16 melanoma cells with different pigmentation were employed in this investigation. Tyrosine content dynamics in tumors and multiple organs during the melanoma progression was monitored, and tyrosine-based melanin synthesis of melanoma cells derived from multi-organ was determined. Additionally, we also adopted RNA-seq, flow cytometry, real-time PCR and composite metastasis mouse model to analyze organotropic colonization and to validate designed therapeutic strategies. Results: B16 melanoma cells with high activity of tyrosinase and sensitivity of tyrosine utilization for melanin synthesis (Tyr-H cells) easily colonized in the lung, while B16 melanoma cells lacking above characteristics (Tyr-L cells) exhibited potent proliferation in the brain. Mechanistically, Tyr-H cells recruited and trained neutrophils and macrophages to establish pulmonary metastatic niche dependent on highly secreted CXCL1 and CXCL2 and an excessive melanosome accumulation-induced cell death. Tyr-L cells enhanced PD-L1 expression in tumor-infiltrated macrophages when they are progressing in the brain. Accordingly, intervention of tyrosinase activity (2-Ethoxybenzamide or hydroquinone) in combination with inhibitors of phagocytosis (GSK343) or chemotaxis (SB225002) suppressed organotropic colonization and significantly improved the survival of melanoma- bearing mice treated with immune checkpoint blockade (PD1 antibody). Conclusions: The heterogeneity of melanoma cells in utilization of tyrosine is associated with organotropic colonization, providing the basis for developing new strategies to combat melanoma.


Assuntos
Melaninas , Melanoma Experimental , Animais , Linhagem Celular Tumoral , Humanos , Pulmão/patologia , Camundongos , Monofenol Mono-Oxigenase/metabolismo , Microambiente Tumoral , Tirosina
8.
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
9.
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
10.
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.

11.
Exp Cell Res ; 390(1): 111953, 2020 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-32156601

RESUMO

MUC1 is a tumor-associated antigen (TAA) overexpressed in many tumor types, which makes it an attractive target for cancer immunotherapy. However, this marker is a non-mutated antigen without high immunogenicity. In this study, we designed several new altered peptides by replacing amino acids in their sequences, which were derived from a low-affinity MUC1 peptide, thus bypassing immune tolerance. Compared to the wild-type (WT) peptide, the altered MUC1 peptides (MUC11081-1089L2, MUC11156-1164L2, MUC11068-1076Y1) showed higher affinity to the HLA-A0201 molecule and stronger immunogenicity. Furthermore, these altered peptides resulted in the generation of more cytotoxic T lymphocytes (CTLs) that could cross-recognize gastric cancer cells expressing WT MUC1 peptides, in an HLA-A0201-restricted manner. In addition, M1.1 (MUC1950-958), a promising antitumor peptide that has been tested in multiple tumors, was not able to induce stronger antitumor responses. Collectively, our results demonstrated that altered peptides from MUC1, as potential HLA-A0201-restricted CTL epitopes, could serve as peptide vaccines or constitute components of peptide-loaded dendritic cell vaccines for gastric cancer treatment.


Assuntos
Epitopos/imunologia , Antígeno HLA-A2/imunologia , Mucina-1/imunologia , Neoplasias Gástricas/imunologia , Linhagem Celular Tumoral , Humanos , Imunoterapia/métodos , Mucina-1/química , Fragmentos de Peptídeos/imunologia , Neoplasias Gástricas/terapia , Linfócitos T Citotóxicos/imunologia
12.
Cell Immunol ; 349: 104042, 2020 03.
Artigo em Inglês | MEDLINE | ID: mdl-32061376

RESUMO

Cytotoxic T lymphocytes (CTLs) play a major role in cancer immunotherapy. A potent tumor immunotherapy may not only require activation of anti-tumor effector cells but also rely on the use of cytokines to create a controlled environment for the development of anti-tumor T cells. In this study, we fabricated a dual-target immunonanoparticle, e.g. poly(d,l-lactide-co-glycolide) nanoparticle, by loading Interleukin-12 (IL-12) and modifying with CD8 and Glypican-3 antibodies on the surface. Our results demonstrate that the fabricated targeting immunonanoparticles bind specifically to the two target cells of interest, i.e. CD8+ T cells and HepG-2 cells via the antibody-antigen interactions and form T cell-HepG-2 cell clusters, which enhances the cytotoxicity of T cells. IL-12-containing dual-target immunonanoparticles delivered IL-12 specifically to CD8+ T cells, and favored the expansion, activation and cytotoxic activity of CD8+ T lymphocytes. These results suggest that dual-target IL-12-encapsulated nanoparticles are a promising platform for cancer immunotherapy.


Assuntos
Linfócitos T CD8-Positivos/efeitos dos fármacos , Imunoterapia/métodos , Interleucina-12/administração & dosagem , Nanocápsulas/administração & dosagem , Linfócitos T Citotóxicos/efeitos dos fármacos , Linfócitos T CD8-Positivos/imunologia , Divisão Celular/efeitos dos fármacos , Citotoxicidade Imunológica , Sistemas de Liberação de Medicamentos , Ensaios de Seleção de Medicamentos Antitumorais , Estabilidade de Medicamentos , Células Hep G2 , Humanos , Imunofenotipagem , Testes de Liberação de Interferon-gama , Interleucina-12/farmacologia , Ativação Linfocitária/efeitos dos fármacos , Copolímero de Ácido Poliláctico e Ácido Poliglicólico , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/farmacologia , Formação de Roseta , Linfócitos T Citotóxicos/imunologia
13.
Cancer Manag Res ; 11: 6855-6869, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31440083

RESUMO

BACKGROUND: Inhibitor of DNA binding 1 (Id1) is upregulated in multiple cancers, and Id1overexpression correlates with cancer aggressiveness and poor clinical outcomes in cancer patients. However, its roles in cancer stem-like cells (CSCs) and epithelial-mesenchymal transition (EMT) are still elusive. PURPOSE: This study aimed to examine the role of Id1 on the mediation of CRC stemness and explore the underlying mechanisms. METHODS: Id1 and CD133 expression was detected by qPCR assay and immunohistochemistry (IHC) in normal mucosal and primary colorectal cancer (CRC) specimens. Id1 was stably knocked down (KD) in human CRC cell lines. Spheres forming assay and tumorigenic assay were performed to evaluate self-renewal capacity and tumor initiation. Expression of CSC- and EMT-related markers and TCF/LEF activity were assessed in HCT116 cells after Id1 KD. RESULTS: qPCR assay showed higher Id1 and CD133 expression in CRC specimens than in normal mucosal specimens (P<0.05). IHC detected high cytoplasmic Id1 expression in 35 CRC specimens (46.7%), and high CD133 expression in 22 CRC specimens (29.3%) and negative expression in 18 normal mucosal specimens. High Id1 expression positively correlated with poor differentiation (P=0.034), and CD133 expression correlated with T category in CRC patients (P=0.002). Spearman correlation analysis revealed a positive correlation between Id1 and CD133 expression in CRC patients (P<0.05). Id1 KD resulted in suppression of proliferation, cell-colony formation, self-renewal capability and CSC-like features in HCT116 cells, and impaired the tumor-initiating capability in CRC cells. In addition, Id1 maintained the stemness of CRC cells via the Id1-c-Myc-PLAC8 axis through activating the Wnt/ß-catenin and Shh signaling pathways. CONCLUSIONS: Id1 expression significantly correlates with CD133 expression in CRC patients, and Id1 KD impairs CSC-like capacity and reverses EMT traits, partially via the Wnt/ß-catenin signaling. Id1 may be a promising therapeutic target against colon CSCs.

14.
Cancer Lett ; 447: 33-40, 2019 04 10.
Artigo em Inglês | MEDLINE | ID: mdl-30684592

RESUMO

Based on analysis of Epstein-Barr virus (EBV) BART microRNA expression profiles, we previously reported that EBV-encoded miR-BART13 is upregulated in nasopharyngeal carcinoma (NPC) plasma specimens. However, the effects and molecular mechanisms of miR-BART13 in NPC remain largely unknown. We found that miR-BART13 was significantly upregulated in NPC tissue specimens. Ectopic expression of miR-BART13 promoted NPC cell proliferation, epithelial mesenchymal transition, and metastasis in vitro, and facilitated xenograft tumor growth and lung metastasis in vivo. Molecularly, NF-κB inhibitor interacting Ras-like 2 (NKIRAS2), a negative regulator of the NF-κB signaling, was identified to be a direct target of miR-BART13 in NPC cells, and NKIRAS2 mRNA and protein expression was inversely correlated with miR-BART13 in NPC tissues, respecitvely. Furthermore, the NF-κB signaling pathway was activated by miR-BART13. By rescued experiments, reconstitution of NKIRAS2 expression abrogated all the phenotypes upregulated by miR-BART13, and attenuated activity of NF-κB signaling pathway activated by miR-BART13 in NPC cells. Our findings indicated the newly identified miR-BART13/NKIRAS2/NF-κB signaling axis may provide further insights into better understanding of NPC initiation and development, and targeting of this pathway could be further studied as a therapeutic strategy for NPC patients.


Assuntos
Proliferação de Células/genética , Herpesvirus Humano 4/genética , MicroRNAs/genética , Carcinoma Nasofaríngeo/virologia , Metástase Neoplásica/genética , RNA Viral/genética , Transdução de Sinais/genética , Animais , Proteínas de Transporte/genética , Linhagem Celular Tumoral , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/virologia , Feminino , Regulação Neoplásica da Expressão Gênica/genética , Células HEK293 , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , NF-kappa B/genética , Carcinoma Nasofaríngeo/genética , Neoplasias Nasofaríngeas/genética , Neoplasias Nasofaríngeas/virologia , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
15.
J Exp Clin Cancer Res ; 38(1): 34, 2019 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-30678708

RESUMO

Following publication of the original article [1], the authors reported two errors in the article. In the caption of Figure 1c the sentence "The 20 most highly upregulated EBV BART miRNAs identified between NPC specimens and normal nasopharyngeal mucosal specimens" should be corrected as "The highly upregulated EBV BART miRNAs identified between NPC specimens and normal nasopharyngeal mucosal specimens".

16.
J Exp Clin Cancer Res ; 37(1): 283, 2018 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-30477559

RESUMO

BACKGROUND: Epstein-Barr virus (EBV) is ubiquitously associated with nasopharyngeal carcinoma (NPC). EBV encodes two groups of microRNAs (miRNAs) which are divided into BamHI fragment H rightward open reading frame 1 (BHRF1) and BamHI-A rightward transcripts (BART) microRNAs. EBV miR-BART has been found to be involved in the development and progression of NPC. However, so far the role of EBV-miR-BART8-3p in NPC progression remains unknown. This study aimed to investigate the role of EBV-miR-BART8-3p in NPC and explore the underlying mechanisms. METHODS: miRNA expression was profiled in NPC and normal nasopharyngeal mucosal specimens using miRNA sequencing. EBV-miR-BART8-3p and RNF38 expression was quantified with qPCR assay. The migration, invasion and metastasis of NPC cells were evaluated using CCK-8, colony-forming, wound-healing, and migration and invasion assays. The expression levels of epithelial-mesenchymal transition (EMT)-related markers,metastasis-related markers and NF-κB and Erk1/2 signaling proteins were determined using Western blotting. Tumorigenic assay was performed to evaluate the pulmonary metastatic ability of NPC cells in vivo. RESULTS: EBV BART miRNAs were highly over-expressed and co-expressed in NPC and might be associated with deactivated immune response in NPC according to the sequencing analysis. EBV-miR-BART8-3p expression was significantly higher in human NPC specimens than in normal nasopharyngeal mucosal specimens. EBV-miR-BART8-3p was found to promote NPC migration, invasion and metastasis, drove an EMT process and upregulated expression of metastasis-related proteins expression in NPC cells. Our data showed EBV-miR-BART8-3p directly targeted RNF38 in NPC cells. CONCLUSION: The present study demonstrates that EBV-miR-BART8-3p plays a significant role in inducing EMT and promoting metastasis through directly targeting RNF38 in NPC cells via the activation of NF-κB and Erk1/2 signaling pathways. Our findings suggest that EBV-miR-BART8-3p is a potential therapeutic target for NPC.


Assuntos
Herpesvirus Humano 4/genética , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Carcinoma Nasofaríngeo/virologia , Animais , Linhagem Celular Tumoral , Proliferação de Células , Transição Epitelial-Mesenquimal , Infecções por Vírus Epstein-Barr/genética , Infecções por Vírus Epstein-Barr/metabolismo , Infecções por Vírus Epstein-Barr/patologia , Infecções por Vírus Epstein-Barr/virologia , Feminino , Herpesvirus Humano 4/metabolismo , Humanos , Camundongos , Camundongos Nus , MicroRNAs/genética , MicroRNAs/metabolismo , Carcinoma Nasofaríngeo/genética , Carcinoma Nasofaríngeo/metabolismo , Carcinoma Nasofaríngeo/patologia , Metástase Neoplásica , Transfecção
17.
Cancer Manag Res ; 10: 4945-4957, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30464595

RESUMO

BACKGROUND: Dendritic cell (DC)-derived exosomes (Dexs) have been proved to induce and enhance antigen-specific T cell responses in vivo, and previous clinical trials have shown the feasibility and safety of Dexs in multiple human cancers. However, there is little knowledge on the efficacy of Dexs against hepatocellular carcinoma (HCC) until now. METHODS: In this study, human peripheral blood-derived DCs were loaded with recombinant adeno-associated viral vector (rAAV)-carrying alpha-fetoprotein (AFP) gene (rAAV/AFP), and high-purity Dexs were generated. Then naive T cells were stimulated with Dexs to investigate the specific T cell-mediated immune responses against HCC. RESULTS: Our findings showed that Dexs were effective to stimulate naive T cell proliferation and induce T cell activation to become antigen-specific cytotoxic T lymphocytes (CTLs), thereby exhibiting antitumor immune responses against HCC. In addition, Dex-sensitized DC precursors seemed more effective to trigger major histocompatibility complex class I (MHC I)-restricted CTL response and allow DCs to make full use of the minor antigen peptides, thereby maximally activating specific immune responses against HCC. CONCLUSION: It is concluded that Dexs, which combine the advantages of DCs and cell-free vectors, are promising to completely, or at least in part, replace mature DCs (mDCs) to function as cancer vaccines or natural antitumor adjuvant.

18.
Clin Exp Med ; 18(4): 523-533, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29855744

RESUMO

Myosin light chain kinase (MYLK) is found to catalyze the phosphorylation of myosin light chains (MLC) and regulate invasion and metastasis in some malignancies. However, there is little knowledge on the role of MYLK in hepatocellular carcinoma (HCC), and no studies have been conducted to investigate the mechanisms underlying MYLK-mediated promotion of HCC invasion and metastasis until now. In this study, we investigated the expression of MYLK in 50 pairs of human HCC and adjacent liver specimens. High MYLK expression was significantly correlated with aggressive clinicopathological features including tumor encapsulation, microvascular invasion and metastasis. In vitro assays showed that shRNA-induced MYLK knockdown significantly inhibited the wound-healing ability of HCC cells and the ability to migrate and invade through Matrigel. We next uncovered that MYLK knockdown resulted in a reduction in the number of F-actin stress fibers, disorganization of F-actin architectures and morphological alterations of HCC cells. Phosphorylated MLC, rather than total MLC, was found to be markedly reduced in response to downregulation of MYLK expression, and MYLK-regulated actin cytoskeleton through phosphorylating MLC in HCC cells. In addition, Western blotting assay revealed downregulation of the epithelial marker E-cadherin and upregulation of mesenchymal markers Vimentin, N-cadherin and Snail. Taken together, our findings indicate that MYLK promotes HCC progression by altering cytoskeleton to enhance epithelial-mesenchymal transition (EMT).


Assuntos
Carcinoma Hepatocelular/enzimologia , Carcinoma Hepatocelular/patologia , Transição Epitelial-Mesenquimal , Neoplasias Hepáticas/enzimologia , Neoplasias Hepáticas/patologia , Quinase de Cadeia Leve de Miosina/biossíntese , Citoesqueleto/enzimologia , Citoesqueleto/genética , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Quinase de Cadeia Leve de Miosina/genética , Metástase Neoplásica
19.
J Exp Clin Cancer Res ; 36(1): 134, 2017 09 29.
Artigo em Inglês | MEDLINE | ID: mdl-28969700

RESUMO

BACKGROUND: Kindlin-2 is a member of the focal adhesion protein family that regulates invasion and metastasis in multiple malignancies; however, little is known about the role of Kindlin-2 in hepatocellular carcinoma (HCC) progression. METHODS: Immunohistochemistry was used to investigate Kindlin-2 expression in 177 pairs of human HCC and adjacent liver tissue samples. The role of Kindlin-2 in the in vitro invasion and migration of HCC cell lines was evaluated in MHCC97H, LM3 and SMMC7721 cells. Microarray expression analysis was applied to explore the molecular mechanism through which Kindlin-2 promoted HCC progression. Quantitative real-time PCR and Western blotting were performed to verify the microarray results. RESULTS: High Kindlin-2 expression was found to significantly correlate with aggressive HCC clinicopathological features including tumor encapsulation, microvascular invasion, extrahepatic metastasis and poor prognosis. In vitro, Kindlin-2 knockout or knockdown inhibited HCC cell adhesion, migration and invasion, while ectopic Kindlin-2 expression promoted these processes. Importantly, Kindlin-2 activated Wnt/ß-catenin signaling and increased ß-catenin expression, especially levels of non-phosphorylated ß-catenin, as well as two Wnt/ß-catenin signaling pathway targets, Axin2 and MMP7. Kindlin-2 also induced a change in the expression profile of HCC cells, suggesting the cells underwent epithelial-mesenchymal transition. For example, the expression of the epithelial marker E-cadherin was downregulated, while the mesenchymal markers Vimentin, N-cadherin and Snail were upregulated. CONCLUSION: Kindlin-2 promotes HCC invasion, metastasis and epithelial-mesenchymal transition through Wnt/ß-catenin signaling.


Assuntos
Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Regulação para Cima , Via de Sinalização Wnt , Idoso , Carcinoma Hepatocelular/genética , Linhagem Celular Tumoral , Transição Epitelial-Mesenquimal , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células Hep G2 , Humanos , Neoplasias Hepáticas/genética , Masculino , Proteínas de Membrana/genética , Pessoa de Meia-Idade , Invasividade Neoplásica , Metástase Neoplásica , Proteínas de Neoplasias/genética
20.
Mol Med Rep ; 16(2): 2075-2088, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28656315

RESUMO

Cervical cancer is the fourth leading cause of cancer mortality in women worldwide. High­risk human papillomavirus infection is a major cause of cervical cancer. A previous study revealed the role of different oncogenes and tumor suppressors in cervical cancer initiation and progression. However, the complicated genetic network regulating cervical cancer remains largely unknown. The present study reported transcriptome sequencing analysis of three cervical squamous cell cancer tissues and paired normal cervical tissues. Transcriptomic analysis revealed that 2,519 genes were differently expressed between cervical cancer tissues and their corresponding normal tissues. Among these, 236 differentially expressed genes (DEGs) were statistically significant, including many DEGs that were novel in cervical cancer, including gastrulation brain homeobox 2,5­hydroxytryptamine receptor 1D and endothelin 3. These 236 significant DEGs were highly enriched in 28 functional gene ontology categories. The Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis suggested involvement of these DEGs in multiple pathways. The present study provides a transcriptome landscape of cervical cancer in Chinese patients and an improved understanding of the genetic regulatory network in cervical cancer tumorigenesis.


Assuntos
Carcinoma de Células Escamosas/genética , Perfilação da Expressão Gênica , Sequenciamento de Nucleotídeos em Larga Escala , Neoplasias do Colo do Útero/genética , Adulto , Feminino , Regulação Neoplásica da Expressão Gênica , Ontologia Genética , Humanos , Pessoa de Meia-Idade , Anotação de Sequência Molecular , Reprodutibilidade dos Testes
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA