Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Toxicol Appl Pharmacol ; 475: 116612, 2023 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-37463651

RESUMEN

The metabolite of organophosphate pesticide chlorpyrifos (CPF), 3,5,6-Trichloro-2-pyridinol (TCP), is persistent and mobile toxic substance in soil and water environments, exhibiting cytotoxic, genotoxic, and neurotoxic properties. However, little is known about its effects on the peripheral auditory system. Herein, we investigated the effects of TCP exposure on mouse postnatal day 3 (P3) cochlear culture and an auditory cell line HEI-OC1 to elucidate the underlying molecular mechanisms of ototoxicity. The damage of TCP to outer hair cells (OHC) and support cells (SC) was observed in a dose and time-dependent manner. OHC and SC were a significant loss from basal to apical turn of the cochlea under exposure over 800 µM TCP for 96 h. As TCP concentrations increased, cell viability was reduced whereas reactive oxygen species (ROS) generation, apoptotic cells, and the extent of DNA damage were increased, accordingly. TCP-induced phosphorylation of the p38 and JNK MAPK are the downstream effectors of ROS. The antioxidant agent, N-acetylcysteine (NAC), could reverse TCP-mediated intracellular ROS generation, inhibit the expressive level of cleaved-caspase 3 and block phosphorylation of p38/JNK. Overall, this is the first demonstration of TCP damaging to peripheral sensory HCs and SC in organotypic cultures from the postnatal cochlea. Data also showed that TCP exposure induced oxidase stress, cell apoptosis and DNA damage in the HEI-OC1 cells. These findings serve as an important reference for assessing the risk of TCP exposure.


Asunto(s)
Antineoplásicos , Ototoxicidad , Animales , Ratones , Especies Reactivas de Oxígeno/metabolismo , Sistemas Microfisiológicos , Antineoplásicos/farmacología , Piridinas/farmacología , Apoptosis , Cisplatino/farmacología
2.
J Gene Med ; 25(7): e3500, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-36942488

RESUMEN

BACKGROUND: Exosomal circular RNAs (circRNAs) played critical roles in tumor development and progression and might be novel biomarkers in the diagnosis and treatment of various cancers. However, the biological functions and clinical implications of exosomal circRNAs in breast cancer are unclear. METHODS: Expression profiles of exosomal circRNAs the in exoRBase 2.0 database were used to identify differentially expressed exosomal circRNAs in breast cancer. The LASSO and SVM-RFE algorithms followed by multivariate logistic regression analysis were performed to construct the diagnostic model. The target genes of circRNAs were selected by combing differential expression analysis and CSCD, TargetScan and ENCORI databases. Univariate and multivariate survival analysis were conducted to construct a survival-associated exosomal circRNA-miRNA-mRNA network. GSVA and CIBERSORT algorithms were used to evaluate the cancer hallmarks and immune cells in breast cancer and Spearman correlation analysis was used to investigate their correlations with the circRNA-miRNA-mRNA network. RESULTS: In total, 347 upregulated and three downregulated exosomal circRNAs were identified in breast cancer patients. The diagnostic model based on 14 exosomal circRNAs showed a high area under the curve (AUC) value in both the training (AUC = 0.98) and validation (AUC = 0.94) dataset. In total, 70 miRNAs and 1147 mRNAs were selected as the downstream targets of circRNAs and were revealed to participate in tumor-associated pathways, including the PI3K-AKT, MAPK, RAS and RAP1 pathways, as well as calcium signaling pathways, in addition to transcriptional misregulations. The constructed survival-associated exosomal circRNA-miRNA-mRNA network contained nine exosomal circRNAs, 12 miRNAs and 10 mRNAs, and showed complicated correlations and interactions within networks. Cancer hallmark pathways, including the TGF-ß, KRAS and MYC signaling pathways, tumor angiogenesis, epithelial-mesenchymal transition, DNA repair and G2M checkpoint, as well as immune cells, including CD4+ and CD8+ T cells, dendritic cells, mast cells, macrophage cells, memory B cells and natural killer cells, were closely correlated with the circRNA-miRNA-mRNA network. CONCLUSIONS: The present study is the first to systematically analyze the exosomal circRNAs in breast cancer. We established an exosomal circRNA diagnostic model and constructed a survival-associated exosomal circRNA-miRNA-mRNA network. Our results revealed the complicated functions and potential mechanisms of the exosomal circRNA-miRNA-mRNA network in breast cancer, which need to be validated further in future studies.


Asunto(s)
Neoplasias de la Mama , MicroARNs , Humanos , Femenino , MicroARNs/genética , Neoplasias de la Mama/genética , ARN Circular/genética , Linfocitos T CD8-positivos , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-myc , Transducción de Señal/genética
3.
Front Genet ; 13: 981030, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36267418

RESUMEN

Long non-coding RNAs (lncRNAs) have been extensively studied as important regulators of tumor development in various cancers. Tumor protein 53 target gene 1 (TP53TG1) is a newly identified lncRNA in recent years, and several studies have shown that TP53TG1 may play oncogenic or anti-oncogenic roles in different cancers. Nevertheless, the role of TP53TG1 in the development of cervical cancer is unclear. In our study, pan-cancer analysis showed that high expression of TP53TG1 was significantly associated with a better prognosis. We then constructed a TP53TG1 overexpression model in HeLa cell line to explore its functions and molecular targets. We found that TP53TG1 overexpression significantly inhibited cell proliferation and induced apoptosis, demonstrating that TP53TG1 may be a novel anti-oncogenic factor in cervical cancer. Furthermore, overexpression of TP53TG1 could activate type I interferon signaling pathways and inhibit the expression of genes involved in DNA damage responses. Meanwhile, TP53TG1 could affect alternative splicing of genes involved in cell proliferation or apoptosis by regulating the expression of many RNA-binding protein genes. Competing endogenous RNA (ceRNA) network analysis demonstrated that TP53TG1 could act as the sponge of several miRNAs to regulate the expression level of target genes. In conclusion, our study highlights the essential role of lncRNA TP53TG1 in the development of cervical cancer and suggests the potential regulatory mechanisms.

4.
J Oncol ; 2022: 1285632, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35859664

RESUMEN

RNA modification has become an exciting underexplored field in recent years. In lung adenocarcinoma (LUAD), m6A was the best characterized and most studied RNA modification, while knowledge about other kinds of RNA modifications in LUAD is limited. In our study, we included a total of 100 RNA modification regulators of eight types of cancer-related RNA modifications (m6A, m1A, m5C, Nm, m7G, Ψ, A-to-I, and mcm5s2U) to systematically profile their specific roles in LUAD. By gene mutation and expression analysis, we identified extensive dysregulations and complicated interactions of 100 RNA modification regulators in LUAD. Based on unsupervised clustering analysis, gene set variation analysis (GSVA), and single-sample gene-set enrichment analysis (ssGSEA), two RNA modification patterns in LUAD were defined to show distinct biological characteristics. The favorable prognostic pattern was enriched with infiltrated immune cells, including activated B cells, CD8 T cells, eosinophil cells, dendritic cells, and natural killer cells, while the unfavorable prognostic pattern was enriched with cancer hallmarks, including hypoxia, epithelial-mesenchymal transition (EMT), angiogenesis, PI3K-AKT-mTOR pathway, MYC pathway, and glycolysis pathway. We also constructed an RNA modification score (RMScore) based on five critical genes (CYP17A1, NTSR1, PITX3, KRT6A, and ANLN) to evaluate the RNA modification status of individual LUAD patients. RMScore was revealed to be related to the infiltrated immune cells and cancer hallmarks and was an independent prognostic factor in the TCGA-LUAD cohort and two external GEO-LUAD cohorts. Our study was the first to comprehensively investigate the dysregulations, crosstalk, and potential prognostic value of eight types of RNA modifications in LUAD. Our results highlighted the significance of eight types of RNA modifications in tumor microenvironments and cancer hallmarks and provided novel prognostic biomarkers and potential therapeutic targets in the management of LUAD patients in the future.

5.
Transl Lung Cancer Res ; 10(7): 3155-3172, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34430355

RESUMEN

BACKGROUND: High incidence of metastasis is the main cause of death for small cell lung cancer (SCLC), with its underlying molecular mechanisms remain unclear. Exosomal miRNAs are important regulators in metastatic processes of various tumors, but their specific role in SCLC metastasis is unknown. METHODS: Small RNA sequencing followed by qRT-PCR verification was used to screen the potential exosomal miRNAs that might mediate SCLC metastasis. SCLC-cell-secreted exosomes were labeled followed by incubating with vascular endothelial cells to evaluate exosome-mediated communication between SCLC cells and vascular endothelial cells. In vitro permeability assay and transendothelial migration assay were applied to investigate the function of exosomal miRNA on vascular endothelial cells. In vivo permeability assay and mouse lung colonization assay were used to verify the effects of exosomal miRNA on vascular barriers and SCLC metastasis in vivo. Proteomics technology, dual-luciferase reporter system together with rescue assays were conducted to excavate the downstream pathways of miRNA. RESULTS: Compared with 57 healthy volunteers and 46 non-small cell lung cancer patients, we identified that the level of exosomal miR-375-3p in 126 SCLC patients was obviously higher and was positively correlated with patient TNM stages. In vitro functional experiments found that SCLC-cell-secreted exosomal miR-375-3p could increase the permeability of vascular endothelial cells and facilitate the transendothelial migration of SCLC cells. In vivo, miR-375-3p-enriched exosomes also destroyed the barrier structure of lung, liver and brain tissues of mice, leaded to an increased blood vessel permeability and finally promoted SCLC metastasis. Mechanistically, SCLC-cell-secreted exosomal miR-375-3p was transferred to vascular endothelial cells. The internalized miR-375-3p broke the tight junction of vascular endothelial cells by directedly binding to the 3'UTR of tight junction protein claudin-1 and negatively regulating its expression. Overexpressing claudin-1 in vascular endothelial cells could rescue the broken vascular barriers induced by miR-375-3p. CONCLUSIONS: Our findings underline the crucial roles of exosomal miRNA-375-3p in regulating vascular endothelial barrier integrity and SCLC metastasis. miRNA-375-3p has a great potential to be a novel biomarker monitoring metastasis and guiding clinical therapeutics of SCLC patients.

6.
Clin Transl Med ; 11(7): e459, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-34323409

RESUMEN

Dysregulated expression of S100A7 is found in several cancers and plays an important role in tumor progression; however, its carcinogenic role in esophageal squamous carcinoma (ESCC) is still poorly understood. Here, we identified that the levels of S100A7 were remarkably upregulated in 341 tumor tissues (P < .001) and 274 serum samples (P < .001) of ESCC patients compared with normal control. It was an independent prognostic factor (P = .026). Furthermore, a new diagnostic model for ESCC based on serum S100A7, SCC, and crfra21-1 was established with area under curve (AUC) up to 0.863 (95% CI: 0.802-0.925). Mechanically, we found upregulated S100A7 could promote cell migration and proliferation through intracellular binding to JAB1 and paracrine interaction with RAGE receptors and then activates the downstream signaling pathways. In addition, exocrine S100A7 could promote M2 macrophage infiltration and polarization by up-regulating M2 macrophage associated proteins, and tumor angiogenesis by enhancing the activation of p-ErK and p-FAK pathways. Further animal experiments confirmed the role of S100A7 in promoting M2 macrophage infiltration and angiogenesis in ESCC. In conclusion, these findings highlighted the potential diagnostic and prognostic value of S100A7 in patients with ESCC. Meanwhile, our results reveal that S100A7 promotes tumor progression by activating oncogenic pathways and remodeling tumor microenvironment, which paving the way for the progress of S100A7 as a therapeutic target for cancer treatment.


Asunto(s)
Neoplasias Esofágicas/diagnóstico , Carcinoma de Células Escamosas de Esófago/diagnóstico , Macrófagos/inmunología , Proteína A7 de Unión a Calcio de la Familia S100/metabolismo , Animales , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/metabolismo , Complejo del Señalosoma COP9/metabolismo , Proliferación Celular , Neoplasias Esofágicas/irrigación sanguínea , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/irrigación sanguínea , Carcinoma de Células Escamosas de Esófago/patología , Femenino , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Macrófagos/citología , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Neovascularización Patológica , Péptido Hidrolasas/metabolismo , Pronóstico , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptor para Productos Finales de Glicación Avanzada/metabolismo , Proteína A7 de Unión a Calcio de la Familia S100/antagonistas & inhibidores , Proteína A7 de Unión a Calcio de la Familia S100/sangre , Proteína A7 de Unión a Calcio de la Familia S100/genética , Transducción de Señal
7.
Cancer Biol Med ; 2021 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-33856140

RESUMEN

OBJECTIVE: The proportion of patients with stage I lung adenocarcinoma (LUAD) has dramatically increased with the prevalence of low-dose computed tomography use for screening. Up to 30% of patients with stage I LUAD experience recurrence within 5 years after curative surgery. A robust risk stratification tool is urgently needed to identify patients who might benefit from adjuvant treatment. METHODS: In this first investigation of the relationship between metabolic reprogramming and recurrence in stage I LUAD, we developed a recurrence-associated metabolic signature (RAMS). This RAMS was based on metabolism-associated genes to predict cancer relapse and overall prognoses of patients with stage I LUAD. The clinical significance and immune landscapes of the signature were comprehensively analyzed. RESULTS: Based on a gene expression profile from the GSE31210 database, functional enrichment analysis revealed a significant difference in metabolic reprogramming that distinguished patients with stage I LUAD with relapse from those without relapse. We then identified a metabolic signature (i.e., RAMS) represented by 2 genes (ACADM and RPS8) significantly related to recurrence-free survival and overall survival times of patients with stage I LUAD using transcriptome data analysis of a training set. The training set was well validated in a test set. The discriminatory power of the 2 gene metabolic signature was further validated using protein values in an additional independent cohort. The results indicated a clear association between a high risk score and a very poor patient prognosis. Stratification analysis and multivariate Cox regression analysis showed that the RAMS was an independent prognostic factor. We also found that the risk score was positively correlated with inflammatory response, the antigen-presenting process, and the expression levels of many immunosuppressive checkpoint molecules (e.g., PD-L1, PD-L2, B7-H3, galectin-9, and FGL-1). These results suggested that high risk patients had immune response suppression. Further analysis revealed that anti-PD-1/PD-L1 immunotherapy did not have significant benefits for high risk patients. However, the patients could respond better to chemotherapy. CONCLUSIONS: This study is the first to highlight the relationship between metabolic reprogramming and recurrence in stage I LUAD, and is the first to also develop a clinically feasible signature. This signature may be a powerful prognostic tool and help further optimize the cancer therapy paradigm.

8.
Clin Transl Immunology ; 10(1): e1231, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33425353

RESUMEN

OBJECTIVES: The tumor mutational burden (TMB) is closely related to immunotherapy outcome. However, the cost of TMB detection is extremely high, which limits its use in clinical practice. A new indicator of genomic instability, the average copy number variation (CNVA), calculates the changes of 0.5-Mb chromosomal fragments and requires extremely low sequencing depth. METHODS: In this study, 50 samples (23 of which were from patients who received immunotherapy) were subjected to low-depth (10X) chromosome sequencing on the MGI platform. CNVA was calculated by the formula avg (abs (copy number-2)). In addition, CNVA and TMB were compared with regard to their ability to predict immune infiltration in 509 patients from TCGA. RESULTS: The high-CNVA group had higher expression levels of PD-L1, CD39 and CD19 and a higher degree of infiltration of CD8+ T cells and CD3 + T cells. Among the 23 patients treated with immunotherapy, the average CNVA value of the stable disease/partial response group was higher than that of the progressive disease group (P < 0.05). Whole-genome sequencing data of 509 patients from TCGA and RT-PCR results of 22 frozen specimens showed that CNVA is more effective than TMB in indicating infiltration of CD8+ T cells and expression of PD-L1, and CNVA also showed a specific positive correlation with TMB (r = 0.2728, P < 0.0001). CONCLUSIONS: Copy number variation can be a good indicator of immune infiltration and immunotherapy efficacy, and with its low cost, it is expected to become a substitute for TMB.

9.
Cell Death Dis ; 11(10): 933, 2020 10 30.
Artículo en Inglés | MEDLINE | ID: mdl-33127882

RESUMEN

Melanotransferrin (MFI2) is a newly identified tumor-associated protein, which consists of two forms of proteins, membrane-bound (mMFI2) and secretory (sMFI2). However, little is known about the expression pattern and their relevance in lung cancer. Here, we found that both two forms of MFI2 are highly expressed in lung cancer. The expression of MFI2 in lung cancer was detected by using the public database and qRT-PCR. Overexpression and knockdown cell lines and recombinant sMFI2 protein were used to study the function of mMFI2 and sMFI2. RNA-seq, protein chip, ChIP assay, Immunoprecipitation, ELISA, and immunofluorescence were used to study the molecular biological mechanism of mMFI2 and sMFI2. We found that mMFI2 promoted the expression of EMT's common marker N-cadherin by downregulating the transcription factor KLI4, which in turn promoted tumor metastasis; sMFI2 could promote the metastasis of autologous tumor cells in an autocrine manner but the mechanism is different from that of mMFI2. In addition, sMFI2 was proved could inhibit the migration of vascular endothelial cells and subsequently enhance angiogenic responses in a paracrine manner. We propose that the expressions and functions of the two forms of MFI2 in lung cancer are relatively independent. Specifically, mMFI2 was a potential lung cancer therapeutic target, while sMFI2 was highly enriched in advanced lung cancer, and could be used as a tumor staging index.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias Pulmonares/genética , Glicoproteínas de Membrana/metabolismo , Animales , Femenino , Humanos , Ratones , Transfección
10.
Cell Death Dis ; 11(10): 853, 2020 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-33056994

RESUMEN

The abnormal secretion of CA125, a classic tumor marker, is usually related to a poor prognosis in various tumors. Thus, this study aimed to explore the potential mechanisms that promote CA125 secretion in lung cancer. By querying the database, the gene endoplasmic reticulum oxidoreductase 1L (ERO1L) was identified and chosen as the research subject. The antibody chips were used to screen the lung cancer cell supernatant and found that the most obvious secreted protein was CA125. ERO1L was found to promote the secretion of IL6R by affecting the formation of disulfide bonds. IL6R bound to IL6 and triggered the activation of the NF-κB signaling pathway. Then, NF-κB bound to the promoter of MUC16, resulting in overexpression of MUC16. The extracellular segment of MUC16 was cleaved to form CA125, while the C terminus of MUC16 promoted the EMT phenotype and the release of IL6, forming a positive feedback pathway. In conclusion, ERO1L might affect the secretion of CA125 through the IL6 signaling pathway and form a positive feedback loop to further promote the development of lung cancer. This might expand the application scope of CA125 in lung cancer.


Asunto(s)
Antígeno Ca-125/metabolismo , Interleucina-6/metabolismo , Neoplasias Pulmonares/metabolismo , Glicoproteínas de Membrana/metabolismo , Proteínas de la Membrana/biosíntesis , Oxidorreductasas/metabolismo , Receptores de Interleucina-6/metabolismo , Animales , Antígeno Ca-125/biosíntesis , Detección Precoz del Cáncer , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Glicoproteínas de Membrana/genética , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Metástasis de la Neoplasia , Oxidorreductasas/genética , Pronóstico , Transducción de Señal
11.
J Exp Clin Cancer Res ; 39(1): 193, 2020 Sep 21.
Artículo en Inglés | MEDLINE | ID: mdl-32958011

RESUMEN

BACKGROUND: Angiogenesis, a basic requirement for tumor cell survival, is considered to be a malignant characteristic of small cell lung cancer (SCLC) and is closely related to the poor outcomes of SCLC patients. miR-141 has been found to play pro- and antiangiogenic roles in different cancers, but its role in SCLC angiogenesis has never been explored. METHODS: Total RNA was isolated from plasm exosomes and serum of SCLC patients to examine the expression of miR-141 by qRT-PCR. Cell proliferation, invasion, migration, tube formation assay, aortic ring assay and mouse tumor model were used to investigate the effect of exosomal miR-141 in angiogenesis in vitro and in vivo. Dual-luciferase assay was conducted to explore the target gene of miR-141. RESULTS: Circulating miR-141 was upregulated in samples from 122 SCLC patients compared with those from normal volunteers and that the increase in miR-141 was significantly associated with advanced TNM stages, implying the potential oncogenic role of miR-141 in SCLC malignancy. In vitro, miR-141 that was packaged into SCLC cell-secreted exosomes and delivered to human umbilical vein vascular endothelial cells (HUVECs) via exosomes facilitated HUVEC proliferation, invasion, migration and tube formation and promoted microvessel sprouting from mouse aortic rings. Matrigel plug assays demonstrated that SCLC cell-derived exosomal miR-141 induced neoangiogenesis in vivo. Furthermore, mouse subcutaneous tumor nodules that were developed from miR-141-overexpressing SCLC cells had a higher microvessel density (MVD) and grew faster than those developed from negative control cells. KLF12 was found to be the direct target gene of miR-141 and that the proangiogenic effect of miR-141 on HUVECs was abrogated by KLF12 overexpression. CONCLUSIONS: Our results demonstrate the specific function of the exosomal miR-141/KLF12 pathway in SCLC angiogenesis for the first time and provide potential novel targets for antiangiogenic therapies for SCLC patients.


Asunto(s)
Factores de Transcripción de Tipo Kruppel/genética , MicroARNs/genética , Neovascularización Patológica/genética , Carcinoma Pulmonar de Células Pequeñas/genética , Animales , Movimiento Celular/genética , Proliferación Celular/genética , Exosomas/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Xenoinjertos , Células Endoteliales de la Vena Umbilical Humana , Humanos , Factores de Transcripción de Tipo Kruppel/sangre , Masculino , Ratones , MicroARNs/sangre , Microvasos/crecimiento & desarrollo , Microvasos/patología , Persona de Mediana Edad , Neovascularización Patológica/sangre , Neovascularización Patológica/patología , Transducción de Señal/genética , Carcinoma Pulmonar de Células Pequeñas/sangre , Carcinoma Pulmonar de Células Pequeñas/patología
12.
Clin Transl Med ; 10(3): e128, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32659050

RESUMEN

BACKGROUND: Many tumor-derived endothelial cells (TECs) are shed into the blood and turn into circulating TECs (CTECs). Rare circulating non-hematologic aneuploid cells contain CTCs and CTECs, which are biologically and functionally different from each other. CD31 is one of the most representative endothelial cell (EC) markers, yet CD31 alone is not sufficient to detect malignant CTECs due to the existence of abundant normal ECs in circulation. Aneuploidy of chromosome 8 (CEP8) is an important criterion for the identification of malignant cells. Combined in situ phenotypic and karyotypic characterization, which includes an examination of both protein expression and aneuploid chromosomes, has demonstrated its unique advantage for both effective distinguishing and comprehensive detection of CTCs and CTECs. METHODS: A total of 98 subjects were recruited in the current study, including healthy donors and patients with benign disease and early-stage non-small-cell lung cancer (NSCLC). SE-iFISH was performed to quantitatively analyze diverse subtypes of aneuploid CD31+ CTECs and CD31- CTCs classified upon the ploidy of chromosome 8 and tumor marker expression in the specimens collected from the recruited subjects. RESULTS: CD31- CTCs primarily consist of triploid CTCs with a small cell size (≤5 µm) and large hyperploid CTCs (≥ pentaploid), whereas CD31+ CTECs are mainly comprised of large hyperploid cells. Enumeration of the total numbers of both CTCs and CTECs might help identify malignant nodules with a high sensitivity, whereas quantification of tetraploid CTCs and CTECs specifically exhibited a high specificity for the identification of malignant nodules. CONCLUSIONS: Combined detection of the specific subtypes of aneuploid CD31+ CTECs and CD31- CTCs may help to effectively identify malignant nodules with a higher sensitivity and specificity in early stage NSCLC patients.

13.
Biofabrication ; 12(4): 045014, 2020 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-32599574

RESUMEN

Towards the development of in vivo-mimicking tumor model for extensive study of tumorigenesis and establishment of personalized therapy, patient-derived primary tumor cells were employed in this work for three-dimensional (3D) bioprinting. Intrahepatic cholangiocarcinoma cells isolated from patient were bioprinted using a composite hydrogel system of gelatin-alginate-MatrigelTM into pre-designed grid architecture. ICC cells were observed to process a colony forming ability with high survival rate and active proliferation. Expression levels of tumor markers, cancer stem cell markers, matrix metalloproteinase protein, index of tumor fibrosis, index of liver function, and epithelial-mesenchymal transition regulatory proteins confirmed the development of the invasive and metastatic phenotype of the intrahepatic cholangiocarcinoma cells in the 3D printed tumor microenvironment. Similar results were obtained in anti-cancer drug resistance of the intrahepatic cholangiocarcinoma cells in the 3D bioprinted construct that demonstrated stem-like properties, which suggested the promising potential of current 3D printed tumor model in the development of personalized therapy, especially for discovery of more conducive targeted drugs.


Asunto(s)
Antineoplásicos/uso terapéutico , Neoplasias de los Conductos Biliares/tratamiento farmacológico , Bioimpresión , Colangiocarcinoma/tratamiento farmacológico , Ensayos de Selección de Medicamentos Antitumorales , Modelos Biológicos , Alginatos/química , Antineoplásicos/farmacología , Neoplasias de los Conductos Biliares/patología , Biomarcadores de Tumor/metabolismo , Colangiocarcinoma/patología , Colágeno/química , Combinación de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fibrosis , Gelatina/química , Humanos , Tinta , Laminina/química , Hígado/fisiopatología , Metaloproteinasas de la Matriz/metabolismo , Impresión Tridimensional , Proteoglicanos/química , Reología
14.
Cell Oncol (Dordr) ; 43(4): 681-694, 2020 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-32462608

RESUMEN

PURPOSE: Lung adenocarcinoma (LUAD) is the predominant subtype of lung cancer, with increasing evidence showing clinical benefits of immunotherapy. However, a lack of integrated profiles of complex LUAD immune microenvironments hampers the application of immunotherapy, resulting in limited eligible patient populations as well as drug resistance problems. Here, we aimed to systematically profile the immune signatures of LUADs and to assess the role of the immune microenvironment in patient outcome. METHODS: We systematically profiled the immune signatures of LUADs deposited in the TCGA and GEO databases using a total of 730 immune-related genes. Differential expression analysis was used to identify dysregulated genes. Univariate Cox analysis followed by robust likelihood-based survival analysis and multivariate Cox analysis were applied to construct an immune-related prognostic model. RESULTS: We found that differentially expressed immune genes were mainly enriched in immune cell proliferation, migration, activation and the NF-κB and TNF signaling pathways. The 10-immune gene predictive model that we constructed could differentiate LUAD patients with different overall survival times in several datasets, with areas under the curve (AUCs) of 0.67, 0.69, 0.72 and 0.74. LUAD patients with high- or low-risk scores exhibited distinct immune cell compositions, which may explain the prognostic significance of our model. CONCLUSIONS: Our results add to the current knowledge of immune processes in LUADs and underscore the critical role of the immune microenvironment in LUAD patient outcome.


Asunto(s)
Adenocarcinoma del Pulmón/inmunología , Biomarcadores de Tumor/inmunología , Neoplasias Pulmonares/inmunología , Microambiente Tumoral/inmunología , Adulto , Anciano , Biomarcadores de Tumor/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Transcriptoma , Microambiente Tumoral/genética
15.
Biofabrication ; 12(4): 042001, 2020 07 09.
Artículo en Inglés | MEDLINE | ID: mdl-32470967

RESUMEN

Studying biological characteristics of tumors and evaluating the treatment effects require appropriate in vitro tumor models. However, the occurrence, progression, and migration of tumors involve spatiotemporal changes, cell-microenvironment and cell-cell interactions, and signal transmission in cells, which makes the construction of in vitro tumor models extremely challenging. In the past few years, advances in biomaterials and tissue engineering methods, especially development of the bioprinting technology, have paved the way for innovative platform technologies for in vitro cancer research. Bioprinting can accurately control the distribution of cells, active molecules, and biomaterials. Furthermore, this technology recapitulates the key characteristics of the tumor microenvironment and constructs in vitro tumor models with bionic structures and physiological systems. These models can be used as robust platforms to study tumor initiation, interaction with the microenvironment, angiogenesis, motility and invasion, as well as intra- and extravasation. Bioprinted tumor models can also be used for high-throughput drug screening and validation and provide the possibility for personalized cancer treatment research. This review describes the basic characteristics of the tumor and its microenvironment and focuses on the importance and relevance of bioprinting technology in the construction of tumor models. Research progress in the bioprinting of monocellular, multicellular, and personalized tumor models is discussed, and comprehensive application of bioprinting in preclinical drug screening and innovative therapy is reviewed. Finally, we offer our perspective on the shortcomings of the existing models and explore new technologies to outline the direction of future development and application prospects of next-generation tumor models.


Asunto(s)
Bioimpresión , Neoplasias/terapia , Medicina de Precisión , Animales , Humanos , Microfluídica , Impresión Tridimensional , Microambiente Tumoral
16.
Cancer Sci ; 111(6): 1876-1886, 2020 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-32187778

RESUMEN

The tumor microenvironment (TME) is a vital component of tumor tissue. Increasing evidence suggests their significance in predicting outcomes and guiding therapies. However, no studies have reported a systematic analysis of the clinicopathologic significance of TME in lung adenocarcinoma (LUAD). Here, we inferred tumor stromal cells in 1184 LUAD patients using computational algorithms based on bulk tumor expression data, and evaluated the clinicopathologic significance of stromal cells. We found LUAD patients showed heterogeneous abundance in stromal cells. Infiltration of stromal cells was influenced by clinicopathologic features, such as age, gender, smoking, and TNM stage. By clustering stromal cells, we identified 2 clinically and molecularly distinct LUAD subtypes with immune active and immune repressed features. The immune active subtype is characterized by repressed metabolism and repressed proliferation of tumor cells, while the immune repressed subtype is characterized by active metabolism and active proliferation of tumor cells. Differentially expressed gene analysis of the two LUAD subtypes identified an immune activation signature. To diagnose TME subtypes practically, we constructed a TME score using principal component analysis based on the immune activation signature. The TME score predicted TME subtypes effectively in 3 independent datasets with areas under the receiver operating characteristic curves of 0.960, 0.812, and 0.819, respectively. In conclusion, we proposed 2 clinically and molecularly distinct LUAD subtypes based on tumor microenvironment that could be valuable in predicting clinical outcome and guiding immunotherapy.


Asunto(s)
Adenocarcinoma del Pulmón/clasificación , Neoplasias Pulmonares/clasificación , Microambiente Tumoral/fisiología , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/metabolismo , Algoritmos , Humanos , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/metabolismo , Sensibilidad y Especificidad
17.
Cancer Sci ; 111(5): 1739-1749, 2020 May.
Artículo en Inglés | MEDLINE | ID: mdl-32167618

RESUMEN

We aimed to verify the expression status and diagnostic significance of isocitrate dehydrogenase 1 (IDH1) in non-small-cell lung cancer (NSCLC), especially during early stages. Serum IDH1 levels were measured by ELISA. A total of 1223 participants (660 patients with NSCLC, 276 healthy controls [HCs], 95 patients with benign pulmonary conditions [BPCs], 135 patients with other cancers [OCs], and 57 samples with interfering factors) were divided into a training cohort and a validation cohort according to 3 testing centers. The IDH1 concentrations in the NSCLC group were obviously higher than those in the control groups (P < .001). Area under the receiver operating characteristic curves (AUCs) for discriminating NSCLC patients from controls (HC, BPC, and OC) were 0.870 and 0.745 (sensitivity, 63.3% and 55.0%; specificity, 86.8% and 86.3%) in the training cohort and validation cohort, respectively. The AUCs for discriminating stage 0-IA lung cancer patients from HCs were 0.907 and 0.788 (sensitivity, 58.6% and 59.1%; specificity, 92.9% and 89.3%) in 2 cohorts, respectively. Isocitrate dehydrogenase 1 showed specificity for NSCLC and had no diagnostic value for other common cancers. Furthermore, IDH1 was significantly reduced in postoperative serum. Isocitrate dehydrogenase 1 shows clinical utility as a serum protein biomarker for the early diagnosis of NSCLC.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/diagnóstico , Isocitrato Deshidrogenasa/sangre , Neoplasias Pulmonares/diagnóstico , Adulto , Anciano , Anciano de 80 o más Años , Área Bajo la Curva , Biomarcadores de Tumor/sangre , Carcinoma de Pulmón de Células no Pequeñas/sangre , Carcinoma de Pulmón de Células no Pequeñas/patología , Detección Precoz del Cáncer , Femenino , Humanos , Neoplasias Pulmonares/sangre , Neoplasias Pulmonares/patología , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados , Sensibilidad y Especificidad
18.
Cancer Lett ; 470: 95-105, 2020 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-31644929

RESUMEN

Immune checkpoint inhibitors against PD-1/PD-L1 yield improved survival rates of KRAS-mutant NSCLC patients, who conferred a poor prognosis without effective targeted therapy until now. Yet, the underlying association between KRAS mutations and immune responses remains unclear. We performed an integrated analysis of the data from publicly available repositories and from clinical center cohorts to explore the association between KRAS mutation status and tumor immunity-associated features, including PD-L1 expression, CD8+ tumor-infiltrating lymphocytes (TILs) and tumor mutational burden (TMB). Our results revealed that KRAS mutations are correlated with an inflammatory tumor microenvironment and tumor immunogenicity, resulting in superior patient response to PD-1/PD-L1 inhibitors. Meanwhile, three-pool analysis further confirmed that KRAS-mutant NSCLC patients show remarkable clinical benefit from anti-PD-1/PD-L1 immunotherapy. In addition, a KRAS-mutant lung adenocarcinoma mouse model was established to estimate the relative efficacy of anti-PD-L1 monoclonal antibody monotherapy or combination treatment with docetaxel versus docetaxel alone. Most surprisingly, we found that PD-L1 blockade combined with docetaxel did not promote an anti-tumor response. These findings uncover that PD-1/PD-L1 blockade monotherapy may be the optimal therapeutic schedule in NSCLC patients harboring KRAS mutations.


Asunto(s)
Adenocarcinoma del Pulmón/tratamiento farmacológico , Antineoplásicos Inmunológicos/uso terapéutico , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Proteínas Proto-Oncogénicas p21(ras)/genética , Adenocarcinoma del Pulmón/genética , Adenocarcinoma del Pulmón/inmunología , Adenocarcinoma del Pulmón/mortalidad , Animales , Antineoplásicos Inmunológicos/farmacología , Antígeno B7-H1/antagonistas & inhibidores , Antígeno B7-H1/inmunología , Linfocitos T CD8-positivos/inmunología , Carcinoma de Pulmón de Células no Pequeñas/genética , Carcinoma de Pulmón de Células no Pequeñas/inmunología , Carcinoma de Pulmón de Células no Pequeñas/mortalidad , Línea Celular Tumoral/trasplante , Estudios de Cohortes , Conjuntos de Datos como Asunto , Modelos Animales de Enfermedad , Femenino , Humanos , Pulmón/inmunología , Pulmón/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/inmunología , Neoplasias Pulmonares/mortalidad , Linfocitos Infiltrantes de Tumor/inmunología , Masculino , Ratones , Persona de Mediana Edad , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Receptor de Muerte Celular Programada 1/inmunología , Supervivencia sin Progresión , Ensayos Clínicos Controlados Aleatorios como Asunto , Escape del Tumor/genética , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
19.
Oncoimmunology ; 8(10): e1625689, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31646071

RESUMEN

Objectives: Although low-dose computed tomography has been confirmed to have meaningful diagnostic utility, lung cancer is still the leading cause of cancer-related deaths for both genders worldwide. Thus, a novel panel with a stronger diagnostic performance for lung cancer is needed. This study aimed to investigate the efficacy of a new panel in lung cancer diagnosis. Materials and Methods: The serum levels of carcinoembryonic antigen (CEA), cancer antigen 125 (CA125) and seven autoantibodies were measured and statistically analyzed in samples from healthy controls and patients with lung cancer. The 316 candidates enrolled in this study were randomly assigned into two groups for the training and validation of a diagnostic panel. Results: An optimal panel with four biomarkers (CEA, CA125, Annexin A1-Ab, and Alpha enolase-Ab) was established, with an area under the receiver operator characteristic (ROC) curve (AUC) of 0.897, a sensitivity of 86.5%, a specificity of 82.3%, a positive predictive value (PPV) of 88.3%, a negative predictive value (NPV) of 79.7%, and a diagnostic accuracy of 84.8% for the training group. The panel was validated, with an AUC of 0.856 and a sensitivity of 87.5% for the validation group. Furthermore, the new panel performed significantly better in lung cancer screening than did CEA and CA125 in all of the cohorts (p< .05). Conclusion: The diagnostic performance of CEA and CA125 was significantly enhanced through their combination with two autoantibodies (Annexin A1-Ab, and Alpha enolase-Ab). Optimization of the measured autoantibodies is critical for generating a panel to detect lung cancer in patients.

20.
Cancer Lett ; 459: 86-99, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31173852

RESUMEN

Interferons (IFNs) play crucial roles in the development and treatment of cancer. Long non-coding RNAs (lncRNAs) are emerging molecules involved in cancer progression. Here, we identified and characterized an IFN-inducible nuclear lncRNA IRF1-AS (Interferon Regulatory Factor 1 Antisense RNA) which was positively correlated with IRF1 expression. IFNs upregulate IRF1-AS via the JAK-STAT pathway. Knockdown and overexpression of IRF1-AS revealed that IRF1-AS inhibits oesophageal squamous cell carcinoma (ESCC) proliferation and promotes apoptosis in vitro and in vivo. Mechanistically, IRF1-AS activates IRF1 (Interferon Regulatory Factor 1) transcription through interacting with ILF3 (Interleukin Enhancer Binding Factor 3) and DHX9 (DExH-Box Helicase 9). In turn, IRF1 binds to the IRF1-AS promoter directly and activates IRF1-AS transcription. Global analysis of IRF1-AS-regulated genes indicated that IRF1-AS activates the IFN response in vitro and in vivo. IRF1 knockdown in IRF1-AS-overexpressing cells abolished the antiproliferative effect and activation of the IFN response. Furthermore, IRF1-AS was downregulated in ESCC tissues, and low expression correlated with poor prognosis. In conclusion, the interferon-inducible lncRNA IRF1-AS represses esophageal squamous cell carcinoma progression by promoting interferon response through a positive regulatory loop with IRF1.


Asunto(s)
Neoplasias Esofágicas/tratamiento farmacológico , Carcinoma de Células Escamosas de Esófago/tratamiento farmacológico , Factor 1 Regulador del Interferón/genética , Interferón beta/farmacología , Interferón gamma/farmacología , ARN sin Sentido/genética , ARN Largo no Codificante/genética , Animales , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/patología , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Humanos , Factor 1 Regulador del Interferón/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , ARN sin Sentido/biosíntesis , ARN sin Sentido/metabolismo , ARN Largo no Codificante/biosíntesis , ARN Largo no Codificante/metabolismo , Activación Transcripcional , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA