Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Biol Macromol ; 275(Pt 2): 133618, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38971275

RESUMEN

There have been notable irregularities in CMTM6 expression observed in hepatocellular carcinoma (HCC), with an evident correlation between CMTM6 dysregulation and patient prognosis. The cell cycle progression came to a halt at the G2/M phase. In-depth RNA-sequencing analysis of CMTM6 knockdown Hep3B cells revealed that the most prominent effect of CMTM6 perturbation was on the expression of CXCL8, a chemokine involved in immune responses, particularly through the interleukin-17F (IL-17F) signaling pathway. By carefully examining the RNA-sequencing data obtained from CMTM6 knockdown Hep3B cells and cross-referencing it with the TCGA-LIHC database, we were able to discern that CMTM6 and programmed death-ligand 1 (PD-L1) collaboratively partake in immune regulation within T cells. Furthermore, CMTM6 exerted an influential role in modulating the infiltration of CD4+ and CD8+ T cells in the HCC microenvironment, thereby impacting the overall immune response. Our investigation found that HCC cases characterized by an elevated co-expression of CMTM6 and PD-L1, along with augmented CD4+ T cell infiltration, demonstrated comparatively longer overall and progression-free survival rates when contrasted with those displaying lower CD4+ T cell infiltration.


Asunto(s)
Carcinoma Hepatocelular , Regulación Neoplásica de la Expresión Génica , Neoplasias Hepáticas , Proteínas con Dominio MARVEL , Microambiente Tumoral , Humanos , Carcinoma Hepatocelular/inmunología , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Carcinoma Hepatocelular/metabolismo , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/inmunología , Neoplasias Hepáticas/patología , Neoplasias Hepáticas/metabolismo , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Proteínas con Dominio MARVEL/genética , Proteínas con Dominio MARVEL/metabolismo , Pronóstico , Línea Celular Tumoral , Proteínas de la Mielina/genética , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Masculino , Femenino , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo
2.
J Gastrointest Oncol ; 15(3): 1082-1100, 2024 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-38989413

RESUMEN

Background: Hepatocellular carcinoma (HCC) poses a global threat to life; however, numerical tools to predict the clinical prognosis of these patients remain scarce. The primary objective of this study is to establish a clinical scoring system for evaluating the overall survival (OS) rate and cancer-specific survival (CSS) rate in HCC patients. Methods: From the Surveillance, Epidemiology, and End Results (SEER) Program, we identified 45,827 primary HCC patients. These cases were randomly allocated to a training cohort (22,914 patients) and a validation cohort (22,913 patients). Univariate and multivariate Cox regression analyses, coupled with Kaplan-Meier methods, were employed to evaluate prognosis-related clinical and demographic features. Factors demonstrating prognostic significance were used to construct the model. The model's stability and accuracy were assessed through C-index, receiver operating characteristic (ROC) curves, calibration curves, and clinical decision curve analysis (DCA), while comparisons were made with the American Joint Committee on Cancer (AJCC) staging. Ultimately, machine learning (ML) quantified the variables in the model to establish a clinical scoring system. Results: Univariate and multivariate Cox regression analyses identified 11 demographic and clinical-pathological features as independent prognostic indicators for both CSS and OS using. Two models, each incorporating the 11 features, were developed, both of which demonstrated significant prognostic relevance. The C-index for predicting CSS and OS surpassed that of the AJCC staging system. The area under the curve (AUC) in time-dependent ROC consistently exceeded 0.74 in both the training and validation sets. Furthermore, internal and external calibration plots indicated that the model predictions aligned closely with observed outcomes. Additionally, DCA demonstrated the superiority of the model over the AJCC staging system, yielding greater clinical net benefit. Ultimately, the quantified clinical scoring system could efficiently discriminate between high and low-risk patients. Conclusions: A ML clinical scoring system trained on a large-scale dataset exhibits good predictive and risk stratification performance in the cohorts. Such a clinical scoring system is readily integrable into clinical practice and will be valuable in enhancing the accuracy and efficiency of HCC management.

3.
J Int Med Res ; 52(5): 3000605241247695, 2024 May.
Artículo en Inglés | MEDLINE | ID: mdl-38775372

RESUMEN

Giant choledochal cysts are rare, and so little data exist on the best surgical treatment method. We present here, a case of a giant choledochal cyst that was successfully excised by laparoscopic resection. A 37-year-old female presented with right upper abdominal pain and mild jaundice. On examination she had a right upper abdominal mass which on imaging was observed to be a giant choledochal cyst of type IVa, measuring approximately 129 mm × 190 mm. Her blood test results showed abnormal liver function. We successfully performed laparoscopic resection of the cyst, the patient recovered well and was discharged from hospital eight days post-operation without any complications. We wish to share the experience of this rare case and provide some clinical basis for future diagnosis and use of laparoscopic resection in the treatment of giant choledochal cysts.


Asunto(s)
Quiste del Colédoco , Laparoscopía , Humanos , Quiste del Colédoco/cirugía , Quiste del Colédoco/diagnóstico , Quiste del Colédoco/diagnóstico por imagen , Femenino , Adulto , Laparoscopía/métodos , Tomografía Computarizada por Rayos X
4.
BMC Gastroenterol ; 24(1): 142, 2024 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-38654165

RESUMEN

OBJECTIVES: Cuproptosis represents an innovative type of cell death, distinct from apoptosis, driven by copper dependency, yet the involvement of copper apoptosis-associated long non-coding RNAs (CRLncRNAs) in hepatocellular carcinoma (HCC) remains unclear. This study is dedicated to unveiling the role and significance of these copper apoptosis-related lncRNAs within the context of HCC, focusing on their impact on both the development of the disease and its prognosis. METHODS: We conducted an analysis of gene transcriptomic and clinical data for HCC cases by sourcing information from The Cancer Genome Atlas database. By incorporating cuproptosis-related genes, we established prognostic features associated with cuproptosis-related lncRNAs. Furthermore, we elucidated the mechanism of cuproptosis-related lncRNAs in the prognosis and treatment of HCC through comprehensive approaches, including Lasso and Cox regression analyses, survival analyses of samples, as well as examinations of tumor mutation burden and immune function. RESULTS: We developed a prognostic model featuring six cuproptosis-related lncRNAs: AC026412.3, AC125437.1, AL353572.4, MKLN1-AS, TMCC1-AS1, and SLC6A1-AS1. This model demonstrated exceptional prognostic accuracy in both training and validation cohorts for patients with tumors, showing significantly longer survival times for those categorized in the low-risk group compared to the high-risk group. Additionally, our analyses, including tumor mutation burden, immune function, Gene Ontology, Kyoto Encyclopedia of Genes and Genomes pathway enrichment, and drug sensitivity, further elucidated the potential mechanisms through which cuproptosis-associated lncRNAs may influence disease outcome. CONCLUSIONS: The model developed using cuproptosis-related long non-coding RNAs (lncRNAs) demonstrates promising predictive capabilities for both the prognosis and immunotherapy outcomes of tumor patients. This could play a crucial role in patient management and the optimization of immunotherapeutic strategies, offering valuable insights for future research.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , ARN Largo no Codificante , Humanos , ARN Largo no Codificante/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/patología , Pronóstico , Cobre , Apoptosis/genética , Masculino , Biomarcadores de Tumor/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Transcriptoma , Análisis de Supervivencia
5.
J Cell Mol Med ; 28(5): e18106, 2024 03.
Artículo en Inglés | MEDLINE | ID: mdl-38239038

RESUMEN

Hepatocellular carcinoma (HCC) is the fifth most frequently diagnosed cancer and ranks third in cancer-related fatalities. The recognized involvement of long noncoding RNAs (lncRNAs) in several cancer types, including HCC, inspired this study to explore a novel lncRNA's functional importance in the progression of HCC. To achieve this, lncRNA microarray analysis was conducted on three distinct sets of HCC tissues, revealing LINC00707 as the most significantly upregulated lncRNA. Further research into its biological functions has revealed that LINC00707 acts as an oncogene, driving HCC progression by enhancing the proliferation, migration and invasion of HCC cells. Mechanistic insights were provided, demonstrating that LINC00707 interacts with YTH N6-methyladenosine RNA-binding protein 2 (YTHDF2), thus facilitating the ubiquitination-dependent degradation of the YTHDF2 protein. Furthermore, LINC00707 was found to influence the cytotoxicity of NK-92MI cells against HCC cells through its interactions with YTHDF2. These findings significantly contribute to a deeper understanding of the role played by LINC00707 in the progression of HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , ARN Largo no Codificante , Humanos , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Células Asesinas Naturales/metabolismo , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética , Línea Celular Tumoral , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
6.
J Cell Mol Med ; 28(1): e18018, 2024 01.
Artículo en Inglés | MEDLINE | ID: mdl-37944063

RESUMEN

Metabolic pathways exert a significant influence on the onset and progression of cancer. Public data on hepatocellular carcinoma (HCC) patients were obtained from The Cancer Genome Atlas (TCGA) and International Cancer Genome Consortium (ICGC) databases. Analysis was performed in R software using different R packages. Here, we integrated the data from multiple independent HCC cohorts, including TCGA-LIHC, ICGC-FR and ICGC-JP. Then, the enrichment score of 21 metabolism-related pathways was quantified using the ssGSEA algorithm. Next, univariate Cox regression analysis was applied to identify the metabolic terms with significant correlation to patient survival. Finally, a prognosis model based on linoleic acid metabolism, sphingolipid metabolism and regulation of lipolysis in adipocytes was established, which showed good performance in predicting patients' survival. Furthermore, we conducted a biological enrichment analysis to delineate the biological disparities between high- and low-risk patients. Notably, we discerned differences in the microenvironments between these two patient groups. We also found that low-risk patients could potentially respond better to immunotherapy. Drug sensitivity analysis suggested that low-risk patients are more susceptible to bexarotene and erlotinib, yet exhibit resistance to ATRA and bleomycin. Furthermore, through the use of LASSO logistic regression analysis, we identified 19 characteristic genes, which could robustly indicate the risk groups. Our research underscores the role of linoleic acid metabolism, sphingolipid metabolism and the regulation of lipolysis in adipocytes in HCC, pointing towards potential avenues for future research.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Ácido Linoleico , Inmunoterapia , Esfingolípidos , Microambiente Tumoral
7.
Cancer Sci ; 114(9): 3649-3665, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37400994

RESUMEN

As an epitranscriptomic modulation manner, N6 -methyladenosine (m6 A) modification plays important roles in various diseases, including hepatocellular carcinoma (HCC). m6 A modification affects the fate of RNAs. The potential contributions of m6 A to the functions of RNA still need further investigation. In this study, we identified long noncoding RNA FAM111A-DT as an m6 A-modified RNA and confirmed three m6 A sites on FAM111A-DT. The m6 A modification level of FAM111A-DT was increased in HCC tissues and cell lines, and increased m6 A level was correlated with poor survival of HCC patients. m6 A modification increased the stability of FAM111A-DT transcript, whose expression level showed similar clinical relevance to that of the m6 A level of FAM111A-DT. Functional assays found that only m6 A-modified FAM111A-DT promoted HCC cellular proliferation, DNA replication, and HCC tumor growth. Mutation of m6 A sites on FAM111A-DT abolished the roles of FAM111A-DT. Mechanistic investigations found that m6 A-modified FAM111A-DT bound to FAM111A promoter and also interacted with m6 A reader YTHDC1, which further bound and recruited histone demethylase KDM3B to FAM111A promoter, leading to the reduction of the repressive histone mark H3K9me2 and transcriptional activation of FAM111A. The expression of FAM111A was positively correlated with the m6 A level of FAM111A-DT, and the expression of methyltransferase complex, YTHDC1, and KDM3B in HCC tissues. Depletion of FAM111A largely attenuated the roles of m6 A-modified FAM111A-DT in HCC. In summary, the m6 A-modified FAM111A-DT/YTHDC1/KDM3B/FAM111A regulatory axis promoted HCC growth and represented a candidate therapeutic target for HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , ARN Largo no Codificante , Humanos , Carcinoma Hepatocelular/patología , Neoplasias Hepáticas/patología , Línea Celular Tumoral , Activación Transcripcional , Proliferación Celular/genética , ARN , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Histona Demetilasas con Dominio de Jumonji/genética , Histona Demetilasas con Dominio de Jumonji/metabolismo , Receptores Virales/genética
8.
Comput Math Methods Med ; 2023: 8958962, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36785674

RESUMEN

Background: Immune microenvironment implicated in liver cancer development. Nevertheless, previous studies have not fully investigated the immune microenvironment in liver cancer. Methods: The open-access data used for analysis were obtained from The Cancer Genome Atlas (TCGA-LIHC) and the International Cancer Genome Consortium databases (ICGC-JP and ICGC-FR). R program was employed to analyze all the data statistically. Results: First, the TCGA-LIHC, ICGC-FR, and ICGC-JP cohorts were selected for our analysis, which were merged into a combined cohort. Then, we quantified 53 immune terms in this combined cohort with large populations using the ssGSEA algorithm. Next, a prognostic approach was established based on five immune principles (CORE.SERUM.RESPONSE.UP, angiogenesis, CD8.T.cells, Th2.cells, and B.cells) was established, which showed great prognostic prediction efficiency. Clinical correlation analysis demonstrated that high-risk patients could reveal higher progressive clinical features. Next, to examine the inherent biological variations in high- and low-risk patients, pathway enrichment tests were conducted. DNA repair, E2F targets, G2M checkpoints, HEDGEHOG signaling, mTORC1 signaling, and MYC target were positively correlated with the risk score. Examination of genomic instability revealed that high-risk patients may exhibit a higher tumor mutation burden score. Meanwhile, the risk score showed a strong positive correlation with the tumor stemness index. In addition, the Tumor Immune Dysfunction and Exclusion outcome indicated that high-risk patients could be higher responsive to immunotherapy, whereas low-risk patients may be higher responsive to Erlotinib. Finally, six characteristic genes DEPDC1, DEPDC1B, NGFR, CALCRL, PRR11, and TRIP13 were identified for risk group prediction. Conclusions: In summary, our study identified a signature as a useful tool to indicate prognosis and therapy options for liver cancer patients.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Humanos , Proteínas Hedgehog , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/terapia , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/terapia , Pronóstico , Inmunoterapia , Microambiente Tumoral/genética , ATPasas Asociadas con Actividades Celulares Diversas , Proteínas de Ciclo Celular , Proteínas de Neoplasias , Proteínas Activadoras de GTPasa
9.
Pathol Res Pract ; 237: 153955, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35841693

RESUMEN

BACKGROUND: Hepatocellular Carcinoma (HCC) is recognized as the second leading cause of cancer-associated deaths globally. Hypoxia-inducible factor 1alpha (HIF1A) has been documented to promote HCC cell migration, invasion and cell cycle. Dual specificity phosphatase 18 (DUSP18) has been predicted to be up-regulated in hypoxia and its expression is positively linked to HIF1A expression in HCC cells. However, their function and molecular mechanism have not been investigated in HCC in depth. PURPOSE: This study aimed to uncover the functional roles of HIF1A and DUSP18, as well as relevant mechanisms underlying their regulation in HCC cells. METHODS: RT-qPCR and western blot were performed to examine gene expression. Functional assays were implemented to reveal the regulatory impact of target genes on HCC cells. Mechanism experiments were conducted to analyze gene interaction. RESULTS: DUSP18 was found to have significantly high expression in hypoxia-induced HCC cells. HIF1A promoted HCC cell migration, invasion and cell cycle by transcriptionally activating DUSP18. DUSP18 mediated MAPK14 dephosphorylation to weaken MAPK14 activity, which further inhibited MAPK14-mediated TP53 phosphorylation, consequently promoting multiple biological behaviors of HCC cells. CONCLUSION: Hypoxia-induced HIF1A activates DUSP18 transcription to further promote MAPK14 dephosphorylation, thereby suppressing TP53 phosphorylation and functionally promoting malignant behaviors of HCC cells.


Asunto(s)
Carcinoma Hepatocelular , Fosfatasas de Especificidad Dual , Subunidad alfa del Factor 1 Inducible por Hipoxia , Neoplasias Hepáticas , Proteína Quinasa 14 Activada por Mitógenos , Humanos , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Fosfatasas de Especificidad Dual/genética , Fosfatasas de Especificidad Dual/metabolismo , Regulación Neoplásica de la Expresión Génica , Hipoxia , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/patología , Proteína Quinasa 14 Activada por Mitógenos/genética , Transducción de Señal/genética
10.
Cell Death Dis ; 13(3): 223, 2022 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-35273149

RESUMEN

Targeting key genes that play dominant roles in T cell dysfunction is an efficient strategy for cancer immunotherapy. Here, we aimed to investigate the role of TPX2 in the antitumor effect of CD8 + T cells in hepatocellular carcinoma (HCC). Flow cytometry was used to assay the level of cell surface markers and cytokines in T cells, through which we found that TPX2 was downregulated in HCC-infiltrating CD8 + T cells. TPX2 depletion restricted the antitumor activity of CD8 + T cells, and TPX2 overexpression increased the antitumor effect of CD8 + T cells in tumor-bearing Cd8-/- mice. TPX2 overexpression improved the antitumor function of human CD8 + T cells and response to anti-PD-1 therapy in an HCC patient-derived xenograft (PDX) mouse model with or without anti-PD-1 therapy. In mechanism, TPX2 promotes the phosphorylation of P65, thus increases the level of p-P65 in nuclear, and p-P65 binds to the promoter of CXCR5, activates its transcription, and increases the level of CXCR5 on CD8 + T cells in a TPX2-dependent way. In conclusion, TPX2 maintains the antitumor effect of CD8 + T cells in HCC by regulating CXCR5 via the NF-κB signaling pathway. Increased TPX2 expression in CD8 + T cells exerts synergistic effects with anti-PD-1 therapy, suggesting a promising immunotherapeutic method in HCC.


Asunto(s)
Carcinoma Hepatocelular , Neoplasias Hepáticas , Animales , Linfocitos T CD8-positivos/metabolismo , Carcinoma Hepatocelular/patología , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Regulación hacia Abajo , Humanos , Inmunoterapia , Neoplasias Hepáticas/patología , Ratones , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Proteínas Nucleares
11.
Cell Death Dis ; 13(2): 102, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-35110549

RESUMEN

Hepatocellular carcinoma (HCC) is one of the leading lethal malignancies and a hypervascular tumor. Although some long non-coding RNAs (lncRNAs) have been revealed to be involved in HCC. The contributions of lncRNAs to HCC progression and angiogenesis are still largely unknown. In this study, we identified a HCC-related lncRNA, CMB9-22P13.1, which was highly expressed and correlated with advanced stage, vascular invasion, and poor survival in HCC. We named this lncRNA Progression and Angiogenesis Associated RNA in HCC (PAARH). Gain- and loss-of function assays revealed that PAARH facilitated HCC cellular growth, migration, and invasion, repressed HCC cellular apoptosis, and promoted HCC tumor growth and angiogenesis in vivo. PAARH functioned as a competing endogenous RNA to upregulate HOTTIP via sponging miR-6760-5p, miR-6512-3p, miR-1298-5p, miR-6720-5p, miR-4516, and miR-6782-5p. The expression of PAARH was significantly positively associated with HOTTIP in HCC tissues. Functional rescue assays verified that HOTTIP was a critical mediator of the roles of PAARH in modulating HCC cellular growth, apoptosis, migration, and invasion. Furthermore, PAARH was found to physically bind hypoxia inducible factor-1 subunit alpha (HIF-1α), facilitate the recruitment of HIF-1α to VEGF promoter, and activate VEGF expression under hypoxia, which was responsible for the roles of PAARH in promoting angiogenesis. The expression of PAARH was positively associated with VEGF expression and microvessel density in HCC tissues. In conclusion, these findings demonstrated that PAARH promoted HCC progression and angiogenesis via upregulating HOTTIP and activating HIF-1α/VEGF signaling. PAARH represents a potential prognostic biomarker and therapeutic target for HCC.


Asunto(s)
Carcinoma Hepatocelular/patología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Neoplasias Hepáticas/patología , Neovascularización Patológica/patología , ARN Largo no Codificante/fisiología , Factor A de Crecimiento Endotelial Vascular/genética , Animales , Apoptosis/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Regulación Neoplásica de la Expresión Génica , Humanos , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Ratones , MicroARNs/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , ARN Largo no Codificante/genética , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo
12.
Artif Cells Nanomed Biotechnol ; 50(1): 10-16, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35086395

RESUMEN

To explore the association between the single nucleotide polymorphism (SNP) of leptin receptor (LEPR) gene and the susceptibility to osteoporosis (OP) among Chinese Mulao people. A total of 738 people were involved. Bone mineral density (BMD) was examined by calcaneus ultrasound attenuation measurement. Six SNPs of LEPR were detected. The genotypes, allele frequencies, linkage disequilibrium, and haplotype were analyzed. BMD decreased with age and males had higher BMD than women. The proportion of normal bone mass decreased with age, and morbidity of OP increased. Three out of six SNPs showed a difference between OP and normal group. Individuals with AA genotype of rs1137100 in OP group outnumber the normal group, AA increased the risk of OP. In rs2767485, CT increased the risk of OP, C allele may be susceptible to OP. TT genotype of rs465555 was susceptible genotype of OP, T locus may be associated with OP. Strong linkage disequilibrium was detected among rs1137100, rs1137101, and rs4655555. Four haplotypes were constructed, among which, AACGCT and GGTGTA increased the risk of OP by 3.9 and 4.2 times, respectively, whereas, GGCGTA reduced 74% of OP susceptibility. The rs1137100, rs2767485, and rs465555 of LEPR were associated with OP in Chinese Mulao people.


Asunto(s)
Pueblo Asiatico , Osteoporosis , Pueblo Asiatico/genética , China/epidemiología , Femenino , Frecuencia de los Genes , Genotipo , Humanos , Masculino , Osteoporosis/diagnóstico por imagen , Osteoporosis/genética , Polimorfismo de Nucleótido Simple/genética , Receptores de Leptina
13.
Cell Death Discov ; 7(1): 385, 2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34897283

RESUMEN

In solid tumors, hypoxia facilitates malignant progression of cancer cells by triggering epithelial-mesenchymal transition (EMT) and cancer stemness. Fascin-1, an actin-bundling protein, takes part in the formation of many actin-based cellular structures. In the present study, we explored the potential functions of hypoxia-induced upregulation of Fascin-1 in liver cancer. Transcriptome RNA-sequencing was conducted to identify hypoxia-related genes. The potential functions of Fascin-1 were evaluated by western blot, transwell migration and invasion assays, sphere-formation assay, tumor xenograft growth, gelatin zymography analysis, immunofluorescence, cell viability assay, soft agar assay, and flow cytometry. We found that Fascin-1 was upregulated by hypoxia in liver cancer cell lines, elevated in liver cancer patients and correlated with larger tumor size, lymph node metastasis, distant metastasis, and shorter overall survival. Knockdown of Fascin-1 suppressed migration, invasion, EMT, stemness, and tumor xenograft growth of liver cancer cells under both normoxia and hypoxia conditions, while forced Fascin-1 expression showed opposite effects. Moreover, hypoxia-induced upregulation of Fascin-1 was regulated by the Akt/Rac1 signaling, and inhibition of Akt/Rac1 signaling by EHop-016 and MK-2206 restrained migration, invasion, EMT, and stemness of liver cancer cells under hypoxia. Furthermore, Fascin-1 knockdown suppressed MMP-2 and MMP-9 expression, impaired actin cytoskeleton rearrangement, inactivated Hippo/YAP signaling, and increased Sorafenib sensitivity in liver cancer cells. Our study provided a novel insight of Fascin-1 in regulating migration, invasion, EMT, and stemness of liver cancer cells under normoxia and hypoxia conditions.

14.
J Immunol Res ; 2021: 3759879, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34722778

RESUMEN

Accumulating evidence indicates that circular RNAs (circRNAs) can interact with microRNAs to modulate gene expression in various cancers, including hepatocellular carcinoma (HCC). Although the significant role of circRNAs has been well documented in HCC, the complex mechanisms of circRNAs still need to be elucidated. Our current study is aimed at investigating the function of circ_0001588 in HCC, which was observed to significantly increase in HCC tissues and cells. We demonstrated that the knockdown of circ_0001588 resulted in repressed cell proliferation, migration, and invasion. In vivo studies using a nude mouse model showed that circ_0001588 downregulation reduced tumor size. Moreover, miR-874 was predicted as a target of circ_0001588. Using luciferase binding assays, we proved that circ_0001588 functions as a molecular ceRNA of miR-874 and that CDK4 acts as a downstream target of miR-874 in HCC. It was confirmed that overexpression of miR-874 decreased the proliferation, migration, and invasion triggered by the increase in circ_0001588. In summary, our results indicate that circ_0001588 acts as a ceRNA and promotes HCC progression by targeting the miR-874/CDK4 signaling pathway. Hence, we propose that circ_0001588 may be a promising target for HCC treatment.


Asunto(s)
Carcinoma Hepatocelular/genética , Quinasa 4 Dependiente de la Ciclina/genética , Neoplasias Hepáticas/genética , MicroARNs/metabolismo , ARN Circular/metabolismo , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Neoplasias Hepáticas/patología , Ratones , ARN Circular/genética , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Cell Death Dis ; 12(12): 1103, 2021 11 23.
Artículo en Inglés | MEDLINE | ID: mdl-34815380

RESUMEN

The crosstalk between cancer cells and tumor microenvironment plays critical roles in hepatocellular carcinoma (HCC). The identification of long non-coding RNAs (lncRNAs) mediating the crosstalk might promote the development of new therapeutic strategies against HCC. Here, we identified a lncRNA, HOMER3-AS1, which is over-expressed in HCC and correlated with poor survival of HCC patients. HOMER3-AS1 promoted HCC cellular proliferation, migration, and invasion, and reduced HCC cellular apoptosis. Furthermore, HOMER3-AS1 promoted macrophages recruitment and M2-like polarization. In vivo, HOMER3-AS1 significantly facilitated HCC progression. Mechanism investigations revealed that HOMER3-AS1 activated Wnt/ß-catenin signaling via upregulating HOMER3. Functional rescue experiments revealed that HOMER3/Wnt/ß-catenin axis mediated the roles of HOMER3-AS1 in promoting HCC cellular malignant phenotypes. Furthermore, colony stimulating factor-1 (CSF-1) was also identified as a critical downstream target of HOMER3-AS1. HOMER3-AS1 increased CSF-1 expression and secretion. Blocking CSF-1 reversed the roles of HOMER3-AS1 in inducing macrophages recruitment and M2 polarization. Furthermore, positive correlations between HOMER3-AS1 and HOMER3 expression, HOMER3-AS1 and CSF-1 expression, and HOMER3-AS1 expression and M2-like macrophages infiltration were found in human HCC tissues. In summary, our findings demonstrated that HOMER3-AS1 drives HCC progression via modulating the behaviors of both tumor cells and macrophages, which are dependent on the activation of HOMER3/Wnt/ß-catenin axis and CSF-1, respectively. HOMER3-AS1 might be a promising prognostic and therapeutic target for HCC.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Macrófagos/metabolismo , ARN Largo no Codificante/genética , Animales , Carcinoma Hepatocelular/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Femenino , Humanos , Neoplasias Hepáticas/patología , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Oncogenes , Pronóstico , Células Tumorales Cultivadas
16.
Front Oncol ; 11: 754835, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34733789

RESUMEN

BACKGROUND: Hepatocellular carcinoma (HCC) is one of the most aggressive malignancies. Increasing evidence revealed that long noncoding RNAs (lncRNAs) were frequently involved in various malignancies. Here, we explored the clinical significances, roles, and mechanisms of lncRNA ADORA2A antisense RNA 1 (ADORA2A-AS1) in HCC. METHODS: The clinical significances of ADORA2A-AS1 in HCC were analyzed using RNA sequencing (RNA-seq) data from The Cancer Genome Atlas (TCGA) project. The expressions of ADORA2A-AS1, Fascin Actin-Bundling Protein 1 (FSCN1), Matrix Metallopeptidase 2 (MMP2), and Baculoviral IAP Repeat Containing 7 (BIRC7) in HCC tissues and cells were measured by qRT-PCR. Cell Counting Kit-8 (CCK-8), 5-ethynyl-2'-deoxyuridine (EdU), caspase-3 activity assay, transwell migration and invasion assays, and xenograft growth and metastasis experiments were performed to evaluate the roles of ADORA2A-AS1 in HCC. RNA pull-down, RNA immunoprecipitation, qRT-PCR, Western blot, and RNA stability assay were performed to elucidate the mechanisms of ADORA2A-AS1 in HCC. RESULTS: ADORA2A-AS1 was identified as an HCC-related lncRNA, whose low expression was correlated with advanced stage and poor outcome in HCC. Gain- and loss-of functional experiments demonstrated that ADORA2A-AS1 inhibited HCC cell proliferation, induced cell apoptosis, repressed cell migration and invasion, and repressed xenograft growth and metastasis in vivo. Mechanistically, ADORA2A-AS1 competitively bound HuR (Hu Antigen R), repressed the binding of HuR to FSCN1 transcript, decreased FSCN1 transcript stability, and downregulated FSCN1 expression. The expression of FSCN1 was negatively correlated with ADORA2A-AS1 in HCC tissues. Through downregulating FSCN1, ADORA2A-AS1 repressed AKT pathway activation. Functional rescue assays showed that blocking of FSCN1/AKT axis abrogated the roles of ADORA2A-AS1 in HCC. CONCLUSION: Low-expression ADORA2A-AS1 is correlated with poor survival of HCC patients. ADORA2A-AS1 exerts tumor-suppressive roles in HCC via binding HuR and repressing FSCN1/AKT axis.

18.
J Oncol ; 2021: 5547345, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34484334

RESUMEN

INTRODUCTION: Our work was aimed to explore the mechanisms of MAGI2 antisense RNA 3 (MAGI2-AS3) in regulating hepatocellular carcinoma (HCC) carcinogenesis. METHODS: MAGI2-AS3, microRNA-519c-3p (miR-519c-3p), and thioredoxin interacting protein (TXNIP) levels in HCC were detected by the RT-qPCR method. Cell proliferation and apoptosis rate were measured using Cell Counting Kit-8 assay and flow cytometry assay. Relationship between MAGI2-AS3, TXNIP, and miR-519c-3p were analyzed via luciferase activity assay, RNA pull-down assay, and RNA immunoprecipitation assay. Mouse xenograft models of HCC were conducted to explore the roles of MAGI2-AS3 in vivo. RESULTS: MAGI2-AS3 levels were elevated, and miR-519c-3p decreased in HCC. MAGI2-AS3 overexpression inhibits while its knockdown stimulates HCC cell growth through miR-519c-3p. Moreover, miR-519c-3p overexpression stimulates HCC cell growth. MAGI2-AS3 serves as competing endogenous RNA (ceRNA) of miR-519c-3p to regulate TXNIP in HCC. And, TXNIP upregulation weakened the influence of MAGI2-AS3 knockdown on HCC cell behaviors. Additionally, MAGI2-AS3 overexpression suppressed HCC tumor growth in vivo. CONCLUSION: MAGI2-AS3 inhibits HCC tumorigenesis through miR-519c-3p/TXNIP axis in vitro and in vivo, indicating MAGI2-AS3 plays a crucial role in HCC development.

19.
Aging (Albany NY) ; 13(14): 18740-18756, 2021 07 29.
Artículo en Inglés | MEDLINE | ID: mdl-34351868

RESUMEN

In this study, we investigated the mechanistic role and prognostic significance of IGSF10 in lung adenocarcinoma. Oncomine database analysis showed that IGSF10 expression was significantly reduced in most cancer types, including lung adenocarcinoma (LUAD). In the TCGA-LUAD dataset, IGSF10 expression correlated positively with proportions of tumor-infiltrated B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages, and dendritic cells. Kaplan-Meier survival analysis showed that overall survival of patients with low IGSF10 expression was significantly shorter than those with high IGSF10 expression. MiRWalk2.0 database analysis and dual luciferase reporter assays confirmed that miR-106b-5p suppressed IGSF10 expression by binding to its 3'UTR. MiR-106b-5p levels inversely correlated with IGSF10 expression in the TCGA-LUAD dataset. Moreover, inhibition of miR-106b-5p significantly decreased in vitro proliferation, migration, and invasion by LUAD cells, whereas miR-106b-5p overexpression reversed those effects. These results demonstrate that IGSF10 is an independent prognostic factor for LUAD. Furthermore, miR-106b-5p suppressed IGSF10 expression in LUAD tissues by binding to its 3'UTR, which makes IGSF10 and miR-106b-5p potential prognostic biomarkers and therapeutic targets in LUAD patients.


Asunto(s)
Adenocarcinoma del Pulmón/genética , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Inmunoglobulinas/genética , Neoplasias Pulmonares/genética , MicroARNs/genética , Adenocarcinoma del Pulmón/metabolismo , Línea Celular Tumoral , Proliferación Celular , Bases de Datos Factuales , Conjuntos de Datos como Asunto , Femenino , Humanos , Inmunoglobulinas/metabolismo , Leucocitos/metabolismo , Neoplasias Pulmonares/metabolismo , Masculino , MicroARNs/metabolismo , Invasividad Neoplásica , Pronóstico , Análisis de Supervivencia
20.
Cell Death Discov ; 7(1): 182, 2021 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-34282135

RESUMEN

Hepatocellular carcinoma (HCC) is a common malignancy. CD8+ T cell-mediated immune response is critical for the inhibition of HCC progression. M2 macrophages participate in HCC progression. This study set out to investigate the effect of M2 macrophage-derived extracellular vesicles (EVs) on CD8+ T cell exhaustion in HCC. M2 macrophage-derived EVs were isolated and identified. The murine model of primary HCC was established through DEN/CCl4 induction, and model mice were injected with EVs. Peripheral blood mononuclear cells (PBMCs) were isolated from the mouse liver and CD8+ T cells were sorted. The expressions of immune checkpoint inhibitory receptors and effector cytokines on CD8+ T cells were detected, followed by the evaluation of CD8+ T cell proliferation and killing function. miR-21-5p expression in M2 macrophage-derived EVs was detected. The binding relationship between miR-21-5p and YOD1 was verified. The activation of the YAP/ß-catenin pathway was detected. Consequently, M2 macrophage-derived EVs promoted CD8+ T cell exhaustion in HCC mice. miR-21-5p expression was upregulated in M2 macrophage-derived EVs, and EVs carried miR-21-5p into HCC tissues. miR-21-5p targeted YOD1. Inhibition of miR-21-5p or overexpression of YOD1 annulled the promoting effect of EVs on CD8+ T cell exhaustion. YOD1 inactivated the YAP/ß-catenin pathway. In conclusion, M2 macrophage-derived EVs facilitated CD8+ T cell exhaustion via the miR-21-5p/YOD1/YAP/ß-catenin axis. This study may confer novel insights into the immunotherapy of HCC.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA