Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
1.
Mol Cancer ; 23(1): 143, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38992675

ABSTRACT

BACKGROUND: Emerging evidence indicates the pivotal involvement of circular RNAs (circRNAs) in cancer initiation and progression. Understanding the functions and underlying mechanisms of circRNAs in tumor development holds promise for uncovering novel diagnostic indicators and therapeutic targets. In this study, our focus was to elucidate the function and regulatory mechanism of hsa-circ-0003764 in hepatocellular carcinoma (HCC). METHODS: A newly discovered hsa-circ-0003764 (circPTPN12) was identified from the circbase database. QRT-PCR analysis was utilized to assess the expression levels of hsa-circ-0003764 in both HCC tissues and cells. We conducted in vitro and in vivo experiments to examine the impact of circPTPN12 on the proliferation and apoptosis of HCC cells. Additionally, RNA-sequencing, RNA immunoprecipitation, biotin-coupled probe pull-down assays, and FISH were employed to confirm and establish the relationship between hsa-circ-0003764, PDLIM2, OTUD6B, P65, and ESRP1. RESULTS: In HCC, the downregulation of circPTPN12 was associated with an unfavorable prognosis. CircPTPN12 exhibited suppressive effects on the proliferation of HCC cells both in vitro and in vivo. Mechanistically, RNA sequencing assays unveiled the NF-κB signaling pathway as a targeted pathway of circPTPN12. Functionally, circPTPN12 was found to interact with the PDZ domain of PDLIM2, facilitating the ubiquitination of P65. Furthermore, circPTPN12 bolstered the assembly of the PDLIM2/OTUD6B complex by promoting the deubiquitination of PDLIM2. ESRP1 was identified to bind to pre-PTPN12, thereby fostering the generation of circPTPN12. CONCLUSIONS: Collectively, our findings indicate the involvement of circPTPN12 in modulating PDLIM2 function, influencing HCC progression. The identified ESRP1/circPTPN12/PDLIM2/NF-κB axis shows promise as a novel therapeutic target in the context of HCC.


Subject(s)
Carcinoma, Hepatocellular , Cell Proliferation , Gene Expression Regulation, Neoplastic , LIM Domain Proteins , Liver Neoplasms , NF-kappa B , RNA, Circular , RNA-Binding Proteins , Signal Transduction , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/metabolism , Humans , Liver Neoplasms/genetics , Liver Neoplasms/pathology , Liver Neoplasms/metabolism , RNA, Circular/genetics , LIM Domain Proteins/genetics , LIM Domain Proteins/metabolism , NF-kappa B/metabolism , Mice , Animals , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Cell Line, Tumor , Disease Progression , Apoptosis/genetics , Prognosis , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Male , Female , Mice, Nude
2.
J Hepatol ; 80(1): 82-98, 2024 01.
Article in English | MEDLINE | ID: mdl-37838036

ABSTRACT

BACKGROUND & AIMS: Hepatocellular carcinoma (HCC) is among the most prevalent and lethal cancers worldwide. The tumor microenvironment (TME) contributes to the poor response of patients with HCC to current therapies, while tumor vascular endothelial cells (ECs) are fundamental TME components that significantly contribute to tumor progression. However, the specific functions and mechanisms of tumor vascular ECs in HCC remain unclear. METHODS: We screened and validated diacylglycerol kinase gamma (DGKG) hyper-expression specifically in HCC tumor vascular ECs. Single-cell RNA-sequencing, cytometry by time-of-flight, and in vitro and in vivo studies were performed to investigate the functions of endothelial DGKG. Multiplexed immunohistochemistry staining and flow cytometry were used to evaluate changes in the TME. RESULTS: Functionally, endothelial DGKG promotes tumor angiogenesis and immunosuppressive regulatory T-cell differentiation in HCC. Of significance, we found that HIF-1α activates DGKG transcription by directly binding to its promoter region under hypoxia. Upregulated DGKG promotes HCC progression by recruiting ubiquitin specific peptidase 16 to facilitate ZEB2 deubiquitination, which increases TGF-ß1 secretion, thus inducing tumor angiogenesis and regulatory T-cell differentiation. Importantly, targeting endothelial DGKG potentiated the efficiency of dual blockade of PD-1 and VEGFR-2. CONCLUSION: Hypoxia-induced EC-specific DGKG hyper-expression promotes tumor angiogenesis and immune evasion via the ZEB2/TGF-ß1 axis, suggesting EC-specific DGKG as a potential therapeutic target for HCC. IMPACT AND IMPLICATIONS: Here, we reported that hypoxia-induced endothelial cell-specific DGKG hyper-expression promotes angiogenesis and immune evasion in HCC by recruiting USP16 for K48-linked deubiquitination and inducing the subsequent stabilization of ZEB2, leading to increased TGF-ß1 secretion. Most importantly, endothelial DGKG inhibition greatly improved the efficacy of the dual combination of anti-VEGFR2 and anti-PD-1 treatment in a mouse HCC model, significantly inhibiting the malignant progression of HCC and improving survival. This preclinical study supports the targeting of endothelial DGKG as a potential strategy for precision HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Animals , Humans , Carcinoma, Hepatocellular/pathology , Liver Neoplasms/pathology , Transforming Growth Factor beta1/metabolism , Endothelial Cells/metabolism , Immune Evasion , Angiogenesis , Cell Line, Tumor , Neovascularization, Pathologic/metabolism , Hypoxia/metabolism , Disease Models, Animal , Tumor Microenvironment
3.
Nat Commun ; 13(1): 7677, 2022 12 12.
Article in English | MEDLINE | ID: mdl-36509766

ABSTRACT

Programmed death receptor-1 (PD-1) blockade have achieved some efficacy but only in a fraction of patients with hepatocellular carcinoma (HCC). Programmed cell death 1 ligand 1 (PD-L1) binds to its receptor PD1 on T cells to dampen antigen-tumor immune responses. However, the mechanisms underlying PD-L1 regulation are not fully elucidated. Herein, we identify that tumoral Prdm1 overexpression inhibits cell growth in immune-deficient mouse models. Further, tumoral Prdm1 overexpression upregulates PD-L1 levels, dampening anti-tumor immunity in vivo, and neutralizes the anti-tumor efficacy of Prdm1 overexpression in immune-competent mouse models. Mechanistically, PRDM1 enhances USP22 transcription, thus reducing SPI1 protein degradation through deubiquitination, which enhances PD-L1 transcription. Functionally, PD-1 mAb treatment reinforces the efficacy of Prdm1-overexpressing HCC immune-competent mouse models. Collectively, we demonstrate that the PRDM1-USP22-SPI1 axis regulates PD-L1 levels, resulting in infiltrated CD8+ T cell exhaustion. Furthermore, PRDM1 overexpression combined with PD-(L)1 mAb treatment provides a therapeutic strategy for HCC treatment.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Mice , Animals , B7-H1 Antigen , Programmed Cell Death 1 Receptor , Immune Evasion , CD8-Positive T-Lymphocytes , Positive Regulatory Domain I-Binding Factor 1/genetics , Positive Regulatory Domain I-Binding Factor 1/metabolism
4.
Front Oncol ; 12: 965711, 2022.
Article in English | MEDLINE | ID: mdl-36276134

ABSTRACT

Objective: Immune checkpoint inhibitors (ICIs) have recently been increasingly used in cancer treatment, whereas their clinical application in biliary tract cancer (BTC) patients is uncommon. This study aimed to evaluate the efficacy and safety of ICIs plus capecitabine and oxaliplatin (CAPOX) in the treatment of BTC patients. Methods: This retrospective study reviewed 26 unresectable or advanced BTC patients who received ICIs plus CAPOX. The treatment continued until disease progression, uncontrollable adverse event (AE) occurrence, intolerable toxicity occurrence, or voluntary withdrawal. Results: The median treatment cycles were 5.5 [interquartile range (IQR): 3.8-8.0]. Complete response, partial response, stable disease, and progressive disease rates were 0.0%, 46.2%, 23.1%, and 30.8%, respectively. Objective response rate (ORR) and disease control rate (DCR) were 46.2% and 69.2%, correspondingly. Regarding survival, the median progression-free survival (PFS) and overall survival (OS) were 6.1 (95% CI: 4.4-7.7) months and 16.5 (95% CI: 5.0-28.0) months; moreover, the 1-year PFS and OS rates were 21.5% and 54.3%, respectively. An Eastern Cooperative Oncology Group (ECOG) score of 1-3 (vs. 0) was associated with declined DCR, PFS, and OS (all p < 0.050). The most common AEs of ICIs plus CAPOX were thrombocytopenia (61.5%), neutropenia (26.9%), and reactive cutaneous capillary endothelial proliferation (RCCEP) (23.1%). Moreover, 13 (50.0%) patients suffered from grade 3-4 AEs, including thrombocytopenia (50.0%), neutropenia (7.7%), liver dysfunction (7.7%), and RCCEP (3.8%). Notably, the majority of AEs were controllable. Conclusion: ICIs plus CAPOX chemotherapy exhibit a good efficacy and a manageable safety profile in the treatment of patients with unresectable or advanced BTC.

5.
J Exp Clin Cancer Res ; 41(1): 281, 2022 Sep 22.
Article in English | MEDLINE | ID: mdl-36131287

ABSTRACT

BACKGROUND: Circular RNA (circRNA), a new class of non-coding RNA, has obvious correlations with the occurrence and development of many diseases, including tumors. This study aimed to investigate the potential roles of circPAK1 in hepatocellular carcinoma (HCC). METHODS: High-throughput sequencing was performed on 3 pairs of HCC and matched normal tissues to determine the upregulated circRNAs. The expression level of circPAK1 was detected by qRT-PCR in HCC and paired with normal liver tissue samples. The effects of circPAK1 on proliferation, invasion, metastasis and apoptosis of HCC cells were evaluated by in vitro and in vivo experiments. We also constructed Chitosan/si-circPAK1 (CS/si-circPAK1) nanocomplexes using Chitosan material to evaluate its in vivo therapeutic effect on HCC. High-throughput sequencing, RNA-sequencing, RNA probe pull-down, RNA immunoprecipitation and Co-Immunoprecipitation assays were performed to explore the relationship between circPAK1, 14-3-3ζ, p-LATS1 and YAP. Exosomes isolated from lenvatinib-resistant HCC cell lines were used to evaluate the relationship between exosomal circPAK1 and lenvatinib resistance. RESULTS: CircPAK1, a novel circRNA, is highly expressed in HCC tumor tissues and cell lines as well as correlated with poor outcomes in HCC patients. Functionally, circPAK1 knockdown inhibited HCC cell proliferation, migration, invasion and angiogenesis while circPAK1 overexpression promoted HCC progression. The tumor-promoting phenotypes of circPAK1 on HCC were also confirmed by animal experiments. Importantly, the application of CS/si-circPAK1 nanocomplexes showed a better therapeutic effect on tumor growth and metastasis. Mechanistically, circPAK1 enhanced HCC progression by inactivating the Hippo signaling pathway, and this kind of inactivation is based on its competitively binding of 14-3-3 ζ with YAP, which weakens the recruitment and cytoplasmic fixation of 14-3-3 ζ to YAP, thus promoting YAP nucleus localization. Additionally, circPAK1 could be transported by exosomes from lenvatinib-resistant cells to sensitive cells and induce lenvatinib resistance of receipt cells. CONCLUSION: CircPAK1 exerts its oncogenic function by competitively binding 14-3-3 ζ with YAP, thus promoting YAP nucleus localization, leading to the inactivation of a Hippo signaling pathway. Exosomal circPAK1 may drive resistance to lenvatinib, providing a potential therapeutic target for HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Chitosan , Liver Neoplasms , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Animals , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Cell Proliferation , Chitosan/metabolism , Chitosan/pharmacology , Gene Expression Regulation, Neoplastic , Liver Neoplasms/pathology , Phenylurea Compounds , Protein Serine-Threonine Kinases , Quinolines , RNA, Circular/genetics , RNA, Untranslated , Transcription Factors/genetics , Transcription Factors/metabolism
6.
Front Oncol ; 12: 956225, 2022.
Article in English | MEDLINE | ID: mdl-35898867

ABSTRACT

Polo-like kinase (PLK) 2 is an evolutionarily conserved serine/threonine kinase that shares the n-terminal kinase catalytic domain and the C-terminal Polo Box Domain (PBD) with other members of the PLKs family. In the last two decades, mounting studies have focused on this and tried to clarify its role in many aspects. PLK2 is essential for mitotic centriole replication and meiotic chromatin pairing, synapsis, and crossing-over in the cell cycle; Loss of PLK2 function results in cell cycle disorders and developmental retardation. PLK2 is also involved in regulating cell differentiation and maintaining neural homeostasis. In the process of various stimuli-induced stress, including oxidative and endoplasmic reticulum, PLK2 may promote survival or apoptosis depending on the intensity of stimulation and the degree of cell damage. However, the role of PLK2 in immunity to viral infection has been studied far less than that of other family members. Because PLK2 is extensively and deeply involved in normal physiological functions and pathophysiological mechanisms of cells, its role in diseases is increasingly being paid attention to. The effect of PLK2 in inhibiting hematological tumors and fibrotic diseases, as well as participating in neurodegenerative diseases, has been gradually recognized. However, the research results in solid organ tumors show contradictory results. In addition, preliminary studies using PLK2 as a disease predictor and therapeutic target have yielded some exciting and promising results. More research will help people better understand PLK2 from principle to practice.

7.
PLoS One ; 17(5): e0269249, 2022.
Article in English | MEDLINE | ID: mdl-35639708

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is the cause of the coronavirus disease (COVID-19), which poses a major threat to humans worldwide. With the continuous progress of the pandemic, a growing number of people are infected with SARS-CoV-2, including hepatocellular carcinoma (HCC) patients. However, the relationship between COVID-19 and HCC has not been fully elucidated. In order to provide better treatment for HCC patients infected with SARS-CoV-2, it's urgently needed to identify common targets and find effective drugs for both. In our study, transcriptomic analysis was performed on both selected lung epithelial cell datasets of COVID-19 patients and the datasets of HCC patients to identify the synergistic effect of COVID-19 in HCC patients. What's more, common differentially expressed genes were identified, and a protein-protein interactions network was designed. Then, hub genes and basic modules were detected based on the protein-protein interactions network. Next, functional analysis was performed using gene ontology terminology and the Kyoto Encyclopedia of Genes and Genomes pathway. Finally, protein-protein interactions revealed COVID-19 interaction with key proteins associated with HCC and further identified transcription factor (TF) genes and microRNAs (miRNA) with differentially expressed gene interactions and transcription factor activity. This study reveals that COVID-19 and HCC are closely linked at the molecular level and proposes drugs that may play an important role in HCC patients with COVID-19. More importantly, according to the results of our research, two critical drugs, Ilomastat and Palmatine, may be effective for HCC patients with COVID-19, which provides clinicians with a novel therapeutic idea when facing possible complications in HCC patients with COVID-19.


Subject(s)
COVID-19 Drug Treatment , COVID-19 , Carcinoma, Hepatocellular , Liver Neoplasms , COVID-19/complications , Carcinoma, Hepatocellular/complications , Carcinoma, Hepatocellular/genetics , Humans , Liver Neoplasms/complications , Liver Neoplasms/genetics , SARS-CoV-2 , Transcription Factors
8.
Front Oncol ; 12: 878513, 2022.
Article in English | MEDLINE | ID: mdl-35530358

ABSTRACT

Objectives: Hepatocellular carcinoma (HCC) is the most common primary liver cancer and characterized by high aggressiveness and extremely poor prognosis. Increasing evidence has suggested that circular RNAs (circRNAs), which are highly stable, play crucial roles in the progression of multiple malignancies. However, the roles of circRNAs in HCC remain elusive. Materials and Methods: The expression patterns of circRNAs in HCC were identified by qRT-PCR. A series of functional experiments both in vivo and in vitro were used to determine the role of circERBIN in HCC proliferation. Bioinformatics and an RNA pulldown assay were used to identify potential downstream targets of circERBIN. Results: The expression of circERBIN was upregulated in HCC cell lines and tissues, which was predictive of a poor prognosis in HCC patients. Elevated circERBIN promoted G1/S transition of HCC cells, thus facilitating the proliferation and tumorigenesis of HCC cells. Mechanistic investigations revealed that circERBIN regulated HCC proliferation by acting as a sponge of miR-1263, which subsequently targeted cyclin dependent kinase 6 and controlled G1/S transition. Conclusion: Taken together, these results determined that circERBIN functions as an important epigenetic regulator in HCC development, highlighting that circERBIN is a promising target for treatment of HCC.

9.
Int J Biol Sci ; 18(6): 2345-2361, 2022.
Article in English | MEDLINE | ID: mdl-35414780

ABSTRACT

Matrix Gla protein (MGP) was originally reported as a physiological suppressor of ectopia calcification and has also been reported to be associated with cancer. However, the relation between the biological functions of MGP and the immune response in colorectal cancer (CRC) remains unclear. Here, we investigated the regulatory role of MGP in the immune microenvironment of CRC. MGP expression in CRC samples was assessed by single-cell RNA sequencing and the Gene Expression Omnibus (GEO) database, and confirmed by quantitative real-time Polymerase Chain Reaction (qRT-PCR) and immunohistochemistry analysis of human CRC samples. The effect of MGP on proliferation and invasion of CRC cells was evaluated by in vitro assays involving MGP knockdown and overexpression. Luciferase reporter assay and chromatin immunoprecipitation (ChIP)-qPCR assay were performed to identify transcriptional regulatory sites of the nuclear factor kappa-B (NF-κB) and programmed cell death ligand 1 (PD-L1). In vivo experiments were performed in mouse model of CRC liver metastasis established via spleen injection. The results revealed that MGP was significantly upregulated in cancer cell clusters from the primary CRC or liver metastases, compared with that in the corresponding paracancerous tissues via single-cell RNA sequencing. MGP enriched intracellular free Ca2+ levels and promoted NF-κB phosphorylation, thereby activated PD-L1 expression to promote CD8+ T cell exhaustion in CRC. The luciferase reporter assay and ChIP-qPCR assay indicated that the transcriptional regulation of NF-κB upregulated PD-L1 expression. In vivo, MGP inhibition significantly decreased the rate of CRC liver metastasis, which was further reduced after combined therapy with αPD1 (anti-PD1). In conclusions, this study revealed that MGP can facilitate CD8+ T cell exhaustion by activating the NF-κB pathway, leading to liver metastasis of CRC. The combination of MGP knockdown and αPD1 can synergistically resist liver metastasis of CRC.


Subject(s)
Colorectal Neoplasms , Liver Neoplasms , Animals , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/metabolism , Calcium-Binding Proteins , Cell Line, Tumor , Cell Proliferation/genetics , Colorectal Neoplasms/metabolism , Extracellular Matrix Proteins , Gene Expression Regulation, Neoplastic/genetics , Humans , Liver Neoplasms/pathology , Mice , NF-kappa B/metabolism , Neoplasm Metastasis/pathology , Signal Transduction/genetics , Tumor Microenvironment , Matrix Gla Protein
10.
J Immunother Cancer ; 10(4)2022 04.
Article in English | MEDLINE | ID: mdl-35379737

ABSTRACT

OBJECTIVE: This study aimed to assess the efficacy and safety of camrelizumab plus apatinib in patients with resectable hepatocellular carcinoma (HCC) as neoadjuvant therapy. METHODS: Initially, 20 patients with HCC were screened and 18 patients with resectable HCC were enrolled in this open-label, single-arm, phase II clinical trial. Patients received three cycles of neoadjuvant therapy including three doses of camrelizumab concurrent with apatinib for 21 days followed by surgery. Four to 8 weeks after surgery, patients received eight cycles of adjuvant therapy with camrelizumab in combination with apatinib. Major pathological reactions (MPR), complete pathological reactions (pCR), objective response rate (ORR), relapse-free survival (RFS), and adverse events (AE) were assessed. In addition, cancer tissue and plasma samples were collected before and after treatment, and genetic differences between responding and non-responding lesions were compared by tumor immune microenvironment (TIME) analysis, circulating tumor DNA (ctDNA) analysis and proteomics analysis. RESULTS: In 18 patients with HCC who completed neoadjuvant therapy, 3 (16.7%) and 6 (33.3%) patients with HCC reached ORR based on Response Evaluation Criteria in Solid Tumors (RECIST) V.1.1 and modified RECIST criteria, respectively. Of the 17 patients with HCC who received surgical resection, 3 (17.6%) patients with HCC reported MPR and 1 (5.9%) patient with HCC achieved pCR. The 1-year RFS rate of the enrolled patients was 53.85% (95% CI: 24.77% to 75.99%). Grade 3/4 AEs were reported in 3 (16.7%) of the 18 patients, with the most common AEs being rash (11.1%), hypertension (5.6%), drug-induced liver damage (5.6%), and neutropenia (5.6%) in the preoperative phase. The 289 NanoString panel RNA sequencing showed that TIME cell infiltration especially dendritic cells (DCs) infiltration was better in responding tumors than in non-responding tumors. Our results of ctDNA revealed a higher positive rate (100%) among patients with HCC with stage IIb-IIIa disease. When comparing patients with pCR/MPR and non-MPR, we observed more mutations in patients who achieved pCR/MPR at baseline (6 mutations vs 2.5 mutations, p=0.025). Patients who were ctDNA positive after adjuvant therapy presented a trend of shorter RFS than those who were ctDNA negative. Proteomic analysis suggested that abnormal glucose metabolism in patients with multifocal HCC might be related to different sensitivity of treatment in different lesions. CONCLUSION: Perioperative camrelizumab plus apatinib displays a promising efficacy and manageable toxicity in patients with resectable HCC. DCs infiltration might be a predictive marker of response to camrelizumab and apatinib as well as patients' recurrence. ctDNA as a compose biomarker can predict pathological response and relapse. Abnormal glucose metabolism in patients with multifocal HCC may be related to different sensitivity of treatment in different lesions. TRIAL REGISTRATION NUMBER: NCT04297202.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Antibodies, Monoclonal, Humanized , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/pathology , Humans , Liver Neoplasms/pathology , Neoplasm Recurrence, Local , Perioperative Period , Proteomics , Pyridines , Tumor Microenvironment
11.
Sensors (Basel) ; 22(2)2022 Jan 11.
Article in English | MEDLINE | ID: mdl-35062497

ABSTRACT

Group-based sparse representation (GSR) uses image nonlocal self-similarity (NSS) prior to grouping similar image patches, and then performs sparse representation. However, the traditional GSR model restores the image by training degraded images, which leads to the inevitable over-fitting of the data in the training model, resulting in poor image restoration results. In this paper, we propose a new hybrid sparse representation model (HSR) for image restoration. The proposed HSR model is improved in two aspects. On the one hand, the proposed HSR model exploits the NSS priors of both degraded images and external image datasets, making the model complementary in feature space and the plane. On the other hand, we introduce a joint sparse representation model to make better use of local sparsity and NSS characteristics of the images. This joint model integrates the patch-based sparse representation (PSR) model and GSR model, while retaining the advantages of the GSR model and the PSR model, so that the sparse representation model is unified. Extensive experimental results show that the proposed hybrid model outperforms several existing image recovery algorithms in both objective and subjective evaluations.

12.
Front Cell Dev Biol ; 9: 796686, 2021.
Article in English | MEDLINE | ID: mdl-34869393

ABSTRACT

Mounting evidence has demonstrated that circular RNAs have an important function in tumorigenesis and cancer evolvement. CircCRIM1 has been shown to be a poor prognostic element in multiple human malignancies. However, the clinical significance and mechanism of circCRIM1 in hepatocellular carcinoma (HCC) is still unclear. The present study confirmed the expression level of circCRIM1 using quantitative real-time PCR. In addition, circCRIM1 siRNA and overexpression vectors were used for transfection into LM3 or Huh7 cells to down- or up-regulate the expression of circCRIM1. In vitro and in vivo experiments were performed to explore the function of circCRIM1 in HCC. RNA pull-down, RNA immunoprecipitation, fluorescent in situ hybridization, and luciferase reporter assays were conducted to confirm the relationship between miR-378a-3p and circCRIM1 or S-phase kinase-associated protein 2 (SKP2) in HCC. Then, circCRIM1 was up-regulated in HCC and its expression level was significantly associated with poor prognosis and clinicopathologic characteristics. CircCRIM1 enhanced the proliferation and angiogenesis of HCC cells in vitro and promoted xenograft growth in vivo. Moreover, circCRIM1 upregulated the expression of SKP2 by functioning as a sponge for miR-378a-3p. These findings suggest that circCRIM1 boosts the HCC progression via the miR-378-3p/SKP2 axis and may act as a crucial epigenetic therapeutic molecule target in HCC.

13.
Cell Death Discov ; 7(1): 270, 2021 Oct 01.
Article in English | MEDLINE | ID: mdl-34599150

ABSTRACT

Circular RNAs (circRNAs), continuous loops of single-stranded RNA, regulate gene expression during the development of various cancers. However, the function of circRNAs in hepatocellular carcinoma (HCC) is rarely discussed. Quantitative real-time polymerase chain reaction (qRT-PCR) was used to determine the mRNA levels of circ_0011385, miR-361-3p, and STC2 in 96 pairs of HCC tissues (tumor tissues and adjacent normal tissues), HCC cell lines, and L02 (human normal liver cell line) cells. The relationships between circ_0011385 expression and clinical features of HCC were evaluated. Functional experiments in vitro or in vivo were used to evaluate the biological function of circ_0011385. Bioinformatics analysis was performed to predict miRNAs and mRNAs sponged by circ_0011385. RNA immunoprecipitation (RIP) and dual-luciferase reporter gene assays were used to elucidate the interactions among circ_0011385, miR-361-3p, and STC2 (stanniocalcin 2). ChIP and dual-luciferase reporter gene assays were used to identify the upstream regulator of circ_0011385. High expression of circ_0011385 was observed in HCC tissues and cell lines and was significantly associated with tumor size, TNM stage, and prognosis. In addition, inhibition of circ_0011385 expression prevented the proliferation of HCC cells in vitro and in vivo. Circ_0011385 sponged miR-361-3p, thereby regulating the mRNA expression of STC2. In addition, the transcription of circ_0011385 was regulated by SP3. Circ_0011385 knockdown suppressed cell proliferation and tumor activity in HCC. Circ_0011385 may therefore serve as a new biomarker in the diagnosis and treatment of HCC.

14.
Mol Ther Nucleic Acids ; 25: 67-82, 2021 Sep 03.
Article in English | MEDLINE | ID: mdl-34188972

ABSTRACT

As next-generation sequencing (NGS) is leaping forward, more than 160 covalent RNA modification processes have been reported, and they are widely present in every organism and overall RNA type. Many modification processes of RNA introduce a new layer to the gene regulation process, resulting in novel RNA epigenetics. The commonest RNA modification includes pseudouridine (Ψ), N 7-methylguanosine (m7G), 5-hydroxymethylcytosine (hm5C), 5-methylcytosine (m5C), N 1-methyladenosine (m1A), N 6-methyladenosine (m6A), and others. In this study, we focus on non-coding RNAs (ncRNAs) to summarize the epigenetic consequences of RNA modifications, and the pathogenesis of cancer, as diagnostic markers and therapeutic targets for cancer, as well as the mechanisms affecting the immune environment of cancer. In addition, we summarize the current status of epigenetic drugs for tumor therapy based on ncRNA modifications and the progress of bioinformatics methods in elucidating RNA modifications in recent years.

15.
Ann Transl Med ; 8(19): 1250, 2020 Oct.
Article in English | MEDLINE | ID: mdl-33178782

ABSTRACT

Obstructive jaundice is characterized by an obstruction of the intrahepatic or extrahepatic biliary system, and the most common causes include pancreatic and duodenal periampullary cancer. There have been some cases reporting obstructive jaundice caused by infection. Deep tissue infection usually develops in the individuals who are immunologically compromised or chronically ill, while a few cases reported in the immunocompetent patients. Those cases were diagnosed by fungal culture or percutaneous biopsy. Here, we presented an interesting case of obstructive jaundice secondary to fungal infection confirmed by postoperative pathological examination. A 79 years old man complaint about upper abdominal discomfort, darkened urine, and skin itch, with a history of esophageal cancer operation 5 years ago. The serology for hepatitis virus and human immunodeficiency virus (HIV) was negative. Imaging examinations showed a nodular located at distal common bile duct. As evidenced by increased level of cancer antigen 19-9 (CA19-9), the patient was highly suspected to be malignant obstructive jaundice. Thus, pylorus preserving pancreaticoduodenectomy (PPPD) was conducted. To our surprise, the ultimate diagnosis was fungal infection at the site of duodenum ampulla by the postoperative pathological examination, with no evidence of malignance. Anti-infective therapy was conducted subsequently, combined by fluconazole, sulperazone and tinidazole. Three weeks later, the patient was generally in good condition and discharged from hospital. During the 2-year follow-up, no fungal infection or tumor recurrence was observed. This case reminded us that fungal infection could be the cause of obstructive jaundice in an elderly person.

16.
Ann Transl Med ; 7(22): 628, 2019 Nov.
Article in English | MEDLINE | ID: mdl-31930029

ABSTRACT

BACKGROUND: This study aimed to investigate the mechanism of galectin (Gal)-1 of regulating Treg/Th17 in pathogenesis of acute rejection after liver transplantation in rat. METHODS: Mononuclear cells were induced to immature dendritic cells (imDCs), which were transfected with or without NF-κB/RelB. Western Blot was performed to detect the expression of NF-κB/RelB. the expression of CD11c, CD45RB, CD80 and MHC II were detected by flow cytometry. Enzyme-linked immunosorbent assay (ELISA) was employed to detect cytokines IL-27 and TGF-ß. Lewis and dark agouti (DA) rats were generally anaesthetized by isoflurane inhalation to establish liver transplant models. RESULTS: We demonstrate that Gal-1 disturbs maturation of imDCs by downregulating NF-κB/RelB expression, and Gal-1 negatively controls CD4+ proliferation though IL-27 pathway. CONCLUSIONS: In aggregate, Gal-1 promotes Treg differentiation in CD4+ T cells though NF-κB/RelB-IL-27 pathway. These findings suggest a new therapeutic target to mediate Treg population.

17.
Cell Death Dis ; 9(3): 290, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29459719

ABSTRACT

Thymic-derived regulatory T cell (tTreg) clinical trials show therapeutic promise in the prevention of acute graft-versus-host disease (GVHD) in allogeneic hematopoietic stem cell transplantation patients. However, strategies are needed to improve tTreg proliferative ability and survival as a means to improve tTreg therapy and reduce the requirement for producing large numbers of Treg cells for adoptive tTreg transfer. Autophagy is a self-degradative process for cytosolic components, which is involved in cells death, differentiation, lymphocyte homeostasis, and tTreg function. Studies have shown that mice with tTreg cells that have a disrupted autophagy process have defective tTreg cell generation and function, resulting in autoimmune disease and failed GVHD prevention by adoptively transferred tTreg cells. We found the attenuated autophagy status during ex vivo expansion, which leads us to determine whether tTreg cell survival could be augmented by miR-142-3p, the miRNA which is highly expressed in tTreg cells and potentially targets autophagy-related protein (ATG)-1, ATG16L1. We demonstrate that miR-142-3p downregulates ATG16L1 mRNA and production of ATG16L1, that has been linked to autoimmune diseases. Conversely, miR-142-3p knock-down improved tTreg cell expansion, survival and function in vitro and vivo. In aggregate, these studies provide a new approach that uses miR-142-3p knockdown to increase tTreg cell efficacy by increasing ATG16L1 mRNA and protein and the autophagy process.


Subject(s)
Autophagy , Carrier Proteins/immunology , MicroRNAs/immunology , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology , Animals , Autophagy-Related Proteins , Carrier Proteins/genetics , Cell Differentiation , Cell Proliferation , Cells, Cultured , Humans , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , MicroRNAs/genetics , MicroRNAs/metabolism , T-Lymphocytes, Regulatory/immunology , Thymus Gland/immunology
18.
Hepatobiliary Pancreat Dis Int ; 16(4): 364-369, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28823365

ABSTRACT

BACKGROUND: Fast-track surgery and enhanced recovery after surgery have been applied to many surgical procedures; however, data on fast-track surgery and enhanced recovery after surgery following liver transplantation is limited. This study aimed to conduct a prospective study to determine the effects of fast-track surgery on prognosis after liver transplantation. METHODS: This was a prospective, single-blinded, randomized study. One hundred twenty-eight patients undergoing liver transplantation were selected for the fast-track (FT group, n=54) or conventional process (NFT group, n=74). The primary endpoints were intensive care unit (ICU) stay and hospital stay. The secondary endpoints were as follows: operative time, anhepatic phase time, intraoperative blood loss, intraoperative blood transfusion volume, postoperative complications, readmission rate, and postoperative mortality. RESULTS: There was no significant difference in preoperative demographics between the two groups. The median ICU stay was 2 days (range 1-7 days) in the FT group and 5 days (range 3-12 days) in the NFT group (P<0.01). Furthermore, the hospital stay was also significantly reduced in the FT group (P<0.01). The operative time, anhepatic phase time, intraoperative blood loss, and intraoperative blood transfusion volume were decreased in the FT group compared with the NFT group (P<0.05). Based on Spearman correlation analysis, the ICU stay and hospital stay may be positively correlated with operative time, anhepatic phase time and intraoperative blood loss. There were no differences in the incidence of postoperative complications, readmissions, and postoperative mortality between the two groups. CONCLUSION: Fast-track procedures effectively reduce the ICU stay and hospital stay without adversely affecting prognosis. This study demonstrated that fast-track protocols are safe and feasible in liver transplantation.


Subject(s)
End Stage Liver Disease/surgery , Length of Stay , Liver Transplantation/methods , Adult , Aged , Blood Loss, Surgical , China , End Stage Liver Disease/diagnosis , End Stage Liver Disease/etiology , End Stage Liver Disease/mortality , Female , Humans , Intensive Care Units , Liver Transplantation/adverse effects , Liver Transplantation/mortality , Male , Middle Aged , Operative Time , Patient Readmission , Postoperative Complications/etiology , Prospective Studies , Recovery of Function , Single-Blind Method , Time Factors , Treatment Outcome
19.
Cell Physiol Biochem ; 41(3): 1189-1198, 2017.
Article in English | MEDLINE | ID: mdl-28472799

ABSTRACT

BACKGROUND/AIMS: Immunosuppression is one of the hallmarks of cancer; however, its molecular mechanism remains unknown. In the present study, we sought to investigate the expression and activation of yes-associated protein 1 (YAP-1) and its roles in T cells within hepatocellular carcinoma (HCC). METHODS: The expression and activation of YAP-1 were accessed by real-time PCR, immunohistochemistry staining, western blot, and flow cytometry. The potential regulation effect of YAP-1 on Regulatory T cells (Tregs) differentiation was predicted using bioinformatics tools and verified by in vitro studies. RESULTS: Significant overexpression and activation of YAP-1 was detected within peripheral blood mononuclear cells and showed positive linear correlation to Treg percentage; it may serve as a valuable indicator of a bad prognosis. Using in vitro studies, we found that overexpression and activation of YAP-1 can promote naïve T cell polarization stimulation to Tregs by increasing the expression of TGFBR2. The YAP-1/TEADs DNA binding site was spotted within the promoter region of TGFBR2 and related to its transcription activity. YAP-1 acted as a co-activator of TGFBR2 transcription by binding directly to the TGFBR2 promoter through TEADs. CONCLUSION: Overexpression and activation of YAP-1 in HCC T cells can induce immunosuppression by promoting Treg differentiation via transcriptional enhancement of TGFBR2.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Carcinoma, Hepatocellular/genetics , Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Receptors, Transforming Growth Factor beta/genetics , T-Lymphocytes, Regulatory/immunology , Adaptor Proteins, Signal Transducing/immunology , Adult , Binding Sites , Carcinoma, Hepatocellular/immunology , Carcinoma, Hepatocellular/pathology , Case-Control Studies , Cell Differentiation , Cell Proliferation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/immunology , Female , Humans , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/pathology , Liver Neoplasms/immunology , Liver Neoplasms/pathology , Male , Middle Aged , Nuclear Proteins/genetics , Nuclear Proteins/immunology , Phosphoproteins/immunology , Primary Cell Culture , Promoter Regions, Genetic , Protein Binding , Protein Serine-Threonine Kinases/immunology , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/immunology , Signal Transduction , T-Lymphocytes, Regulatory/pathology , TEA Domain Transcription Factors , Transcription Factors/genetics , Transcription Factors/immunology , Transcription, Genetic , YAP-Signaling Proteins
20.
Tumour Biol ; 37(11): 14629-14635, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27619679

ABSTRACT

STAT3 is an oncogene which mainly capable of regulating various biological characters by its transcriptional factor features driving thousands of genes. However, its upstream noncoding RNA-related regulations still remain unknown. In this study, we focused on the microRNA (miRNA)-associated single nucleotide polymorphisms (SNPs) in the 3' unstranslated region (UTR) of STAT3 to investigate the further relationship of the SNPs with miRNAs among Chinese patients with hepatocellular carcinoma (HCC). We found that patients suffering from HBV infection indicated to be the susceptible population by comparing with controls. Besides, SNP rs111904020 (U/G) in STAT3, 3'UTR was involved in the occurrence of HCC by acting as tumor promoter factors. SNP rs111904020 (U/G) could be regulated by miR-214 which caused an upregulation of STAT3. Furthermore, the carriers of G genotype was related to high expression of STAT3, and larger tumor size as well as high probability of metastasis. Moreover, the SNP of U/G was associated with poor survival and high reoccurrence of HCC with a 5-year follow-up study. In conclusion, our findings have shown that the SNP rs111904020 (U/G) in STAT3 3'UTR acted as promotion factors in the HCC development through disrupting the regulatory role of miR-214 in STAT3 expression.


Subject(s)
3' Untranslated Regions/genetics , Carcinoma, Hepatocellular/secondary , Liver Neoplasms/pathology , MicroRNAs/genetics , Polymorphism, Single Nucleotide/genetics , STAT3 Transcription Factor/genetics , Biomarkers, Tumor , Carcinoma, Hepatocellular/epidemiology , Carcinoma, Hepatocellular/genetics , Case-Control Studies , China/epidemiology , Female , Follow-Up Studies , Humans , Liver Neoplasms/epidemiology , Liver Neoplasms/genetics , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Prevalence , Prognosis , Real-Time Polymerase Chain Reaction , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...