Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 61
Filtrer
1.
Heliyon ; 10(14): e34203, 2024 Jul 30.
Article de Anglais | MEDLINE | ID: mdl-39104492

RÉSUMÉ

Objective: The present study aimed to explore the function of human bone marrow mesenchymal stem cells (hBMMSCs)-derived exosomal long noncoding RNA histocompatibility leukocyte antigen complex P5 (HCP5) in the osteogenic differentiation of human periodontal ligament stem cells (hPDLSCs) to improve chronic periodontitis (CP). Methods: Exosomes were extracted from hBMMSCs. Alizarin red S staining was used to detect mineralised nodules. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) was used to measure HCP5 and miR-24-3p expression. The mRNA and protein levels of alkaline phosphatase (ALP), osteocalcin, osterix, runt-related transcription factor 2, bone morphogenetic protein 2, osteopontin, fibronectin, collagen 1, heme oxygenase 1 (HO1), P38, and ETS transcription factor ELK1 (ELK1) were detected using RT-qPCR and Western blot. Enzyme-linked immunosorbent assay (ELISA) kits were used to determine the HO1 and carbon monoxide concentrations. Heme, biliverdin, and Fe2+ levels were determined using detection kits. Micro-computed tomography, hematoxylin and eosin staining, ALP staining, tartrate-resistant acid phosphatase staining, ELISA, and RT-qPCR were conducted to evaluate the effect of HCP5 on CP mice. Dual luciferase, RNA immunoprecipitation, and RNA pulldown experiments were performed to identify the interactions among HCP5, miR-24-3p, and HO1. Results: The osteogenic ability of hPDLSCs significantly increased when co-cultured with hBMMSCs or hBMMSCs exosomes. Overexpression of HCP5 and HO1 in hBMMSCs exosomes promoted the osteogenic differentiation of hPDLSCs, and knockdown of HCP5 repressed the osteogenic differentiation of hPDLSCs. HCP5 knockdown enhanced the inflammatory response and repressed osteogenesis in CP mice. MiR-24-3p overexpression diminished the stimulatory effect of HCP5 on the osteogenic ability of hPDLSCs. Mechanistically, HCP5 acted as a sponge for miR-24-3p and regulated HO1 expression, and HO1 activated the P38/ELK1 pathway. Conclusion: HBMMSCs-derived exosomal HCP5 promotes the osteogenic differentiation of hPDLSCs and alleviates CP by regulating the miR-24-3p/HO1/P38/ELK1 signalling pathway.

2.
Pathol Res Pract ; 257: 155286, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38599044

RÉSUMÉ

In spite of the decrease in breast cancer (BC) death rates, it has remained a significant public health concern. Dysregulation of the Hippo pathway contributes to breast cancer development and progression by enhancing cancerous cell proliferation, survival, invasion, and migration. Investigating the connection between specific lncRNAs (SNHG15, HCP5, and LINC01433) and YAP and WWTR1, and the impact of these lncRNAs on the expression of YAP and WWTR1 proteins in the Hippo pathway, may offer valuable understanding for BC diagnosis and treatment. Forty BC tissue samples were acquired from the Tumor Bank and utilized for RNA and protein extraction. Real-time PCR and western blotting techniques were performed to assess the gene and protein expressions, respectively. Correlations between variables and their associations with clinicopathological features in BC were evaluated using Mann-Whitney U or Student's t-test. Additionally, the analysis of the GEO database was utilized to validate the findings. In cancerous tissue, the up-regulation of YAP, WWTR1, HCP5, SNHG15, and Linc01433 at both the mRNA and protein levels corresponds to the findings in GEO datasets. A significant association was found between YAP and histological grade, while WWTR1 showed a correlation with family history and HER-2. The distinct and notable expression of YAP, WWTR1, SNHG15, HCP5, and Linc01433 in BC tissues, together with the results of combined ROC curve analysis derived from our finding and GEO database suggest that a combined panel of these 5 RNAs may have great potential in predicting of BC and its management.


Sujet(s)
Protéines adaptatrices de la transduction du signal , Tumeurs du sein , Régulation de l'expression des gènes tumoraux , ARN long non codant , Facteurs de transcription , Transcriptional coactivator with PDZ-binding motif proteins , Protéines de signalisation YAP , Femelle , Humains , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Tumeurs du sein/génétique , Tumeurs du sein/anatomopathologie , Tumeurs du sein/métabolisme , Régulation de l'expression des gènes tumoraux/génétique , Protéines et peptides de signalisation intracellulaire/génétique , Protéines et peptides de signalisation intracellulaire/métabolisme , ARN long non codant/génétique , Transactivateurs/génétique , Facteurs de transcription/génétique , Protéines de signalisation YAP/génétique , Protéines de signalisation YAP/métabolisme
3.
Cancer Cell Int ; 24(1): 142, 2024 Apr 20.
Article de Anglais | MEDLINE | ID: mdl-38643145

RÉSUMÉ

BACKGROUND: Hepatocellular carcinoma (HCC) is widely recognized for its unfavorable prognosis. Increasing evidence has revealed that LGALS3 has an essential function in initiating and developing several malignancies in humans. Nevertheless, thorough analysis of the expression profile, clinical prognosis, pathway prediction, and immune infiltration of LGALS3 has not been fully explored in HCC. METHODS: In this study, an initial pan-cancer analysis was conducted to investigate the expression and prognosis of LGALS3. Following a comprehensive analysis, which included expression analysis and correlation analysis, noncoding RNAs that contribute to the overexpression of LGALS3 were subsequently identified. This identification was further validated using HCC clinical tissue samples. TIMER2 and GEPIA2 were employed to examine the correlation between LGALS3 and HCP5 with immunological checkpoints, cell chemotaxis, and immune infiltration in HCC. The R program was applied to analyze the expression distribution of immune score in in HCC patients with high and low LGALS3 expression. The expression profiles of immune checkpoints were also analyzed. Use R to perform GSVA analysis in order to explore potential signaling pathways. RESULTS: First, we conducted pan-cancer analysis for LGALS3 expression level through an in-depth analysis of public databases and found that HCC has a high LGALS3 gene and protein expression level, which were then verified in clinical HCC specimens. Meanwhile, high LGALS3 gene expression is related to malignant progression and poor prognosis of HCC. Univariate and multivariate analyses confirmed that LGALS3 could serve as an independent prognostic marker for HCC. Next, by combining comprehensive analysis and validation on HCC clinical tissue samples, we hypothesize that the HCP5/hsa-miR-27b-3p axis could serve as the most promising LGALS3 regulation mechanism in HCC. KEGG and GO analyses highlighted that the LGALS3-related genes were involved in tumor immunity. Furthermore, we detected a significant positive association between LGALS3 and HCP5 with immunological checkpoints, cell chemotaxis, and immune infiltration. In addition, high LGALS3 expression groups had significantly higher immune cell scores and immune checkpoint expression levels. Finally, GSVA analysis was performed to predict potential signaling pathways linked to LGALS3 and HCP5 in immune evasion and metabolic reprogramming of HCC. CONCLUSIONS: Our findings indicated that the upregulation of LGALS3 via the HCP5/hsa-miR-27b-3p axis is associated with unfavorable prognosis and increased tumor immune infiltration in HCC.

4.
Funct Integr Genomics ; 24(2): 61, 2024 Mar 20.
Article de Anglais | MEDLINE | ID: mdl-38507114

RÉSUMÉ

This research provides a glimmer of hope that the knockout of HCP5 leads to a therapy response to considerably prolong the life of patients with OC. RT-PCR evaluated the expression of lncRNA HCP5 in the ovarian cancer OVCAR-3 cell line. CRISPR knockout cell lines validated by western blot. Small genomic deletions at the targeted locus were induced. CCK-8 colony formation assays were used to analyze the effect of HCP5 knockout on the proliferation capacity of OVCAR-3 cells. Transwell migration and invasion assayed. Furthermore, the Sphere-formation assay isolated the most aggressive population of cancer stem cells. Bioinformatic analysis showed a significant correlation between lncRNA HCP5 up-regulation and OVCAR-3 cell proliferation. The ChIP technique assesses specific sites of interaction between transcription factors and DNA. Real-time PCR assays explored the relationship between HCP5, Hsa-miR-9-5p, CXCR4, CDH1, caspase-3, p53, bcl2 and survivin. PCR carried out amplification of the 448-bp band for sgRNA1 and sgRNA2 after the use of particular primers for HCP5. the number of breast cancer cells that moved to the bottom chamber reduced considerably after transfection with PX461-sgRNA1/2 vectors compared to the Blank control groups (P < 0.05). MTT assay designated growth curves that showed the rate of OVCAR-3 growth was significantly repressed (***P < 0.001) when compared with control OVCAR-3 cells after HCP5 knockdown. Also, the survival results of W.T cells in 24, 48 and 72 h showed 92%, 87% and 85%, respectively. This is while the cells of the CRISPR/Cas9 group in which LncRNA HCP5 was knocked out had 42% (*P < 0.05), 23%(**P < 0.01) and 14% (**P < 0.01) survival, respectively. The expression levels of caspase-3, Hsa-miR-9-5p, P53 genes in the HCP5 deletion of CRISPR/Cas9 group significantly increased than the W.T. control group; the deletion group showed a considerable reduction in HCP5 expression compared to the blank control group (3.6-fold, p < 0.01). Whereas BCL2, SURVIVIN, CXCR4, CDH1 genes expression markedly increased than in HCP5 knockout cells (5.8-fold, p < 0.05). These results indicate that CRISPR/Cas9-mediated HCP5 disruption on OVCAR-3 cell lines promotes anti-tumor biomarkers, suppressing ovarian cancer progression. Consistent with these results, HCP5 is one of the most critical lnc for the efficient proliferation and migration of OVCAR-3 cell lines.


Sujet(s)
microARN , Tumeurs de l'ovaire , ARN long non codant , Humains , Femelle , Tumeurs de l'ovaire/génétique , Tumeurs de l'ovaire/anatomopathologie , ARN long non codant/génétique , ARN long non codant/métabolisme , Survivine/génétique , Survivine/métabolisme , Caspase-3/génétique , Caspase-3/métabolisme , Apoptose/génétique , Lignée cellulaire tumorale , Régulation positive , microARN/génétique , Prolifération cellulaire/génétique , Protéines proto-oncogènes c-bcl-2/génétique , Protéines proto-oncogènes c-bcl-2/métabolisme , Régulation de l'expression des gènes tumoraux
5.
Diabetes Metab Syndr Obes ; 17: 157-163, 2024.
Article de Anglais | MEDLINE | ID: mdl-38222030

RÉSUMÉ

Purpose: LncRNA HCP5 has been reported to participate in high glucose-induced pathological processes, whereas its role in gestational diabetes mellitus (GDM) is unclear. This study aimed to explore the role of HCP5 in GDM. Methods: This study enrolled a total of 220 pregnant women (gestational age = 1 month). A follow-up study was performed until delivery. The occurrence of GDM was checked every month during follow-up. Plasma samples were collected from all participants and expression of HCP5 was determined with RT-qPCR. The 220 patients were divided into high and low GDM groups, and GDM-free curves were plotted for both groups and compared. The ROC curve was plotted to explore the predictive value of plasma HCP5 on the day of admission for GDM. INS-1 cells were transfected with HCP5 expression vector or siRNA, and cell viability under high glucose was determined by the MTT assay. An ELISA was applied to determine insulin levels in the cell culture medium. Results: During follow-up, the level of HCP5 was increased during pregnancy and the high HCP5 level group showed a significantly higher incidence of GDM. Plasma levels of HCP5 on the day of admission effectively separated GDM patients from healthy controls. HCP5 negatively regulated cell viability and insulin secretion under high glucose treatment. Conclusion: HCP5 may act as a predictor for GDM, and it negatively regulated INS-1 cell viability and insulin secretion under high glucose conditions.

6.
Biochem Genet ; 2023 Dec 10.
Article de Anglais | MEDLINE | ID: mdl-38071681

RÉSUMÉ

Ovarian cancer (OC) is a major gynecological malignancy with an annually increasing morbidity that poses a significant threat to the health of women worldwide. Most OC patients are diagnosed at an advanced stage. It is an urgent task to search for biomarkers for the diagnosis and treatment of OC. The lncRNA HCP5 (HCP5) was recently identified as an oncogene in several malignant tumors. However, the function of HCP5 in OC has rarely been reported. Herein, the levels of HCP5 and PTBP1 were found to be markedly increased in malignant OC tumor tissues and OC cell lines. In HCP5-silenced SKOV-3 and HEY cells, cell viability was markedly decreased, and the apoptosis rate was significantly increased, with more cells exhibiting G0/G1 arrest and increased expression of cleaved caspase-3 and cleaved caspase-9. Furthermore, the number of migrated cells, number of invaded cells, and migration distance were notably decreased by the knockdown of HCP5 in SKOV-3 cells and HEY cells. In the xenograft model established with SKOV-3 cells, the number of lung metastases, tumor growth, and Ki67 expression in tumor tissues were markedly decreased by the knockdown of HCP5, accompanied by an increased percentage of TUNEL-positive cells. HCP5 was found to be localized in the nucleus, and the interaction between HCP5 and PTBP1 was verified by RNA pull-down and RNA immunoprecipitation assays. Furthermore, in HCP5-overexpressing OC cells, the impacts of HCP5 on cell proliferation and apoptosis were significantly attenuated by the knockdown of PTBP1. Collectively, these results indicate that HCP5 facilitates the progression of OC by interacting with the PTBP1 protein.

7.
Cancers (Basel) ; 15(19)2023 Sep 22.
Article de Anglais | MEDLINE | ID: mdl-37835376

RÉSUMÉ

As immune checkpoint inhibitors (ICI) emerge as a paradigm-shifting treatment option for patients with advanced or metastatic cancer, there is a growing demand for biomarkers that can distinguish which patients are likely to benefit. In the case of triple-negative breast cancer (TNBC), characterized by a lack of therapeutic targets, pembrolizumab approval for high-risk early-stage disease occurred regardless of PD-L1 status, which keeps the condition in a biomarker limbus. In this review, we highlight the participation of long non-coding RNAs (lncRNAs) in the regulation of the PD-1/PD-L1 pathway, as well as in the definition of prognostic immune-related signatures in many types of tumors, aiming to shed light on molecules that deserve further investigation for a potential role as biomarkers. We also conducted a bioinformatic analysis to investigate lncRNAs already investigated in PD-1/PDL-1 pathways in other cancer types, considering the TNBC molecular context. In this sense, from the generated data, we evidence here two lncRNAs, UCA1 and HCP5, which have not yet been identified in the context of the tumoral immune response in breast cancer. These candidates can be further explored to verify their use as biomarkers for ICI response. In this article, we present an updated review regarding the use of lncRNA as biomarkers of response to ICI, highlighting the versatility of using these molecules.

8.
Cancers (Basel) ; 15(6)2023 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-36980766

RÉSUMÉ

BACKGROUND: Long non-coding RNAs (lncRNAs) are a class of RNA molecules that are longer than 200 nucleotides and were initially believed to lack encoding capability. However, recent research has found open reading frames (ORFs) within lncRNAs, suggesting that they may have coding capacity. Despite this discovery, the mechanisms by which lncRNA-encoded products are involved in cancer are not well understood. The current study aims to investigate whether lncRNA HCP5-encoded products promote triple-negative breast cancer (TNBC) by regulating ferroptosis. METHODS: We used bioinformatics to predict the coding capacity of lncRNA HCP5 and conducted molecular biology experiments and a xenograft assay in nude mice to investigate the mechanism of its encoded products. We also evaluated the expression of the HCP5-encoded products in a breast cancer tissue microarray. RESULTS: Our analysis revealed that the ORF in lncRNA HCP5 can encode a protein with 132-amino acid (aa), which we named HCP5-132aa. Further experiments showed that HCP5-132aa promotes TNBC growth by regulating GPX4 expression and lipid ROS level through the ferroptosis pathway. Additionally, we found that the breast cancer patients with high levels of HCP5-132aa have poorer prognosis. CONCLUSIONS: Our study suggests that overexpression of lncRNA HCP5-encoded protein is a critical oncogenic event in TNBC, as it regulates ferroptosis. These findings could provide new therapeutic targets for the treatment of TNBC.

9.
Mol Ther ; 31(2): 552-568, 2023 02 01.
Article de Anglais | MEDLINE | ID: mdl-36245126

RÉSUMÉ

Inducing cancer cell apoptosis through cytotoxic reagents is the main therapeutic strategy for diverse cancer types. However, several antiapoptotic factors impede curative cancer therapy by driving cancer cells to resist cytotoxic agent-induced apoptosis, thus leading to refractoriness and relapse. To define critical antiapoptotic factors that contribute to chemoresistance in esophageal squamous cell carcinoma (ESCC), we generated two pairs of parental and apoptosis-resistant cell models through cisplatin (DDP) induction and then performed whole-transcriptome sequencing. We identified the long noncoding RNA (lncRNA) histocompatibility leukocyte antigen complex P5 (HCP5) as the chief culprit for chemoresistance. Mechanistically, HCP5 interacts with UTP3 small subunit processome component (UTP3) and prevents UTP3 degradation from E3 ligase tripartite motif containing 29 (TRIM29)-mediated ubiquitination. UTP3 then recruits c-Myc to activate vesicle-associated membrane protein 3 (VAMP3) expression. Activated VAMP3 suppresses caspase-dependent apoptosis and eventually leads to chemoresistance. Accordingly, the expression level of the HCP5/UTP3/c-Myc/VAMP3 axis in chemoresistant patients is significantly higher than that in chemosensitive patients. Thus, our study demonstrated that the HCP5/UTP3/c-Myc/VAMP3 axis plays an important role in the inhibition of cancer cell apoptosis and that HCP5 may be a promising chemosensitivity target for cancer treatment.


Sujet(s)
Antinéoplasiques , Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , microARN , ARN long non codant , Humains , Antinéoplasiques/pharmacologie , Antinéoplasiques/usage thérapeutique , Apoptose/génétique , Lignée cellulaire tumorale , Prolifération cellulaire , Protéines de liaison à l'ADN/métabolisme , Tumeurs de l'oesophage/génétique , Carcinome épidermoïde de l'oesophage/traitement médicamenteux , Régulation de l'expression des gènes tumoraux , microARN/génétique , Récidive tumorale locale/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , Facteurs de transcription/génétique , Ubiquitination , Synaptobrévine-3/génétique , Synaptobrévine-3/métabolisme
10.
Article de Chinois | WPRIM (Pacifique Occidental) | ID: wpr-1025530

RÉSUMÉ

Objective:To investigate the effect of long non-coding RNA (lncRNA) human histocompatibility leukocyte antigen complex P5 (HCP5) on the neuronal injury induced by oxygen glucose deprivation/reoxygenation (OGD/R), and to analyze its potential mechanism.Methods:SH-SY5Y cells were divided into control group (CON group, normal medium culture), model group (Model group, OGD/R), interference control group (si-NC group, OGD/R after HCP5 small interfering RNA negative control (si-NC)), HCP5 interference group (si-HCP5 group, OGD/R after HCP5 small interfering RNA (si-HCP5)), HCP5 interference+ inhibitor control group (si-HCP5+ anti-NC group, OGD/R after transfection of si-HCP5, miR-525-5p inhibitor negative control (anti-NC)), HCP5 interference+ miR-525-5p inhibitor group (si-HCP5+ anti-miR-525-5p group, OGD/R after transfection of si-HCP5, miR-525-5p inhibitor). qRT-PCR was used to detect the expression of lncRNA HCP5 and miR-525-5p in cells.The activity of SH-SY5Y cells was detected by MTT.The level of reactive oxygen species (ROS) in the cells was detected by fluorescent probe of dichlorofluorescein diacetate (DCFH-DA). The apoptosis of SH-SY5Y cells was detected by flow cytometry.Western blot was used to detect the expression of BTG2, Bcl-2 related X protein (Bax), B lymphocyte tumor 2 (Bcl-2) and cleaved caspase-3 protein.SPSS 25.0 software was used to analyze the data, and one-way ANOVA was used for comparison between multiple groups, and SNK- q test was used for further comparison between two groups. Results:There were statistically significant differences in lncRNA HCP5, miR-525-5p RNA levels and BTG2 protein expression levels among the 6 groups ( F=28.853, 59.241, 13.731, all P<0.001). Compared with the CON group, the Model group had higher level of lncRNA HCP5, lower level of miR-525-5p, and higher level of BTG2 protein (all P<0.05). Compared with the Model group, the si-HCP5 group had lower level of lncRNA HCP5, higher level of miR-525-5p, and lower level of BTG2 protein (all P<0.05). Compared with the si-HCP5+ anti-NC group, the si-HCP5+ anti-miR-525-5p group had higher level of lncRNA HCP5, lower level of miR-525-5p, and higher level of BTG2 protein (all P<0.05). There were statistically significant differences in cell activity and ROS levels among the six groups of cells ( F=16.180, 59.950, both P<0.001). The cell activity of the Model group was lower than that of the CON group (0.33±0.12, 0.63±0.11) ( P<0.05), and the ROS level was higher than that of the CON group (224.62±23.27, 100.00±0.00) ( P<0.05). The cell activity of the si-HCP5+ anti-miR-525-5p group was lower than that of the si-HCP5+ anti-NC group (0.38±0.08, 0.58±0.08) ( P<0.05), and the ROS level was higher than that of the si-HCP5+ anti-NC group (207.83±19.39, 135.27±14.36) ( P<0.05). There were statistically significant differences in the apoptosis rate and expression levels of apoptotic proteins Bcl-2, Bax, and cleared Caspase-3 among the six groups of cells ( F=27.994, 29.660, 45.000, 52.983, all P<0.001). There were no statistically significant difference in Bax, Bcl-2, cleared Caspase-3 protein levels, and apoptosis rate in SH-SY5Y cells between the Model group and the si-NC group, as well as between the si-HCP5 group and the si-HCP5+ anti-NC group (all P>0.05). Compared with the CON group, the apoptosis rate, levels of Bax and cleared Casase-3 protein in the Model group were significantly upregulated (all P<0.05), while the Bcl-2 protein level was significantly downregulated ( P<0.05). Compared with the Model group and si-NC group, the si-HCP5 group showed significant downregulation of cell apoptosis rate and levels of Bax and cleared Caspase-3 protein (all P<0.05), while the Bcl-2 protein level was upregulated ( P<0.05). Compared with the si-HCP5 group and si-HCP5+ anti-NC group, the si-HCP5+ anti-miR-525-5p group showed significant upregulation of cell apoptosis rate and levels of Bax and cleared Caspase-3 protein levels (all P<0.05), and significant downregulation of Bcl-2 protein levels ( P<0.05). Conclusion:lncRNA HCP5 may inhibit the expression of BTG2 by targeting up-regulation of miR-525-5p, thus leading to apoptosis of nerve cells in OGD/R models.

11.
Am J Chin Med ; 50(6): 1529-1551, 2022.
Article de Anglais | MEDLINE | ID: mdl-35931662

RÉSUMÉ

Our study aimed to explore the function and mechanism of Dexmedetomidine (Dex) in regulating myocardial ischemia/reperfusion (I/R)-induced mitochondrial apoptosis through lncRNA HCP5. We demonstrated Dex suppressed I/R-induced myocardial infarction and mitochondrial apoptosis in vivo. Dex induced the expression of lncRNA HCP5 and MCL1, inhibited miR-29a expression and activated the JAK2/STAT3 signaling. Dex attenuated hypoxia/reoxygenation (H/R)-induced mitochondrial apoptosis by upregulating lncRNA HCP5 in cardiomyocytes. Overexpression of lncRNA HCP5 sponged miR-29a to suppress H/R-induced mitochondrial apoptosis. Knockdown of miR-29a also alleviated cardiomyocyte apoptosis by upregulating MCL1. Overexpression of lncRNA HCP5 activated the JAK2/STAT3 signaling through sponging miR-29a and enhancing MCL1 expression in cardiomyocytes. Dex mitigated myocardial I/R-induced mitochondrial apoptosis through the lncRNA HCP5/miR-29a/MCL1 axis and activation of the JAK2/STAT3 signaling.


Sujet(s)
Dexmédétomidine , microARN , Ischémie myocardique , Lésion de reperfusion myocardique , ARN long non codant , Apoptose , Dexmédétomidine/pharmacologie , Humains , microARN/génétique , Mitochondries/effets des médicaments et des substances chimiques , Protéine Mcl-1/métabolisme , Ischémie myocardique/génétique , Lésion de reperfusion myocardique/traitement médicamenteux , Lésion de reperfusion myocardique/génétique , Lésion de reperfusion myocardique/métabolisme , Myocytes cardiaques/métabolisme , ARN long non codant/génétique
12.
Kaohsiung J Med Sci ; 38(10): 960-970, 2022 Oct.
Article de Anglais | MEDLINE | ID: mdl-35899856

RÉSUMÉ

Osteosarcoma (OS) is the second most common primary malignant bone tumors in adolescents that causes cancer-related deaths. Previous studies have confirmed the promoting role of lncRNA HCP5 in the development of OS, but the specific mechanism is still not well understood. MiRNA levels were measured via RT-qPCR and protein expression was detected via western blotting. Cell proliferation was analyzed by CCK-8 assays and colony formations assay were conducted to measure colony formation ability. Dual-luciferase reporter assay was performed to detect the targeting relationship between HCP5 and miR-29b-3p, and between miR-29b-3p and LOXL2. Wound healing assays and Transwell assays were conducted to verify the migration and invasion abilities of OS cells. Correlations between the levels of HCP5 and miR-29b-3p, and between miR-29b-3p and LOXL2 were determined by Pearson correlation coefficient analysis. MiR-29b-3p expression was decreased and HCP5 and LOXL2 levels were increased in OS tissues and cell lines. MiR-29b-3p could directly act on LOXL2 and knockdown of LOXL2 restrained the proliferation, migration, and invasion of OS cells. Moreover, transfection with sh-HCP5-1 and sh-HCP5-2 suppressed the malignant biological behavior of OS cells. HCP5 directly targeted miR-29b-3p, and promoted OS proliferation, migration, and invasion via the miR-29b-3p/LOXL2 axis. The lncRNA HCP5 may upregulate LOXL2 expression by targeting miR-29b-3p, thereby promoting OS proliferation, migration, and invasion.


Sujet(s)
Tumeurs osseuses , microARN , Ostéosarcome , ARN long non codant , Amino-acid oxidoreductases/génétique , Amino-acid oxidoreductases/métabolisme , Tumeurs osseuses/génétique , Lignée cellulaire tumorale , Mouvement cellulaire/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux , Humains , microARN/génétique , microARN/métabolisme , Ostéosarcome/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme
13.
Front Neurol ; 13: 878947, 2022.
Article de Anglais | MEDLINE | ID: mdl-35693013

RÉSUMÉ

Objectives: Present study aims to identify the essential mRNAs responsible for the development of brain neurovascular-related metastases (BNM) among lung adenocarcinoma (LUAD) patients. Further, we attempted to predict brain metastases more accurately and prevent their development in LUAD patients. Methods: Transcriptome data analysis was used to identify differentially expressed mRNAs (DEMs) associated with brain metastasis, and thereby the ferroptosis index (FPI) is calculated using a computational model. Meanwhile, the DEmRNAs linked with FPI, and brain metastasis were derived by the intersection of these two groups of DEMs. We also constructed a ceRNA network containing these DEmRNAs, identifying the HCP5 /hsa-miR-17-5p/HOXA7 axis for analysis. Further, a clinical cohort was employed to validate the regulatory roles of molecules involved in the ceRNA regulatory axis. Results: Here we report the development of a ceRNA network based on BNM-associated DEMs and FPI-associated DEmRNAs which includes three core miRNAs (hsa-miR-338-3p, hsa-miR-429, and hsa-miR-17-5p), three mRNAs (HOXA7, TBX5, and TCF21), and five lncRNAs (HCP5, LINC00460, TP53TG1). Using gene set enrichment analysis (GSEA) and survival analysis, the potential axis of HCP5 /hsa-miR-17-5p/HOXA7 was further investigated. It is found that HOXA7 and ferroptosis index are positively correlated while inhibiting tumor brain metastasis. It may be that HCP5 binds competitively with miR-17-5p and upregulates HOXA7 to increase iron death limiting brain cancer metastases. Conclusions: The expression of both HOXA7 and HCP5 is positively correlated with FPI, indicating a possible link between ferroptosis and BNM. According to the results of our study, the ferroptosis-related ceRNA HCP5 /hsa-miR-17-5p/HOXA7 axis may contribute to the development of BNM in LUAD patients.

14.
Endocr J ; 69(9): 1117-1129, 2022 Sep 28.
Article de Anglais | MEDLINE | ID: mdl-35545536

RÉSUMÉ

HCP5 has been reported to be downregulated in ovarian granulosa cells (OGCs) and to facilitate cell proliferation. Human umbilical cord mesenchymal stem cell exosome (hucMSCs-exo) treatment can prevent OGCs apoptosis in vitro. However, the functional mechanism of HCP5 and hucMSCs-exo requires further exploration. Fluorescence-activated cell sorting (FACS) was performed to measure the expression of markers related to hucMSCs. The osteogenic and adipogenic potential of hucMSCs was measured by alkaline phosphatase (ALP) and Alizarin red and by oil red-O staining, respectively. Real-time quantitative polymerase chain reaction (RT-qPCR) and Western blotting were used to detect the mRNA and protein levels, respectively. Cell proliferation and apoptosis were measured by Cell Counting Kit-8 (CCK-8) assay, colony formation assay and flow cytometry. The interaction of HCP5/musashi RNA-binding protein 2 (MSI2) and oestrogen receptor alpha 1 (ESR1) mRNA was analysed using RNA pulldown and RIP assays. HucMSCs and exosomes were successfully isolated and identified. HucMSC-derived exosomes promoted the proliferation of OGCs and ESR1 expression and inhibited cell apoptosis. HCP5 overexpression in exosomes further enhanced these effects. MSI2 knockdown led to the opposite results. HCP5 targeted MSI2, and MSI2 knockdown reduced the decreases in HCP5 and ESR1 expression. Mechanistically, HCP5 in HucMSC-derived exosomes promoted ESR1 expression by binding to MSI2, which promoted the proliferation of OGCs.


Sujet(s)
Exosomes , Cellules souches mésenchymateuses , microARN , ARN long non codant , Phosphatase alcaline/métabolisme , Apoptose/génétique , Prolifération cellulaire , Récepteur alpha des oestrogènes/génétique , Récepteur alpha des oestrogènes/métabolisme , Exosomes/génétique , Exosomes/métabolisme , Femelle , Cellules de la granulosa/métabolisme , Humains , microARN/génétique , ARN long non codant/génétique , ARN long non codant/métabolisme , ARN messager/métabolisme , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme , Protéines de liaison à l'ARN/pharmacologie , Cordon ombilical/métabolisme
15.
Bioengineered ; 13(4): 9301-9311, 2022 04.
Article de Anglais | MEDLINE | ID: mdl-35389828

RÉSUMÉ

Esophageal squamous cell carcinoma (ESCC) is a deadly malignant tumor that threatens human health. Long noncoding RNA (lncRNA) is widely expressed in eukaryotes and is closely associated with human disease progression. However, its role in ESCC remains incompletely understood. In this study, we analyzed the results of three gene expression omnibus (GEO) databases containing lncRNA expression data of ESCC and normal tissues. The results showed that HCP5 was significantly overexpressed in ESCC tissues, which was further verified in our collected ESCC samples. The functional study suggested that HCP5 knockdown inhibited ESCC cell proliferation and invasion. Regarding the mechanism, HCP5 was able to directly interact with YTHDF1, a N6-methyladenosine (m6A) reader, enhancing the binding of YTHDF1 to m6A-modified HK2 mRNA, leading to increasing HK2 stability, thereby promoting the Warburg effect (aerobic glycolysis) of ESCC cells. The nude mice model showed that the knockdown of HCP5 in vivo remarkably reduced tumor size. Clinically, high HCP5 was positively correlated with larger tumor volume, higher TNM stage and lymph node metastasis. Moreover, ESCC patients with high HCP5 exerted shorter survival time than patients with low HCP5. These findings uncover the importance of HCP5 in human ESCC progression; the turbulence of HCP5/YTHDF1/HK2 axis may be responsible for ESCC carcinogenicity.


Sujet(s)
Tumeurs de l'oesophage , Carcinome épidermoïde de l'oesophage , ARN long non codant , Animaux , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire/génétique , Tumeurs de l'oesophage/métabolisme , Carcinome épidermoïde de l'oesophage/métabolisme , Régulation de l'expression des gènes tumoraux , Humains , Souris , Souris nude , Pronostic , ARN long non codant/génétique , ARN long non codant/métabolisme , Régulation positive/génétique
16.
J Cancer ; 13(6): 1882-1894, 2022.
Article de Anglais | MEDLINE | ID: mdl-35399723

RÉSUMÉ

To investigate the important roles of the cancer-promoting long non-coding RNAs (lncRNAs) in cervical cancer, the up-regulated lncRNAs and prognostic analysis were identified through Lnc2Cancer and Lncar. LncRNA-regulated miRNA and miRNA-target mRNA were analyzed based on starBase v2.0 and miTarbase to predict the lncRNA-miRNA-mRNA ceRNA network. Based on the above findings, the abnormally expressed histocompatibility leukocyte antigen complex P5 (HCP5) was identified in 31 cervical cancer patients through RT-qPCR. The stable cell lines were constructed to explore the effect of HCP5 on the promotion of cervical cancer and the regulatory role on the expression of miR-216a-5p and CDC42. Cell Counting Kit-8 (CCK8) assay, cell clone formation, and transwell assay were used to examine proliferation and migration ability of cervical cancer cells. The results displayed that the overexpression of HCP5 promoted cervical cancer cell proliferation and migration in vitro, and the elevated HCP5 can also promote tumor growth in vivo. Besides, RT-qPCR and western blot assay revealed that elevated HCP5 suppressed miR-216a-5p expression and then up-regulated the expression of CDC42. In contrast, knocking down HCP5 resulted in increased expression of miR-216a-5p and then downregulated the expression of CDC42. Rescue experiments also demonstrated that miR-216a-5p could in part intercept in promotion impact caused by HCP5 on cervical cancer cells. Above all, HCP5, as an oncogene, can promote proliferation and migration ability of cervical cancer via the regulation of the miR-216a-5p/CDC42 axis.

17.
Inflammation ; 45(3): 1281-1297, 2022 Jun.
Article de Anglais | MEDLINE | ID: mdl-35122570

RÉSUMÉ

Allergic rhinitis (AR) is an allergic disease characterized as (immunoglobulin, IgE)-mediated type I hypersensitivity disorder. Regulatory T cells (Tregs) play a crucial role in AR. In the present study, we aimed to investigate the mechanism of how Tregs are regulated by long noncoding RNA HCP5 and the regulatory role of HCP5 in IL-13-induced inflammatory response in nasal epithelial cells (NECs) from AR patients. Peripheral blood mononuclear cells (PBMCs) and NECs were obtained from collected blood samples and nasal epithelial tissues. CD4+ T cells and Tregs were purified using certain cell isolation kits from PBMCs and Tregs were also differentiated from CD4+ T cells using recombinant human IL-2 and TGF-ß. The expression levels of HCP5, miR-16, ATXN2L, GM-CSF, eotaxin, and MUC5AC were detected by real-time PCR and western blot. The concentrations of inflammatory cytokines were detected by enzyme-linked immunosorbent assay (ELISA). The interaction among HCP5, miR-16, and ATXN2L were verified by dual-luciferase reporter assay. lncRNA HCP5 expression dramatically downregulated in PBMCs, CD4+ T cells, Tregs, and nasal tissues of AR patients, as well as in IL-13-treated NECs. HCP5 promoted Tregs differentiation and proliferation via targeting miR-16/ATXN2L axis. Additionally, HCP5 inhibited IL-13-induced GM-CSF, eotaxin, and MUC5AC production in NECs. HCP5 sponged miR-16 and negatively regulated its expression, and miR-16 targeted ATXN2L and inhibition of miR-16 suppressed IL-13-induced GM-CSF, eotaxin, and MUC5AC expression. HCP5/miR-16/ATXN2L axis mediated Tregs proliferation and functions in AR. Besides, the regulation of IL-13-induced dysfunction of NECs by lncRNA HCP5 depended on miR-16/ATXN2L in the inflammatory response of AR.


Sujet(s)
microARN , ARN long non codant/génétique , Rhinite allergique , Cellules épithéliales/métabolisme , Facteur de stimulation des colonies de granulocytes et de macrophages , Humains , Interleukine-13/métabolisme , Agranulocytes/métabolisme , microARN/métabolisme , Muqueuse nasale/métabolisme , ARN long non codant/métabolisme
18.
Bioengineered ; 13(1): 884-893, 2022 01.
Article de Anglais | MEDLINE | ID: mdl-34969363

RÉSUMÉ

Recently, long noncoding RNAs (lncRNAs) have been revealed to participate in cancer therapy. Especial in tumor radiotherapy, lncRNAs usually could enhance or restrict the radiosensitivity in different ways. LncRNA HCP5 is highly expressed in esophageal cancer and influenced the malignant behaviors of esophageal cancer cells. However, this study dedicates to clarify if lncRNA HCP5 affects the radiosensitivity of esophageal carcinoma. The expression levels of HCP5 in esophageal cancer and adjacent noncancerous tissue were first analyzed on the TCGA database and then detected by qRT-PCR. The related functional experiments were used to investigate whether the radiosensitivity of esophageal squamous cell carcinoma was affected by the inhibition of HCP5. The expression results showed HCP5 is upregulated in esophageal cancers compared to the normal tissues. Meanwhile, knockdown HCP5 further suppressed the proliferation and promoted the apoptosis of esophageal cancer cells treated with a 2 Gy dose of radiotherapy. Moreover, we uncovered that knockdown HCP5 eliminated radiotherapy resistance by modulating the miR-216a-3p/PDK1 axis to inhibit the AKT activation. Finally, rescue experiments pointed that lowering the miR-216a-3p expression weakened the inhibition effect of knockdown HCP5 on cells treated with radiotherapy. To summary, our results indicate that HCP5 is involved in esophageal carcinoma radiotherapy and knockdown HCP5 enhances the radiosensitivity of esophageal carcinoma by modulating AKT signaling activation.


Sujet(s)
Tumeurs de l'oesophage/génétique , microARN/génétique , Protéines proto-oncogènes c-akt/métabolisme , Pyruvate dehydrogenase acetyl-transferring kinase/métabolisme , ARN long non codant/génétique , Radiotolérance , Régulation positive , Études cas-témoins , Lignée cellulaire tumorale , Mouvement cellulaire , Prolifération cellulaire , Survie cellulaire , Bases de données génétiques , Tumeurs de l'oesophage/métabolisme , Régulation de l'expression des gènes tumoraux , Humains , Transduction du signal
19.
Cell Biol Toxicol ; 38(6): 979-993, 2022 12.
Article de Anglais | MEDLINE | ID: mdl-34331612

RÉSUMÉ

BACKGROUND: Although long non-coding RNA (lncRNA) HCP plays essential roles in human cancers, its function and mechanism in multiple myeloma (MM) have not crystallized. METHODS: HCP5 level in MM was assessed through qRT-PCR. A series of functional investigations were conducted to evaluate the influences of HCP5 on proliferation and apoptosis. Bioinformatics analysis and RIP/RNA pull-down assays were carried out to determine the relationships among HCP5, miR-128-3p, and PLAGL2. Relative protein level was determined through Western blot. A xenograft tumor model was applied for validating the roles of HCP5/miR-128-3p/PLAGL2 axis in vivo. RESULTS: HCP5 was significantly increased in MM. HCP5 knockdown effectively thwarted the proliferative rate and cell cycle of MM cell lines and suppressed tumor growth. HCP5 regulated PLAGL2 expression by sponging miR-128-3p. PLAGL2 overexpression effectively rescued cells from influences by sh-HCP5 on cell proliferative and apoptotic rates. Additionally, HCP5 knockdown significantly inhibited Wnt/ß-catenin/cyclin D1 signaling, and these effects were eliminated by PLAGL2 overexpression. CONCLUSION: Our study revealed that HCP5/miR-128-3p/PLAGL2 is closely correlated to MM development by modulating Wnt/ß-catenin/cyclin D1 signaling. HCP5 promoted cell proliferation and tumor formation of MM cells by activating the Wnt/ß-catenin/CCND1 signaling pathway by sponging miR-128-3p to increase PLAGL2 expression.


Sujet(s)
microARN , Myélome multiple , ARN long non codant , Humains , ARN long non codant/génétique , ARN long non codant/métabolisme , bêta-Caténine/métabolisme , Cycline D1/génétique , Cycline D1/métabolisme , microARN/génétique , microARN/métabolisme , Myélome multiple/génétique , Lignée cellulaire tumorale , Voie de signalisation Wnt/génétique , Prolifération cellulaire/génétique , Régulation de l'expression des gènes tumoraux/génétique , Mouvement cellulaire , Protéines de liaison à l'ADN/génétique , Facteurs de transcription/génétique , Protéines de liaison à l'ARN/métabolisme
20.
Cancer Cell Int ; 21(1): 686, 2021 Dec 19.
Article de Anglais | MEDLINE | ID: mdl-34923990

RÉSUMÉ

BACKGROUND: Accumulating studies indicated that dysregulated long non-coding RNA human histocompatibility leukocyte antigen (HLA) Complex P5 (HCP5) may functions as an potential prognostic predictor in multiple cancers. This meta-analysis was performed to systematically collect studies and conduct an evidence-based evaluation of the prognostic role of HCP5 in malignancies. METHODS: Four databases (PubMed, Web of Science, Embase and Cochrane library) were comprehensively retrieved from their initiation date to November 9, 2021. Hazard ratio (HR) or odds ratio (OR) with 95% confidence interval (CI) were used to assess the associations between the expression level of HCP5 and prognosis or clinical characteristics. Moreover, results were validated by Gene Expression Profiling Interactive Analysis 2 (GEPIA2) and the National Genomics Data Center (NGDC). Subsequently, the molecular mechanism of HCP5 was predicted based on MEM and StarBase databases. The study protocol was registered at PROSPERO (ID: CRD42021274208). RESULTS: 9 studies, containing 641 patients, were included in this meta-analysis. Our results revealed that HCP5 overexpression was associated with poor overall survival (OS), tumor type, histological differentiation, and lymph node metastasis in most cancers, but was not associated with age, gender and tumor size; down-regulation of HCP5 was associated with worse OS, advanced tumor stage, positive distal metastasis and lymph node metastasis in skin cutaneous melanoma (SKCM). HCP5 was significantly up-regulated in four cancers and down-regulated in SKCM, which was validated by the GEPIA2 cohort. HCP5 expression in various types of cancer was also verified in NGDC. Further functional prediction revealed that HCP5 may participate in some cancer-related pathways. CONCLUSION: There is a significantly association between dysregulation of HCP5 and both prognosis and clinicopathological features in various cancers. HCP5 may be functions as a novel potential prognostic biomarker and therapeutic target in multiple human cancers.

SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE