Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 8.398
Filter
1.
J Clin Invest ; 134(9)2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38530366

ABSTRACT

Aberrant expression of the E26 transformation-specific (ETS) transcription factors characterizes numerous human malignancies. Many of these proteins, including EWS:FLI1 and EWS:ERG fusions in Ewing sarcoma (EwS) and TMPRSS2:ERG in prostate cancer (PCa), drive oncogenic programs via binding to GGAA repeats. We report here that both EWS:FLI1 and ERG bind and transcriptionally activate GGAA-rich pericentromeric heterochromatin. The respective pathogen-like HSAT2 and HSAT3 RNAs, together with LINE, SINE, ERV, and other repeat transcripts, are expressed in EwS and PCa tumors, secreted in extracellular vesicles (EVs), and are highly elevated in plasma of patients with EwS with metastatic disease. High human satellite 2 and 3 (HSAT2,3) levels in EWS:FLI1- or ERG-expressing cells and tumors were associated with induction of G2/M checkpoint, mitotic spindle, and DNA damage programs. These programs were also activated in EwS EV-treated fibroblasts, coincident with accumulation of HSAT2,3 RNAs, proinflammatory responses, mitotic defects, and senescence. Mechanistically, HSAT2,3-enriched cancer EVs induced cGAS-TBK1 innate immune signaling and formation of cytosolic granules positive for double-strand RNAs, RNA-DNA, and cGAS. Hence, aberrantly expressed ETS proteins derepress pericentromeric heterochromatin, yielding pathogenic RNAs that transmit genotoxic stress and inflammation to local and distant sites. Monitoring HSAT2,3 plasma levels and preventing their dissemination may thus improve therapeutic strategies and blood-based diagnostics.


Subject(s)
DNA Damage , Extracellular Vesicles , Oncogene Proteins, Fusion , Proto-Oncogene Protein c-fli-1 , RNA-Binding Protein EWS , Transcriptional Regulator ERG , Humans , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Transcriptional Regulator ERG/genetics , Transcriptional Regulator ERG/metabolism , Male , RNA-Binding Protein EWS/genetics , RNA-Binding Protein EWS/metabolism , Proto-Oncogene Protein c-fli-1/genetics , Proto-Oncogene Protein c-fli-1/metabolism , Sarcoma, Ewing/genetics , Sarcoma, Ewing/pathology , Sarcoma, Ewing/metabolism , Sarcoma, Ewing/immunology , Cell Line, Tumor , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Inflammation/genetics , Inflammation/metabolism , Inflammation/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/metabolism , Mice , Animals , Heterochromatin/metabolism , Heterochromatin/genetics
2.
Exp Hematol ; 133: 104175, 2024 May.
Article in English | MEDLINE | ID: mdl-38311165

ABSTRACT

Acute myeloid leukemia (AML) is a common hematological cancer. Cancer cells exchange information with the surrounding microenvironment, which can be transmitted by extracellular vesicles (EVs). In recent years, the genetic materials transported by EVs have attracted attention due to their important roles in different pathological processes. EV-derived ncRNAs (EV-ncRNAs) regulate physiological functions and maintain homeostasis, mainly including microRNAs, long noncoding RNAs, and circular RNAs. However, the mechanism of involvement and potential clinical application of EV-ncRNAs in AML have not been reported. Given the unique importance of the bone marrow microenvironment (BMME) for AML, a greater understanding of the communication between leukemic cells and the BMME is needed to improve the prognosis of patients and reduce the incidence of recurrence. Additionally, studies on leukemic EV-ncRNA transport guide the design of new diagnostic and therapeutic tools for AML. This review systematically describes intercellular communication in the BMME of AML and emphasizes the role of EVs. More importantly, we focus on the information transmission of EV-ncRNAs in the BMME to explore their clinical application as potential biomarkers and therapeutic targets.


Subject(s)
Bone Marrow , Cell Communication , Extracellular Vesicles , Leukemia, Myeloid, Acute , Tumor Microenvironment , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Extracellular Vesicles/metabolism , Extracellular Vesicles/genetics , Bone Marrow/metabolism , Bone Marrow/pathology , Animals , RNA, Untranslated/genetics , RNA, Untranslated/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
3.
Anal Biochem ; 661: 114986, 2023 01 15.
Article in English | MEDLINE | ID: mdl-36384188

ABSTRACT

MicroRNAs (miRNAs) associated with lung cancer are diversifying. MiR-21, Let-7, and miR-141 are common diagnostic targets. Some new lung cancer miRNAs, such as miR-25, miR-145, and miR-126, have received increasing attention. Although various techniques are available for the analysis of lung cancer miRNAs, electrochemistry has been recognized for its high sensitivity, low cost, and rapid response. However, how to realize the signal amplification is one of the most important contents in the design of electrochemical biosensors. Herein, we mainly introduce the amplification strategy based on enzyme-free amplification and signal conversion, including non-linear HCR, catalytic hairpin assembly (CHA), electrochemiluminescence (ECL), and Faraday cage. Furthermore, new progress has emerged in the fields of nanomaterials, low oxidation potential, and simultaneous detection of multiple targets. Finally, we summarize some new challenges that electrochemical techniques may encounter in the future, such as improving single-base discrimination ability, shortening electrochemical detection time, and providing real body fluid samples assay.


Subject(s)
Biomarkers, Tumor , Lung Neoplasms , MicroRNAs , RNA, Neoplasm , Humans , Electrochemistry , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , MicroRNAs/analysis , MicroRNAs/genetics , RNA, Neoplasm/analysis , RNA, Neoplasm/genetics , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics
4.
Nature ; 608(7921): 199-208, 2022 08.
Article in English | MEDLINE | ID: mdl-35859180

ABSTRACT

Circulating tumour DNA (ctDNA) in blood plasma is an emerging tool for clinical cancer genotyping and longitudinal disease monitoring1. However, owing to past emphasis on targeted and low-resolution profiling approaches, our understanding of the distinct populations that comprise bulk ctDNA is incomplete2-12. Here we perform deep whole-genome sequencing of serial plasma and synchronous metastases in patients with aggressive prostate cancer. We comprehensively assess all classes of genomic alterations and show that ctDNA contains multiple dominant populations, the evolutionary histories of which frequently indicate whole-genome doubling and shifts in mutational processes. Although tissue and ctDNA showed concordant clonally expanded cancer driver alterations, most individual metastases contributed only a minor share of total ctDNA. By comparing serial ctDNA before and after clinical progression on potent inhibitors of the androgen receptor (AR) pathway, we reveal population restructuring converging solely on AR augmentation as the dominant genomic driver of acquired treatment resistance. Finally, we leverage nucleosome footprints in ctDNA to infer mRNA expression in synchronously biopsied metastases, including treatment-induced changes in AR transcription factor signalling activity. Our results provide insights into cancer biology and show that liquid biopsy can be used as a tool for comprehensive multi-omic discovery.


Subject(s)
Circulating Tumor DNA , Drug Resistance, Neoplasm , Genome, Human , Genomics , High-Throughput Nucleotide Sequencing , Mutation , Prostatic Neoplasms , Androgen Receptor Antagonists/pharmacology , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Circulating Tumor DNA/blood , Circulating Tumor DNA/genetics , Clone Cells/metabolism , Clone Cells/pathology , Disease Progression , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Genetic Markers/genetics , Genome, Human/genetics , Genomics/methods , Humans , Liquid Biopsy/methods , Male , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Nucleosomes/genetics , Nucleosomes/metabolism , Prostatic Neoplasms/blood , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , RNA, Messenger/analysis , RNA, Messenger/genetics , RNA, Neoplasm/analysis , RNA, Neoplasm/genetics , Receptors, Androgen/metabolism
5.
Comput Math Methods Med ; 2022: 2857022, 2022.
Article in English | MEDLINE | ID: mdl-35756415

ABSTRACT

Oral squamous cell carcinoma (OSCC) has increasingly become a worldwide health concern, and its survival rate has not been much improved partially due to a deficiency of precise molecular markers. Dysregulation of LINC01116, a long noncoding RNA sequence, has been observed in several types of cancer. However, the role played by LINC01116 in OSCC has not yet been fully elaborated. This study explored how LINC01116 was involved in the regulation of OSCC progression by analyzing expressions of LINC01116 in OSCC patients. The findings demonstrated upregulation of LINC01116 in OSCC tissues as opposed to regular oral mucosa, and overexpression of LINC01116 was correlated with advanced tumor status. LINC01116 knockdown using shRNA markedly reduced the OSCC cell invasion and migration in vitro. Moreover, the expression of LINC01116 was negatively correlated with that of microRNA-9-5p (miR-9). Luciferase reporter and loss-of-function assays demonstrated that LINC01116 functioned as a competing endogenous RNA (ceRNA) that could effectively sponge miR-9, thus regulating the derepression of matrix metalloproteinase 1 (MMP1). Furthermore, we confirmed that LINC01116 knockdown did not affect the expression of MMP1 messenger RNA (mRNA). Collectively, it is demonstrated in this study that overexpression of LINC01116 can promote the OSCC progression. The LINC01116-miR-9-MMP1 axis provides a novel insight into the OSCC pathogenesis and offers potential therapeutic targets against OSCC.


Subject(s)
Matrix Metalloproteinase 1 , MicroRNAs , Mouth Neoplasms , RNA, Long Noncoding , RNA, Neoplasm , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Humans , Matrix Metalloproteinase 1/biosynthesis , Matrix Metalloproteinase 1/genetics , Matrix Metalloproteinase 1/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/pathology
6.
Med Sci Monit ; 28: e935055, 2022 Mar 12.
Article in English | MEDLINE | ID: mdl-35277469

ABSTRACT

BACKGROUND Tongue cancer is the most prevalent of head and neck squamous cell carcinomas, including base of tongue cancer (BOT) and oral squamous cell carcinoma of the mobile tongue (OTSCC). We aimed to investigate the role of RIPOR3 in tumorigenesis and its development as a potential prognostic biomarker for tongue cancer, especially OTSCC. MATERIAL AND METHODS Associations of expression, clinical pathologic features, and overall survival were analyzed by logistic regression, multivariate Cox analysis, and Kaplan-Meier methods. Gene set enrichment analysis (GSEA) and the CIBERSORT algorithm were performed to determine the correlation between RIPOR3 and tumor immune infiltration. cBioPortal was used for methylation and copy number variation (CNV) analysis. The Human Protein Atlas (HPA) and GSE31056 dataset were used for further external validation. RESULTS RIPOR3 expression in OTSCC was significantly associated with various clinicopathological parameters. Kaplan-Meier survival analysis showed that OTSCC with low RIPOR3 expression had a worse prognosis than that with high RIPOR3 expression. Multivariate analysis revealed that lower RIPOR3 expression was an independent prognostic factor for poor prognosis. GSEA and Neighbor Gene Network analysis showed RIPOR3 expression was related with the modulation and function of the immune-related pathway. Methylation level and CNV analysis showed that the downregulated expression of RIPOR3 was significantly related to hypermethylation but not to CNV. Finally, high RIPOR3 expression was validated at the protein level using the HPA database and GSE31056 dataset. CONCLUSIONS These findings suggested that RIPOR3 might serve as a promising prognostic biomarker and is related to the immune cell infiltration of OTSCC.


Subject(s)
Carcinogenesis/genetics , Down-Regulation , Gene Expression Regulation, Neoplastic , Mouth Neoplasms/genetics , RNA, Neoplasm/genetics , rac GTP-Binding Proteins/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/pathology , DNA Copy Number Variations , DNA Methylation , Female , Humans , Male , Middle Aged , Prognosis , Tongue Neoplasms/genetics , rac GTP-Binding Proteins/biosynthesis
7.
Med Sci Monit ; 28: e935071, 2022 Mar 17.
Article in English | MEDLINE | ID: mdl-35296631

ABSTRACT

BACKGROUND As the second most frequent factor of brain metastasis worldwide, breast cancer and its pathogenesis have been researched intensively. Nevertheless, the molecular mechanisms of brain metastasis from breast cancer (BMBC) remain uncertain. The purpose of this study was to explore the key genes concerning the prognosis of BMBC and identify their predictive value. MATERIAL AND METHODS Obtained from the Gene Expression Omnibus (GEO) database, microarray datasets GSE125989, GSE52604, and GSE159956 were used to identify the differentially expressed genes (DEGs) and perform function enrichment analysis. RESULTS Of a total of 240 DEGs, 113 genes were upregulated and 127 genes were downregulated. The protein-protein interaction (PPI) was performed through STRING, and 29 hub genes were screened through Cytoscape. After being examined through the cBioportal online platform and the Oncomine database, 8 key genes were finally obtained, including COL14A1, COL3A1, COL6A3, THY1, MMP14, GAP43, PTPRN, and SNAP25. In the validation dataset GSE46928, COL14A1 was shown to have predictive significance of brain metastasis in breast cancer. CONCLUSIONS The key genes explored in this article could assist in identifying the molecular mechanism of BMBC. Also, COL14A1, COL3A1, COL6A3, THY1, MMP14, GAP43, PTPRN, and SNAP25 might be candidate targets for diagnosis and treatment of BMBC, and COL3A1 might have predictive value.


Subject(s)
Brain Neoplasms/genetics , Breast Neoplasms/genetics , Computational Biology/methods , Down-Regulation , Gene Expression Regulation, Neoplastic , Oncogenes/genetics , Up-Regulation , Brain Neoplasms/diagnosis , Brain Neoplasms/secondary , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Female , Gene Expression Profiling , Gene Regulatory Networks , Humans , Neoplasm Metastasis , Prognosis , RNA, Neoplasm/genetics
8.
PLoS One ; 17(2): e0264025, 2022.
Article in English | MEDLINE | ID: mdl-35213597

ABSTRACT

Experimental breakthroughs have provided unprecedented insights into the genes involved in cancer. The identification of such cancer driver genes is a major step in gaining a fuller understanding of oncogenesis and provides novel lists of potential therapeutic targets. A key area that requires additional study is the posttranscriptional control mechanisms at work in cancer driver genes. This is important not only for basic insights into the biology of cancer, but also to advance new therapeutic modalities that target RNA-an emerging field with great promise toward the treatment of various cancers. In the current study we performed an in silico analysis on the transcripts associated with 800 cancer driver genes (10,390 unique transcripts) that identified 179,190 secondary structural motifs with evidence of evolutionarily ordered structures with unusual thermodynamic stability. Narrowing to one transcript per gene, 35,426 predicted structures were subjected to phylogenetic comparisons of sequence and structural conservation. This identified 7,001 RNA secondary structures embedded in transcripts with evidence of covariation between paired sites, supporting structure models and suggesting functional significance. A select set of seven structures were tested in vitro for their ability to regulate gene expression; all were found to have significant effects. These results indicate potentially widespread roles for RNA structure in posttranscriptional control of human cancer driver genes.


Subject(s)
Evolution, Molecular , Neoplasms , Nucleic Acid Conformation , Phylogeny , RNA Processing, Post-Transcriptional , RNA Stability , RNA, Neoplasm , Humans , Neoplasms/genetics , Neoplasms/metabolism , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism
9.
Clin Cancer Res ; 28(3): 498-506, 2022 02 01.
Article in English | MEDLINE | ID: mdl-35105718

ABSTRACT

PURPOSE: Previously, clinical trials of experimental virotherapy for recurrent glioblastoma multiforme (GBM) demonstrated that inoculation with a conditionally replication-competent Δγ134.5 oncolytic herpes simplex virus (oHSV), G207, was safe. Following the initial safety study, a phase Ib trial enrolled 6 adult patients diagnosed with GBM recurrence from which tumor tissue was banked for future studies. PATIENTS AND METHODS: Here, we analyzed tumor RNA sequencing (RNA-seq) data obtained from pre- and posttreatment (collected 2 or 5 days after G207 injection) biopsies from the phase Ib study patients. RESULTS: Using a Spearman rank-order correlation analysis, we identified approximately 500 genes whose expression pattern correlated with survival duration. Many of these genes were enriched for the intrinsic IFN-mediated antiviral and adaptive immune functional responses, including immune cell chemotaxis and antigen presentation to T-cells. Furthermore, we show that the expression of several T-cell-related genes was highest in the patient with the longest survival after G207 inoculation. CONCLUSIONS: Our data support that the oHSV-induced type I IFN production and the subsequent recruitment of an adaptive immune response differed between enrolled patients and showed association with survival duration in patients with recurrent malignant glioma after treatment with an early generation oHSV.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/therapy , Clinical Trials, Phase I as Topic , Gene Expression Profiling/methods , Glioblastoma/genetics , Glioblastoma/therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/therapy , Oncolytic Virotherapy/methods , Oncolytic Viruses , RNA, Neoplasm/genetics , Simplexvirus , Adult , Aged , Brain Neoplasms/immunology , Brain Neoplasms/mortality , Female , Glioblastoma/immunology , Glioblastoma/mortality , Humans , Male , Middle Aged , Neoplasm Recurrence, Local/mortality , Survival Rate
10.
Proc Natl Acad Sci U S A ; 119(6)2022 02 08.
Article in English | MEDLINE | ID: mdl-35105806

ABSTRACT

The protumor roles of alternatively activated (M2) tumor-associated macrophages (TAMs) have been well established, and macrophage reprogramming is an important therapeutic goal. However, the mechanisms of TAM polarization remain incompletely understood, and effective strategies for macrophage targeting are lacking. Here, we show that miR-182 in macrophages mediates tumor-induced M2 polarization and can be targeted for therapeutic macrophage reprogramming. Constitutive miR-182 knockout in host mice and conditional knockout in macrophages impair M2-like TAMs and breast tumor development. Targeted depletion of macrophages in mice blocks the effect of miR-182 deficiency in tumor progression while reconstitution of miR-182-expressing macrophages promotes tumor growth. Mechanistically, cancer cells induce miR-182 expression in macrophages by TGFß signaling, and miR-182 directly suppresses TLR4, leading to NFκb inactivation and M2 polarization of TAMs. Importantly, therapeutic delivery of antagomiR-182 with cationized mannan-modified extracellular vesicles effectively targets macrophages, leading to miR-182 inhibition, macrophage reprogramming, and tumor suppression in multiple breast cancer models of mice. Overall, our findings reveal a crucial TGFß/miR-182/TLR4 axis for TAM polarization and provide rationale for RNA-based therapeutics of TAM targeting in cancer.


Subject(s)
Cellular Reprogramming , Mammary Neoplasms, Animal/metabolism , MicroRNAs/metabolism , RNA, Neoplasm/metabolism , Signal Transduction , Tumor-Associated Macrophages/metabolism , Animals , Female , Gene Expression Regulation, Neoplastic , HeLa Cells , Humans , Mammary Neoplasms, Animal/genetics , Mice , Mice, Knockout , MicroRNAs/genetics , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , RNA, Neoplasm/genetics , Toll-Like Receptor 4/biosynthesis , Toll-Like Receptor 4/genetics , Transforming Growth Factor beta/biosynthesis , Transforming Growth Factor beta/genetics
11.
Eur J Med Res ; 27(1): 14, 2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35101137

ABSTRACT

BACKGROUND: Aberrant Wnt signalling, regulating cell development and stemness, influences the development of many cancer types. The Aryl hydrocarbon receptor (AhR) mediates tumorigenesis of environmental pollutants. Complex interaction patterns of genes assigned to AhR/Wnt-signalling were recently associated with lung cancer susceptibility. AIM: To assess the association and predictive ability of AhR/Wnt-genes with lung cancer in cases and controls of European descent. METHODS: Odds ratios (OR) were estimated for genomic variants assigned to the Wnt agonist and the antagonistic genes DKK2, DKK3, DKK4, FRZB, SFRP4 and Axin2. Logistic regression models with variable selection were trained, validated and tested to predict lung cancer, at which other previously identified SNPs that have been robustly associated with lung cancer risk could also enter the model. Furthermore, decision trees were created to investigate variant × variant interaction. All analyses were performed for overall lung cancer and for subgroups. RESULTS: No genome-wide significant association of AhR/Wnt-genes with overall lung cancer was observed, but within the subgroups of ever smokers (e.g., maker rs2722278 SFRP4; OR = 1.20; 95% CI 1.13-1.27; p = 5.6 × 10-10) and never smokers (e.g., maker rs1133683 Axin2; OR = 1.27; 95% CI 1.19-1.35; p = 1.0 × 10-12). Although predictability is poor, AhR/Wnt-variants are unexpectedly overrepresented in optimized prediction scores for overall lung cancer and for small cell lung cancer. Remarkably, the score for never-smokers contained solely two AhR/Wnt-variants. The optimal decision tree for never smokers consists of 7 AhR/Wnt-variants and only two lung cancer variants. CONCLUSIONS: The role of variants belonging to Wnt/AhR-pathways in lung cancer susceptibility may be underrated in main-effects association analysis. Complex interaction patterns in individuals of European descent have moderate predictive capacity for lung cancer or subgroups thereof, especially in never smokers.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Gene Expression Regulation, Neoplastic , Genetic Predisposition to Disease , Genome-Wide Association Study/methods , Lung Neoplasms/genetics , RNA, Neoplasm/genetics , Receptors, Aryl Hydrocarbon/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Female , Genotype , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Receptors, Aryl Hydrocarbon/metabolism , Wnt Signaling Pathway
12.
Signal Transduct Target Ther ; 7(1): 40, 2022 02 14.
Article in English | MEDLINE | ID: mdl-35153295

ABSTRACT

Circular RNAs (circRNAs) were shown to play an important role in the occurrence and progression of tumors. However, the functions of nuclear genome-derived circRNAs localized in mitochondria of tumor cells remain largely elusive. Here, we report that circPUM1, a circular RNA derived from back-splicing of pre-mRNAs of nuclear genome PUM1, localizes in mitochondria. The expression level of circPUM1 is positively correlated with HIF1α accumulation under CoCl2-induced intracellular hypoxic-like condition in esophageal squamous cell carcinoma (ESCC) cell lines. Importantly, circPUM1 acts as a scaffold for the interaction between UQCRC1 and UQCRC2 in ESCC cell lines. Knock-down of circPUM1 would result in lower intracellular oxygen concentration, downregulated oxidative phosphorylation, decrease of mitochondrial membrane potential, increase of ROS generation and shrinking of mitochondria, respectively. CircPUM1 depletion induces dysfunction of the mitochondrial complex III and the cleavage of caspase3 spontaneously. Interestingly, disruption of circPUM1 led to pyroptosis that initiates the cell death of ESCC cell lines. Therefore, we conclude that circPUM1 plays a critical role in maintaining the stability of mitochondrial complex III to enhance oxidative phosphorylation for ATP production of ESCC cells and moreover propose that ESCC cells exploit circPUM1 during cell adaptation.


Subject(s)
Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Mitochondria/metabolism , Oxidative Phosphorylation , RNA, Circular/metabolism , RNA, Neoplasm/metabolism , Animals , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Humans , Male , Mice , Mice, Nude , Mitochondria/genetics , RNA, Circular/genetics , RNA, Neoplasm/genetics
13.
Mol Cell Biochem ; 477(4): 1281-1293, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35129780

ABSTRACT

Triple-Negative Breast Cancer (TNBC) is one of the most aggressive and hot BC subtypes. Our research group has recently shed the light on the utility of natural compounds as effective immunotherapeutic agents. The aim of this study is to investigate the role of a methoxylated quercetin glycoside (MQG) isolated from Cleome droserifolia in harnessing TNBC progression and tuning the tumor microenvironment and natural killer cells cytotoxicity. Results showed that MQG showed the highest potency (IC50 = 12 µM) in repressing cellular proliferation, colony-forming ability, migration, and invasion capacities. Mechanistically, MQG was found to modulate a circuit of competing endogenous RNAs where it was found to reduce the oncogenic MALAT-1 lncRNA and induce TP53 and its downstream miRNAs; miR-155 and miR-146a. Accordingly, this leads to alteration in several downstream signaling pathways such as nitric oxide synthesizing machinery, natural killer cells' cytotoxicity through inducing the expression of its activating ligands such as MICA/B, ULBP2, CD155, and ICAM-1 and trimming of the immune-suppressive cytokines such as TNF-α and IL-10. In conclusion, this study shows that MQG act as a compelling anti-cancer agent repressing TNBC hallmarks, activating immune cell recognition, and alleviating the immune-suppressive tumor microenvironment experienced by TNBC patients.


Subject(s)
Glycosides/pharmacology , MicroRNAs/immunology , RNA, Long Noncoding/immunology , RNA, Neoplasm/immunology , Triple Negative Breast Neoplasms/immunology , Tumor Suppressor Protein p53/immunology , Female , Humans , MCF-7 Cells , MicroRNAs/genetics , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics , Triple Negative Breast Neoplasms/drug therapy , Triple Negative Breast Neoplasms/genetics , Tumor Suppressor Protein p53/genetics
14.
Mol Cell Biochem ; 477(4): 1295-1308, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35137328

ABSTRACT

Previous studies have reported that exosomes bearing certain microRNAs (miRNAs) are related to the physiological functions of different types of cancer cells. Our study aimed to elucidate the role of miR-200a in esophageal squamous cell carcinoma (ESCC). We observed that miR-200a expression is higher in esophageal carcinoma cells, tissues, and exosomes than in normal cells and healthy tissues. We showed that exosome-shuttled miR-200a promotes the proliferation, migration, and invasion of esophageal cells and inhibits apoptosis, thereby leading to the progression of ESCC. We showed that miR-200a exerts its effects through its interaction with Keap1, thus altering the Keap1/Nrf2 signaling pathway. Our results suggest that exosome-shuttled miR-200a might be useful as a biomarker for prognosis in patients with ESCC.


Subject(s)
Cell Movement , Cell Proliferation , Esophageal Neoplasms/metabolism , Exosomes/metabolism , Gene Expression Regulation, Neoplastic , Kelch-Like ECH-Associated Protein 1/biosynthesis , MicroRNAs/metabolism , Neoplasm Proteins/biosynthesis , RNA, Neoplasm/metabolism , Aged , Cell Line, Tumor , Esophageal Neoplasms/genetics , Exosomes/genetics , Female , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Proteins/genetics , RNA, Neoplasm/genetics
15.
Elife ; 112022 02 11.
Article in English | MEDLINE | ID: mdl-35147498

ABSTRACT

Lung cancer (LC) prognosis is closely linked to the stage of disease when diagnosed. We investigated the biomarker potential of serum RNAs for the early detection of LC in smokers at different prediagnostic time intervals and histological subtypes. In total, 1061 samples from 925 individuals were analyzed. RNA sequencing with an average of 18 million reads per sample was performed. We generated machine learning models using normalized serum RNA levels and found that smokers later diagnosed with LC in 10 years can be robustly separated from healthy controls regardless of histology with an average area under the ROC curve (AUC) of 0.76 (95% CI, 0.68-0.83). Furthermore, the strongest models that took both time to diagnosis and histology into account successfully predicted non-small cell LC (NSCLC) between 6 and 8 years, with an AUC of 0.82 (95% CI, 0.76-0.88), and SCLC between 2 and 5 years, with an AUC of 0.89 (95% CI, 0.77-1.0), before diagnosis. The most important separators were microRNAs, miscellaneous RNAs, isomiRs, and tRNA-derived fragments. We have shown that LC can be detected years before diagnosis and manifestation of disease symptoms independently of histological subtype. However, the highest AUCs were achieved for specific subtypes and time intervals before diagnosis. The collection of models may therefore also predict the severity of cancer development and its histology. Our study demonstrates that serum RNAs can be promising prediagnostic biomarkers in an LC screening setting, from early detection to risk assessment.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , MicroRNAs , RNA, Neoplasm , Biomarkers, Tumor/blood , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/genetics , Early Detection of Cancer , Humans , Lung Neoplasms/blood , Lung Neoplasms/diagnosis , Lung Neoplasms/genetics , Lung Neoplasms/pathology , MicroRNAs/blood , MicroRNAs/genetics , RNA, Neoplasm/blood , RNA, Neoplasm/genetics , ROC Curve
16.
Sci Rep ; 12(1): 1789, 2022 02 02.
Article in English | MEDLINE | ID: mdl-35110572

ABSTRACT

Despite the recent precipitous decline in the cost of genome sequencing, library preparation for RNA-seq is still laborious and expensive for applications such as high throughput screening. Limited availability of RNA generated by some experimental workflows poses an additional challenge and increases the cost of RNA library preparation. In a search for low cost, automation-compatible RNA library preparation kits that maintain strand specificity and are amenable to low input RNA quantities, we systematically tested two recent commercial technologies-Swift RNA and Swift Rapid RNA, presently offered by Integrated DNA Technologies (IDT) -alongside the Illumina TruSeq stranded mRNA, the de facto standard workflow for bulk transcriptomics. We used the Universal Human Reference RNA (UHRR) (composed of equal quantities of total RNA from 10 human cancer cell lines) to benchmark gene expression in these kits, at input quantities ranging between 10 to 500 ng. We found normalized read counts between all treatment groups to be in high agreement. Compared to the Illumina TruSeq stranded mRNA kit, both Swift RNA library kits offer shorter workflow times enabled by their patented Adaptase technology. We also found the Swift RNA kit to produce the fewest number of differentially expressed genes and pathways directly attributable to input mRNA amount.


Subject(s)
Biomarkers, Tumor/genetics , Gene Library , Neoplasms/genetics , RNA, Neoplasm/analysis , RNA-Seq/methods , RNA-Seq/standards , Transcriptome , Gene Expression Profiling , Humans , Neoplasms/pathology , RNA, Neoplasm/genetics , Sequence Analysis, RNA/methods , Tumor Cells, Cultured
17.
Eur J Med Res ; 27(1): 29, 2022 Feb 24.
Article in English | MEDLINE | ID: mdl-35209947

ABSTRACT

BACKGROUND: The incidence of thyroid cancer, a most common tumor in the endocrine system, has increased in recent years. A growing number of studies have focused on the molecular mechanisms of thyroid cancer subtypes, aiming to identify effective therapeutic targets. Endocytosis is of vital significance in the malignant development of tumors, although its involvement in thyroid cancer has been rarely reported. METHODS: HIP1R expressions in thyroid cancer from the TCGA database were analyzed by UALCAN software. Thyroid epithelial and cancer cell lines were cultured in vitro. Western blotting and quantitative PCR were used to analyze protein and mRNA levels, respectively. Cell viability was measured by CCK-8 assay. Immunofluorescence staining indicated protein distribution in cell. Co-immunoprecipitation was used to study protein-protein interaction. Immunohistochemical staining was used to analyze protein expression in clinical tissues. Differences between groups were compared using the two-tailed Student's t test, and those among three or more groups were compared by one-way or two-way ANOVA. RESULTS: In the present study, HIP1R (Huntingtin Interacting Protein 1 Related) was found upregulated in thyroid cancer tissues and cell lines compared with that in the controls, while knockdown of HIP1R significantly inhibited the proliferation of thyroid cancer cells. Since HIP1R is essential for the clathrin-dependent endocytic process, we thereafter explored the effect of HIP1R on the endocytosis of thyroid cancer cells. Interestingly, knockdown of HIP1R significantly reduced the number of clathrin-coated pits (CCPs) in thyroid cancer cells. In addition, the interaction between HIP1R and PTEN (phosphatase and tensin homolog) was identified in thyroid cancer cells. Knockdown of HIP1R downregulated intracellular PTEN in thyroid cancer cells, but upregulated membrane-binding PTEN. Notably, flurbiprofen, a commonly used analgesic, significantly inhibited the proliferation of thyroid cancer cells and interfered with the interaction between HIP1R and PTEN, thereby enhancing the binding of PTEN to cell membrane. However, the proliferation inhibitory effect of flurbiprofen was attenuated when knocking down HIP1R or PTEN. CONCLUSIONS: Upregulated HIP1R in thyroid cancer cells promotes cell proliferation and mediates the endocytosis of PTEN. Flurbiprofen may exert an anti-tumor effect on thyroid cancer by blocking the interaction between HIP1R and PTEN.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Flurbiprofen/pharmacology , Gene Expression Regulation, Neoplastic , Microfilament Proteins/genetics , RNA, Neoplasm/genetics , Thyroid Neoplasms/genetics , Adaptor Proteins, Signal Transducing/biosynthesis , Cell Proliferation , Cells, Cultured , Cyclooxygenase Inhibitors/pharmacology , Endocytosis/drug effects , Endocytosis/genetics , Humans , Microfilament Proteins/biosynthesis , Signal Transduction , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/pathology
18.
Int J Mol Sci ; 23(3)2022 Jan 27.
Article in English | MEDLINE | ID: mdl-35163397

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most common types of cancer diagnosed worldwide with high morbidity; drug resistance is often responsible for treatment failure in CRC. Non-coding RNAs (ncRNAs) play distinct regulatory roles in tumorigenesis, cancer progression and chemoresistance. METHODS: A literature search was conducted in PubMed database in order to sum up and discuss the role of exosomal ncRNAs (ex-ncRNAs) in CRC drug resistance/response and their possible mechanisms. RESULTS: Thirty-six (36) original research articles were identified; these included exosome or extracellular vesicle (EV)-containing microRNAs (miRNAs), long non-coding RNAs (lncRNAs), circular RNAs (circRNAs) and small-interfering (siRNAs). No studies were found for piwi-interacting RNAs. CONCLUSIONS: Exosomal transfer of ncRNAs has been documented as a new mechanism of CRC drug resistance. Despite being in its infancy, it has emerged as a promising field for research in order to (i) discover novel biomarkers for therapy monitoring and/or (ii) reverse drug desensitization.


Subject(s)
Colorectal Neoplasms , Drug Resistance, Neoplasm/genetics , Exosomes , RNA, Neoplasm , RNA, Untranslated , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Exosomes/genetics , Exosomes/metabolism , Humans , RNA, Neoplasm/genetics , RNA, Neoplasm/metabolism , RNA, Untranslated/genetics , RNA, Untranslated/metabolism
19.
Int J Mol Sci ; 23(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35163589

ABSTRACT

Hepatocellular carcinoma (HCC) is the most common primary malignancy of the liver and is one of the leading causes of cancer-related deaths worldwide. Regorafenib, a multi-kinase inhibitor, is used as a second-line treatment for advanced HCC. Here, we aimed to investigate the mechanism of the antitumor effect of regorafenib on HCC and evaluate altered microRNA (miRNA) expression. Cell proliferation was examined in six HCC cell lines (HuH-7, HepG2, HLF, PLC/PRF/5, Hep3B, and Li-7) using the Cell Counting Kit-8 assay. Xenografted mouse models were used to assess the effects of regorafenib in vivo. Cell cycle analysis, western blotting analysis, and miRNA expression analysis were performed to identify the antitumor inhibitory potential of regorafenib on HCC cells. Regorafenib suppressed proliferation in HuH-7 cell and induced G0/G1 cell cycle arrest and cyclin D1 downregulation in regorafenib-sensitive cells. During miRNA analysis, miRNA molecules associated with the antitumor effect of regorafenib were found. Regorafenib suppresses cell proliferation and tumor growth in HCC by decreasing cyclin D1 via alterations in intracellular and exosomal miRNAs in HCC.


Subject(s)
Carcinoma, Hepatocellular/drug therapy , Gene Expression Regulation, Neoplastic/drug effects , Liver Neoplasms/drug therapy , MicroRNAs/biosynthesis , Phenylurea Compounds/pharmacology , Pyridines/pharmacology , RNA, Neoplasm/biosynthesis , Carcinoma, Hepatocellular/genetics , Carcinoma, Hepatocellular/metabolism , G1 Phase Cell Cycle Checkpoints/drug effects , G1 Phase Cell Cycle Checkpoints/genetics , Hep G2 Cells , Humans , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , MicroRNAs/genetics , RNA, Neoplasm/genetics , Resting Phase, Cell Cycle/drug effects , Resting Phase, Cell Cycle/genetics
20.
Biomed Res Int ; 2022: 1987519, 2022.
Article in English | MEDLINE | ID: mdl-35059460

ABSTRACT

Radioresistance of breast cancer is a major reason for therapeutic failure and limits further increases in the dose of radiation due to severe adverse effects. Recently, long noncoding RNAs (lncRNAs) have been shown to regulate cancer proliferation, chemoresistance, and radioresistance. Among these lncRNAs, lncRNA GAS5 expression was shown to be downregulated in breast cancer and related to trastuzumab resistance. However, its role in the radiation response is unclear. In this study, we demonstrated that lncRNA GAS5 expression was reduced in irradiated cells and that overexpression of GAS5 reduced cell viability and promoted cell apoptosis after irradiation. Moreover, overexpression of GAS5 resulted in increased G2/M arrest and unrepaired DNA damage, indicating a radiosensitizing role of GAS5 in breast cancer cells. Finally, we found that a GAS5-interacting miRNA, miR-21, reversed the radiosensitizing effects of GAS5 by inhibiting the apoptotic pathway. In conclusion, we found that lncRNA GAS5 sensitized breast cancer cells to ionizing radiation by inhibiting DNA repair and suppressing miR-21, identifying novel targets for breast cancer radiosensitization.


Subject(s)
Breast Neoplasms/metabolism , DNA Repair , DNA, Neoplasm/metabolism , RNA, Long Noncoding/metabolism , RNA, Neoplasm/metabolism , X-Rays , Breast Neoplasms/genetics , DNA, Neoplasm/genetics , Female , Humans , MCF-7 Cells , RNA, Long Noncoding/genetics , RNA, Neoplasm/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...