Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Mol Cancer ; 23(1): 128, 2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38890620

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) play important roles in cancer progression and metastasis. However, the expression profiles and biological roles of circRNAs in non-small cell lung cancer (NSCLC) remain unclear. METHODS: In this study, we identified a novel circRNA, hsa_circ_0006834 (termed circ6834), in NSCLC by RNA-seq and investigated the biological role of circ6834 in NSCLC progression in vitro and in vivo. Finally, the molecular mechanism of circ6834 was revealed by tagged RNA affinity purification (TRAP), western blot, RNA immunoprecipitation, dual luciferase reporter gene assays and rescue experiments. RESULTS: Our results showed that circ6834 was downregulated in NSCLC tumor tissues and cell lines. Circ6834 overexpression inhibited NSCLC cell growth and metastasis both in vitro and in vivo, while circ6834 knockdown had the opposite effect. We found that TGF-ß treatment decreased circ6834 expression, which was associated with the QKI reduction in NSCLC cells and circ6834 antagonized TGF-ß-induced EMT and metastasis in NSCLC cells. Mechanistically, circ6834 bound to AHNAK protein, a key regulator of TGF-ß/Smad signaling, and inhibited its stability by enhancing TRIM25-mediated ubiquitination and degradation. In addition, circ6834 acted as a miRNA sponge for miR-873-5p and upregulated TXNIP gene expression, which together inactivated the TGF-ß/Smad signaling pathway in NSCLC cells. CONCLUSION: In conclusion, circ6834 is a tumor-suppressive circRNA that inhibits NSCLC progression by forming a negative regulatory feedback loop with the TGF-ß/Smad signaling pathway and represents a novel therapeutic target for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Carrier Proteins , Cell Proliferation , Gene Expression Regulation, Neoplastic , Lung Neoplasms , MicroRNAs , RNA, Circular , Humans , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/metabolism , RNA, Circular/genetics , MicroRNAs/genetics , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Lung Neoplasms/metabolism , Animals , Mice , Cell Line, Tumor , Carrier Proteins/genetics , Carrier Proteins/metabolism , Disease Progression , Cell Movement/genetics , Signal Transduction , Female , Transforming Growth Factor beta/metabolism , Male , Epithelial-Mesenchymal Transition/genetics
2.
Adv Sci (Weinh) ; 11(24): e2309298, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38639382

ABSTRACT

M2-polarized tumor-associated macrophages (M2 TAMs) promote cancer progression. Exosomes mediate cellular communication in the tumor microenvironment (TME). However, the roles of exosomes from M2 TAMs in gastric cancer progression are unclear. Herein, it is reported that M2 TAMs-derived exosomes induced aerobic glycolysis in gastric cancer cells and enhanced their proliferation, metastasis, and chemoresistance in a glycolysis-dependent manner. It is identified that MALAT1 (metastasis-associated lung adenocarcinoma transcript 1) is enriched in M2 TAM exosomes and confirmed that MALAT1 transfer from M2 TAMs to gastric cancer cells via exosomes mediates this effect. Mechanistically, MALAT1 interacted with the δ-catenin protein and suppressed its ubiquitination and degradation by ß-TRCP. In addition, MALAT1 upregulated HIF-1α expression by acting as a sponge for miR-217-5p. The activation of ß-catenin and HIF-1α signaling pathways by M2 TAM exosomes collectively led to enhanced aerobic glycolysis in gastric cancer cells. Finally, a dual-targeted inhibition of MALAT1 in both gastric cancer cells and macrophages by exosome-mediated delivery of siRNA remarkably suppressed gastric cancer growth and improved chemosensitivity in mouse tumor models. Taken together, these results suggest that M2 TAMs-derived exosomes promote gastric cancer progression via MALAT1-mediated regulation of glycolysis. The findings offer a potential target for gastric cancer therapy.


Subject(s)
Disease Progression , Exosomes , Glycolysis , RNA, Long Noncoding , Stomach Neoplasms , Tumor-Associated Macrophages , Stomach Neoplasms/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/immunology , Exosomes/metabolism , Exosomes/genetics , Humans , Mice , Animals , Tumor Microenvironment/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , Cell Line, Tumor , Disease Models, Animal , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Cell Proliferation/genetics
3.
Article in English | MEDLINE | ID: mdl-38504413

ABSTRACT

Circular RNAs are a class of noncoding RNAs with covalently linked 5' and 3' ends that arise from backsplicing events. The absence of a 5' cap and a 3' poly(A) tail makes circular RNAs relatively more stable than their linear counterparts. They are evolutionary conserved and tissue-specific, and some show disease-specific expression patterns. Although their biological functions remain largely unknown, circular RNAs have been shown to play regulatory roles by acting as microRNA sponges, regulators of RNA-binding proteins, alternative splicing, and parental gene expression, and they could even encode proteins. Over the past few decades, circular RNAs have attracted wide attention in oncology owing to their implications in various tumors. Many circular RNAs have been characterized as key players in gastrointestinal cancers and influence cancer growth, progression, metastasis, and therapeutic resistance. Accumulating evidence reveals that their unique characteristics, coupled with their critical roles in tumorigenesis, make circular RNAs promising non-invasive clinical biomarkers for gastrointestinal cancers. In the present review, we summarized the biological roles of the emerging circular RNAs and their potential as biomarkers and therapeutic targets, which may help better understand their clinical significance in the management of gastrointestinal cancers.

4.
Gene ; 910: 148331, 2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38438055

ABSTRACT

Compelling evidence has identified circRNAs as crucial regulators in initiation and progression of various cancers, including gastric cancer (GC). However, the function and regulatory mechanisms of circRNAs in GC remain largely unknown. In this study, attention is paid to a novel circular RNA circ1811, which exerts significant downregulated expression in GC tissues compared with adjacent non-cancerous tissues. The expression of circ1811 in GC tumor tissues is negatively correlated with the extent of lymphatic metastasis in GC patients. Overexpression of circ1811 inhibited GC cell proliferation, migration and invasion while promoting apoptosis, whereas knockdown of circ1811 led to the opposite effects. AGO2 RIP and dual luciferase reporter assays indicated that circ1811 directly sponges miR-632 to upregulate the expression of DAPK1. Collectively, circ1811 acts as a tumor-suppressor for GC progression by regulating the miR-632/DAPK1 axis. Our findings suggest the potential of circ1811 as ideal biomarker and therapeutic target for GC.


Subject(s)
MicroRNAs , Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , Lymphatic Metastasis , Cell Proliferation/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Death-Associated Protein Kinases/genetics , Death-Associated Protein Kinases/metabolism
5.
Adv Mater ; 36(19): e2310318, 2024 May.
Article in English | MEDLINE | ID: mdl-38320755

ABSTRACT

Neutrophils are the most abundant white blood cells in the circulation and act as the first line of defense against infections. Increasing evidence suggests that neutrophils possess heterogeneous phenotypes and functional plasticity in human health and diseases, including cancer. Neutrophils play multifaceted roles in cancer development and progression, and an N1/N2 paradigm of neutrophils in cancer is proposed, where N1 neutrophils exert anti-tumor properties while N2 neutrophils display tumor-supportive and immune-suppressive functions. Selective activation of beneficial neutrophil population and targeted inhibition or re-polarization of tumor-promoting neutrophils has shown an important potential in tumor therapy. In addition, due to the natural inflammation-responsive and physical barrier-crossing abilities, neutrophils and their derivatives (membranes and extracellular vesicles (EVs)) are regarded as advanced drug delivery carriers for enhanced tumor targeting and improved therapeutic efficacy. In this review, the recent advances in engineering neutrophils for drug delivery and targeting neutrophils for remodeling tumor microenvironment (TME) are comprehensively presented. This review will provide a broad understanding of the potential of neutrophils in cancer therapy.


Subject(s)
Neoplasms , Neutrophils , Tumor Microenvironment , Humans , Neutrophils/metabolism , Neoplasms/drug therapy , Neoplasms/therapy , Animals , Tumor Microenvironment/drug effects , Drug Delivery Systems , Antineoplastic Agents/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Extracellular Vesicles/metabolism , Drug Carriers/chemistry
6.
Clin Chim Acta ; 554: 117773, 2024 Feb 01.
Article in English | MEDLINE | ID: mdl-38199579

ABSTRACT

BACKGROUND: Gastric cancer (GC) is the third leading cause of cancer-related death worldwide. Sensitive and accurate biomarkers can greatly aid in early diagnosis and favorable prognosis. Neutrophils are the most abundant immune cells in human circulation and play a critical role in tumor progression. Neutrophil-derived exosomes (Neu-Exo) contain abundant bioactive molecules and are critically involved in disease progression. METHODS: We proposed a Dynabeads-based (CD66b antibody-coupled) separation and detection system for Neu-Exo analysis. Dual antibody-assisted fluorescent Dynabeads was established to detect Neu-Exo abundance. MiRNA signature of Neu-Exo was identified by RNA sequencing. QRT-PCR and droplet digital PCR (ddPCR) were used for candidate miRNA detection and the potential of Neu-Exo miRNAs in the diagnosis of gastric cancer was evaluated. RESULTS: Dual antibody-assisted fluorescent Dynabeads obtained a detection limit of 7.8 × 105 particles/mL of Neu-Exo and a recovery rate of 81 % under optimized conditions. ROC curve indicated that the abundance of CD66b+ Neu-Exo could well distinguish GC patients from healthy controls (HC) (AUC > 0.8). Additionally, miR-223-3p was found among the top differentially expressed miRNAs in Neu-Exo and presented superior diagnostic value in gastric cancer. Droplet digital PCR (ddPCR) significantly improved the diagnostic efficiency to differentiate GC patients from HC and benign gastric diseases (BGD) patients (AUC > 0.9). CONCLUSION: The Dynabeads-based separation and detection system, assisted with ddPCR analysis, provides a promising platform to enrich Neu-Exo and analyze miRNA profile for gastric cancer liquid biopsy.


Subject(s)
Exosomes , MicroRNAs , Stomach Neoplasms , Humans , MicroRNAs/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Neutrophils/pathology , Biomarkers, Tumor/genetics , Polymerase Chain Reaction , Exosomes/genetics , Exosomes/pathology
7.
Mol Cancer ; 22(1): 122, 2023 08 04.
Article in English | MEDLINE | ID: mdl-37537569

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide. Long non-coding RNAs (lncRNAs) have emerged as key regulators of cancer development and progression, and as promising biomarkers for the diagnosis and prognosis of cancer. In this study, we identified a new lncRNA (LINC02159) that was upregulated in the tumor tissues and serum of non-small cell lung cancer (NSCLC) patients. We demonstrated that knockdown of LINC02159 inhibited NSCLC cell proliferation, migration, and invasion, but induced cell apoptosis and cell cycle arrest in vitro and retarded tumor growth in vivo, while overexpression of LINC02159 led to the opposite effect. We discovered that LINC02159 was highly correlated with cancer growth and metastasis-related pathways by using transcriptomic analysis and that YAP1 was a potential target gene of LINC02159. Mechanistically, LINC02159 bound to the Aly/REF export factor (ALYREF) to enhance the stability of YAP1 messenger RNA (mRNA) via m5C modification, which led to the overexpression of YAP1 and the activation of the Hippo and ß-catenin signaling pathways in NSCLC cells. Rescue experiments showed that LINC01259 promoted NSCLC progression in a YAP1- and ALYREF-dependent manner. In conclusion, LINC02159 plays an oncogenic role in NSCLC progression by regulating ALYREF/YAP1 signaling, and it has the potential to be utilized as a diagnostic marker and therapeutic target for NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , RNA, Long Noncoding , Humans , Carcinoma, Non-Small-Cell Lung/pathology , Lung Neoplasms/pathology , Cell Line, Tumor , Signal Transduction , RNA, Long Noncoding/metabolism , Cell Proliferation/genetics , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Nuclear Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , RNA-Binding Proteins/metabolism
8.
J Cancer ; 14(10): 1736-1750, 2023.
Article in English | MEDLINE | ID: mdl-37476194

ABSTRACT

Exosomes are a typical subset of extracellular vesicles (EVs) that can be transmitted from parent cells to recipient cells via human bodily fluids. Exosomes perform a vital role in mediating intercellular communication by shuttling bioactive cargos, such as nucleic acids, proteins and lipids. Long noncoding RNAs (lncRNAs) are transcripts longer than 200 nucleotides without protein translation ability and can be selectively packaged into exosomes. Accumulating evidence indicates that exosomal lncRNAs have a critical role in tumor initiation and progression through regulating tumor proliferation, apoptosis, invasion, metastasis, angiogenesis, treatment resistance and tumor microenvironment. Increasing studies suggest that exosomal lncRNAs have great potential to be served as novel targets and non-invasive biomarkers for diagnosis and prognosis in non-small cell lung cancer (NSCLC). In this review, we provide an overview of current research on the disordered functions of exosomal lncRNAs in NSCLC and summarize their potential clinical applications as diagnostic and prognostic biomarkers and therapeutic targets for NSCLC.

9.
Cancer Cell Int ; 23(1): 149, 2023 Jul 31.
Article in English | MEDLINE | ID: mdl-37525212

ABSTRACT

BACKGROUND: Spalt-like protein 4 (SALL4) is a stemness-related transcription factor whose abnormal re-expression contributes to cancer initiation and progression. However, the role of SALL4 in cancer angiogenesis remains unknown. METHODS: Analyses of clinical specimens via TCGA datasets were performed to determine the expression level and clinical significance of SALL4 in STAD (Stomach Adenocarcinoma). SALL4 knockdown, knockout, and overexpression were achieved by siRNA, CRISPR/Cas9, and plasmid transfection. The effects of conditioned medium (CM) from SALL4 knockdown or overexpression of gastric cancer cells on endothelial cell proliferation, migration, and tube formation were investigated by CCK-8 assay, transwell migration assay, and tube formation assay. The regulation of VEGF gene expression by SALL4 was studied by qRT-PCR, western blot, chromatin immunoprecipitation (ChIP) assay, and electrophoretic mobility shift assay (EMSA). Engineered exosomes from 293T cells loaded with si-SALL4-B and thalidomide were produced to test their therapeutic effect on gastric cancer progression. RESULTS: SALL4 expression was increased in STAD and positively correlated with tumor progression and poor prognosis. SALL4-B knockdown or knockout decreased while over-expression increased the promotion of human umbilical vein endothelial cells (HUVEC) cell proliferation, migration, and tube formation by gastric cancer cell-derived CM. Further investigation revealed a widespread association of SALL4 with angiogenic gene transcription through the TCGA datasets. Additionally, SALL4-B knockdown reduced, while over-expression enhanced the expression levels of VEGF-A, B, and C genes. The results of ChIP and EMSA assays indicated that SALL4 could directly bind to the promoters of VEGF-A, B, and C genes and activate their transcription, which may be associated with increased histone H3-K79 and H3-K4 modifications in their promoter regions. Furthermore, si-SALL4-B and thalidomide-loaded exosomes could be efficiently uptaken by gastric cancer cells and significantly reduced SALL4-B and Vascular Endothelial Growth Factor (VEGF) expression levels in gastric cancer cells, thus inhibiting the pro-angiogenic role of their derived CM. CONCLUSION: These findings suggest that SALL4 plays an important role in angiogenesis by transcriptionally regulating VEGF expression. Co-delivery of the functional siRNA and anticancer drug via exosomes represents a useful approach to inhibiting cancer angiogenesis by targeting SALL4/VEGF pathway.

10.
J Clin Lab Anal ; 37(9-10): e24927, 2023 May.
Article in English | MEDLINE | ID: mdl-37337914

ABSTRACT

SALL4 (split-like protein 4) is a member of the mammalian homologs of the Drosophila homoeotic gene spalt (sal) and acts as a zinc finger transcription factor to govern the self-renewal and pluripotency of embryonic stem cells. SALL4 expression gradually decreases during development and is even absent in most adult tissues. However, increasing evidence suggests that SALL4 expression is restored in human cancers and its aberrant expression is associated with the progression of many hematopoietic malignancies and solid tumors. The potent roles of SALL4 in regulating cancer cell proliferation, apoptosis, metastasis, and drug resistance have been reported. SALL4 plays a dual role in epigenetic modulation by acting as either an activator or a repressor of its target genes. Furthermore, SALL4 interacts with other partners to control the expression of many downstream genes and the activation of various key signaling transduction pathways. SALL4 is considered as a promising diagnostic and prognostic biomarker and therapeutic target for cancer. In this review, we highlighted the major advances in the roles and mechanisms of SALL4 in cancer and the therapeutic strategies for targeting SALL4 to treat cancer.


Subject(s)
Neoplasms , Transcription Factors , Adult , Animals , Humans , Transcription Factors/genetics , Transcription Factors/metabolism , Neoplasms/genetics , Gene Expression Regulation , Cell Proliferation , Mammals/metabolism
11.
Mol Cancer ; 22(1): 30, 2023 02 13.
Article in English | MEDLINE | ID: mdl-36782290

ABSTRACT

tRNA-derived fragments (tRFs) are an emerging category of small non-coding RNAs that are generated from cleavage of mature tRNAs or tRNA precursors. The advance in high-throughput sequencing has contributed to the identification of increasing number of tRFs with critical functions in distinct physiological and pathophysiological processes. tRFs can regulate cell viability, differentiation, and homeostasis through multiple mechanisms and are thus considered as critical regulators of human diseases including cancer. In addition, increasing evidence suggest the extracellular tRFs may be utilized as promising diagnostic and prognostic biomarkers for cancer liquid biopsy. In this review, we focus on the biogenesis, classification and modification of tRFs, and summarize the multifaceted functions of tRFs with an emphasis on the current research status and perspectives of tRFs in cancer.


Subject(s)
Neoplasms , RNA, Transfer , Humans , RNA, Transfer/genetics , Neoplasms/genetics , Neoplasms/pathology
12.
Front Physiol ; 13: 988773, 2022.
Article in English | MEDLINE | ID: mdl-36160866

ABSTRACT

Background: Aerobic exercise could produce a positive effect on the brain by releasing brain-derived neurotrophic factor (BDNF). In untrained healthy humans there seems to be a linear correlation between exercise duration and the positive effect of acute aerobic exercise on brain-derived neurotrophic factor levels. Therefore, we performed two different duration of high-intensity interval training protocols (HIIT), both known to improve cardiovascular fitness, to determine whether then have a similar efficacy in affecting brain-derived neurotrophic factor levels. Methods: 12 untrained young males (aged 23.7 ± 1.8 years), participated in a randomized controlled cross-over trial. They underwent two different work-to-rest ratio high-intensity interval training protocols: high-intensity interval training 1 (30 min, 15 intervals of 1 min efforts at 85%-90% VO2max with 1 min of active recovery at 50%-60% VO2max) and HIIT2 (30 min, 10 intervals of 2 min efforts at 85%-90% VO2max with 1 min of active recovery at 50%-60% VO2max). Serum cortisol, brain-derived neurotrophic factor were collected at baseline, immediately following intervention, and 30 min into recovery for measurements using a Sandwich ELISA method, blood lactate was measured by using a portable lactate analyzer. Results: Our results showed that the similar serum brain-derived neurotrophic factor change in both high-intensity interval training protocols, with maximal serum brain-derived neurotrophic factor levels being reached toward the end of intervention. There was no significant change in serum brain-derived neurotrophic factor from baseline after 30 min recovery. We then showed that both high-intensity interval training protocols significantly increase blood lactate and serum cortisol compared with baseline value (high-intensity interval training p < 0.01; high-intensity interval training 2 p < 0.01), with high-intensity interval training 2 reaching higher blood lactate levels than high-intensity interval training 1 (p = 0.027), but no difference was observed in serum cortisol between both protocols. Moreover, changes in serum brain-derived neurotrophic factor did corelate with change in blood lactate (high-intensity interval training 1 r = 0.577, p < 0.05; high-intensity interval training 2 r = 0.635, p < 0.05), but did not correlate with the change in serum cortisol. Conclusions: brain-derived neurotrophic factor levels in untrained young men are significantly increased in response to different work-to-rest ratio of high-intensity interval training protocols, and the magnitude of increase is exercise duration independent. Moreover, the higher blood lactate did not raise circulating brain-derived neurotrophic factor. Therefore, given that prolonged exercise causes higher levels of cortisol. We suggest that the 1:1work-to-rest ratio of high-intensity interval training protocol might represent a preferred intervention for promoting brain health.

13.
Mol Cancer ; 21(1): 141, 2022 07 02.
Article in English | MEDLINE | ID: mdl-35780119

ABSTRACT

BACKGROUND: Increasing studies suggest that circular RNAs (circRNAs) are critical regulators of cancer development and progression. However, the biological roles and mechanisms of circRNAs in gastric cancer (GC) remain largely unknown. METHODS: We identified the differentially expressed circRNAs in GC by analyzing Gene Expression Omnibus (GEO) datasets. We explored the biological roles of circRNAs in GC by in vitro functional assays and in vivo animal studies. We performed tagged RNA affinity purification (TRAP), RNA immunoprecipitation (RIP), mass spectrometry (MS), RNA sequencing, luciferase reporter assays, and rescue experiments to investigate the mechanism of circRNAs in GC. RESULTS: Downregulated expression of circular RNA EIF4G3 (circEIF4G3; hsa_circ_0007991) was found in GC and was associated with poor clinical outcomes. Overexpression of circEIF4G3 suppressed GC growth and metastasis through the inhibition of ß-catenin signaling, whereas knockdown of circEIF4G3 showed the opposite effects. Mechanistic studies revealed that circEIF4G3 bound to δ-catenin protein to promote its TRIM25-mediated ubiquitin degradation and interacted with miR-4449 to upregulate SIK1 expression. CONCLUSION: Our findings uncovered a tumor suppressor function of circEIF4G3 in GC through the regulation of δ-catenin protein stability and miR-4449/SIK1 axis. CircEIF4G3 may act as a promising prognostic biomarker and therapeutic target for GC.


Subject(s)
MicroRNAs , Stomach Neoplasms , Animals , Catenins , Cell Line, Tumor , Cell Proliferation/genetics , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , Stomach Neoplasms/pathology , Ubiquitin , beta Catenin/genetics , Delta Catenin
14.
Front Cell Dev Biol ; 10: 862524, 2022.
Article in English | MEDLINE | ID: mdl-35300408

ABSTRACT

Bone marrow-derived mesenchymal stem cell (BMSC) is one crucial component of the multiple myeloma (MM) microenvironment and supports the malignant progression of MM. Whether BMSCs act on MM cells via exosomes has not been well characterized. Herein, we used microarrays to screen out differentially expressed miRNAs in BMSCs from patients with MM (MM-MSCs) or benign diseases (BD-MSCs). We found that miR-483-5p was highly expressed in MM-MSCs, which may be transported through exosomes from MM-MSCs to MM cells to increase miR-483-5p expression in them. We then investigated the role and mechanism of miR-483-5p in the aggressive progression of MM in vitro. We verified that miR-483-5p promoted MM cell proliferation and reduced apoptosis. Then we predicted and validated that TIMP2, a tumor suppressor gene, is the downstream target of miR-483-5p in MM. In summary, our study indicated that MM-MSCs promote MM malignant progression via the release of exosomes and regulation of miR-483-5p/TIMP2 axis, suggesting an essential role of BMSCs derived exosomal miRNA in MM and a potential marker for MM diagnosis and therapy.

15.
Mol Cancer ; 21(1): 56, 2022 02 18.
Article in English | MEDLINE | ID: mdl-35180868

ABSTRACT

Liquid biopsy, characterized by minimally invasive detection through biofluids such as blood, saliva, and urine, has emerged as a revolutionary strategy for cancer diagnosis and prognosis prediction. Exosomes are a subset of extracellular vesicles (EVs) that shuttle molecular cargoes from donor cells to recipient cells and play a crucial role in mediating intercellular communication. Increasing studies suggest that exosomes have a great promise to serve as novel biomarkers in liquid biopsy, since large quantities of exosomes are enriched in body fluids and are involved in numerous physiological and pathological processes. However, the further clinical application of exosomes has been greatly restrained by the lack of high-quality separation and component analysis methods. This review aims to provide a comprehensive overview on the conventional and novel technologies for exosome isolation, characterization and content detection. Additionally, the roles of exosomes serving as potential biomarkers in liquid biopsy for the diagnosis, treatment monitoring, and prognosis prediction of cancer are summarized. Finally, the prospects and challenges of applying exosome-based liquid biopsy to precision medicine are evaluated.


Subject(s)
Exosomes , Neoplasms , Biomarkers, Tumor/analysis , Exosomes/pathology , Humans , Liquid Biopsy/methods , Neoplasms/diagnosis , Neoplasms/pathology , Precision Medicine , Prognosis
16.
Front Cell Dev Biol ; 9: 753706, 2021.
Article in English | MEDLINE | ID: mdl-34722539

ABSTRACT

Long non-coding RNA (lncRNA) DANCR (also known as ANCR)-differentiation antagonizing non-protein coding RNA, was first reported in 2012 to suppress differentiation of epithelial cells. Emerging evidence demonstrates that DANCR is a cancer-associated lncRNA abnormally expressed in many cancers (e.g., lung cancer, gastric cancer, breast cancer, hepatocellular carcinoma). Increasing studies suggest that the dysregulation of DANCR plays critical roles in cancer cell proliferation, apoptosis, migration, invasion, and chemoresistance in vitro and tumor growth and metastasis in vivo. Mechanistic analyses show that DANCR can serve as miRNA sponges, stabilize mRNAs, and interact with proteins. Recent research reveals that DANCR can be detected in many body fluids such as serum, plasma, and exosomes, providing a quick and convenient method for cancer monitor. Thus DANCR can be used as a promising diagnostic and prognostic biomarker and therapeutic target for various types of cancer. This review focuses on the role and mechanism of DANCR in cancer progression with an emphasis on the clinical significance of DANCR in human cancers.

17.
Mol Cancer ; 20(1): 101, 2021 08 12.
Article in English | MEDLINE | ID: mdl-34384442

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) play important roles in cancer development and progression. The purpose of this study is to identify aberrantly expressed circRNAs in gastric cancer (GC), unravel their roles in GC progression, and provide new targets for GC diagnosis and therapy. METHODS: Bioinformatic analyses were performed to identify the aberrantly expression of hsa_circ_0061137 (termed as circDIDO1) in GC. Gain- and loss-of-function studies were performed to examine the biological roles of circDIDO1 in GC progression. Tagged RNA affinity purification, mass spectrometry, immunofluorescence, co-immunoprecipitation, and Western blot were used to identify circRNA-interacting and circRNA-encoded proteins. RNA sequencing, qRT-PCR, and Western blot were performed to analyze circRNA-regulated downstream target genes and signaling pathways. Mouse tumor models were used to analyze the effects of circDIDO1 on GC growth and metastasis. RESULTS: CircDIDO1 was transcribed from human DIDO1 (death-inducer obliterator 1) gene and formed by back-splicing of exons 2-6 of the linear transcript. circDIDO1 was down-regulated in GC tissues and its low levels were associated with larger tumor size, distal metastasis, and poor prognosis. CircDIDO1 overexpression inhibited while knockdown promoted GC cell proliferation, migration and invasion. CircDIDO1 overexpression suppressed GC growth and metastasis in mouse tumor models. Mechanistically, circDIDO1 encoded a novel 529aa protein that directly interacted with poly ADP-ribose polymerase 1 (PARP1) and inhibited its activity. CircDIDO1 also specifically bound to peroxiredoxin 2 (PRDX2) and promoted RBX1-mediated ubiquitination and degradation of PRDX2, which led to the inactivation of its downstream signaling pathways. CONCLUSIONS: CircDIDO1 is a new circRNA that has tumor suppressor function in GC and it may serve as a potential prognostic biomarker and therapeutic target for GC.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Neoplastic , Peroxiredoxins/metabolism , RNA, Circular/genetics , Stomach Neoplasms/metabolism , Animals , Cell Line, Tumor , DNA-Binding Proteins/chemistry , Disease Models, Animal , Gene Expression Profiling , Genes, Tumor Suppressor , Heterografts , Humans , Immunohistochemistry , Mice , Models, Biological , Peroxiredoxins/chemistry , Poly (ADP-Ribose) Polymerase-1/metabolism , Protein Binding , Protein Stability , Proteomics/methods , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
18.
J Clin Lab Anal ; 34(10): e23433, 2020 Oct.
Article in English | MEDLINE | ID: mdl-32608539

ABSTRACT

BACKGROUND: Increasing evidence indicates that circular RNAs (circRNAs) are dysregulated in human cancers. The biological roles of circRNAs in gastric cancer (GC) have not been well-characterized. METHODS: The GEO database was used to analyze circRNA expression profile in GC. The expression level of target circRNA in tumor tissues and adjacent non-tumor tissues was detected by reverse transcription-quantitative PCR. Gene transfection was used to manipulate the expression of circRNAs. The biological roles of circRNAs in cell proliferation, migration, and invasion were determined by cell counting, colony formation, transwell migration, Matrigel invasion, and mouse xenograft tumor assays. The interactions between circRNAs and miRNAs were verified by RNA immunoprecipitation and luciferase reporter assays. RESULTS: We found that circHN1 was upregulated in GC tissues and cell lines compared to adjacent non-tumor tissues and normal gastric epithelial cells. Additionally, circHN1 silencing significantly promoted GC cell growth, colony formation, migration, and invasion, whereas circHN1 overexpression had the opposite effects. CircHN1 overexpression also suppressed gastric cancer growth in the mouse xenograft tumor model. CircHN1 was mainly localized in the cytoplasm of GC cells and could bind to AGO2. MiR-1248 and miR-375 were predicted to interact with circHN1 by bioinformatic analyses. MiR-1248 and miR-375 overexpression inhibited the activity of the circHN1 luciferase reporter. CONCLUSION: CircHN1 is aberrantly expressed in GC and affects the proliferation and migration of gastric cancer cells by acting as miRNA sponge.


Subject(s)
Cell Movement/genetics , RNA, Circular/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Animals , Base Sequence , Cell Line, Tumor , Cell Proliferation/genetics , Exons/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice, Inbred BALB C , MicroRNAs/genetics , MicroRNAs/metabolism , Middle Aged , Neoplasm Invasiveness , RNA Splicing/genetics , RNA, Circular/genetics , Up-Regulation/genetics
19.
Hum Cell ; 33(3): 652-662, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32350750

ABSTRACT

The tumor microenvironment (TM) is an essential factor of tumor progression. Mesenchymal stem cells (MSCs) are important components of the TM and play critical roles in cancer metastasis. Resveratrol (RES) is a potential antitumor drug that has attracted extensive attention. However, it remains unclear whether RES can exert its antitumor activity by targeting MSCs located in the TM. In this study, we demonstrated that the conditioned medium of gastric-cancer-derived MSCs (GC-MSCs) promoted gastric cancer (GC) metastasis and facilitated the progression of epithelialmesenchymal transition (EMT) of GC cells. However, after pretreatment with RES, the prometastatic effect of GC-MSCs on GC cells was reversed. Furthermore, RES reduced GC-MSC (IL-6, IL-8, MCP-1, VEGF) gene expression and protein secretion, and counteracted the activation of the GC-MSC-induced Wnt/ß-catenin signaling of GC cells, with less ß-catenin nuclear transport and declined expression of ß-catenin, CD44, and CyclinD3 in GC cells. Re-expression of ß-catenin impaired the inhibitory effect of RES on GC cells. In conclusion, RES restricted the mobility increase of GC cells and reversed the progress of EMT induced by GC-MSCs by inactivating the Wnt/ß-catenin signaling. GC-MSCs are promising target for RES in the inhibition of GC metastasis.


Subject(s)
Mesenchymal Stem Cells/physiology , Neoplasm Metastasis/drug therapy , Resveratrol/therapeutic use , Stomach Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic , Cell Line, Tumor , Chemokine CCL2/genetics , Chemokine CCL2/metabolism , Gene Expression/drug effects , Humans , Interleukin-6/genetics , Interleukin-6/metabolism , Interleukin-8/genetics , Interleukin-8/metabolism , Mesenchymal Stem Cells/pathology , Molecular Targeted Therapy , Phytotherapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Tumor Microenvironment , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...