Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 45
Filter
1.
J Exp Clin Cancer Res ; 43(1): 181, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38937855

ABSTRACT

BACKGROUND: This study aimed to develop a novel six-gene expression biomarker panel to enhance the early detection and risk stratification of peritoneal recurrence and micrometastasis in locally advanced gastric cancer (LAGC). METHODS: We used genome-wide transcriptome profiling and rigorous bioinformatics to identify a six-gene expression biomarker panel. This panel was validated across multiple clinical cohorts using both tissue and liquid biopsy samples to predict peritoneal recurrence and micrometastasis in patients with LAGC. RESULTS: Through genome-wide expression profiling, we identified six mRNAs and developed a risk prediction model using 196 samples from a surgical specimen training cohort. This model, incorporating a 6-mRNA panel with clinical features, demonstrated high predictive accuracy for peritoneal recurrence in gastric cancer patients, with an AUC of 0.966 (95% CI: 0.944-0.988). Transitioning from invasive surgical or endoscopic biopsy to noninvasive liquid biopsy, the model retained its predictive efficacy (AUC = 0.963; 95% CI: 0.926-1.000). Additionally, the 6-mRNA panel effectively differentiated patients with or without peritoneal metastasis in 95 peripheral blood specimens (AUC = 0.970; 95% CI: 0.936-1.000) and identified peritoneal micrometastases with a high efficiency (AUC = 0.941; 95% CI: 0.874-1.000). CONCLUSIONS: Our study provides a novel gene expression biomarker panel that significantly enhances early detection of peritoneal recurrence and micrometastasis in patients with LAGC. The RSA model's predictive capability offers a promising tool for tailored treatment strategies, underscoring the importance of integrating molecular biomarkers with clinical parameters in precision oncology.


Subject(s)
Biomarkers, Tumor , Gene Expression Profiling , Neoplasm Micrometastasis , Neoplasm Recurrence, Local , Peritoneal Neoplasms , Stomach Neoplasms , Humans , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Liquid Biopsy/methods , Female , Neoplasm Micrometastasis/genetics , Male , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/genetics , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Biomarkers, Tumor/genetics , Gene Expression Profiling/methods , Middle Aged , Transcriptome , Aged
2.
Mol Carcinog ; 63(7): 1378-1391, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38656643

ABSTRACT

Accumulating evidence suggests that lymphangiogenesis plays a crucial role in lymphatic metastasis, leading to tumor immune tolerance. However, the specific mechanism remains unclear. In this study, miR-431-5p was markedly downregulated in both gastric cancer (GC) tissues and plasma exosomes, and its expression were correlated negatively with LN metastasis and poor prognosis. Mechanistically, miR-431-5p weakens the TGF-ß1/SMAD2/3 signaling pathway by targeting ZEB1, thereby suppressing the secretion of VEGF-A and ANG2, which in turn hinders angiogenesis, lymphangiogenesis, and lymph node (LN) metastasis in GC. Experiments using a popliteal LN metastasis model in BALB/c nude mice demonstrated that miR-431-5p significantly reduced popliteal LN metastasis. Additionally, miR-431-5p enhances the efficacy of anti-PD1 treatment, particularly when combined with galunisertib, anti-PD1 treatment showing a synergistic effect in inhibiting GC progression in C57BL/6 mice. Collectively, these findings suggest that miR-431-5p may modulate the TGF-ß1/SMAD2/3 pathways by targeting ZEB1 to impede GC progression, angiogenesis, and lymphangiogenesis, making it a promising therapeutic target for GC management.


Subject(s)
Gene Expression Regulation, Neoplastic , Lymphangiogenesis , Lymphatic Metastasis , Mice, Inbred BALB C , MicroRNAs , Neovascularization, Pathologic , Signal Transduction , Smad2 Protein , Smad3 Protein , Stomach Neoplasms , Transforming Growth Factor beta1 , Zinc Finger E-box-Binding Homeobox 1 , Stomach Neoplasms/pathology , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Humans , Animals , MicroRNAs/genetics , Lymphangiogenesis/genetics , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/metabolism , Transforming Growth Factor beta1/metabolism , Transforming Growth Factor beta1/genetics , Zinc Finger E-box-Binding Homeobox 1/genetics , Zinc Finger E-box-Binding Homeobox 1/metabolism , Smad3 Protein/metabolism , Smad3 Protein/genetics , Smad2 Protein/metabolism , Smad2 Protein/genetics , Mice, Nude , Male , Female , Cell Line, Tumor , Mice, Inbred C57BL , Prognosis , Middle Aged , Angiogenesis
3.
Eur J Clin Invest ; 54(8): e14201, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38533747

ABSTRACT

BACKGROUND: Robotic gastrectomy is increasingly utilized for gastric cancer, but high morbidity remains a concern. Myosteatosis or low skeletal muscle density reflecting fatty infiltration, associates with complications after other cancer surgeries but has not been evaluated for robotic gastrectomy. METHODS: This retrospective study analysed 381 patients undergoing robotic gastrectomy for gastric cancer from September 2019 to October 2022. Myosteatosis was quantified on preoperative computed tomography (CT) images at lumbar 3 (L3). Propensity score matching addressed potential confounding between myosteatosis and non-myosteatosis groups. Outcomes were postoperative complications, 30 days mortality, 30 days readmissions and survival. RESULTS: Myosteatosis was present in 33.6% of patients. Myosteatosis associated with increased overall (47.7% vs. 26.5%, p < 0.001) and severe complications (12.4% vs. 4.9%, p < 0.001). After matching, myosteatosis remained associated with increased overall complications, major complications, intensive care unit (ICU) transfer and readmission (all p < 0.05). Myosteatosis independently predicted overall [odds ratio (OR) = 2.86, 95% confidence interval (CI): 1.57-5.20, p = 0.001] and severe complications (OR = 4.81, 95% CI: 1.51-15.27, p = 0.008). Myosteatosis also associated with reduced overall (85.0% vs. 93.2%, p = 0.015) and disease-free survival (80.3% vs. 88.4%, p=0.029). On multivariate analysis, myosteatosis independently predicted poorer survival [hazard ratio (HR) = 2.83, 95% CI: 1.32-6.08, p=0.012] and disease-free survival (HR = 1.83, 95% CI: 1.01-3.30, p=0.032). CONCLUSION: Preoperative CT-defined myosteatosis independently predicts increased postoperative complications and reduced long-term survival after robotic gastrectomy for gastric cancer. Assessing myosteatosis on staging CT could optimize preoperative risk stratification.


Subject(s)
Gastrectomy , Postoperative Complications , Propensity Score , Robotic Surgical Procedures , Stomach Neoplasms , Tomography, X-Ray Computed , Humans , Stomach Neoplasms/surgery , Stomach Neoplasms/mortality , Gastrectomy/adverse effects , Male , Female , Postoperative Complications/epidemiology , Middle Aged , Aged , Retrospective Studies , Patient Readmission/statistics & numerical data , Sarcopenia/diagnostic imaging , Muscle, Skeletal/diagnostic imaging
4.
Nutrition ; 123: 112408, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38513525

ABSTRACT

BACKGROUND: Sarcopenia, defined as decreased muscle mass and function, correlates with postoperative morbidity and mortality in cancer surgery. However, sarcopenia's impact specifically following robotic gastrectomy for gastric cancer has not been clearly defined. This study aimed to determine the influence of sarcopenia on short- and long-term clinical outcomes after robotic gastrectomy for gastric cancer. METHODS: This retrospective study analyzed 381 gastric cancer patients undergoing robotic gastrectomy. Sarcopenia was diagnosed by preoperative computed tomography (CT) body composition analysis. Propensity score matching created 147 pairs of sarcopenia and nonsarcopenia patients for comparison. Outcomes included postoperative complications, survival, inflammatory markers, length of stay, intensive care unit (ICU) transfer, and readmissions. RESULTS: Sarcopenia patients exhibited significantly higher rates of overall (53.7% versus 21.1%, P < 0.001), serious (12.9% versus 4.1%, P = 0.007), and grade III-IV complications compared to nonsarcopenia pairs after matching. Sarcopenia independently predicted reduced 3-years overall (HR = 2.53, 95% CI: 1.19-5.40, P = 0.016) and disease-free survival (HR = 1.99, 95% CI: 1.09-3.66, P = 0.026). Sarcopenia patients also showed heightened postoperative leukocyte, neutrophil, platelet, platelet to lymphocyte ratio (PLR), systemic immune-inflammation index (SII), and monocyte to lymphocyte ratio (MLR) levels alongside suppressed lymphocytes, monocytes, and neutrophil to lymphocyte ratio (NLR). CONCLUSION: Preoperative sarcopenia is correlated with increased postoperative complications and poorer long-term survival in gastric cancer patients undergoing robotic gastrectomy. Sarcopenia assessment can optimize preoperative risk stratification and perioperative management in this population.


Subject(s)
Gastrectomy , Postoperative Complications , Propensity Score , Robotic Surgical Procedures , Sarcopenia , Stomach Neoplasms , Humans , Sarcopenia/etiology , Stomach Neoplasms/surgery , Stomach Neoplasms/complications , Male , Female , Retrospective Studies , Gastrectomy/adverse effects , Gastrectomy/methods , Postoperative Complications/epidemiology , Postoperative Complications/etiology , Robotic Surgical Procedures/methods , Robotic Surgical Procedures/adverse effects , Aged , Middle Aged , Prognosis , Preoperative Period , Length of Stay/statistics & numerical data
5.
Cell Death Dis ; 15(1): 3, 2024 01 04.
Article in English | MEDLINE | ID: mdl-38177102

ABSTRACT

Current treatment strategies for cancer, especially advanced cancer, are limited and unsatisfactory. One of the most substantial advances in cancer therapy, in the last decades, was the discovery of a new layer of immunotherapy approach, immune checkpoint inhibitors (ICIs), which can specifically activate immune cells by targeting immune checkpoints. Immune checkpoints are a type of immunosuppressive molecules expressed on immune cells, which can regulate the degree of immune activation and avoid autoimmune responses. ICIs, such as anti-PD-1/PD-L1 drugs, has shown inspiring efficacy and broad applicability across various cancers. Unfortunately, not all cancer patients benefit remarkably from ICIs, and the overall response rates to ICIs remain relatively low for most cancer types. Moreover, the primary and acquired resistance to ICIs pose serious challenges to the clinical application of cancer immunotherapy. Thus, a deeper understanding of the molecular biological properties and regulatory mechanisms of immune checkpoints is urgently needed to improve clinical options for current therapies. Recently, circular RNAs (circRNAs) have attracted increasing attention, not only due to their involvement in various aspects of cancer hallmarks, but also for their impact on immune checkpoints in shaping the tumor immune microenvironment. In this review, we systematically summarize the current status of immune checkpoints in cancer and the existing regulatory roles of circRNAs on immune checkpoints. Meanwhile, we also aim to settle the issue in an evidence-oriented manner that circRNAs involved in cancer hallmarks regulate the effects and resistance of ICIs by targeting immune checkpoints.


Subject(s)
Neoplasms , RNA, Circular , Humans , RNA, Circular/genetics , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Immunotherapy , Tumor Microenvironment/genetics
6.
Stem Cell Rev Rep ; 20(1): 329-346, 2024 01.
Article in English | MEDLINE | ID: mdl-37889447

ABSTRACT

BACKGROUND: Although mesenchymal stem cells (MSCs) are used as therapeutic agents for skin injury therapy, few studies have reported the effects of dosing duration and delivery frequency on wound healing. In addition, before the clinical application of MSCs, it is important to assess whether their usage might influence tumor occurrence. METHODS: We described the metabolic patterns of subcutaneous injection of hUC-MSCs using fluorescence tracing and qPCR methods and applied them to the development of drug delivery strategies for promoting wound healing. RESULTS: (i) We developed cGMP-compliant hUC-MSC products with critical quality control points for wound healing; (ii) The products did not possess any tumorigenic or tumor-promoting/inhibiting ability in vivo; (iii) Fluorescence tracing and qPCR analyses showed that the subcutaneous application of hUC-MSCs did not result in safety-relevant biodistribution or ectopic migration; (iv) Reinjecting hUC-MSCs after significant consumption significantly improved reepithelialization and dermal regeneration. CONCLUSIONS: Our findings provided a reference for controlling the quality of MSC products used for wound healing and highlighted the importance of delivery time and frequency for designing in vivo therapeutic studies.


Subject(s)
Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Neoplasms , Humans , Tissue Distribution , Mesenchymal Stem Cell Transplantation/methods , Wound Healing , Mesenchymal Stem Cells/metabolism , Umbilical Cord/metabolism , Neoplasms/metabolism
7.
Eur J Med Res ; 28(1): 492, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37936161

ABSTRACT

Immune-checkpoint inhibitors (ICIs), different from traditional cancer treatment models, have shown unprecedented anti-tumor effects in the past decade, greatly improving the prognosis of many malignant tumors in clinical practice. At present, the most widely used ICIs in clinical immunotherapy for a variety of solid tumors are monoclonal antibodies against cytotoxic T lymphocyte antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1) and their ligand PD-L1. However, tumor patients may induce immune-related adverse events (irAEs) while performing immunotherapy, and irAE is an obstacle to the prospect of ICI treatment. IrAE is a non-specific disease caused by immune system imbalance, which can occur in many tissues and organs. For example, skin, gastrointestinal tract, endocrine system and lung. Although the exact mechanism is not completely clear, related studies have shown that irAE may develop through many ways. Such as excessive activation of autoreactive T cells, excessive release of inflammatory cytokines, elevated levels of autoantibodies, and common antigens between tumors and normal tissues. Considering that the occurrence of severe IrAE not only causes irreversible damage to the patient's body, but also terminates immunotherapy due to immune intolerance. Therefore, accurate identification and screening of sensitive markers of irAE are the main beneficiaries of ICI treatment. Additionally, irAEs usually require specific management, the most common of which are steroids and immunomodulatory therapies. This review aims to summarize the current biomarkers for predicting irAE in gastric cancer and their possible mechanisms.


Subject(s)
Antineoplastic Agents, Immunological , Stomach Neoplasms , Humans , Antineoplastic Agents, Immunological/adverse effects , Antibodies, Monoclonal/adverse effects , Cytokines , Autoantibodies
8.
Eur J Med Res ; 28(1): 405, 2023 Oct 07.
Article in English | MEDLINE | ID: mdl-37803421

ABSTRACT

Increased lymphangiogenesis and lymph node (LN) metastasis are thought to be important steps in cancer metastasis, and are associated with patient's poor prognosis. There is increasing evidence that the lymphatic system may play a crucial role in regulating tumor immune response and limiting tumor metastasis, since tumor lymphangiogenesis is more prominent in tumor metastasis and diffusion. Lymphangiogenesis takes place in embryonic development, wound healing, and a variety of pathological conditions, including tumors. Tumor cells and tumor microenvironment cells generate growth factors (such as lymphangiogenesis factor VEGF-C/D), which can promote lymphangiogenesis, thereby inducing the metastasis and diffusion of tumor cells. Nevertheless, the current research on lymphangiogenesis in gastric cancer is relatively scattered and lacks a comprehensive understanding. Therefore, in this review, we aim to provide a detailed perspective on molecules and signal transduction pathways that regulate gastric cancer lymphogenesis, which may provide new insights for the diagnosis and treatment of cancer.


Subject(s)
Lymphangiogenesis , Stomach Neoplasms , Humans , Lymphangiogenesis/physiology , Stomach Neoplasms/metabolism , Lymphatic Metastasis , Signal Transduction , Tumor Microenvironment
11.
Mol Oncol ; 17(7): 1437-1452, 2023 07.
Article in English | MEDLINE | ID: mdl-37014625

ABSTRACT

Previous studies have uncovered the key role of circular RNAs (circRNAs) in various diseases, including cancer. However, the growth-inhibitory effects of circRNAs on esophageal squamous cell carcinoma (ESCC) have not been completely elucidated. This study characterized a newly identified circRNA derived from exons 9-13 of TNRC6B (named circ-TNRC6B). The expression of circ-TNRC6B in ESCC tissues was markedly downregulated when compared to that in non-tumor tissues. In 53 ESCC cases, circ-TNRC6B expression was negatively correlated with the T stage. Multivariate Cox regression analysis showed that circ-TNRC6B upregulation was an independent protective factor for ESCC patients' prognosis. Overexpression and knockdown functional experiments demonstrated that circ-TNRC6B inhibited ESCC cell proliferation, migration, and invasion. RNA immunoprecipitation and dual-luciferase reporter assays demonstrated that circ-TNRC6B sponges oncogenic miR-452-5p to upregulate the expression and activity of DAG1. Treatment with miR-452-5p inhibitor partially reversed the circ-TNRC6B-induced changes in the biological behavior of ESCC cells. These findings demonstrated that circ-TNRC6B exerts a tumor-suppressing effect in ESCC through the miR-452-5p/DAG1 axis. Thus, circ-TNRC6B is a potential prognostic biomarker for the clinical management of ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , MicroRNAs , Humans , RNA, Circular/genetics , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Neoplasms/genetics , Cell Proliferation/genetics , MicroRNAs/genetics , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Cell Movement/genetics , Dystroglycans , RNA-Binding Proteins/genetics
12.
Front Nutr ; 10: 1037200, 2023.
Article in English | MEDLINE | ID: mdl-36819699

ABSTRACT

Skeletal muscle is the foundation of human function and plays a key role in producing exercise, bone protection, and energy metabolism. Sarcopenia is a systemic disease, which is characterized by degenerative changes in skeletal muscle mass, strength, and function. Therefore, sarcopenia often causes weakness, prolonged hospitalization, falls and other adverse consequences that reduce the quality of life, and even lead to death. In recent years, sarcopenia has become the focus of in-depth research. Researchers have suggested some molecular mechanisms for sarcopenia according to different muscle physiology. These mechanisms cover neuromuscular junction lesion, imbalance of protein synthesis and breakdown, satellite cells dysfunction, etc. We summarize the latest research progress on the molecular mechanism of sarcopenia in this review in order to provide new ideas for future researchers to find valuable therapeutic targets and develop relevant prevention strategies.

13.
BMC Gastroenterol ; 23(1): 23, 2023 Jan 23.
Article in English | MEDLINE | ID: mdl-36690935

ABSTRACT

BACKGROUND: Previous studies have confirmed that preoperative nutritional-inflammatory indicators can predict prognosis in various malignancies. However, to the best of our knowledge, no study has investigated the assessment of systemic inflammatory immunity index (SII) combined with prognostic nutritional index (PNI) scores to predict prognosis after neoadjuvant treatment with imatinib in locally advanced gastrointestinal stromal tumours (LA-GIST). The aim of this study was to evaluate the predictive value of pretreatment SII-PNI scores in predicting recurrence after neoadjuvant therapy with imatinib in patients with LA-GIST. METHODS: We retrospectively analyzed 57 patients with LA-GIST who received imatinib neoadjuvant from January 2013 to March 2019. Patients were divided into recurrence and non-recurrence groups according to their follow-up status, and SII and PNI cut-offs were calculated by receiver operating characteristic. The SII-PNI score ranged from 0 to 2 and were categorized into the following: score of 2, high SII (≥ 544.6) and low PNI (≤ 47.2); score of 1, either high SII (≥ 544.6) or low PNI (≤ 47.2); score of 0, no high SII (≥ 544.6) nor low PNI (≤ 47.2). RESULTS: All patients received imatinib neoadjuvant therapy for a median treatment period of 8.5 months (ranging from 3.2 to 12.6 months), with 8 patients (14.04%) and 49 patients (85.96%) developing recurrence and non-recurrence, respectively. Patients with a high SII-PNI score had a significantly worse recurrence-free survival time than those with a low SII-PNI score (P = 0.022, 0.046), and had a poorer pathological response (P = 0.014). Multivariate analysis demonstrated that the SII-PNI score was an independent prognostic factor for prediction of recurrence-free survival (P = 0.002). CONCLUSION: The pre-treatment SII-PNI score can be used to predict the efficacy after neoadjuvant treatment with imatinib in patients with LA-GIST, which may be a promising predictor of recurrence-free survival time for patients.


Subject(s)
Gastrointestinal Stromal Tumors , Nutritional Status , Humans , Prognosis , Imatinib Mesylate , Gastrointestinal Stromal Tumors/pathology , Neoadjuvant Therapy , Retrospective Studies , Inflammation/pathology , Nutrition Assessment
15.
Biochem Biophys Res Commun ; 645: 61-70, 2023 02 19.
Article in English | MEDLINE | ID: mdl-36680938

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a common malignancy with high morbidity and mortality. Although circular RNAs (circRNAs) play important roles in various cancers including ESCC, the role of the circRNA mannosidase alpha class 1A member 2 (circMAN1A2) in ESCC has been rarely studied. This study aimed to explore the role of circMAN1A2 in ESCC. CircMAN1A2 expression in ESCC tissues and cells was evaluated, and the relationship between circMAN1A2 expression and prognosis in patients with ESCC was analyzed. C-C chemokine ligand 5 (CCL5) was found to be a downstream target of circMAN1A2 by analysing the Agilent Microarray. Next, we performed in vitro and in vivo xenotransplantation assays to explore the role of circMAN1A2 in ESCC. We observed that high circMAN1A2 expression is associated with poor prognosis in patients with ESCC. Suppression of circMAN1A2 expression inhibits the proliferation, migration, and invasiveness of ESCC via regulating CCL5. Our results suggest that circMAN1A2 can promote the progression of ESCC by regulating CCL5. Thus, circMAN1A2 might be a novel diagnostic biomarker of ESCC, and targeting circMAN1A2 using inhibitors could be a potential therapeutic strategy to treat ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , MicroRNAs , Humans , Esophageal Squamous Cell Carcinoma/pathology , RNA, Circular/genetics , RNA, Circular/metabolism , Esophageal Neoplasms/pathology , Ligands , Mannosidases/metabolism , Cell Proliferation/genetics , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics
16.
Cancers (Basel) ; 14(23)2022 Nov 28.
Article in English | MEDLINE | ID: mdl-36497344

ABSTRACT

Circular RNA (circRNA), a type of non-coding RNA, has received a great deal of attention with regard to the initiation and progression of tumors. However, the molecular mechanism and function of circRNAs in breast cancer (BC) remain unclear. In the current study, we discovered that hsa_circ_0028899 (also called circRNF10) was significantly reduced in BC tissues, and a higher level of circRNF10 was markedly related to a favorable prognosis. The results of CCK8, colony formation, Transwell, ELISA, and NK cell-mediated cytotoxicity assays indicated that increased circRNF10 expression could significantly repress the proliferation, invasion, and migration of BC cells and enhance the killing efficiency of NK cells against BC cells. According to these biological functions, the possible role and molecular mechanism of circRNF10 in BC cells were further investigated. We used bioinformatics prediction tools to predict circRNF10-bound miRNAs, which were verified by many experimental studies, including FISH, luciferase reporter assays, RIP, and Western blots. These data suggest that circRNF10 serves as a molecular sponge for miR-934 to further regulate PTEN expression and PI3k/Akt/MICA signaling in vitro and tumor growth in vivo. Altogether, these findings reveal that circRNF10 functions as a novel anti-oncogene in BC via sponging miR-934 and suppressing the PI3K/Akt/MICA pathway.

17.
Mol Cancer ; 21(1): 217, 2022 12 13.
Article in English | MEDLINE | ID: mdl-36514094

ABSTRACT

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is a common gastrointestinal malignant tumor, while the molecular mechanisms have not been fully elucidated. Multiple circular RNAs have been reported to involve in the onset and progression of malignant tumors through various molecular mechanisms. However, the clinical significance and functional mechanism of most circRNAs involved in the progression of ESCC remains obscure. METHODS: RNA-Seq was used to explore potential circRNAs in participated in 5 pairs of ESCC and their corresponding normal esophageal tissues. The up-regulated circCYP24A1 was selected. Fluorescence in situ hybridization was cunducted to verificated the expression and intracellular localization of circCYP24A1 by using the tissue microarray. The Kaplan-Meier method and Cox proportional hazards model was used to examine the potential prognostic value of circCYP24A1 on overall survival of ESCC patients. The biological function were confirmed by gain- and loss-of-function approaches in vivo. mRNA expression profile microarray was proformed to investigate the downstream signaling pathways involved in circCYP24A1. RNA pull-down assay and mass spectrometry were performed to identify the proteins associated with circCYP24A1. Rescue experiments were carried out to identified hypothetical regulatory role of circCYP24A1 on ESCC progression in vivo and in virto. RESULTS: In this study, we identified circCYP24A1 in ESCC tissues by RNA sequencing, which is up-regulated in 114 cases of ESCC tissues and acts as a novel prognosis-related factor. Moreover, circCYP24A1 promoted the ability of proliferation, migration, invasion and clone formation in vitro, as well as tumor growth in vivo. Mechanistically, chemokine (C-Cmotif) ligand 5 (CCL5) is functional downstream mediator for circCYP24A1, which is screened by mRNA microarray. Moreover, circCYP24A1 physically interacts with M2 isoform of pyruvate kinase (PKM2). Rescue experiments showed that PKM2 knockdown partly reverses the promotional effects of circCYP24A1. It was revealed that circCYP24A1 increases secretion of CCL5 through the mechanism mainly by interacting with PKM2, an activator of NF-κB pathway, and thereby accelerate malignant progression of ESCC. CONCLUSIONS: Up-regulated circCYP24A1 could activate NF-κB pathway by binding PKM2, which promotes the secretion of CCL5 and accelerate malignant progression of ESCC. Our fndings recommended a novel function for circCYP24A1 as a potential effective biomarker for judging prognosis and a therapeutic target in ESCC.


Subject(s)
Chemokine CCL5 , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , RNA, Circular , Humans , Cell Line, Tumor , Cell Movement , Cell Proliferation , Chemokine CCL5/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Regulation, Neoplastic , In Situ Hybridization, Fluorescence , NF-kappa B/genetics , NF-kappa B/metabolism , RNA, Circular/genetics , RNA, Messenger , Thyroid Hormone-Binding Proteins
18.
Cancer Cell Int ; 22(1): 264, 2022 Aug 22.
Article in English | MEDLINE | ID: mdl-35996149

ABSTRACT

Interaction between tumor cells and tumor microenvironment (TME) is critical to promote tumor progression and metastasis. As the most abundant immune cells in TME, macrophages can be polarized into M2-like tumor-associated macrophages (TAMs) which further promote tumor progression. However, to date, the molecular mechanisms of TAM polarization in TME are still largely unknown. In the present study, we revealed that circular RNA circWWC3 could up-regulate the expression and secretion of IL-4 in breast cancer cells. Enhanced secretion of IL-4 from breast cancer cells could augment the M2-like polarization of macrophages in TME, which further promotes the migration of breast cancer cells. In addition, increased secretion of IL-4 from breast cancer cells could induce the expression PD-L1 in M2 macrophages. Moreover, up-regulated IL-4 also enhanced the expression of PD-L1 in breast cancer cells, which further facilitates breast cancer immune evasion. Though analyzing the expression of circWWC3, IL-4, PD-L1, and CD163 in 140 cases of breast cancer tissues, we found that high expression of circWWC3 was associated with poor overall survival and disease-free survival of breast cancer patients. Breast cancer patients with circWWC3high/PD-L1high breast cancer cells and CD163high macrophages had a poorer overall survival and disease-free survival. Conclusively, circWWC3 might augment breast cancer progression through promoting M2 macrophage polarization and tumor immune escape via regulating the expression and secretion of IL-4. CircWWC3 might be a potential therapeutic target in breast cancer.

19.
Sci Rep ; 12(1): 13757, 2022 08 12.
Article in English | MEDLINE | ID: mdl-35962061

ABSTRACT

Circular RNAs (circRNAs) are a type of noncoding RNA, which play a vital role in the occurrence and development of esophageal squamous cell carcinoma (ESCC). While the role of novel circADAMTS6 in ESCC remains unknown. We assessed circADAMTS6 expression in ESCC tissues and cells, and the relationship between circADAMTS6 expression and overall survival of ESCC patients. Functional experiments in vitro and xenograft in vivo assay were applied to explore the functions and mechanisms of circADAMTS6 in ESCC. Results found that up-regulation of circADAMTS6 was associated with poor overall survival and may acted as an independent risk factor for ESCC prognosis. Knockdown of circADAMTS6 significantly inhibited the proliferation, migration and invasion of ESCC cells and growth of xenograft tumors in vivo. Induced AGR2 expression was able to rescue the loss of function induced by si-circADAMTS6 in KYSE150 cell. CircADAMTS6 may acts as oncogene by activating AGR2 and the Hippo signaling pathway coactivator YAP in ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , MicroRNAs , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Gene Expression Regulation, Neoplastic , Humans , MicroRNAs/genetics , Mucoproteins/genetics , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Oncogene Proteins/metabolism , Prognosis , RNA, Circular/genetics
20.
Thorac Cancer ; 13(11): 1548-1557, 2022 06.
Article in English | MEDLINE | ID: mdl-35474604

ABSTRACT

BACKGROUND: Circular RNAs (circRNAs) are involved in the tumorigenesis and progression of lung adenocarcinoma (LUAD). This study aimed to determine the role of circ_0072088 in LUAD. METHODS: The existence and expression of circ_0072088 in human LUAD tissues and cell lines were determined through Sanger sequencing, quantitative reverse transcription-polymerase chain reaction, and fluorescence in situ hybridization (FISH). Subsequently, the biological role of circ_0072088 was examined using loss-of-function assays in H1299 cells. Moreover, circ_0072088/miR-1261/PIK3CA pathway-mediated biological effects in H1299 were verified using bioinformatic prediction and experiments, including interaction analysis (FISH, luciferase reporter, and RNA-pulldown assays), and tumor biological function test (CCK8 and colony formation, wound healing, and transwell assays). Finally, miR-1261 and PIK3CA expression and LUAD patient survival were further analyzed using FISH, immunohistochemical staining, and the Kaplan-Meier plotter database, respectively. RESULTS: First, an increase in circ_0072088 was confirmed in human LUAD tissues. Thereafter, it was mainly localized in the cytoplasm and was found to enhance cell proliferation, migration, and invasion of H1299 cells. Mechanistically, circ_0072088 directly downregulated miR-1261 expression, whereas increased PIK3CA gene expression was associated with poor overall survival of LUAD patients. The activation of the circ_0072088/miR-1261/PIK3CA regulatory pathway may play a significant role in the tumorigenesis and progression of LUAD. CONCLUSIONS: Circ_0072088-dependent regulation of miR-1261/PIK3CA is important for cell proliferation, migration, and invasion during the tumorigenesis and progression of LUAD, warranting the need to consider the circ_0072088/miR-1261/PIK3CA regulatory pathway as a potential therapeutic target in patients with lung adenocarcinoma.


Subject(s)
Adenocarcinoma of Lung , Class I Phosphatidylinositol 3-Kinases , Lung Neoplasms , MicroRNAs , RNA, Circular , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Adenocarcinoma of Lung/pathology , Carcinogenesis , Cell Line, Tumor , Cell Proliferation/physiology , Class I Phosphatidylinositol 3-Kinases/genetics , Disease Progression , Humans , In Situ Hybridization, Fluorescence , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Circular/genetics , RNA, Circular/metabolism , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL
...