1
|
Alkharsan AMHMS, Safaralizadeh R, Khalaj-Kondori M, HosseinpourFeizi M. Examination of the effects of capecitabine treatment on the HT-29 colorectal cancer cell line and HCG 11, HCG 15, and HCG 18 lncRNAs in CRC patients before and after chemotherapy. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:6929-6940. [PMID: 39702604 DOI: 10.1007/s00210-024-03674-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 11/24/2024] [Indexed: 12/21/2024]
Abstract
Colorectal cancer (CRC) is the third most common malignancy worldwide. Long noncoding RNAs (lncRNAs) are involved in several pathogenic pathways related to CRC. This study aimed to compare the expression profiles of HCG11, HCG15, and HCG18 genes in CRC patients before and after chemotherapy. Moreover, capecitabine's effects, which is a chemotherapeutic agent, were investigated on apoptosis, cell cycle, and the lncRNA expression in CRC using HT-29 cells. qRT-PCR was used to measure lncRNAs expression in patient and healthy tissues, and the HT-29 CRC cell line. Additionally, the diagnostic and prognostic utility of these lncRNAs were assessed using the ROC curve analysis. The MTT assay was used to evaluate the cytotoxicity of capecitabine, and by using flow cytometry, apoptosis induction and cell cycle progression were investigated. CRC patients expressed higher levels of HCG11 and HCG15 and lower levels of HCG18. Furthermore, those receiving capecitabine demonstrated a decrease in HCG11 and an increase in HCG18 expression. In the HT-29 cell line, capecitabine can also increase the expression of HCG18 and decrease the expression of HCG11 and HCG15. However, no statistically significant variations were determined in the expression of these lncRNAs in clinical variables. Additionally, the data show that HCG18 is a poor prognostic biomarker, and HCG11 and HCG18 are poor diagnostic biomarkers. Treatment with capecitabine caused an accumulation of sub-G1 cells, indicating a potent apoptotic effect on HT-29 cells. These findings confirmed capecitabine's anticancer effects and showed that it can increase HCG18 and reduce HCG11 and HCG15 expression.
Collapse
Affiliation(s)
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran.
| | - Mohammad Khalaj-Kondori
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | | |
Collapse
|
2
|
Kan F, Wang D, Li S, Gao Y, Wang J. ITGA5 drives angiogenesis in diabetic retinopathy via TAK-1/NF-kB activation. Hum Cell 2025; 38:105. [PMID: 40410450 DOI: 10.1007/s13577-025-01233-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 05/11/2025] [Indexed: 05/25/2025]
Abstract
Diabetic retinopathy is a retinal damage, which causes vision impairment and blindness. Integrin Subunit Alpha 5 (ITGA5) regulates angiogenic response, but its roles in diabetic retinopathy remain unclear. In this work, diabetes mellitus was induced in rats by streptozotocin. ITGA5 interference was achieved by intravitreal delivery of adeno-associated virus. Upregulation of ITGA5 was found in diabetic rat retinal tissues. ITGA5 knockdown decreased the neovascularization, acellular capillary formation, and pericytes. The protein expression of vascular endothelial growth factor (VEGFA), vascular adhesion molecule-1(VCAM-1), and intercellular adhesion molecule-1 (ICAM-1) was reduced after ITGA5 interference. Besides, ITGA5 knockdown decreased the phosphorylation level of FAK, TAK-1, and p65. In vitro, rat retinal microvascular endothelial cells (RRMECs) were cultured under high glucose condition to stimulate diabetic environment. ITGA5 knockdown inhibited VEGFA secretion, tube formation, cell invasion, and migration. Upregulation of VCAM-1 and ICAM-1 that induced by high glucose was reversed by ITGA5 silencing. ITGA5 knockdown blocked the activation of TAK-1/NF-kB pathway in RRMECs. Additionally, in oxygen-induced retinopathy model, ITGA5 interference inhibited pathological neovascularization. These results demonstrate that ITGA5 contributes to the angiogenesis in diabetic retinopathy by activating TAK-1/NF-kB pathway.
Collapse
Affiliation(s)
- Feifei Kan
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Di Wang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Sijia Li
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Yi Gao
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Jianwen Wang
- Department of Ophthalmology, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
3
|
Su Y, Jin Y. A narrative review of papillary thyroid carcinoma-related long non-coding RNAs and their relevance to malignant tumors. Transl Cancer Res 2025; 14:2125-2149. [PMID: 40224997 PMCID: PMC11985200 DOI: 10.21037/tcr-24-1038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Accepted: 01/14/2025] [Indexed: 04/15/2025]
Abstract
Background and Objective In recent years, research on the relationship between papillary thyroid carcinoma (PTC) and long non-coding RNAs (lncRNAs) has been burgeoning. However, there has not been an analysis of the regulatory mechanisms of these lncRNAs in all tumors, nor a comprehensive categorization and comparison of these mechanisms. This review aims to uncover whether PTC-related lncRNAs also play an important role in other tumors and to identify a common pattern of action. Methods We conducted a statistical analysis of lncRNAs related to PTC that have been reported during the period from Jan 2022 to May 2024 through searching in the Embase, Web of Science, and PubMed databases, focusing on those with greater research value. Using them as the focal points of our study, we compiled data on their different regulatory mechanisms across various malignant tumors, emphasizing key findings. Key Content and Findings This comprehensive analysis not only provides valuable insights into potential regulatory mechanisms of these lncRNAs in PTC but also serves as a reference for exploring their broader regulatory networks within cancer. The principal discovery is that lncRNAs associated with PTC can competitively interact with microRNAs (miRNAs). This interaction influences miRNA-targeted messenger RNA (mRNA) and the expression of cancer-related proteins, ultimately facilitating the progression of PTC as well as other malignant tumors. Conclusions The lncRNAs associated with PTC exert regulatory functions in other malignancies as well and possess similar regulatory mechanisms. This provides a molecular basis for the future development of relevant targeted therapies.
Collapse
Affiliation(s)
- Yuanhao Su
- Department of General Surgery, The Second Affiliated Hospital, Xi’an Jiaotong University, Xi’an, China
| | - Yi Jin
- Cell and Gene Research Therapy Institute, The First Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
4
|
Lv J, Chen F, Lv L, Zhang L, Zou H, Liu Y, Bai Y, Fang R, Qin T, Deng Z. LncRNA ABHD11-AS1 Elevates CALM2 to Promote Metastasis of Thyroid Cancer Through Sponging miR-876-5p. Biochem Genet 2025:10.1007/s10528-025-11072-9. [PMID: 40117023 DOI: 10.1007/s10528-025-11072-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 02/21/2025] [Indexed: 03/23/2025]
Abstract
In the past decade, the treatment of thyroid cancer (TC) has been brought to a new era, but tumor metastasis still is an intractable difficulty in clinical. LncRNA ABHD11-AS1 has been confirmed to be involved in TC progression. However, its specific mechanism remains unknown. Tissues from TC patients and TC cells served as mainly experimental subjects in this study. The migration of TC cells was assessed using the scratch assay, and the ability of cell invasion was evaluated by transwell assay. RT-qPCR and western blot were conducted to determine the levels of related genes and proteins. The dual-luciferase reporter assay was used to validate the relationships among ABHD11-AS1, miR-876-5p and CALM2. ABHD11-AS1 and CALM2 are elevated in TC cancer samples and cells, while the expression of miR-876-5p is reduced. Subsequently, the ability of TC cells to migrate, invade and EMT was inhibited by both ABHD11-AS1 knockdown or miR-876-5p overexpression. Moreover, the suppression of malignant characteristics in TC cells resulting from ABHD11-AS knockdown was counteracted by the inhibition of miR-876-5p or the upregulation of CALM2. On the mechanism, ABHD11-AS1 elevated CALM2 and promoted the malignant development of TC cells by acting as a miR-876-5p sponge. ABHD11-AS1 mediated the miR-876-5p/CALM2 axis to drive the metastasis of thyroid cancer.
Collapse
Affiliation(s)
- Juan Lv
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Fukun Chen
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Ling Lv
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Lu Zhang
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Huangren Zou
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Yanlin Liu
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Yuke Bai
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Ruotong Fang
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Tiantian Qin
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China
| | - Zhiyong Deng
- Department of Nuclear Medicine, Yunnan Cancer Hospital, the Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, No. 519 Kunzhou Road, Xishan District, Kunming, 650118, Yunnan Province, China.
| |
Collapse
|
5
|
Tan S, Li S, Xia L, Jiang X, Ren Z, Peng Q, Peng M, Yang W, Xu X, Oyang L, Shen M, Wang J, Li H, Wu N, Tang Y, Liao Q, Lin J, Zhou Y. Long non‑coding RNA ABHD11‑AS1 inhibits colorectal cancer progression through interacting with EGFR to suppress the EGFR/ERK signaling pathway. Int J Oncol 2025; 66:20. [PMID: 39950321 PMCID: PMC11844336 DOI: 10.3892/ijo.2025.5726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 01/15/2025] [Indexed: 02/23/2025] Open
Abstract
Long non‑coding (lnc)RNAs participate in colorectal cancer (CRC) occurrence and progression. The present study aimed to investigate whether lncRNA ABHD11‑AS1 regulates malignant biological behavior of CRC cells. Bioinformatic analysis, reverse transcription‑quantitative PCR and in situ hybridization revealed that ABHD11‑AS1 expression was decreased in CRC samples and associated with an unfavorable prognosis. ABHD11‑AS1 overexpression significantly decreased proliferation, migration and invasion of CRC cells, whereas ABHD11‑AS1 inhibition had the opposite effects. ABHD11‑AS1 interacted with EGFR to inhibit EGFR phosphorylation and attenuate EGFR/ERK signaling, which in turn suppressed the malignant biological behavior of CRC cells. The tumor suppressor function of ABHD11‑AS1 was attenuated by the EGFR agonist NSC228155. Finally, resveratrol (RSV) inhibited CRC cell proliferation, migration and invasion, which may be associated with RSV‑induced decrease in SPT6 homolog, histone chaperone and transcription elongation factor protein expression and increase in ABHD11‑AS1 transcript levels. ABHD11‑AS1 inhibited the phosphorylation of EGFR and decreased EGFR/ERK signaling by interacting with EGFR, thereby delaying the progression of CRC. The ABHD11‑AS1/EGFR/ERK axis may be a novel therapeutic target for preventing CRC progression.
Collapse
Affiliation(s)
- Shiming Tan
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Shizhen Li
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Longzheng Xia
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Xianjie Jiang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Zongyao Ren
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Qiu Peng
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Mingjing Peng
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Wenjuan Yang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
| | - Xuemeng Xu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Linda Oyang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Mengzhou Shen
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Jiewen Wang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Haofan Li
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
| | - Nayiyuan Wu
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Yanyan Tang
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Qianjin Liao
- Department of Oncology, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan 410005, P.R. China
| | - Jinguan Lin
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| | - Yujuan Zhou
- The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University/Hunan Cancer Hospital, Hunan Key Laboratory of Cancer Metabolism, Changsha, Hunan 410013, P.R. China
- Hunan Engineering Research Center of Tumor Organoid Technology and Application, Public Service Platform of Tumor Organoid Technology, Changsha, Hunan 410013, P.R. China
| |
Collapse
|
6
|
Taha SR, Karimi M, Mahdavi B, Yousefi Tehrani M, Bemani A, Kabirian S, Mohammadi J, Jabbari S, Hushmand M, Mokhtar A, Pourhanifeh MH. Crosstalk between non-coding RNAs and programmed cell death in colorectal cancer: implications for targeted therapy. Epigenetics Chromatin 2025; 18:3. [PMID: 39810224 PMCID: PMC11734566 DOI: 10.1186/s13072-024-00560-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 11/13/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) remains one of the most common causes of cancer-related mortality worldwide. Its progression is influenced by complex interactions involving genetic, epigenetic, and environmental factors. Non-coding RNAs (ncRNAs), including microRNAs (miRNAs), long non-coding RNAs (lncRNAs), and circular RNAs (circRNAs), have been identified as key regulators of gene expression, affecting diverse biological processes, notably programmed cell death (PCD). OBJECTIVE This review aims to explore the relationship between ncRNAs and PCD in CRC, focusing on how ncRNAs influence cancer cell survival, proliferation, and treatment resistance. METHODS A comprehensive literature analysis was conducted to examine recent findings on the role of ncRNAs in modulating various PCD mechanisms, including apoptosis, autophagy, necroptosis, and pyroptosis, and their impact on CRC development and therapeutic response. RESULTS ncRNAs were found to significantly regulate PCD pathways, impacting tumor growth, metastasis, and treatment sensitivity in CRC. Their influence on these pathways highlights the potential of ncRNAs as biomarkers for early CRC detection and as targets for innovative therapeutic interventions. CONCLUSION Understanding the involvement of ncRNAs in PCD regulation offers new insights into CRC biology. The targeted modulation of ncRNA-PCD interactions presents promising avenues for personalized cancer treatment, which may improve patient outcomes by enhancing therapeutic effectiveness and reducing resistance.
Collapse
Affiliation(s)
- Seyed Reza Taha
- Department of Pathology and Immunology, Washington University School of Medicine, St. LouisWashington, MO, USA
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Mehdi Karimi
- Faculty of Medicine, Bogomolets National Medical University (NMU), Kiev, Ukraine.
| | - Bahar Mahdavi
- Department of Molecular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | | | - Ali Bemani
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Shahriar Kabirian
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Javad Mohammadi
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Sina Jabbari
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Meysam Hushmand
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Alireza Mokhtar
- Student Research Committee, Kashan University of Medical Sciences, Kashan, Iran
| | - Mohammad Hossein Pourhanifeh
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Iran.
- PAKAN Institute, Tehran, Iran.
| |
Collapse
|
7
|
Lin G, Ye T, Wang J. The association between long noncoding RNA ABHD11-AS1 and malignancy prognosis: a meta-analysis. BMC Cancer 2024; 24:1083. [PMID: 39223500 PMCID: PMC11367821 DOI: 10.1186/s12885-024-12866-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Accumulating evidence has highlighted that lncRNA ABHD11-AS1 plays an essential role in tumorigenesis and is expected to become a new predictive biomarker and ideal target for cancer therapy, whereas some of their findings are conflicting due to the relatively small sample size of individual studies. Thus, this meta-analysis aimed to quantitatively ascertain the association of ABHD11-AS1 with diverse human malignancies. METHODS Eight databases were comprehensively screened for relevant articles on January 1, 2024. The significance of ABHD11-AS1 in malignancies was determined by odds ratios (ORs) or hazard ratios (HRs) with corresponding 95% confidence interval (CI). Subgroup analyses and sensitivity analyses were applied to verify the reliability and robustness of the pooled results. Simultaneously, the GEPIA2021 and UCSC Xena databases were applied to further strengthen the results. RESULTS Fourteen clinical studies comprising eight kinds of malignancies and 1215 malignancy cases were enrolled into this meta-analysis. The pooled results showed that increased ABHD11-AS1 expression was remarkably associated with lymph node metastasis (OR = 2.73, 95%CI [1.97, 3.77], I2 = 0%, p < 0.00001), advanced tumor stage ( OR = 3.14, 95%CI [2.34, 4.21], I2 = 39%, p < 0.00001), and unfavorable overall survival (OS) (HR = 1.81, 95%CI [1.58, 2.06], I2 = 0%, p < 0.00001). Subgroup analyses and sensitivity analyses indicated that the pooled results were reliable and robust. Additionally, ABHD11-AS1 was significantly increased in eight kinds of malignancies according to the validation of the GEPIA2021 database. Meanwhile, the UCSC Xena databases further revealed that elevated ABHD11-AS1 expression was significantly associated with poor prognosis as assessed by progression free interval (PFI), disease free interval (DFI), disease specific survival (DSS), and OS. CONCLUSIONS Current evidence supports the association of elevated ABHD11-AS1 expression with poor prognosis. Thereby, ABHD11-AS1 may be considered as a promising biomarker to screen cancer and predict malignancy prognosis. Also, there is a necessity for larger-scale multicenter studies with uniform study protocols from different countries to further validate the conclusions.
Collapse
Affiliation(s)
- Guangyao Lin
- Department of Gynecology, Longhua Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, 200030, China
| | - Tao Ye
- Department of Gynecology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China
| | - Jing Wang
- Department of Obstetrics, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| |
Collapse
|
8
|
Li P, Ma X, Gu X. The essential roles of lncRNAs/PI3K/AKT axis in gastrointestinal tumors. Front Cell Dev Biol 2024; 12:1442193. [PMID: 39161590 PMCID: PMC11330846 DOI: 10.3389/fcell.2024.1442193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Accepted: 07/25/2024] [Indexed: 08/21/2024] Open
Abstract
The role of long noncoding RNA (lncRNA) in tumors, particularly in gastrointestinal tumors, has gained significant attention. Accumulating evidence underscores the interaction between various lncRNAs and diverse molecular pathways involved in cancer progression. One such pivotal pathway is the PI3K/AKT pathway, which serves as a crucial intracellular mechanism maintaining the balance among various cellular physiological processes for normal cell growth and survival. Frequent dysregulation of the PI3K/AKT pathway in cancer, along with aberrant activation, plays a critical role in driving tumorigenesis. LncRNAs modulate the PI3K/AKT signaling pathway through diverse mechanisms, primarily by acting as competing endogenous RNA to regulate miRNA expression and associated genes. This interaction significantly influences fundamental biological behaviors such as cell proliferation, metastasis, and drug resistance. Abnormal expression of numerous lncRNAs in gastrointestinal tumors often correlates with clinical outcomes and pathological features in patients with cancer. Additionally, these lncRNAs influence the sensitivity of tumor cells to chemotherapy in multiple types of gastrointestinal tumors through the abnormal activation of the PI3K/AKT pathway. These findings provide valuable insights into the mechanisms underlying gastrointestinal tumors and potential therapeutic targets. However, gastrointestinal tumors remain a significant global health concern, with increasing incidence and mortality rates of gastrointestinal tumors over recent decades. This review provides a comprehensive summary of the latest research on the interactions of lncRNA and the PI3K/AKT pathway in gastrointestinal tumor development. Additionally, it focuses on the functions of lncRNAs and the PI3K/AKT pathway in carcinogenesis, exploring expression profiles, clinicopathological characteristics, interaction mechanisms with the PI3K/AKT pathway, and potential clinical applications.
Collapse
Affiliation(s)
- Penghui Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| | - Xiao Ma
- Zhejiang University School of Medicine, Hangzhou, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
9
|
Bian Y, Xu S, Gao Z, Ding J, Li C, Cui Z, Sun H, Li J, Pu J, Wang K. m 6A modification of lncRNA ABHD11-AS1 promotes colorectal cancer progression and inhibits ferroptosis through TRIM21/IGF2BP2/ FOXM1 positive feedback loop. Cancer Lett 2024; 596:217004. [PMID: 38838765 DOI: 10.1016/j.canlet.2024.217004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/20/2024] [Accepted: 05/25/2024] [Indexed: 06/07/2024]
Abstract
Long non-coding RNA (lncRNA) is closely related to a variety of human cancers, which may provide huge potential biomarkers for cancer diagnosis and treatment. However, the aberrant expression of most lncRNAs in colorectal cancer (CRC) remains elusive. This study aims to explore the clinical significance and potential mechanism of lncRNA ABHD11 antisense RNA 1 (ABHD11-AS1) in the colorectal cancer. Here, we demonstrated that lncRNA ABHD11-AS1 is high-expressed in colorectal cancer (CRC) patients, and strongly related with poor prognosis. Functionally, ABHD11-AS1 suppresses ferroptosis and promotes proliferation and migration in CRC both in vitro and in vivo. Mechanically, lncRNA ABHD11-AS1 interacted with insulin-like growing factor 2 mRNA-binding protein 2 (IGF2BP2) to enhance FOXM1 stability, forming an ABHD11-AS1/FOXM1 positive feedback loop. E3 ligase tripartite motif containing 21 (TRIM21) promotes the degradation of IGF2BP2 via the K48-ubiquitin-lysosome pathway and ABHD11-AS1 promotes the interaction between IGF2BP2 and TRIM21 as scaffold platform. Furthermore, N6 -adenosine-methyltransferase-like 3 (METTL3) upregulated the stabilization of ABHD11-AS1 through the m6A reader IGF2BP2. Our study highlights ABHD11-AS1 as a significant regulator in CRC and it may become a potential target in future CRC treatment.
Collapse
Affiliation(s)
- Yibo Bian
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China; State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers and National Clinical Research Center for Digestive Diseases, Xijing Hospital of digestive Disease, Fourth Military Medical University, Xi'an, 710032, China
| | - Shufen Xu
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Zhishuang Gao
- Department of Breast Surgery, Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Jie Ding
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Chao Li
- Department of General Surgery, Zhongshan Hospital, Fudan University School of Medicine, Shanghai, 200032, China
| | - Zhiwei Cui
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Haoyu Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Juan Li
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Juan Pu
- Department of Oncology, Lianshui County People's Hospital, Huai'an, 223400, China.
| | - Keming Wang
- Department of Oncology, Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
| |
Collapse
|
10
|
Zhu HN, Song DL, Zhang SN, Zheng ZJ, Chen XY, Jin X. Progress in long non-coding RNAs as prognostic factors of papillary thyroid carcinoma. Pathol Res Pract 2024; 256:155230. [PMID: 38461693 DOI: 10.1016/j.prp.2024.155230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Revised: 02/25/2024] [Accepted: 02/25/2024] [Indexed: 03/12/2024]
Abstract
Papillary thyroid carcinoma (PTC) is generally recognized as a slow-growing tumor. However, a small subset of patients may still experience relapse or metastasis shortly after therapy, leading to a poor prognosis and raising concerns about excessive medical treatment. One major challenge lies in the inadequacy of effective biomarkers for accurate risk stratification. Long non-coding RNAs (lncRNAs), which are closely related to malignant characteristics and poor prognosis, play a significant role in the genesis and development of PTC through various pathways. The objective of this review is to provide a comprehensive summary of the biological functions of lncRNAs in PTC, identify prognosis-relevant lncRNAs, and explore their potential mechanisms in drug resistance to BRAF kinase inhibitors, tumor dedifferentiation, and lymph node metastasis. By doing so, this review aims to offer valuable references for both basic research and the prediction of PTC prognosis.
Collapse
Affiliation(s)
- Hao-Nan Zhu
- Department of Clinical Medicine, Medical College, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Dong-Liang Song
- Department of Clinical Medicine, Medical College, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Si-Nan Zhang
- Department of Clinical Medicine, Medical College, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Zhao-Jie Zheng
- Department of Clinical Medicine, Medical College, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Xing-Yu Chen
- Department of Clinical Medicine, Medical College, Shaoxing University, Shaoxing, Zhejiang 312000, China
| | - Xin Jin
- Department of Clinical Medicine, Medical College, Shaoxing University, Shaoxing, Zhejiang 312000, China.
| |
Collapse
|
11
|
Yang T, Tian S, Zhao J, Pei M, Zhao M, Yang X. LncRNA ABHD11-AS1 activates EGFR signaling to promote cervical cancer progression by preventing FUS-mediated degradation of ABHD11 mRNA. Cell Cycle 2023; 22:2538-2551. [PMID: 38146687 PMCID: PMC10936639 DOI: 10.1080/15384101.2023.2297591] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 12/15/2023] [Indexed: 12/27/2023] Open
Abstract
Cervical cancer is one of the most common gynecological cancers with high metastasis, poor prognosis and conventional chemotherapy. The long non-coding RNA (lncRNA) ABHD11 antisense RNA 1 (ABHD11-AS1) plays a vital role in tumorigenesis and is involved in cell proliferation, differentiation, and apoptosis. Especially for cervical cancer, the functions and mechanisms of ABHD11-AS1 are still undetermined. In this study, we explored the role and underlying mechanism of ABHD11-AS1 in cervical cancer. We found that ABHD11-AS1 is highly expressed in cervical cancer tissue. The roles of ABHD11-AS1 and EGFR have investigated the loss of function analysis and cell movability in SiHa and Hela cells. Knockdown of ABHD11-AS1 and EGFR significantly inhibited the proliferation, migration, and invasion and promoted apoptosis of SiHa and Hela cells by up-regulating p21 and Bax and down-regulating cyclin D1, Bcl2, MMP9, and Vimentin. ABHD11-AS1 knockdown could decrease the expression of EGFR. In addition, ABHD11-AS1 could regulate the EGFR signaling pathway, including p-EGFR, p-AKT, and p-ERK. Spearman's correlation analysis and cell experiments demonstrated that ABHD11 was highly expressed in tumor tissue and partially offset the effect of shABHD11-AS1 on the proliferation, migration, and invasion of SiHa and Hela cells. Then, RNA pulldown was used to ascertain the mechanisms of ABHD11-AS1 and FUS. ABHD11-AS1 inhibited ABHD11 mRNA degradation by bounding to FUS. A subcutaneous xenograft of SiHa cells was established to investigate the effect of ABHD11-AS1 in tumor tissue. Knockdown of ABDH11-AS1 inhibited tumor growth and decreased the tumor volume. ABHD11-AS1 knockdown inhibited the expression of Ki67 and Vimentin and up-regulated the expression of Tunel. Our data indicated that ABHD11-AS1 promoted cervical cancer progression by activating EGFR signaling, preventing FUS-mediated degradation of ABHD11 mRNA. Our findings provide novel insights into the potential role of lncRNA in cervical cancer therapy.
Collapse
Affiliation(s)
- Ting Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, China
| | - Sijuan Tian
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, China
| | - Juan Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, China
| | - Meili Pei
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, China
| | - Minyi Zhao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an City, Shaanxi Province, China
| | | |
Collapse
|
12
|
Mahdi Khanifar M, Zafari Z, Sheykhhasan M. Crosstalk between long non-coding RNAs and p53 signaling pathway in colorectal cancer: A review study. Pathol Res Pract 2023; 249:154756. [PMID: 37611430 DOI: 10.1016/j.prp.2023.154756] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/08/2023] [Accepted: 08/08/2023] [Indexed: 08/25/2023]
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignancies worldwide and the third leading cause of cancer-related fatalities. Long non-coding RNAs (lncRNAs) are key regulators of diverse physiological processes and are dysregulated in a wide range of pathophysiological circumstances such as CRC. Studies revealed that aberrant expressions of lncRNAs clearly modulate the expression level of p53 gene in CRC, thereby transactivating multiple downstream pathways. P53 is regarded as a crucial tumor suppressor gene which promotes cell-cycle arrest, DNA repair, senescence or apoptosis in response to cellular stresses. P53 is also mutated in CRC as well as various types of human malignancies. Therefore, lncRNAs interact with the p53 signaling pathway in numerus ways and significantly influence CRC-related processes. The current findings in the investigation of the crosstalk between lncRNAs and the P53 pathway in controlling CRC carcinogenesis, tumor progression, and therapeutic resistance are summarized in the this review. A deeper knowledge of CRC carcinogenesis may also have implications in CRC prevention and treatment through more research.
Collapse
Affiliation(s)
- Mohammad Mahdi Khanifar
- School of Molecular Science, University of Western Australia, Perth, Western Australia, Australia; Department of Biology, Shahed University, Tehran, Iran
| | - Zahra Zafari
- Department of Biology, Shahed University, Tehran, Iran.
| | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran; Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran.
| |
Collapse
|
13
|
Verhoeff TJ, Holloway AF, Dickinson JL. Non-coding RNA regulation of integrins and their potential as therapeutic targets in cancer. Cell Oncol (Dordr) 2023; 46:239-250. [PMID: 36512308 PMCID: PMC10060301 DOI: 10.1007/s13402-022-00752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/19/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Integrins are integral to cell signalling and management of the extracellular matrix, and exquisite regulation of their expression is essential for a variety of cell signalling pathways, whilst disordered regulation is a key driver of tumour progression and metastasis. Most recently non-coding RNAs in the form of micro-RNA (miRNA) and long non-coding RNA (lncRNA) have emerged as a key mechanism by which tissue dependent gene expression is controlled. Whilst historically these molecules have been poorly understood, advances in 'omic' technologies and a greater understanding of non-coding regions of the genome have revealed that non-coding RNAs make up a large proportion of the transcriptome. CONCLUSIONS AND PERSPECTIVES This review examines the regulation of integrin genes by ncRNAs, provides and overview of their mechanism of action and highlights how exploitation of these discoveries is informing the development of novel chemotherapeutic agents in the treatment of cancer. MiRNA molecules have been the most extensively characterised and negatively regulate most integrin genes, classically regulating genes through binding to recognition sequences in the mRNA 3'-untranslated regions of gene transcripts. LncRNA mechanisms of action are now being elucidated and appear to be more varied and complex, and may counter miRNA molecules, directly engage integrin mRNA transcripts, and guide or block both transcription factors and epigenetic machinery at integrin promoters or at other points in integrin regulation. Integrins as therapeutic targets are of enormous interest given their roles as oncogenes in a variety of tumours, and emerging therapeutics mimicking ncRNA mechanisms of action are already being trialled.
Collapse
Affiliation(s)
- Tristan Joseph Verhoeff
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart (Tasmania), Australia
| | - Adele F Holloway
- Tasmanian School of Medicine, College of Health and Medicine, University of Tasmania, Hobart (Tasmania), Australia
| | - Joanne L Dickinson
- Menzies Institute for Medical Research, College of Health and Medicine, University of Tasmania, Hobart (Tasmania), Australia.
| |
Collapse
|
14
|
lncRNA HCG11 Promotes Colorectal Cancer Cell Malignant Behaviors via Sponging miR-26b-5p. J Immunol Res 2023; 2023:9011232. [PMID: 36874625 PMCID: PMC9981294 DOI: 10.1155/2023/9011232] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 09/27/2022] [Accepted: 01/20/2023] [Indexed: 02/25/2023] Open
Abstract
Colorectal cancer (CRC) is a type of gastrointestinal cancer with an increasing incidence. Long noncoding RNAs (lncRNAs) have raised great concern because of wide participation in human diseases, including cancers. However, whether lncRNA HLA complex group 11 (HCG11) played a functional role in CRC remained to be elucidated. Herein, we utilized qRT-PCR to analyze the expression of HCG11 and found that HCG11 was highly expressed in CRC cells. Besides, HCG11 knockdown suppressed cell proliferation, migration, and invasion but facilitated cell apoptosis. Furthermore, supported by bioinformatics analyses and mechanism assays, HCG11, mainly located in cell cytoplasm, was confirmed to competitively bind to miR-26b-5p to modulate the expression of the target messenger RNA (mRNA), namely, cAMP-regulated phosphoprotein 19 (ARPP19). ARPP19 was detected to be upregulated in CRC cells, and ARPP19 silence was verified to inhibit the malignant behaviors of CRC cells. Rescue experiments validated that miR-26b-5p inhibition or ARPP19 overexpression could countervail the inhibitory influences of HCG11 silence on CRC cell biological behaviors in vitro. To conclude, HCG11, upregulated in CRC cells, could promote cell proliferation, migration, and invasion and inhibit cell apoptosis via targeting miR-26b-5p/ARPP19 axis.
Collapse
|
15
|
Wang JF, Chen YY, Zhang SW, Zhao K, Qiu Y, Wang Y, Wang JC, Yu Z, Li BP, Wang Z, Chen JQ. ITGA5 Promotes Tumor Progression through the Activation of the FAK/AKT Signaling Pathway in Human Gastric Cancer. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8611306. [PMID: 36193075 PMCID: PMC9526618 DOI: 10.1155/2022/8611306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Accepted: 07/18/2022] [Indexed: 11/17/2022]
Abstract
BACKGROUND ITGA5 is an adhesion molecule that integrates the intracellular structures with the extracellular matrix to perform biological functions. However, ITGA5 is highly expressed in a variety of tumors and is involved in tumor progression by promoting cell proliferation and metastasis. Nevertheless, little research has been performed on its function in gastric cancer. Therefore, the aim of this study was to investigate the role of ITGA5 in gastric cancer, focusing on the mechanism regulating the proliferation, invasion and migration. METHODS The expression of ITGA5 in gastric cancer tissues was assessed by the use of molecular bioinformatics databases and high-throughput sequencing of gastric cancer tissues from patients. Western blot, qPCR, and immunohistochemistry were performed to detect the expression of ITGA5 in samples from gastric cancer patients and gastric cancer cell lines. Furthermore, the ITGA5 gene was silenced and overexpressed in gastric cancer cells, and the effect on proliferation, invasion, migration, and tumorigenic ability was assessed. RESULTS ITGA5 mRNA and protein expression were upregulated in gastric cancer cell lines and tissues from patients, and its expression was closely associated with tumor size, lymph node metastasis, and TNM stage. In vitro and in vivo experiments showed that ITGA5 silencing resulted in the inhibition of proliferation, invasion, migration, and graft growth of gastric cancer cells; conversely, the overexpression resulted in the promotion of these cell functions. Our results finally showed that the effect of ITGA5 on proliferation, invasion, and migration of gastric cancer cells was performed through the activation of the FAK/AKT pathway. CONCLUSIONS ITGA5 promotes proliferation, invasion, and migration of gastric cancer cells through the activation of FAK/AKT signaling pathway, suggesting that ITGA5 may be potentially considered as a new target in gastric cancer therapy.
Collapse
Affiliation(s)
- Jun-fu Wang
- Department of General Surgery, The First Affiliated Hospital of Nanchang University, Nanchang 330031, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
| | - Ye-yang Chen
- Department of Gastrointestinal Surgery, The First People's Hospital of Yulin, Yulin, 537000 Guangxi Zhuang Autonomous Region, China
| | - Si-wen Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Kun Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
| | - Yue Qiu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Ye Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jian-cheng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Zhu Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Bo-pei Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Zheng Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, Guangxi Zhuang Autonomous Region, China
| | - Jun-qiang Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangxi Medical University, 6 Shuangyong Road, Nanning, 530021 Guangxi Zhuang Autonomous Region, China
- Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, Guangxi Zhuang Autonomous Region, China
| |
Collapse
|
16
|
Zhou X, Zhong F, Yan Y, Wu S, Wang H, Liu J, Li F, Cui D, Xu M. Pancreatic Cancer Cell-Derived Exosomes Promote Lymphangiogenesis by Downregulating ABHD11-AS1 Expression. Cancers (Basel) 2022; 14:cancers14194612. [PMID: 36230535 PMCID: PMC9562033 DOI: 10.3390/cancers14194612] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 09/13/2022] [Accepted: 09/19/2022] [Indexed: 11/16/2022] Open
Abstract
Research on pancreatic cancer microbiomes has attracted attention in recent years. The current view is that enriched microbial communities in pancreatic cancer tissues may affect pancreatic cancer metastasis, including lymph node (LN) metastasis. Similar to carriers of genetic information between cells, such as DNA, mRNA, protein, and non-coding RNA, exosomes are of great importance in early LN metastasis in tumors, including pancreatic cancer. Our previous study showed that the long non-coding RNA ABHD11-AS1 was highly expressed in tissues of patients with pancreatic cancer, and was correlated with patient survival time. However, the role of ABHD11-AS1 in pancreatic cancer LN metastasis has rarely been studied. Hence, in this paper we confirmed that exosomes derived from pancreatic cancer cells could promote lymphangiogenesis in vitro and in vivo, and that the mechanism was related to the downregulation of ABHD11-AS1 expression in lymphatic endothelial cells, and to the enhancement of their ability to proliferate, migrate, and form tubes. These findings preliminarily show a new mechanism by which pancreatic cancer cells regulate peripheral lymphangiogenesis, providing a new therapeutic strategy for inhibiting LN metastasis in pancreatic cancer.
Collapse
Affiliation(s)
- Xulin Zhou
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Fengyun Zhong
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou 215025, China
| | - Yongmin Yan
- Department of Laboratory Medicine, Wujin Hospital Affiliated with Jiangsu University, Jiangsu University, Changzhou 213000, China
| | - Sihui Wu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Huizhi Wang
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Junqiang Liu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Feifan Li
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
| | - Dawei Cui
- The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310030, China
- Correspondence: (D.C.); (M.X.)
| | - Min Xu
- Department of Gastroenterology, Affiliated Hospital of Jiangsu University, Zhenjiang 212001, China
- Correspondence: (D.C.); (M.X.)
| |
Collapse
|
17
|
Golla U, Sesham K, Dallavalasa S, Manda NK, Unnam S, Sanapala AK, Nalla S, Kondam S, Kumar R. ABHD11-AS1: An Emerging Long Non-Coding RNA (lncRNA) with Clinical Significance in Human Malignancies. Noncoding RNA 2022; 8:ncrna8020021. [PMID: 35314614 PMCID: PMC8938790 DOI: 10.3390/ncrna8020021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 02/27/2022] [Accepted: 02/28/2022] [Indexed: 12/24/2022] Open
Abstract
The aberrant expression of lncRNAs has been linked to the development and progression of different cancers. One such lncRNA is ABHD11 antisense RNA 1 (ABHD11-AS1), which has recently gained attention for its significant role in human malignancies. ABHD11-AS1 is highly expressed in gastric, lung, breast, colorectal, thyroid, pancreas, ovary, endometrium, cervix, and bladder cancers. Several reports highlighted the clinical significance of ABHD11-AS1 in prognosis, diagnosis, prediction of cancer progression stage, and treatment response. Significantly, the levels of ABHD11-AS1 in gastric juice had been exhibited as a clinical biomarker for the assessment of gastric cancer, while its serum levels have prognostic potential in thyroid cancers. The ABHD11-AS1 has been reported to exert oncogenic effects by sponging different microRNAs (miRNAs), altering signaling pathways such as PI3K/Akt, epigenetic mechanisms, and N6-methyladenosine (m6A) RNA modification. In contrast, the mouse homolog of AHD11-AS1 (Abhd11os) overexpression had exhibited neuroprotective effects against mutant huntingtin-induced toxicity. Considering the emerging research reports, the authors attempted in this first review on ABHD11-AS1 to summarize and highlight its oncogenic potential and clinical significance in different human cancers. Lastly, we underlined the necessity for future mechanistic studies to unravel the role of ABHD11-AS1 in tumor development, prognosis, progression, and targeted therapeutic approaches.
Collapse
Affiliation(s)
- Upendarrao Golla
- Department of Medicine, Division of Hematology and Oncology, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Penn State Cancer Institute, Pennsylvania State University College of Medicine, Hershey, PA 17033, USA
- Correspondence:
| | - Kishore Sesham
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), Mangalagiri 522503, India;
| | - Siva Dallavalasa
- Center of Excellence in Molecular Biology and Regenerative Medicine (CEMR), Department of Biochemistry, JSS Medical College, Mysuru 570015, India;
| | - Naresh Kumar Manda
- Department of Biochemistry, School of Life Sciences, University of Hyderabad, Hyderabad 500046, India;
| | - Sambamoorthy Unnam
- Faculty of Pharmacy, Sree Dattha Institute of Pharmacy, Ibrahimpatnam 501510, India; (S.U.); (A.K.S.)
| | - Arun Kumar Sanapala
- Faculty of Pharmacy, Sree Dattha Institute of Pharmacy, Ibrahimpatnam 501510, India; (S.U.); (A.K.S.)
| | - Sharada Nalla
- Faculty of Pharmacy, University College of Pharmaceutical Sciences, Palamuru University, Mahabubnagar 509001, India; (S.N.); (S.K.)
| | - Susmitha Kondam
- Faculty of Pharmacy, University College of Pharmaceutical Sciences, Palamuru University, Mahabubnagar 509001, India; (S.N.); (S.K.)
| | - Rajesh Kumar
- Department of Anatomy, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India;
| |
Collapse
|
18
|
Hu Y, Luo M. NORAD-sponged miR-378c alleviates malignant behaviors of stomach adenocarcinoma via targeting NRP1. Cancer Cell Int 2022; 22:79. [PMID: 35164743 PMCID: PMC8842946 DOI: 10.1186/s12935-022-02474-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 01/19/2022] [Indexed: 01/25/2023] Open
Abstract
Background Stomach adenocarcinoma (STAD) is the most common type of gastric cancer (GC), with a high recurrence rate and poor prognosis, but the potential indicators for STAD are insufficient. Methods Herein, we found that MicroRNA-378c (miR-378c) was lowly expressed in STAD, and the low expression of miR-378c was highly correlated with poor overall survival (OS), T stage, Reflux history, DSS events and PFI events of STAD patients. Results In addition, univariate analysis displayed that miR-378c was significantly associated with OS (Hazard ratio 0.735; 95% CI, 0.542–0.995; P = 0.046). Furthermore, it was validated that miR-378c inhibition accelerated STAD cell proliferation, migration, invasion and epithelial-mesenchymal transition (EMT), while they were suppressed by miR-378c overexpression. Mechanistically, Neuropilin 1 (NRP1) was confirmed as the target of miR-378c, and Lnc-NORAD was identified as its sponger. More importantly, NORAD-mediated miR-378c inhibited malignant behaviors of STAD both in vitro and in vivo. Conclusions Collectively, these results suggest miR-378c as a promising indicator for the treatment of STAD. Supplementary Information The online version contains supplementary material available at 10.1186/s12935-022-02474-5.
Collapse
Affiliation(s)
- Yongjun Hu
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China
| | - Ming Luo
- Department of General Surgery, The Second Xiangya Hospital, Central South University, No.139 Renmin Middle Road, Changsha, 410011, Hunan, China.
| |
Collapse
|