1
|
Fu Y, Hu P, Hu Y, Fang Y, Zhou Y, Shi Y, Yang K, Fu T, Li W, Gritskevitch ER, Jin L, Lyu J, Zhao Q. Hepatocyte-specific RAP1B deficiency ameliorates high-fat diet-induced obesity and liver inflammation in mice. Diabetes Obes Metab 2025; 27:3036-3049. [PMID: 40083059 DOI: 10.1111/dom.16309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/16/2025]
Abstract
AIM This study investigated the role of RAP1B in hepatic lipid metabolism and its implications in obesity and associated metabolic disorders, focusing on the molecular mechanisms through which RAP1B influences lipid accumulation, inflammation and oxidative stress in liver tissues and hepatocyte cell lines. MATERIALS AND METHODS Liver-specific RAP1B-knockout (LKO) and overexpression (OE) mice were generated and fed a high-fat diet for 18 weeks to evaluate systemic and hepatic metabolic changes. Comprehensive metabolic phenotyping included measurements of body weight, body fat content, activity levels, energy expenditure (EE), respiratory exchange ratio (RER), glucose tolerance test and insulin tolerance test. RAP1B-knockdown AML12 hepatocytes were used for in vitro studies. Comprehensive transcriptome and metabolome analyses identified differentially expressed genes and key metabolic shifts. Biochemical and histological analyses were performed to assess lipid accumulation, oxidative stress and inflammatory markers. RESULTS We found that LKO mice exhibited significant reductions in body weight, fat pad size and liver mass, along with decreased hepatic lipid accumulation due to enhanced lipid breakdown. These mice demonstrated improved glucose tolerance and insulin sensitivity without changes in food intake. Liver histology showed reduced F4/80-positive macrophage infiltration, indicating decreased inflammatory cell recruitment. Additionally, markers of oxidative stress were significantly lower, and molecular analysis revealed downregulation of the MAPK(p38) and NF-κB signaling pathways, further supporting an anti-inflammatory hepatic environment. In contrast, OE mice showed increased liver weight, aggravated hepatic lipid accumulation driven by enhanced lipogenesis, worsened insulin resistance and elevated inflammation. CONCLUSIONS This study highlights RAP1B's pivotal role in hepatic metabolism and positions it as a potential therapeutic target for obesity and related metabolic disorders.
Collapse
Affiliation(s)
- Yinxu Fu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, China
| | - Pingyi Hu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Yanyang Hu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Yu Fang
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Yaping Zhou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yu Shi
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Kaiqiang Yang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
| | - Ting Fu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
| | - Weijia Li
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
- International Sakharov Environmental Institute, Belarusian State University, Minsk, Republic of Belarus
| | | | - Liqin Jin
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Jianxin Lyu
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, China
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Qiongya Zhao
- School of Laboratory Medicine and Bioengineering, Hangzhou Medical College, Hangzhou, China
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, China
- Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| |
Collapse
|
2
|
Fang X, Huang X, Liu W, Lv W, Ying Y, Huang J. Overexpression of FMOD in Thyroid Carcinoma Triggers M1-Like Tumor-Associated Macrophage Polarization by Targeting Rap1B. FASEB J 2025; 39:e70581. [PMID: 40297936 DOI: 10.1096/fj.202403290rr] [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] [Received: 12/18/2024] [Revised: 04/14/2025] [Accepted: 04/18/2025] [Indexed: 04/30/2025]
Abstract
Thyroid carcinoma, with limited efficacy of current treatment, influences the lives and health of many people. It is important to explore potential therapeutic targets for thyroid carcinoma. Fibromodulin (FMOD) has been indicated to be connected with the progression of different kinds of tumors, with unknown functions in thyroid carcinoma. In this study, the potential candidate therapeutic targets for thyroid carcinoma were identified by bioinformatics methods, and FMOD was screened out for verification. Cell counting kit-8, wound healing, transwell, and flow cytometry assays were conducted to determine the role of FMOD overexpression in cell viability, migration, invasion, and apoptotic rate of thyroid carcinoma cells, respectively. Subcutaneous tumor growth was monitored in nude mice. Tumor-associated macrophages (TAMs) were co-cultured with thyroid carcinoma cells, and the surface marker of M1-like TAMs, CD80, was identified by flow cytometry. Ras-association proximate 1B (Rap1B), the downstream target of FMOD, was predicted by bioinformatic techniques and validated by Rap1B overexpression rescue. FMOD was identified as a tumor suppressor gene in thyroid carcinoma through bioinformatic techniques. FMOD overexpression inhibited cell viability, migration, and invasion and stimulated apoptosis of thyroid carcinoma cells. In vivo, FMOD upregulation could suppress the growth of solid tumors. Moreover, FMOD overexpression in thyroid carcinoma cells promoted M1-like TAM polarization. FMOD downregulated Rap1B expression in thyroid carcinoma cells, and Rap1B overexpression rescue reversed the impact of FMOD on tumor progression and TAM polarization. In conclusion, FMOD exhibited an inhibitory effect on thyroid carcinoma by stimulating M1-like TAM polarization via targeting Rap1B.
Collapse
Affiliation(s)
- Xiangnan Fang
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Xuemei Huang
- Department of Endocrinology and Metabolism, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Wenfeng Liu
- Department of Endocrinology and Metabolism, Gannan Medical University, Ganzhou, Jiangxi, China
| | - Weiming Lv
- Department of Endocrinology and Metabolism, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yong Ying
- Department of Thyroid Hernia Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jinchang Huang
- Department of Pathology, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| |
Collapse
|
3
|
Barjasteh AH, Jaseb Mazhar AleKassar R, Al-Asady AM, Latifi H, Avan A, Khazaei M, Ryzhikov M, Hassanian SM. Therapeutic Potentials of MiRNA for Colorectal Cancer Liver Metastasis Treatment: A Narrative Review. IRANIAN JOURNAL OF MEDICAL SCIENCES 2025; 50:202-219. [PMID: 40255223 PMCID: PMC12008659 DOI: 10.30476/ijms.2024.102910.3622] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/01/2024] [Accepted: 11/26/2024] [Indexed: 04/22/2025]
Abstract
Colorectal cancer (CRC) ranks among the most prevalent cancers worldwide and is the fourth leading cause of cancer-related deaths. Metastasis poses a significant obstacle in CRC treatment, as distant metastasis, particularly to the liver, remains the primary cause of mortality. Colorectal liver metastasis (CRLM) occurs frequently due to the liver's direct vascular connection to the colorectal region via the portal vein. Standard treatment approaches for CRLM are limited; only a few patients qualify for surgical intervention, resulting in a persistently low survival rate. Additionally, resistance to chemotherapy is common, emphasizing the need for more effective targeted therapies. Emerging evidence highlights the pivotal role of microRNAs (miRNAs) in modulating critical pathways associated with CRLM, including tumor invasion, epithelial-mesenchymal transition, and angiogenesis. MiRNAs exhibit dual functions as tumor suppressors and oncogenes by targeting multiple genes, thus playing a complex role in both the initiation and progression of metastasis. The regulatory mechanisms of miRNAs could help to identify novel biomarkers for early diagnosis and prognosis of CRLM, as well as promising therapeutic targets to overcome chemoresistance. Despite numerous studies on miRNA involvement in CRC metastasis, dedicated reviews focusing on miRNAs and CRLM remain scarce. This review aims to approach targeted therapies by examining the current understanding of miRNA involvement in CRLM and exploring their potential as diagnostic, prognostic, and therapeutic agents. Through an integrative approach, we aim to provide insights that could transform CRLM management and improve patient outcomes.
Collapse
Affiliation(s)
- Amir Hossein Barjasteh
- Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Rawa Jaseb Mazhar AleKassar
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Abdulridha Mohammed Al-Asady
- Department of Medical Sciences, Faculty of Nursing, Warith Al-Anbiyaa University, Iraq
- Department of Medical Sciences, Faculty of Dentistry, University of Kerbala, Iraq
- Department of Pharmacology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hanieh Latifi
- Metabolic Syndrome Research Center, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, QLD 4059, Australia
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Department of Medical Physiology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Seyed Mahdi Hassanian
- Department of Clinical Biochemistry, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
4
|
Xing XD, Yan XY, Tan YW, Liu Y, Cui YX, Feng CL, Cai YR, Dai HL, Gao W, Zhou P, Wang HY, Li P, Yang H. Deep-DPC: Deep learning-assisted label-free temporal imaging discovery of anti-fibrotic compounds by controlling cell morphology. J Adv Res 2025:S2090-1232(25)00126-2. [PMID: 40010605 DOI: 10.1016/j.jare.2025.02.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/17/2025] [Accepted: 02/23/2025] [Indexed: 02/28/2025] Open
Abstract
INTRODUCTION Fibrosis can damage the normal function of many organs, such as cardiac function, for which no effective clinical therapies exist. However, traditional approaches to anti-fibrosis drug discovery have primarily focused on the final biological indicators, often overlooking the dynamic morphological changes during fibrosis progression. Here, we present a novel approach, deep-DPC, which integrates label-free, time-series digital phase contrast (DPC) imaging with cell morphology analysis and unsupervised machine learning to dynamically control and monitor cell morphology. OBJECTIVES This method enables discrimination between resting and activated fibrocytes and facilitates the discovery of non-invasive labeled anti-fibrotic lead compounds. METHODS The deep-DPC comprises two major steps: (1) preliminary analysis by Harmony 4.9 software and (2) image classification via a neural network. For the experiment dataset, label-free time-series imaging was acquired from each well at 10 × magnification using the high-content imaging system, equipped with a high-speed charge-coupled device (CCD) camera. Dual-channel output images were generated through the imaging system, with one channel for bright-field and the other for DPC imaging, captured at 30-minute intervals. Firstly, applying the anti-fibrotic cell model as a case, a label-free time-series DPC imaging was developed by combining cell morphological analysis and deep learning, and its stability was verified by training with 12,000 images. Furthermore, the application of deep-DPC in the discovery of anti-fibrotic lead compounds. RESULTS Using the deep-DPC platform, over 100,000 images generated from 1,400 compounds were processed, identifying Neo-Przewaquinone A as a potent anti-fibrosis agent. Neo-Przewaquinone A exerts its effects by inhibiting TGF-β receptor I, thereby maintaining cells in a resting state and arresting the cell cycle. CONCLUSION The deep-DPC offers a promising strategy for fibrosis assessment by combining deep learning with dynamic cell morphology analysis based on time-series DPC images. Additionally, the platform holds potential as a novel therapeutic approach for anti-myocardial fibrosis by regulating cell morphology.
Collapse
Affiliation(s)
- Xu-Dong Xing
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Xiang-Yu Yan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Yan-Wei Tan
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Yang Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Yi-Xin Cui
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Chun-Ling Feng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Yu-Ru Cai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Han-Lin Dai
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Wen Gao
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Ping Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Hui-Ying Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China
| | - Ping Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China.
| | - Hua Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, No. 639 Longmian Dadao, Nanjing 211198, China.
| |
Collapse
|
5
|
Shahrokhi H, Asili J, Tayarani-Najaran Z, Boozari M. Signaling pathways behind the biological effects of tanshinone IIA for the prevention of cancer and cardiovascular diseases. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-03857-x. [PMID: 39937254 DOI: 10.1007/s00210-025-03857-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
Tanshinone IIA (Tan IIA) is a well-known fat-soluble diterpenoid found in Salvia miltiorrhiza, recognized for its various biological effects. The molecular signaling pathways of Tan IIA have been investigated in different diseases, including the anti-inflammatory, hepatoprotective, renoprotective, neuroprotective effects, and fibrosis prevention. This article provides a brief overview of the signaling pathways related to anti-cancer and cardioprotective effects of Tan IIA. It shows that Tan IIAs anti-cancer ability has good expectation through multiplicity mechanisms affecting various aspects' tumor biology. The major pathways involved in its anti-cancer effects include inhibition of PI3/Akt, MAPK, and p53/p21 signaling which leads to enhancement of immune responses and increased radiation sensitivity. Some essential pathways responsible for cardioprotective effects induced by Tan IIA are PI3/AKT activation, MAPK, and SIRT1 promoting protection against ischemia/reperfusion injury in myocardial cells as well as inhibiting pathological remodeling processes. Finally, the article underscores the complex and specific signaling pathways influenced by Tan IIA. The PI3/Akt and MAPK pathways play critical roles in the anti-cancer and cardioprotective effects of Tan IIA. Particularly, Tan IIA suppresses the proliferation of malignancies in cancerous cells but stimulates protective mechanisms in normal cardiovascular cells. These findings highlight the importance of investigating molecular signaling pathways in evaluating the therapeutic potential of natural products. Studying about signaling pathways is vital in understanding the therapeutic aspects of Tan IIA and its derivatives as anti-cancer and cardio-protective agents. Further research is necessary to understand these complex mechanisms.
Collapse
Affiliation(s)
- Homa Shahrokhi
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Javad Asili
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zahra Tayarani-Najaran
- Medical Toxicology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Motahareh Boozari
- Department of Pharmacognosy, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
6
|
Sun L, Li X, Xiao Y, Yu W, Chen X, Wang Z, Xia N, Chen X, Chen M, Zhu H, Li J, Wei J, Han S, Pu L. Mfsd2a suppresses colorectal cancer progression and liver metastasis via the S100A14/STAT3 axis. J Transl Med 2025; 23:59. [PMID: 39806334 PMCID: PMC11726956 DOI: 10.1186/s12967-024-05994-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] [Received: 09/22/2024] [Accepted: 12/13/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Colorectal cancer (CRC) exhibits a high incidence globally, with the liver being the most common site of distant metastasis. At the time of diagnosis, 20-30% of CRC patients already present with liver metastases. Colorectal liver metastasis (CRLM) is a major cause of mortality among CRC patients. The pathogenesis of CRLM involves complex molecular mechanisms and the hepatic immune microenvironment, but current clinical prevention and treatment are significantly limited. Recent studies have revealed that the major facilitator superfamily domain containing protein-2a (Mfsd2a) plays a pivotal role in the development and metastasis of various cancers. For instance, Mfsd2a inhibits gastric cancer initiation and progression and may impact angiogenesis. However, the mechanisms by which Mfsd2a influences CRC progression and liver metastasis remain unclear. METHODS In this study, we conducted a survival analysis of Mfsd2a in colorectal cancer using data from the GEPIA and GEO databases, and examined the expression differences between primary tumor (PT) and liver metastasis (LM). We further assessed the clinical significance and prognostic relevance of Mfsd2a through immunohistochemical analysis of tissue samples from 70 CRLM patients. Moreover, Kaplan-Meier analysis was used to perform survival analysis on these patients. The biological function of Mfsd2a in CRLM was confirmed by a series of experiments conducted both in vitro and in vivo. Additionally, we investigated downstream molecular pathways using western blot, Co-immunoprecipitation, immunofluorescence, and mass spectrometry techniques. RESULTS We observed that Mfsd2a expression is reduced in LM compared to PT, and higher Mfsd2a levels are associated with better prognosis in CRLM patients. Furthermore, function assays demonstrated that Mfsd2a suppresses CRC cells proliferation, migration, invasion, and EMT in vitro, while also delaying tumor growth and liver metastasis in vivo. Mechanistically, Mfsd2a interacts with S100A14, enhancing its expression and inhibiting phosphorylation of STAT3. In addition, the STAT3 activator colivelin partially reversed the inhibitory effect of Mfsd2a overexpression on the progression of colorectal cancer and liver metastasis. CONCLUSION In summary, Mfsd2a inhibits colorectal cancer progression and liver metastasis by interacting with S100A14, thereby suppressing the phosphorylation of STAT3. Mfsd2a functions as a tumor suppressor in CRLM and could be a promising therapeutic target for treating CRC patients with liver metastasis.
Collapse
Affiliation(s)
- Linfeng Sun
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Xiangdong Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Yuhao Xiao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Wenjie Yu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Xuyang Chen
- Department of Pathology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
| | - Ziyi Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Nan Xia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Xuejiao Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Minhao Chen
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Haoliang Zhu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Jie Li
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Jie Wei
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China
| | - Sheng Han
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China.
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China.
| | - Liyong Pu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Nanjing, 210029, China.
- Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, Nanjing, 210029, China.
- NHC Key Laboratory of Living Donor Liver Transplantation (Nanjing Medical University), Nanjing, 210029, China.
| |
Collapse
|
7
|
Wang S, Li Y, Lin Y, Li J, Guo L, Wang H, Lin X, Liu Z, Zhang B, Liao Z, Zhang Z. Bioinformatics analysis and experimental verification of the cancer-promoting effect of DHODH in clear cell renal cell carcinoma. Sci Rep 2024; 14:11985. [PMID: 38796629 PMCID: PMC11127953 DOI: 10.1038/s41598-024-62738-0] [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] [Received: 03/25/2024] [Accepted: 05/21/2024] [Indexed: 05/28/2024] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a malignant tumor of the urinary system. To explore the potential mechanisms of DHODH in ccRCC, we analyzed its molecular characteristics using public databases. TCGA pan-cancer dataset was used to analyze DHODH expression in different cancer types and TCGA ccRCC dataset was used to assess differential expression, prognosis correlation, immune infiltration, single-gene, and functional enrichment due to DHODH. The GSCALite and CellMiner databases were employed to explore drugs and perform molecular docking analysis with DHODH. Protein-protein interaction networks and ceRNA regulatory networks of DHODH were constructed using multiple databases. The effect of DHODH on ccRCC was confirmed in vitro. DHODH was highly expressed in ccRCC. Immune infiltration analysis revealed that DHODH may be involved in regulating the infiltration of immunosuppressive cells such as Tregs. Notably, DHODH influenced ccRCC progression by forming regulatory networks with molecules, such as hsa-miR-26b-5p and UMPS and significantly enhanced the malignant characteristics of ccRCC cells. Several drugs, such as lapatinib, silmitasertib, itraconazole, and dasatinib, were sensitive to DHODH expression and exhibited strong molecular binding with it. Thus, DHODH may promote ccRCC progression and is a candidate effective therapeutic target for ccRCC.
Collapse
Affiliation(s)
- Songsong Wang
- Department of Urology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
- School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Yan Li
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Yilong Lin
- School of Medicine, Xiamen University, Xiamen, 361000, China
- The First Affiliated Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China
| | - Junting Li
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Lang Guo
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Haoyu Wang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China
| | - Xinyuan Lin
- College of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, China
| | - Ziming Liu
- Department of Urology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361000, China.
- School of Medicine, Xiamen University, Xiamen, 361000, China.
| | - Bingqi Zhang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| | - Zhengming Liao
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| | - Zhongmin Zhang
- Department of Urology, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, 430061, Hubei, China.
- Hubei Key Laboratory of Theory and Application Research of Liver and Kidney in Traditional Chinese Medicine (Hubei Province Hospital of Traditional Chinese Medicine), Wuhan, 430061, China.
- The First Clinical Medical Institute, Hubei University of Chinese Medicine, Wuhan, 430060, China.
| |
Collapse
|
8
|
Liu M, Tang H, Gao K, Zhang X, Ma Z, Jia Y, Yang Z, Inam M, Gao Y, Wang G, Shan X. Poly (I:C)-Induced microRNA-30b-5p Negatively Regulates the JAK/STAT Signaling Pathway to Mediate the Antiviral Immune Response in Silver Carp ( Hypophthalmichthys molitrix) via Targeting CRFB5. Int J Mol Sci 2024; 25:5712. [PMID: 38891899 PMCID: PMC11172372 DOI: 10.3390/ijms25115712] [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] [Received: 04/20/2024] [Revised: 05/16/2024] [Accepted: 05/22/2024] [Indexed: 06/21/2024] Open
Abstract
In aquaculture, viral diseases pose a significant threat and can lead to substantial economic losses. The primary defense against viral invasion is the innate immune system, with interferons (IFNs) playing a crucial role in mediating the immune response. With advancements in molecular biology, the role of non-coding RNA (ncRNA), particularly microRNAs (miRNAs), in gene expression has gained increasing attention. While the function of miRNAs in regulating the host immune response has been extensively studied, research on their immunomodulatory effects in teleost fish, including silver carp (Hyphthalmichthys molitrix), is limited. Therefore, this research aimed to investigate the immunomodulatory role of microRNA-30b-5p (miR-30b-5p) in the antiviral immune response of silver carp (Hypophthalmichthys molitrix) by targeting cytokine receptor family B5 (CRFB5) via the JAK/STAT signaling pathway. In this study, silver carp were stimulated with polyinosinic-polycytidylic acid (poly (I:C)), resulting in the identification of an up-regulated miRNA (miR-30b-5p). Through a dual luciferase assay, it was demonstrated that CRFB5, a receptor shared by fish type I interferon, is a novel target of miR-30b-5p. Furthermore, it was found that miR-30b-5p can suppress post-transcriptional CRFB5 expression. Importantly, this study revealed for the first time that miR-30b-5p negatively regulates the JAK/STAT signaling pathway, thereby mediating the antiviral immune response in silver carp by targeting CRFB5 and maintaining immune system stability. These findings not only contribute to the understanding of how miRNAs act as negative feedback regulators in teleost fish antiviral immunity but also suggest their potential therapeutic measures to prevent an excessive immune response.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Yunhang Gao
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (M.L.); (H.T.); (K.G.); (X.Z.); (Z.M.); (Y.J.); (Z.Y.); (M.I.); (X.S.)
| | - Guiqin Wang
- Department of Veterinary Medicine, College of Animal Science and Technology, Jilin Agricultural University, Changchun 130118, China; (M.L.); (H.T.); (K.G.); (X.Z.); (Z.M.); (Y.J.); (Z.Y.); (M.I.); (X.S.)
| | | |
Collapse
|
9
|
Andrabi MQ, Kesavan Y, Ramalingam S. Non-coding RNAs as Biomarkers for Survival in Colorectal Cancer Patients. Curr Aging Sci 2024; 17:5-15. [PMID: 36733201 DOI: 10.2174/1874609816666230202101054] [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] [Received: 06/16/2022] [Revised: 11/25/2022] [Accepted: 12/19/2022] [Indexed: 02/04/2023]
Abstract
Colorectal cancer (CRC) has a high incidence and fatality rate worldwide. It ranks second concerning death worldwide. Cancer patients are diagnosed with the disease at a later stage due to the absence of early diagnostic methods, which leads to increased death. With the help of recent advancements in the fields of diagnosis and therapy, the development of novel methods using new targets could be helpful for the long-term survival of CRC patients when CRC is detected early. However, the prognosis for the advanced stage of CRC is abysmal. New biomarkers are emerging as promising alternatives since they can be utilized for early detection of CRC, are simple to use, and non-invasive. Non-coding RNAs (ncRNAs) have been seen to have an aberrant expression in the development of many malignancies, including CRC. In the past two decades, much research has been done on non-coding RNAs, which may be valuable as biomarkers and targets for antitumor therapy. Non-coding RNAs can be employed in detecting and treating CRC. Non-coding RNAs play an essential role in regulating gene expression. This article reviews ncRNAs and their expression levels in CRC patients that could be used as potential biomarkers. Various ncRNAs have been associated with CRC, such as microRNAs, long non-coding RNAs, circular RNAs, etc. The expression of these non-coding RNAs may provide insights into the stages of cancer and the prognosis of cancer patients and therefore proper precautionary measures can be taken to decrease cancer-related deaths.
Collapse
Affiliation(s)
- Mohammad Qasim Andrabi
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Yasodha Kesavan
- Department of Biotechnology, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| | - Satish Ramalingam
- Department of Genetic Engineering, School of Bio-Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu, 603203, India
| |
Collapse
|
10
|
Osei GY, Adu-Amankwaah J, Koomson S, Beletaa S, Asiamah EA, Smith-Togobo C, Razak SRA. MicroRNAs and colorectal cancer: clinical potential and regulatory networks. Mol Biol Rep 2023; 50:9575-9585. [PMID: 37776413 DOI: 10.1007/s11033-023-08810-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 09/08/2023] [Indexed: 10/02/2023]
Abstract
Colorectal cancer (CRC) is a serious global health concern, with a high incidence and mortality rate. Although there have been advancements in the early detection and treatment of CRC, therapy resistance is common. MicroRNAs (miRNAs), a type of small non-coding RNA that regulates gene expression, are key players in the initiation and progression of CRC. Recently, there has been growing attention to the complex interplay of miRNAs in cancer development. miRNAs are powerful RNA molecules that regulate gene expression and have been implicated in various physiological and pathological processes, including carcinogenesis. By identifying current challenges and limitations of treatment strategies and suggesting future research directions, this review aims to contribute to ongoing efforts to enhance CRC diagnosis and treatment. It also provides a comprehensive overview of the role miRNAs play in CRC carcinogenesis and explores the potential of miRNA-based therapies as a treatment option. Importantly, this review highlights the exciting potential of targeted modulation of miRNA function as a therapeutic approach for CRC.
Collapse
Affiliation(s)
- George Yiadom Osei
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Pulau Pinang, 13200, Malaysia
- Department of Medical Laboratory Sciences, University of Health and Allied Sciences, PMB 31, Ho, Ghana
| | - Joseph Adu-Amankwaah
- Department of Physiology, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Selina Koomson
- Department of Medical Laboratory Sciences, University of Health and Allied Sciences, PMB 31, Ho, Ghana
| | - Solomon Beletaa
- Department of Medical Laboratory Sciences, University of Health and Allied Sciences, PMB 31, Ho, Ghana
| | - Emmanuel Akomanin Asiamah
- Department of Medical Laboratory Sciences, University of Health and Allied Sciences, PMB 31, Ho, Ghana
- Discipline of Public Health Medicine, School of Nursing and Public Health, University of KwaZulu-Natal, Durban, 4001, South Africa
- Cancer and Infectious Diseases Epidemiology Research Unit (CIDERU), College of Health Sciences, University of KwaZulu-Natal, Durban, 4001, South Africa
| | - Cecilia Smith-Togobo
- Department of Medical Laboratory Sciences, University of Health and Allied Sciences, PMB 31, Ho, Ghana
| | - Siti Razila Abdul Razak
- Department of Biomedical Sciences, Advanced Medical and Dental Institute, Universiti Sains Malaysia, Kepala Batas, Pulau Pinang, 13200, Malaysia.
| |
Collapse
|
11
|
Chen X, Tu J, Liu C, Wang L, Yuan X. MicroRNA-621 functions as a metastasis suppressor in colorectal cancer by directly targeting LEF1 and suppressing Wnt/β-catenin signaling. Life Sci 2022; 308:120941. [PMID: 36087740 DOI: 10.1016/j.lfs.2022.120941] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 08/28/2022] [Accepted: 09/05/2022] [Indexed: 11/15/2022]
Abstract
AIMS Colorectal liver metastasis (CRLM) is the leading death-causing among colorectal cancer (CRC) patients. Recently, a novel tumor-related microRNA, miR-621, has been identified as a tumor suppressor in diverse tumor types, but its role in CRLM remains unclear and requires further investigation. MAIN METHODS To elucidate novel regulators of CRLM progression, we used a well-established CRLM animal model. After serially transplanting human colon carcinoma cell lines Caco-2 into the liver, we obtained liver metastatic variants that exhibited a strong ability for invasion and metastasis. High-throughput sequencing was conducted on these newly established cell lines. After comparison and prediction between the two cell lines: parental Caco-2 (hereafter referred to as F0) and F3, miR-621 was identified as a candidate regulator for lymphoid enhancer-binding factor 1 (LEF1) expression. Further validation was achieved with dual-luciferase reporter assay. KEY FINDINGS The gain- and loss-of-function validation showed that miR-621 inhibits cell viability, cell cycle progression, colony formation, and proliferation in vitro. Meanwhile, miR-621 could reverse EMT malignant phenotype. LEF1, an important downstream mediator of activated Wnt/β-catenin signaling pathway, was validated as the direct functional target of miR-621. miR-621 interacts directly with the LEF1 3'-UTR and post-transcriptionally suppresses LEF1 expression. Moreover, LEF1 overexpression reversed the effect of miR-621. LEF1 silencing counteracted miR-621 down-regulation-induced effects. Further in vivo experiments revealed that miR-621 over-expression suppressed CRLM, but LEF1 abrogated the inhibitory effect of miR-621. SIGNIFICANCE MiR-621 is a vital tumor suppressor in CRC and could be a promising anti-cancer therapeutic target.
Collapse
Affiliation(s)
- Xinyi Chen
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang road 1095, Wuhan, Hubei Province, China
| | - Jingyao Tu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang road 1095, Wuhan, Hubei Province, China
| | - Chaofan Liu
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang road 1095, Wuhan, Hubei Province, China
| | - Lu Wang
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang road 1095, Wuhan, Hubei Province, China.
| | - Xianglin Yuan
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Jie Fang road 1095, Wuhan, Hubei Province, China.
| |
Collapse
|
12
|
Ge T, Zhang Y. Tanshinone IIA reverses oxaliplatin resistance in colorectal cancer through microRNA-30b-5p/AVEN axis. Open Med (Wars) 2022; 17:1228-1240. [PMID: 35892081 PMCID: PMC9281591 DOI: 10.1515/med-2022-0512] [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: 10/08/2021] [Revised: 05/11/2022] [Accepted: 05/28/2022] [Indexed: 12/13/2022] Open
Abstract
This research aims to explore the role of Tanshinone IIA (Tan IIA) and microRNA (miR)-30b-5p in chemoresistance of colorectal cancer (CRC). The expression levels of miR-30b-5p and apoptosis and caspase activation inhibitor (AVEN) was detected by reverse transcription-quantitative polymerase chain reaction assay. The cell proliferation and apoptosis were examined by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) and flow cytometry assays. The target relationship between miR-30b-5p and AVEN was confirmed by Dual-luciferase reporter assay. Transwell assay was performed to assess CRC cells’ metastasis. Western blot was carried out to measure the apoptosis-related protein. The results showed that miR-30b-5p was lowly expressed in oxaliplatin-resistance CRC cells SW480 (SW480/R) compared to SW480 cells. Overexpression of miR-30b-5p significantly suppressed the malignant biological behaviors of SW480/R cells and significantly promoted the sensitivity of SW480/R cells to oxaliplatin by down-regulated AVEN expression. Besides, Tan IIA treatment upregulated miR-30b-5p expression in SW480/R cells. Moreover, miR-30b-5p upregulation strengthened the promoting effect of Tan IIA on the sensitivity of SW480/R cells to oxaliplatin. In conclusion, Tan IIA and miR-30b-5p could reverse oxaliplatin resistance of CRC cells and may thus be potential treatment strategies for treating patients with CRC.
Collapse
Affiliation(s)
- Tingrui Ge
- Department of Colorectal Surgery, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| | - Yonggang Zhang
- Department of Colorectal Surgery, The First People's Hospital of Lianyungang, Lianyungang, Jiangsu 222002, P.R. China
| |
Collapse
|
13
|
The E6 Oncoprotein of HPV16 AA-c Variant Regulates Cell Migration through the MINCR/miR-28-5p/RAP1B Axis. Viruses 2022; 14:v14050963. [PMID: 35632705 PMCID: PMC9143115 DOI: 10.3390/v14050963] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/30/2022] [Accepted: 05/03/2022] [Indexed: 01/03/2023] Open
Abstract
The E6 oncoprotein of HPV16 variants differentially alters the transcription of the genes involved in migration and non-coding RNAs such as lncRNAs. The role of the lncRNA MINCR in cervical cancer and its relationship with variants of oncogenic HPV remain unknown. Therefore, the objective of this study was to analyze the effect of the E6 oncoprotein of the AA-c variant of HPV16 in cell migration through the MINCR/miR-28-5p/RAP1B axis. To explore the functional role of MINCR in CC, we used an in vitro model of C33-A cells with exogenous expression of the E6 oncoprotein of the AA-c variant of HPV16. Interfering RNAs performed MINCR silencing, and the expression of miR-28-5p and RAP1B mRNA was analyzed by RT-qPCR. We found that C33-A/AA-c cells expressed MINCR 8-fold higher compared to the control cells. There is an inverse correlation between the expression of miR-28-5p and RAP1B in C33-A/AA-c cells. Our results suggest that MINCR might regulate the expression of RAP1B through the inhibition of miR-28-5p in CC cells expressing the E6 oncoprotein of HPV16 AA-c. We report, for the first time, that the MINCR/miR-28-5p/RAP1B axis positively regulates cell migration in CC-derived cells that express the E6 oncoprotein of the AA-c variant of HPV16.
Collapse
|
14
|
Zhou H, Liu Z, Wang Y, Wen X, Amador EH, Yuan L, Ran X, Xiong L, Ran Y, Chen W, Wen Y. Colorectal liver metastasis: molecular mechanism and interventional therapy. Signal Transduct Target Ther 2022; 7:70. [PMID: 35246503 PMCID: PMC8897452 DOI: 10.1038/s41392-022-00922-2] [Citation(s) in RCA: 144] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Revised: 01/25/2022] [Accepted: 02/09/2022] [Indexed: 02/08/2023] Open
Abstract
Colorectal cancer (CRC) is one of the most frequently occurring malignancy tumors with a high morbidity additionally, CRC patients may develop liver metastasis, which is the major cause of death. Despite significant advances in diagnostic and therapeutic techniques, the survival rate of colorectal liver metastasis (CRLM) patients remains very low. CRLM, as a complex cascade reaction process involving multiple factors and procedures, has complex and diverse molecular mechanisms. In this review, we summarize the mechanisms/pathophysiology, diagnosis, treatment of CRLM. We also focus on an overview of the recent advances in understanding the molecular basis of CRLM with a special emphasis on tumor microenvironment and promise of newer targeted therapies for CRLM, further improving the prognosis of CRLM patients.
Collapse
Affiliation(s)
- Hui Zhou
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Zhongtao Liu
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Yongxiang Wang
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Xiaoyong Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Eric H Amador
- Department of Physics, The University of Texas, Arlington, TX, 76019, USA
| | - Liqin Yuan
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China
| | - Xin Ran
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Li Xiong
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China.
| | - Yuping Ran
- Department of Dermatovenereology, West China Hospital, Sichuan University, Chengdu, Sichuan Province, China
| | - Wei Chen
- Department of Physics, The University of Texas, Arlington, TX, 76019, USA.
- Medical Technology Research Centre, Chelmsford Campus, Anglia Ruskin University, Chelmsford, CM1 1SQ, UK.
| | - Yu Wen
- Department of General Surgery, Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan Province, China.
| |
Collapse
|
15
|
Jia Z, An J, Liu Z, Zhang F. Non-Coding RNAs in Colorectal Cancer: Their Functions and Mechanisms. Front Oncol 2022; 12:783079. [PMID: 35186731 PMCID: PMC8847166 DOI: 10.3389/fonc.2022.783079] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/12/2022] [Indexed: 12/12/2022] Open
Abstract
Colorectal cancer (CRC) is a common malignancy with high mortality. However, the molecular mechanisms underlying CRC remain unclear. Controversies over the exact functions of non-coding RNAs (ncRNAs) in the progression of CRC have been prevailing for multiple years. Recently, accumulating evidence has demonstrated the regulatory roles of ncRNAs in various human cancers, including CRC. The intracellular signaling pathways by which ncRNAs act on tumor cells have been explored, and in CRC, various studies have identified numerous dysregulated ncRNAs that serve as oncogenes or tumor suppressors in the process of tumorigenesis through diverse mechanisms. In this review, we have summarized the functions and mechanisms of ncRNAs (mainly lncRNAs, miRNAs, and circRNAs) in the tumorigenesis of CRC. We also discuss the potential applications of ncRNAs as diagnostic and prognostic tools, as well as therapeutic targets in CRC. This review details strategies that trigger the recognition of CRC-related ncRNAs, as well as the methodologies and challenges of studying these molecules, and the forthcoming clinical applications of these findings.
Collapse
Affiliation(s)
- Zimo Jia
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| | - Jiaqi An
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China
| | - Ziyuan Liu
- School of Medicine, Shihezi University, Shihezi, China
| | - Fan Zhang
- Department of Biochemistry and Molecular Biology, Hebei Medical University, Shijiazhuang, China.,The Key Laboratory of Neural and Vascular Biology, Ministry of Education, Shijiazhuang, China
| |
Collapse
|
16
|
Dastmalchi N, Safaralizadeh R, Teimourian S. An updated review of the pre-clinical role of microRNAs and their contribution to colorectal cancer. Curr Mol Med 2021; 22:851-859. [PMID: 34961460 DOI: 10.2174/1566524021666211213122619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 10/18/2021] [Accepted: 10/26/2021] [Indexed: 11/22/2022]
Abstract
Colorectal cancer (CRC) is one of the main causes of malignancy-related mortality worldwide. It was well-identified that microRNAs (miRNAs) decisively participate in cellular biological pathways; in a way that their deregulated expression causes CRC progression. miRNAs can control the translation and degradation of mRNAs by binding to various molecular targets involved in different biological processes, including growth, apoptosis, cell cycle, autophagy, angiogenesis, metastasis, etc. The functions of these dysregulated miRNAs may be either oncogenic or tumor-suppressive. Therefore, these miRNAs can be contributed to prognostic, diagnostic, and therapeutic approaches in CRC. In this study, we reviewed the tumor-suppressive and oncogenic functions of miRNAs in CRC and assessed their molecular activities in CRC development. However, further investigation for the involvement of dysregulated miRNAs in CRC progression is required.
Collapse
Affiliation(s)
- Narges Dastmalchi
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | - Reza Safaralizadeh
- Department of Animal Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz. Iran
| | - Shahram Teimourian
- Department of Medical Genetics, School of medicine, Iran University of Medical Sciences (IUMS), Tehran. Iran
| |
Collapse
|
17
|
Wang Y, Wang C, Fu Z, Zhang S, Chen J. miR-30b-5p inhibits proliferation, invasion, and migration of papillary thyroid cancer by targeting GALNT7 via the EGFR/PI3K/AKT pathway. Cancer Cell Int 2021; 21:618. [PMID: 34819077 PMCID: PMC8611849 DOI: 10.1186/s12935-021-02323-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2021] [Accepted: 11/09/2021] [Indexed: 12/28/2022] Open
Abstract
BACKGROUND Papillary thyroid carcinoma (PTC) is a common endocrine tumor. Increasing evidence has shown that microRNA dysfunction is involved in the occurrence and development of cancer. The expression of MicroRNA-30b-5p (miR-30b-5p) was down-regulated in PTC; however, its role in the development of PTC is not clear. Hence, this study aimed to explore the role and mechanism of miR-30b-5p in the occurrence and development of PTC. METHODS The qRT-PCR assay was used to detect the expression of miR-30b-5p in 60 cases of papillary thyroid carcinoma along with their matched non-cancerous tissues. This study explored the biological function of miR-30b-5p by the functional gain and loss experiments in vitro and vivo. The direct target gene of miR-30b-5p and its signaling pathway was identified through bioinformatics analysis, qRT-PCR, western blot, rescue experiments, and double luciferase 3'-UTR report analysis. RESULTS This study demonstrated that the low expression of miR-30b-5p is related to poor clinicopathological features. Functionally, the overexpression of miR-30b-5p inhibited the proliferation, invasion, and migration of PTC cells. Bioinformatics and luciferase analysis showed that GALNT7 is the direct and functional target of miR-30b-5p. Moreover, miR-30b-5p inhibited the proliferation of PTC in vivo by inhibiting the expression of GALNT7. The studies on the mechanism have shown that GALNT7 promotes cell proliferation and invasion by activating EGFR/PI3K/AKT kinase pathway, which can be attenuated by the kinase inhibitors. CONCLUSIONS Overall, miR-30b-5p inhibited the progression of papillary thyroid carcinoma by targeting GALNT7 and inhibiting the EGFR/PI3K/AKT pathway.
Collapse
Affiliation(s)
- Ye Wang
- The First Affiliated Hospital of Guangxi Medical University, Department of Gastrointestinal Gland Surgery, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, 530021, Guangxi, China
| | - Congjun Wang
- The First Affiliated Hospital of Guangxi Medical University, Department of Gastrointestinal Gland Surgery, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, 530021, Guangxi, China
| | - Zhao Fu
- The First Affiliated Hospital of Guangxi Medical University, Department of Gastrointestinal Gland Surgery, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, 530021, Guangxi, China
| | - Siwen Zhang
- The First Affiliated Hospital of Guangxi Medical University, Department of Gastrointestinal Gland Surgery, Nanning, 530021, Guangxi, China
- Guangxi Medical University, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, 530021, Guangxi, China
| | - Junqiang Chen
- The First Affiliated Hospital of Guangxi Medical University, Department of Gastrointestinal Gland Surgery, Nanning, 530021, Guangxi, China.
- Guangxi Medical University, Guangxi Key Laboratory of Enhanced Recovery after Surgery for Gastrointestinal Cancer, Nanning, 530021, Guangxi, China.
| |
Collapse
|
18
|
Wang Y, Tong D, Sun Y, Sun H, Liu F, Zou M, Luo R, Peng X. DF-1 cells prevent MG-HS infection through gga-miR-24-3p/RAP1B mediated decreased proliferation and increased apoptosis. Res Vet Sci 2021; 141:164-173. [PMID: 34749101 DOI: 10.1016/j.rvsc.2021.10.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 09/13/2021] [Accepted: 10/28/2021] [Indexed: 12/26/2022]
Abstract
Mycoplasma gallisepticum (MG) is a major poultry pathogen that can induce Chronic Respiratory Disease (CRD) in chickens, causing serious economic losses in the poultry industry worldwide. Increasing evidence suggests that microRNAs (miRNAs) act as a vital role in resisting microbial pathogenesis and maintaining cellular mechanism. Our previous miRNAs sequencing data showed gga-miR-24-3p expression level was significantly increased in MG-infected chicken lungs. The aim of this study is to reveal the cellular mechanism behind the MG-HS infection. We found that gga-miR-24-3p was significantly upregulated and Ras-related protein-B (RAP1B) was downregulated in chicken fibroblast cells (DF-1) with MG infection. Dual luciferase reporting assay and rescue assay confirmed that RAP1B was the target gene of gga-miR-24-3p. Meanwhile, overexpressed gga-miR-24-3p increased the levels of tumor necrosis factor alpha (TNF-α) and interleukin-1β (IL-1β), and significantly inhibited cell proliferation as well as promoted MG-infected DF-1 cell apoptosis, whereas inhibition of gga-miR-24-3p had the opposite effect. More importantly, the results of overexpression and knockdown of target gene RAP1B demonstrated that the presence of RAP1B promoted cell proliferation and it saved the reduced or increased cell proliferation caused by overexpression or inhibition of gga-miR-24-3p. Furthermore, the overexpression of gga-miR-24-3p could significantly inhibit the expression of MG-HS adhesion protein. Taken together, these findings demonstrate that DF-1 cells can resist MG-HS infection through gga-miR-24-3p/RAP1B mediated decreased proliferation and increased apoptosis, which provides a new mechanism of resistance to MG infection in vitro.
Collapse
Affiliation(s)
- Yingjie Wang
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Deng Tong
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Yingfei Sun
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Huanling Sun
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Fule Liu
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Mengyun Zou
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Ronglong Luo
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China
| | - Xiuli Peng
- Key Laboratory of Agricultural Animal Genetics, Breeding and Reproduction, Ministry of Education, Huazhong Agricultural University, Wuhan, Hubei Province 430070, China.
| |
Collapse
|
19
|
Qiu H, Shen X, Chen B, Chen T, Feng G, Chen S, Feng D, Xu Q. miR-30b-5p inhibits cancer progression and enhances cisplatin sensitivity in lung cancer through targeting LRP8. Apoptosis 2021; 26:261-276. [PMID: 33779882 DOI: 10.1007/s10495-021-01665-1] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2021] [Indexed: 12/24/2022]
Abstract
Accumulated evidence has demonstrated that miRNAs are closely implicated in lung carcinogenesis. Herein, we explored the expression pattern of miR-30b-5p in lung cancer, and aimed to uncover miR-30b-5p roles in lung cancer progression and drug resistance. miR-30b-5p expression profiles in lung cancer tissues and the matched non-tumor tissues were determined by using qPCR. Cell viability, migration, invasion and in vivo tumorigenesis were determined by using the CCK-8, colony formation, wound healing, transwell chambers experiments and tumor xenograft models. RNA immunoprecipitation (RIP) and dual luciferase reporter experiments were applied to evaluate the relationship between miR-30b-5p and LRP8. The results demonstrated that miR-30b-5p showed a low expression profile in lung cancer tissues and cells, and closely linked to poor prognosis and malignant clinical process. Cell viability, migration, invasiveness and tumorigenesis were significantly weakened following miR-30b-5p overexpression in A549 and NCI-H1299 cells, while cell apoptosis rates were increased. In addition, miR-30b-5p was lowly expressed in A549/DDP (a cisplatin drug resistant cell line) as compared with A549 cells, and miR-30b-5p increased A549/DDP cell sensitivity to DDP. However, these above roles of miR-30b-5p were all significantly impaired following the overexpression of LRP8 which was overexpressed in lung cancer tissues. Collectively, this study demonstrated that miR-30b-5p functions as a tumor suppressor in lung cancer, and re-sensitizes lung cancer cells to DDP by targeting LRP8.
Collapse
Affiliation(s)
- Haitao Qiu
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, No. 42, Baiziting, Xuanwu district, Nanjing, 210009, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, 210009, China
| | - Xiaokang Shen
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, No. 42, Baiziting, Xuanwu district, Nanjing, 210009, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, 210009, China
| | - Bing Chen
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, No. 42, Baiziting, Xuanwu district, Nanjing, 210009, China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, 210009, China
| | - Tianming Chen
- Nanjing Medical University, Third Affiliated Hospital, Nanjing, 210009, China
| | - Guodong Feng
- Department of Interventional Therapy, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210009, China
| | - Shilin Chen
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, No. 42, Baiziting, Xuanwu district, Nanjing, 210009, China.
| | - Dongjie Feng
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, No. 42, Baiziting, Xuanwu district, Nanjing, 210009, China.
| | - Qiaoshu Xu
- State Key Laboratory of Bioelectronics, Jiangsu Key Laboratory of Biomaterials and Devices, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
20
|
Liu J, Li G. Identification and validation of a risk signature based on extracellular matrix-related genes in gliomas. Medicine (Baltimore) 2021; 100:e25603. [PMID: 33879726 PMCID: PMC8078288 DOI: 10.1097/md.0000000000025603] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 03/28/2021] [Accepted: 04/01/2021] [Indexed: 12/13/2022] Open
Abstract
ABSTRACT Gliomas have the highest incidence among primary brain tumors, and the extracellular matrix (ECM) plays a vital role in tumor progression. We constructed a risk signature using ECM-related genes to predict the prognosis of patients with gliomas.mRNA and clinical data from glioma patients were downloaded from The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx) and Chinese Glioma Genome Atlas (CGGA) databases. Differentially expressed ECM-related genes were screened, and a risk signature was built using least absolute shrinkage and selection operator (LASSO) Cox regression. Cell type identification by estimating relative subsets of RNA transcripts (CIBERSORT) was used to assess immune infiltration in different risk groups. Gene set enrichment analysis (GSEA) was performed to explore the molecular mechanisms of the genes employed in the risk score.Differentially expressed ECM-related genes were identified, and their associated regulatory mechanisms were predicted via analysis of protein-protein interaction (PPI), transcription factor (TF) regulatory and TF coexpression networks. The established risk signature considered 17 ECM-related genes. The prognosis of the high-risk group was significantly worse than that of the low-risk group. We used the CGGA database to validate the signature. CIBERSORT indicated that the levels of naive B cells, activated memory CD4 T cells, regulatory T cells, gamma delta T cells, activated NK cells, monocytes, activated dendritic cells and activated mast cells were higher in the high-risk group. The levels of plasma cells, CD8 T cells, naive CD4 T cells, resting memory CD4 T cells, M0 macrophages, M1 macrophages, resting mast cells, and neutrophils were lower in the high-risk group. Ultimately, GSEA showed that the terms intestinal immune network for IgA production, primary immunodeficiency, and ECM receptor interaction were the top 3 terms enriched in the high-risk group. The terms Wnt signaling pathway, ErbB signaling pathway, mTOR signaling pathway, and calcium signaling pathway were enriched in the low-risk group.We built a risk signature to predict glioma prognosis using ECM-related genes. By evaluating immune infiltration and biofunctions, we gained a further understanding of this risk signature. This risk signature could be an effective tool for predicting glioma prognosis.This study did not require ethical approval. We will disseminate our findings by publishing results in a peer-reviewed journal.
Collapse
|
21
|
Adam-Artigues A, Garrido-Cano I, Simón S, Ortega B, Moragón S, Lameirinhas A, Constâncio V, Salta S, Burgués O, Bermejo B, Henrique R, Lluch A, Jerónimo C, Eroles P, Cejalvo JM. Circulating miR-30b-5p levels in plasma as a novel potential biomarker for early detection of breast cancer. ESMO Open 2021; 6:100039. [PMID: 33477007 PMCID: PMC7820029 DOI: 10.1016/j.esmoop.2020.100039] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2020] [Revised: 12/13/2020] [Accepted: 12/17/2020] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND Recently, microRNAs have been demonstrated to be potential non-invasive biomarkers for diagnosis, prognosis assessment or prediction of response to treatment in cancer. In this study, we evaluate the potential of miR-30b-5p as a biomarker for early diagnosis of breast cancer (BC) in tissue and plasma. METHODS Expression of miR-30b-5p was determined in a series of 112 BC and 40 normal breast tissues. Circulating miR-30b-5p levels in plasma samples were determined in a discovery cohort of 38 BC patients and 40 healthy donors and in a validation cohort of 83 BC patients and 83 healthy volunteers. miR-30b-5p expression was measured by quantitative real-time PCR and receiver operating characteristics curve analysis was carried out. RESULTS The miR-30b-5p expression was significantly lower in BC tissue than in healthy breast samples. In contrast, circulating miR-30b-5p levels were significantly higher in BC patients compared with healthy donors. Furthermore, circulating miR-30b-5p levels were significantly higher in patients with positive axillary lymph node and de novo metastatic patients. Receiver operating characteristics curve analysis demonstrated a good diagnostic potential of miR-30b-5p to detect BC even at an early stage of the disease. CONCLUSION Thus, we highlight the potential of miR-30b-5p as a non-invasive, fast, reproducible and cost-effective diagnostic biomarker of BC.
Collapse
Affiliation(s)
| | | | - S Simón
- Biomedical Research Institute INCLIVA, Valencia, Spain; Clinical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - B Ortega
- Biomedical Research Institute INCLIVA, Valencia, Spain; Clinical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - S Moragón
- Biomedical Research Institute INCLIVA, Valencia, Spain; Clinical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain
| | - A Lameirinhas
- Biomedical Research Institute INCLIVA, Valencia, Spain
| | - V Constâncio
- Cancer Biology and Epigenetics Group Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Porto, Portugal
| | - S Salta
- Cancer Biology and Epigenetics Group Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Porto, Portugal
| | - O Burgués
- Biomedical Research Institute INCLIVA, Valencia, Spain; Clinical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - B Bermejo
- Biomedical Research Institute INCLIVA, Valencia, Spain; Clinical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain
| | - R Henrique
- Cancer Biology and Epigenetics Group Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Porto, Portugal; Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar University of Porto (ICBAS-UP), Porto, Portugal
| | - A Lluch
- Clinical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Department of Medicine, Universitat de València, Valencia, Spain
| | - C Jerónimo
- Cancer Biology and Epigenetics Group Research Center, Portuguese Oncology Institute of Porto (CI-IPOP), Porto, Portugal; Department of Pathology, Portuguese Oncology Institute of Porto, Porto, Portugal; Department of Pathology and Molecular Immunology, Institute of Biomedical Sciences Abel Salazar University of Porto (ICBAS-UP), Porto, Portugal
| | - P Eroles
- Biomedical Research Institute INCLIVA, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain; Department of Physiology, Universitat de València, València, Spain.
| | - J M Cejalvo
- Biomedical Research Institute INCLIVA, Valencia, Spain; Clinical Oncology Department, Hospital Clínico Universitario de Valencia, Valencia, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Madrid, Spain.
| |
Collapse
|
22
|
Gao S, Shi P, Tian Z, Yang X, Liu N. Overexpression of miR-1225 promotes the progression of breast cancer, resulting in poor prognosis. Clin Exp Med 2021; 21:287-296. [PMID: 33423149 DOI: 10.1007/s10238-020-00676-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 11/21/2020] [Indexed: 11/26/2022]
Abstract
Breast cancer is the most common cancer among women, with metastasis as the principal cause of mortality. MiR-1225 has been reported to play roles in the progression of various cancers, but its role in breast cancer was unclear. The expression of miR-1225 was investigated in breast cancer tissues and cells by quantitative real-time PCR. The role of miR-1225 in the cell process of OS was analyzed by CCK-8 assay and Transwell assay. The prognostic value of miR-1225 was evaluated by Kaplan-Meier survival curves and Cox regression analysis. miR-1225 was significantly upregulated in breast cancer tissues, which was associated with the TNM stage of breast cancer patients. The prognosis of patients with high miR-1225 expression was worse than that of patients with low miR-1225 expression, which indicated that miR-1225 acted as an independent factor for the prognosis of breast cancer. Additionally, the upregulation of miR-1225 promoted cell proliferation, migration, and invasion of breast cancer, which suggested miR-1225 might be involved in the progression of breast cancer. JAK1 was identified as the direct target of miR-1225, which was also involved in cell proliferation, migration, and invasion of breast cancer. The overexpression of miR-1225 in breast cancer indicates a poor prognosis of patients and promotes the progression of breast cancer by targeting JAK1. miR-1225 may be a biomarker and therapeutic target for the treatment of breast cancer.
Collapse
Affiliation(s)
- Shangfa Gao
- Department of General Surgery, Chengwu People's Hospital Affiliated to Shandong First Medical University, Heze, 274200, Shandong, China
| | - Peng Shi
- Department of Gland Surgery, Shandong Provincial Hospital, Jinan, 250021, Shandong, China
| | - Zhishuai Tian
- Department of General Surgery, Chengwu People's Hospital Affiliated to Shandong First Medical University, Heze, 274200, Shandong, China
| | - Xingwang Yang
- Department of General Surgery, Zibo City Linzi District People's Hospital, No. 139, Huangong Road, Zibo, 255400, Shandong, China.
| | - Ning Liu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, No. 126 Xi'antai Avenue, Changchun City, 130033, Jilin Province, China.
| |
Collapse
|
23
|
Tombolan L, Millino C, Pacchioni B, Cattelan M, Zin A, Bonvini P, Bisogno G. Circulating miR-26a as Potential Prognostic Biomarkers in Pediatric Rhabdomyosarcoma. Front Genet 2020; 11:606274. [PMID: 33362864 PMCID: PMC7758343 DOI: 10.3389/fgene.2020.606274] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
Rhabdomyosarcoma (RMS) arises from myogenic precursors that fail to complete muscle differentiation and represents the most frequent soft tissue sarcoma in children. Two major histological subtypes are recognized: alveolar RMS, characterized by a more aggressive behavior and a greater proneness to metastasis, and embryonal RMS which accounts for the 80% of cases and carries a better prognosis. Despite the survival of patients with localized tumors has progressively improved, RMS remains a challenging disease especially for metastatic patients and in case of progressive or recurrent disease after front-line therapy. MicroRNAs, a class of small non-coding RNA, have emerged as crucial players in cancer development and progression, and their detection in plasma (circulating miRNAs) represents a promising minimally invasive approach that deserve to be exploited in clinical practice. We evaluated the utility of circulating miRNAs as diagnostic and prognostic biomarkers in children with RMS profiling miRNAs from plasma of a small cohort of RMS patients and healthy donors (HD) using a qPCR Cancer Panel. An assessment of hemolysis status of plasma using miR-451/miR-23a ratio was performed as pre-analytical analysis. Statistical analysis revealed that miRNAs expression pattern clearly distinguished RMS patients from HD (p < 0.05). Interestingly, plasma levels of muscle-specific miR-206 were found to be significantly increased in RMS patients compared to HD, whereas levels of three potential tumor-suppressor miRNAs, miR-26a and miR-30b/30c, were found lower. Reduced levels of circulating miR-26a and miR-30b/c were further measured in an independent larger cohort of patients (validation set) by digital droplet PCR. In particular, we evidenced that miR-26a absolute plasma levels were associated with fusion status and adverse outcome (p < 0.05). Taken together, these findings demonstrate the potential of circulating miRNA as diagnostic and prognostic biomarker in children affected by this malignancy and enforced the key role of miR-26a in pediatric rhabdomyosarcoma.
Collapse
Affiliation(s)
- Lucia Tombolan
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, Padua, Italy
| | - Caterina Millino
- Functional Genomics Laboratory, Department of Biology, University of Padua, Padua, Italy
| | - Beniamina Pacchioni
- Functional Genomics Laboratory, Department of Biology, University of Padua, Padua, Italy
| | - Manuela Cattelan
- Department of Statistical Sciences, University of Padua, Padua, Italy
| | - Angelica Zin
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, Padua, Italy
| | - Paolo Bonvini
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, Padua, Italy
| | - Gianni Bisogno
- Institute of Pediatric Research (IRP), Fondazione Città della Speranza, Padua, Italy.,Department of Woman's and Children's Health, Hematology and Oncology Unit, University of Padua, Padua, Italy
| |
Collapse
|
24
|
Shi Q, Sun B, Wang D, Zhu Y, Zhao X, Yang X, Zhang Y. Circ6401, a novel circular RNA, is implicated in repair of the damaged endometrium by Wharton's jelly-derived mesenchymal stem cells through regulation of the miR-29b-1-5p/RAP1B axis. Stem Cell Res Ther 2020; 11:520. [PMID: 33261656 PMCID: PMC7708228 DOI: 10.1186/s13287-020-02027-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/13/2020] [Indexed: 02/08/2023] Open
Abstract
Background Accumulating evidence indicates that mesenchymal stem cells (MSCs) exert tissue repair effects and therapeutic angiogenesis through their noncoding RNAs (ncRNAs). Our previous studies showed that MSCs derived from Wharton’s jelly (WJ-MSCs) can ameliorate damaged human endometrium by promoting angiogenesis. There is limited information on the functions and mechanism of ncRNAs in MSC-induced endometrial repair, and additional studies are needed for more insights. Methods Here, WJ-MSCs were cocultured with or without endometrial stromal cells (ESCs) damaged by mifepristone (cocultured group versus non-cocultured group). TUNEL staining assays, EdU proliferation assays, flow cytometry apoptosis assays, and western blot assays were performed to observe the reparative effect of WJ-MSCs on damaged ESCs. Subsequently, circular RNA (circRNA) and microRNA microarrays were performed between the two groups. A subset of top upregulated circRNAs was validated by qRT-PCR. The functions of circ6401 (hsa_circ_0006401) in WJ-MSCs were investigated using lentivirus-mediated circRNA overexpression assays. The subcellular localization of circ6401 and miR-29b-1-5p in WJ-MSCs was identified by double RNA fluorescence in situ hybridization. Dual-luciferase reporter assays and western blot assays were performed to elucidate the regulatory mechanisms among circ6401, miR-29b-1-5p, and RAP1B. Results WJ-MSCs significantly improved ESC proliferation and upregulated the expression of vascular angiogenesis markers. Circ6401 was upregulated in WJ-MSCs cocultured with damaged ESCs, while miR-29b-1-5p was significantly downregulated. Furthermore, circ6401 was found to bind to miR-29b-1-5p and prevent it from decreasing the level of RAP1B, a crucial protein involved in the VEGF signaling pathway, which promoted angiogenesis and stimulated the proliferation of ESCs. Conclusions Our results showed the abundance and regulation profiles of ncRNAs of WJ-MSCs during repair of damaged ESCs and, for the first time, clarified the underlying mechanism by which circ6401 promotes endometrial repair by WJ-MSCs; thus, demonstrating that circ6401 may serve as a potential therapeutic target.
Collapse
Affiliation(s)
- Qin Shi
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, 19 Xisi Road, Nantong, Jiangsu, 226000, People's Republic of China
| | - Baolan Sun
- Department of Pediatrics, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Di Wang
- Center for Reproductive Medicine, Affiliated Hospital of Nantong University, Nantong, People's Republic of China
| | - Yi Zhu
- Affiliated Maternity and Child Health Care Hospital of Nantong University, Nantong, People's Republic of China
| | - Xinxin Zhao
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, 19 Xisi Road, Nantong, Jiangsu, 226000, People's Republic of China
| | - Xiaoqing Yang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, 19 Xisi Road, Nantong, Jiangsu, 226000, People's Republic of China.
| | - Yuquan Zhang
- Department of Obstetrics and Gynecology, Affiliated Hospital of Nantong University, 19 Xisi Road, Nantong, Jiangsu, 226000, People's Republic of China.
| |
Collapse
|
25
|
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Hashemi F, Samarghandian S, Najafi M. MicroRNAs in cancer therapy: Their involvement in oxaliplatin sensitivity/resistance of cancer cells with a focus on colorectal cancer. Life Sci 2020; 256:117973. [PMID: 32569779 DOI: 10.1016/j.lfs.2020.117973] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/06/2020] [Accepted: 06/10/2020] [Indexed: 02/08/2023]
Abstract
The resistance of cancer cells into chemotherapy has restricted the efficiency of anti-tumor drugs. Oxaliplatin (OX) being an anti-tumor agent/drug is extensively used in the treatment of various cancer diseases. However, its frequent application has led to chemoresistance. As a consequence, studies have focused in finding underlying molecular pathways involved in OX resistance. MicroRNAs (miRs) are short endogenous non-coding RNAs that are able to regulate vital biological mechanisms such as cell proliferation and cell growth. The abnormal expression of miRs occurs in pathological events, particularly cancer. In the present review, we describe the involvement of miRs in OX resistance and sensitivity. The miRs are able to induce the oncogene factors and mechanisms, resulting in stimulation OX chemoresistance. Also, onco-suppressor miRs can enhance the sensitivity of cancer cells into OX chemotherapy and trigger apoptosis and cell cycle arrest, leading to reduced viability and progression of cancer cells. MiRs can also enhance the efficacy of OX chemotherapy. It is worth mentioning that miRs affect various down-stream targets in OX resistance/sensitivity such as STAT3, TGF-β, ATG4B, FOXO1, LATS2, NF-κB and so on. By identification of these miRs and their upstream and down-stream mediators, further studies can focus on targeting them to sensitize cancer cells into OX chemotherapy and induce apoptotic cell death.
Collapse
Affiliation(s)
- Milad Ashrafizadeh
- Department of Basic Science, Faculty of Veterinary Medicine, University of Tabriz, Tabriz, Iran
| | - Ali Zarrabi
- Sabanci University Nanotechnology Research and Application Center (SUNUM), Tuzla 34956, Istanbul, Turkey; Center of Excellence for Functional Surfaces and Interfaces (EFSUN), Faculty of Engineering and Natural Sciences, Sabanci University, Tuzla, Istanbul 34956, Turkey
| | | | - Farid Hashemi
- DVM. Graduated, Young Researcher and Elite Club, Kazerun Branch, Islamic Azad University, Kazeroon, Iran
| | - Fardin Hashemi
- Student Research Committee, Department of Physiotherapy, Faculty of Rehabilitation, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeed Samarghandian
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran
| | - Masoud Najafi
- Radiology and Nuclear Medicine Department, School of Paramedical Sciences, Kermanshah University of Medical Sciences, Kermanshah, Iran.
| |
Collapse
|