1
|
Hou SM, Cheng CY, Chen WL, Chang EM, Lin CY. NGF-TrkA Axis Enhances PDGF-C-Mediated Angiogenesis in Osteosarcoma via miR-29b-3p Suppression: A Potential Therapeutic Strategy Using Larotrectinib. Life (Basel) 2025; 15:99. [PMID: 39860039 PMCID: PMC11766545 DOI: 10.3390/life15010099] [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: 12/10/2024] [Revised: 01/10/2025] [Accepted: 01/12/2025] [Indexed: 01/27/2025] Open
Abstract
Angiogenesis plays a critical role in osteosarcoma (OS) growth and metastasis. While nerve growth factor (NGF) is implicated in cancer progression, its role in OS angiogenesis remains unclear. This study explored NGF's effects on angiogenesis and the underlying molecular mechanisms. Analysis of GEO (GSE16088) data identified five angiogenesis markers significantly upregulated in OS tissues. In vitro experiments demonstrated that NGF enhanced HUVEC tube formation by upregulating platelet-derived growth factor C (PDGF-C) expression and suppressing microRNA-29b-3p (miR-29b-3p). The results of tube formation assays confirmed that NGF stimulation significantly increased the angiogenic capacity of MG63/NGF cells compared to MG63 cells. Furthermore, larotrectinib, a TrkA inhibitor, effectively reduced the migration and invasion abilities of MG63/NGF cells in a dose-dependent manner. These findings suggest that the NGF-TrkA axis promotes PDGF-C-mediated angiogenesis by inhibiting miR-29b-3p signaling. Larotrectinib could serve as a potential therapeutic agent targeting NGF-mediated angiogenesis in OS, offering a promising avenue for treatment.
Collapse
Affiliation(s)
- Sheng-Mou Hou
- Department of Research, Taiwan Blood Services Foundation, Taipei 111, Taiwan;
- The Director’s Office, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
| | - Ching-Yuan Cheng
- Division of Chest Medicine, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan;
| | - Wei-Li Chen
- Graduate Institute of Anatomy and Cell Biology, College of Medicine, National Taiwan University, Taipei 100, Taiwan;
| | - En-Ming Chang
- Department of Respiratory Care, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan;
| | - Chih-Yang Lin
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei 111, Taiwan
| |
Collapse
|
2
|
Chiappetta C, Della Rocca C, Di Cristofano C. Whole-Exome Analysis and Osteosarcoma: A Game Still Open. Int J Mol Sci 2024; 25:13657. [PMID: 39769419 PMCID: PMC11728052 DOI: 10.3390/ijms252413657] [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] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/11/2025] Open
Abstract
Osteosarcoma (OS) is the most prevalent malignant bone tumor in adolescents and young adults. OS cells grow in a permissive local microenvironment which modulates their behavior and facilitates all steps in tumor development (e.g., proliferation/quiescence, invasion/migration, and drug resistance) and contributes to their intrinsic heterogeneity. The lung parenchyma is the most common metastatic site in OS, and metastatic foci are frequently associated with a poor clinical outcome. Although multiple factors may be responsible for the disease, including genetic mutations (e.g., Rb and p53), the molecular mechanism of development of OS remains unclear, and the conventional treatment for OS is still based on a sequential approach that combines chemotherapy and surgery. Also, despite the increase in clinical trials, the survival rates for OS have not improved. Non-specific targeting therapies thus show poor therapeutic effects, along with side effects at high doses. For these reasons, many efforts have been made to characterize the complex genome of OS thanks to the whole-exome analysis, with the aim of identifying predictive biomarkers to give these patients a better therapeutic option. This review aims to summarize and discuss the main recent advances in OS molecular research for precision medicine.
Collapse
Affiliation(s)
| | - Carlo Della Rocca
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy;
| | - Claudio Di Cristofano
- Department of Medico-Surgical Sciences and Biotechnologies, Sapienza University of Rome, 04100 Latina, Italy;
| |
Collapse
|
3
|
Zhang H, Bai Y, Li J, Chen T, Shang G. FBXO22 promotes osteosarcoma progression via regulation of FOXO1 for ubiquitination and degradation. J Cell Mol Med 2024; 28:e70021. [PMID: 39153212 PMCID: PMC11330286 DOI: 10.1111/jcmm.70021] [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: 04/23/2024] [Revised: 07/26/2024] [Accepted: 08/07/2024] [Indexed: 08/19/2024] Open
Abstract
Accumulating evidence has demonstrated that F-box protein 22 (FBXO22) participates in tumour development and progression in various types of human malignancies. However, the functions and detailed molecular mechanisms of FBXO22 in osteosarcoma tumorigenesis and progression remain elusive. In this study, we aimed to determine the effects of FBXO22 on the cell proliferation, migration and invasion of osteosarcoma cells using cell counting kit-8 and Matrigel Transwell approaches. Moreover, we explored the molecular mechanisms by which FBXO22 mediated oncogenesis and progression in osteosarcoma via Western blotting, immunoprecipitation and ubiquitination. We found that FBXO22 depletion inhibited the proliferation, migration and invasion of osteosarcoma cells, whereas FBXO22 overexpression increased the proliferation and motility of osteosarcoma cells. Mechanistically, FBXO22 promoted the ubiquitination and degradation of FoxO1 in osteosarcoma cells. FBXO22 depletion reduced cell proliferation and motility via regulation of FoxO1. Taken together, our findings provide new insight into FBXO22-induced osteosarcoma tumorigenesis. The inhibition of FBXO22 could be a promising strategy for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- He Zhang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Yang Bai
- Department of NursingShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Jiatong Li
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Ting Chen
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| | - Guanning Shang
- Department of OrthopedicsShengjing Hospital of China Medical UniversityShenyangLiaoningChina
| |
Collapse
|
4
|
Fatema K, Wang Y, Pavek A, Larson Z, Nartker C, Plyler S, Jeppesen A, Mehling B, Capecchi MR, Jones KB, Barrott JJ. Arid1a Loss Enhances Disease Progression in a Murine Model of Osteosarcoma. Cancers (Basel) 2024; 16:2725. [PMID: 39123453 PMCID: PMC11311538 DOI: 10.3390/cancers16152725] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/25/2024] [Accepted: 07/28/2024] [Indexed: 08/12/2024] Open
Abstract
Osteosarcoma is an aggressive bone malignancy, molecularly characterized by acquired genome complexity and frequent loss of TP53 and RB1. Obtaining a molecular understanding of the initiating mutations of osteosarcomagenesis has been challenged by the difficulty of parsing between passenger and driver mutations in genes. Here, a forward genetic screen in a genetic mouse model of osteosarcomagenesis initiated by Trp53 and Rb1 conditional loss in pre-osteoblasts identified that Arid1a loss contributes to OS progression. Arid1a is a member of the canonical BAF (SWI/SNF) complex and a known tumor suppressor gene in other cancers. We hypothesized that the loss of Arid1a increases the rate of tumor progression and metastasis. Phenotypic evaluation upon in vitro and in vivo deletion of Arid1a validated this hypothesis. Gene expression and pathway analysis revealed a correlation between Arid1a loss and genomic instability, and the subsequent dysregulation of genes involved in DNA DSB or SSB repair pathways. The most significant of these transcriptional changes was a concomitant decrease in DCLRE1C. Our findings suggest that Arid1a plays a role in genomic instability in aggressive osteosarcoma and a better understanding of this correlation can help with clinical prognoses and personalized patient care.
Collapse
Affiliation(s)
- Kaniz Fatema
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (K.F.); (A.P.); (C.N.); (S.P.); (A.J.); (B.M.)
| | - Yanliang Wang
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132, USA;
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
| | - Adriene Pavek
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (K.F.); (A.P.); (C.N.); (S.P.); (A.J.); (B.M.)
| | - Zachary Larson
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (K.F.); (A.P.); (C.N.); (S.P.); (A.J.); (B.M.)
| | - Christopher Nartker
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (K.F.); (A.P.); (C.N.); (S.P.); (A.J.); (B.M.)
| | - Shawn Plyler
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (K.F.); (A.P.); (C.N.); (S.P.); (A.J.); (B.M.)
| | - Amanda Jeppesen
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (K.F.); (A.P.); (C.N.); (S.P.); (A.J.); (B.M.)
| | - Breanna Mehling
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (K.F.); (A.P.); (C.N.); (S.P.); (A.J.); (B.M.)
| | - Mario R. Capecchi
- Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, UT 84132, USA;
| | - Kevin B. Jones
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132, USA;
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
| | - Jared J. Barrott
- Department of Biomedical and Pharmaceutical Sciences, College of Pharmacy, Idaho State University, Pocatello, ID 83209, USA; (K.F.); (A.P.); (C.N.); (S.P.); (A.J.); (B.M.)
- Department of Oncological Sciences, University of Utah School of Medicine, Salt Lake City, UT 84132, USA;
- Department of Orthopaedics, University of Utah School of Medicine, Salt Lake City, UT 84132, USA
- Huntsman Cancer Institute, Salt Lake City, UT 84112, USA
- Department of Cell Biology and Physiology, Brigham Young University, Provo, UT 84602, USA
- Simmons Center for Cancer Research, Provo, UT 84602, USA
| |
Collapse
|
5
|
Li R, Chen P, Zhou Y, Lang Y, Zhou C, Ren J, Maimaitiyimin A, Chen Z, Liu C, Mainike A, Ding L. LncRNA HOXA-AS3 promotes cell proliferation and invasion via targeting miR-218-5p/FOXP1 axis in osteosarcoma. Sci Rep 2024; 14:16581. [PMID: 39019995 PMCID: PMC11254915 DOI: 10.1038/s41598-024-67596-4] [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: 11/28/2023] [Accepted: 07/12/2024] [Indexed: 07/19/2024] Open
Abstract
Osteosarcoma is an aggressive form of bone cancer and affects the health in children and adolescents. Although conventional treatment improves the osteosarcoma survival, some patients have metastasis and drug resistance, leading to a worse prognosis. Therefore, it is necessary to explore the molecular mechanism of osteosarcoma occurrence and progression, which could discover the novel treatment for osteosarcoma. Long noncoding RNAs (lncRNAs) have been reported to regulate osteosarcoma occurrence and malignant progression. LncRNA HOXA-AS3 facilitates the tumorigenesis and progression in a variety of human cancers. However, the underlying mechanism of lncRNA HOXA-AS3-induced oncogenesis is poorly determined in osteosarcoma. To address this point, we utilized several cellular biological strategies and molecular approaches to explore the biological functions and mechanisms of lncRNA HOXA-AS3 in osteosarcoma cells. We found that lncRNA HOXA-AS3 facilitates cell proliferation and invasion via targeting miR-218-5p/FOXP1 axis in osteosarcoma. In conclusion, lncRNA HOXA-AS3 could be a promising target for osteosarcoma treatment.
Collapse
Affiliation(s)
- Rong Li
- College of Public Health, State Key Laboratory of Special Environment and Health Research in Xinjiang, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Pingbo Chen
- Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
| | - Yubo Zhou
- Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
| | - Yi Lang
- Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, 830017, Xinjiang, China
| | - Changhui Zhou
- College of Public Health, State Key Laboratory of Special Environment and Health Research in Xinjiang, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Jingqin Ren
- College of Public Health, State Key Laboratory of Special Environment and Health Research in Xinjiang, Xinjiang Medical University, Urumqi, Xinjiang, China
| | | | - Zhen Chen
- College of Public Health, State Key Laboratory of Special Environment and Health Research in Xinjiang, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Chengqing Liu
- College of Public Health, State Key Laboratory of Special Environment and Health Research in Xinjiang, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Abasi Mainike
- Xinjiang Medical University Affiliated Fifth Hospital, Urumqi, Xinjiang, China
| | - Lu Ding
- Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, 830017, Xinjiang, China.
- Postdoctoral Research Center on Public Health and Preventive Medicine, Xinjiang Medical University, Urumqi, Xinjiang, China.
| |
Collapse
|
6
|
Huang Y, Pan C, Wu S, Ye F, Yang L. A combination of cuproptosis and lncRNAs predicts the prognosis and tumor immune microenvironment in cervical cancer. Discov Oncol 2024; 15:116. [PMID: 38609663 PMCID: PMC11014842 DOI: 10.1007/s12672-024-00964-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/30/2024] [Indexed: 04/14/2024] Open
Abstract
BACKGROUND Cuproptosis induces proteotoxic stress and eventually leads to cell death. However, the relationship between cuproptosis and lncRNAs in cervical cancer has not been fully elucidated. Therefore, we aim to explore the association among lncRNAs, cuproptosis and clinical features in cervical cancer. METHODS RNA sequencing, genetic mutations, and clinical data of CESC patients were obtained from TCGA. Cuproptosis-associated genes were gathered. WGCNA was used to cluster important modules, and KEGG, GO, GSEA and GSVA were used to explore functional and pathway enrichment. The association between immune microenvironment and cuproptosis-related lncRNAs was performed by using cibersort algorithm and other platforms, including XCELL, TIMER, QUANTISEQ, MCPCOUNTER and EPIC. Fluorescence quantitative PCR was employed to detect the expression of LINC01833 and LINC02321, and CCK-8 and cell scratch assays were used to assess cell proliferation and migration capabilities after LINCRNA interference. RESULTS 202 upregulated and 45 downregulated lncRNAs were selected. The survival analysis showed that there was a statistically significant difference in survival rates between the high-risk and low-risk groups. The prognosis of tumour mutation burden and the degree of immune infiltration were differed noticeably between the high-risk and low-risk groups. BHG712, TL-2-105, FR-180204, Masitinib, TAK-715, ODI-027, JW-7-24-2, and OSI-930 had substantially higher IC50 values in the high-risk group. Notably, we found AL360178.1 was associated with RNF44 E3 ubiquitin ligase expression. In cervical cancer cell lines, LINC01833 and LINC02321 displayed significant upregulation. Efficient siRNA transfection led to a decreased expression of LINC01833 and LINC02321. This knockdown significantly hindered both cell proliferation and migration capabilities in cervical cancer cells compared to the negative control. CONCLUSION In conclusion, we constructed five cuprotosis-related lncRNA prognostic models, which may be new tumor therapeutic targets for the prevention and treatment of cervical cancer.
Collapse
Affiliation(s)
- Yitong Huang
- Department of Gynecological Oncology, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Chenxiang Pan
- Department of Gynecological Oncology, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Suni Wu
- Department of Gynecological Oncology, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Feng Ye
- Department of Gynecological Oncology, Wenzhou Central Hospital, The Dingli Clinical Institute of Wenzhou Medical University, Wenzhou, 325000, Zhejiang, China
| | - Lihua Yang
- Department of Gynecology, Tangshan Maternal and Child Health Hospital, Tangshan, 063000, Hebei, China.
| |
Collapse
|
7
|
Sun M, Shi G, Zhang X, Kan C, Xie S, Peng W, Liu W, Wang P, Zhang R. Deciphering roles of protein post-translational modifications in IgA nephropathy progression and potential therapy. Aging (Albany NY) 2024; 16:964-982. [PMID: 38175721 PMCID: PMC10817402 DOI: 10.18632/aging.205406] [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: 09/05/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024]
Abstract
Immunoglobulin A nephropathy (IgAN), one type of glomerulonephritis, displays the accumulation of glycosylated IgA in the mesangium. Studies have demonstrated that both genetics and epigenetics play a pivotal role in the occurrence and progression of IgAN. Post-translational modification (PTM) has been revealed to critically participate in IgAN development and progression because PTM dysregulation results in impaired degradation of proteins that regulate IgAN pathogenesis. A growing number of studies identify that PTMs, including sialylation, o-glycosylation, galactosylation, phosphorylation, ubiquitination and deubiquitination, modulate the initiation and progression of IgAN. Hence, in this review, we discuss the functions and mechanisms of PTMs in regulation of IgAN. Moreover, we outline numerous compounds that govern PTMs and attenuate IgAN progression. Targeting PTMs might be a useful strategy to ameliorate IgAN.
Collapse
Affiliation(s)
- Mengying Sun
- Department of Nephrology, Zhuhai People’s Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong 519000, China
| | - Guojuan Shi
- Department of Nephrology, Zhuhai People’s Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong 519000, China
| | - Xiaohan Zhang
- Department of Nephrology, Zhuhai People’s Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong 519000, China
| | - Chao Kan
- Department of Nephrology, Zhuhai People’s Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong 519000, China
| | - Shimin Xie
- Department of Nephrology, Zhuhai People’s Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong 519000, China
| | - Weixiang Peng
- Department of Nephrology, Zhuhai People’s Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong 519000, China
| | - Wenjun Liu
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, Zhejiang 310018, China
| | - Peter Wang
- Department of Medicine, Zhejiang Zhongwei Medical Research Center, Hangzhou, Zhejiang 310018, China
| | - Rui Zhang
- Department of Nephrology, Zhuhai People’s Hospital, Zhuhai Clinical Medical College of Jinan University, Zhuhai, Guangdong 519000, China
| |
Collapse
|
8
|
Wang M, Li W, Han G, Bai X, Xie J. MicroRNA-605-3p Inhibited the Growth and Chemoresistance of Osteosarcoma Cells via Negatively Modulating RAF1. Protein Pept Lett 2024; 31:559-568. [PMID: 39076089 PMCID: PMC11497142 DOI: 10.2174/0109298665314658240712051206] [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: 03/05/2024] [Revised: 05/12/2024] [Accepted: 05/30/2024] [Indexed: 07/31/2024]
Abstract
BACKGROUND Osteosarcoma (OS) is the leading cancer-associated mortality in childhood and adolescence. Increasing evidence has demonstrated the key function of microRNAs (miRNAs) in OS development and chemoresistance. Among them, miRNA-605-3p acted as an important tumor suppressor and was frequently down-regulated in multiple cancers. However, the function of miR-650-3p in OS has not been reported. OBJECTIVE The aim of this work is to explore the novel role of miR-605-3p in osteosarcoma and its possible involvement in OS chemotherapy resistance. METHODS The expression levels of miR-605-3p in OS tissues and cells were assessed by reverse transcription quantitative PCR (RT-qPCR). The relevance of miR-605-3p with the prognosis of OS patients was determined by the Kaplan-Meier analysis. Additionally, the influence of miR-605-3p on OS cell growth was analyzed using the cell counting kit-8, colony formation assay, and flow cytometry. The mRNA and protein expression of RAF1 were detected by RT-qPCR and western blot. The binding of miR-605-3p with the 3'-UTR of RAF1 was confirmed by dual-luciferase reporter assay. RESULTS Our results showed that miR-605-3p was markedly decreased in OS tissues and cells. A lower level of miR-605-3p was strongly correlated with lymph node metastasis and poor 5-year overall survival rate of OS patients. In vitro assay found that miR-605-3p suppressed OS cell proliferation and promoted cell apoptosis. Mechanistically, the proto-oncogene RAF1 was seen as a target of miR-605-3p and strongly suppressed by miR-605-3p in OS cells. Restoration of RAF1 markedly eliminated the inhibitory effect of miR-605-3p on OS progression, suggesting RAF1 as a key mediator of miR-605-3p. Consistent with the decreased level of RAF1, miR-605-3p suppressed the activation of both MEK and ERK in OS cells, which are the targets of RAF1. Moreover, lower levels of miR-605-3p were found in chemoresistant OS patients, and downregulated miR-605-3p increased the resistance of OS cells to therapeutic agents. CONCLUSION Our data revealed that miR-605-3p serves as a tumor suppressor gene by regulating RAF1 and increasing the chemosensitivity of OS cells, which provided the novel working mechanism of miR-605-3p in OS. Engineering stable nanovesicles that could efficiently deliver miR-605-3p with therapeutic activity into tumors could be a promising therapeutic approach for the treatment of OS.
Collapse
Affiliation(s)
- Mao Wang
- Department of Bone and Soft Tissue Oncology, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Weina Li
- Department of Radiotherapy, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Guohui Han
- Department of Breast Surgery, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Xiangdong Bai
- Department of Breast Surgery, Shanxi Province Cancer Hospital/ Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences/Cancer Hospital Affiliated to Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jun Xie
- Department of Biochemistry and Molecular Biology, Shanxi Medical University, Taiyuan, 030001, China
| |
Collapse
|
9
|
Zhan J, Li Z, Lin C, Wang D, Yu L, Xiao X. The role of circRNAs in regulation of drug resistance in ovarian cancer. Front Genet 2023; 14:1320185. [PMID: 38152652 PMCID: PMC10751324 DOI: 10.3389/fgene.2023.1320185] [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: 10/11/2023] [Accepted: 12/04/2023] [Indexed: 12/29/2023] Open
Abstract
Ovarian cancer is one of the female reproductive system tumors. Chemotherapy is used for advanced ovarian cancer patients; however, drug resistance is a pivotal cause of chemotherapeutic failure. Hence, it is critical to explore the molecular mechanisms of drug resistance of ovarian cancer cells and to ameliorate chemoresistance. Noncoding RNAs (ncRNAs) have been identified to critically participate in drug sensitivity in a variety of human cancers, including ovarian cancer. Among ncRNAs, circRNAs sponge miRNAs and prevent miRNAs from regulation of their target mRNAs. CircRNAs can interact with DNA or proteins to modulate gene expression. In this review, we briefly describe the biological functions of circRNAs in the development and progression of ovarian cancer. Moreover, we discuss the underneath regulatory molecular mechanisms of circRNAs on governing drug resistance in ovarian cancer. Furthermore, we mention the novel strategies to overcome drug resistance via targeting circRNAs in ovarian cancer. Due to that circRNAs play a key role in modulation of drug resistance in ovarian cancer, targeting circRNAs could be a novel approach for attenuation of chemoresistance in ovarian cancer.
Collapse
Affiliation(s)
- Jun Zhan
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Zhiyi Li
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Changsheng Lin
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Dingding Wang
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Lei Yu
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| | - Xue Xiao
- Department of Obstetrics and Gynecology, West China Second University Hospital, Sichuan University, Chengdu, Sichuan, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
10
|
Wang R, Yang Y, Wang L, Shi Q, Ma H, He S, Feng L, Fang J. SOX2-OT Binds with ILF3 to Promote Head and Neck Cancer Progression by Modulating Crosstalk between STAT3 and TGF-β Signaling. Cancers (Basel) 2023; 15:5766. [PMID: 38136312 PMCID: PMC10742126 DOI: 10.3390/cancers15245766] [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: 10/29/2023] [Revised: 12/01/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Long non-coding RNA (lncRNA) is involved in the progression of head and neck squamous cell carcinoma (HNSCC). The molecular mechanism of lncRNA SOX2-OT in HNSCC remains unclear. Therefore, we aimed to elucidate the oncogenic role of SOX2-OT in HNSCC. QRT-PCR analysis was performed in 61 pairs of HNSCC cancer tissues, adjacent normal tissues, and 68 plasma samples confirmed that lncRNA SOX2-OT was overexpressed in cancer tissues and plasma samples, which served as a poor prognostic factor for HNSCC. The FISH assay demonstrated that SOX2-OT was localized in the nucleus and cytoplasm of HNSCC cell lines. Further, the cell function assay confirmed that SOX2-OT promoted cell proliferation and metastasis in vitro and in vivo. RNA pulldown and RIP assay results revealed that SOX2-OT bonds with ILF3 in HNSCC, and the rescue assay confirmed that SOX2-OT played an oncogenic role depending on ILF3 protein expression. Ingenuity pathway analysis and Western blotting indicated that SOX2-OT regulated HNSCC progression by promoting STAT3 phosphorylation and modulating the crosstalk between STAT3 and TGF-β signaling. These results reveal evidence for the role of SOX2-OT in HNSCC progression and metastasis by binding to ILF3, which may serve as a therapeutic target and prognostic biomarker in HNSCC.
Collapse
Affiliation(s)
- Ru Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Yifan Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Lingwa Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Qian Shi
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Hongzhi Ma
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Shizhi He
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Ling Feng
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| | - Jugao Fang
- Department of Otorhinolaryngology, Head and Neck Surgery, Beijing Tong Ren Hospital, Capital Medical University, 1 Dongjiaominxiang Street, Beijing 100730, China; (R.W.); (Y.Y.); (L.W.); (Q.S.); (H.M.); (S.H.); (L.F.)
- Key Laboratory of Otorhinolaryngology, Head and Neck Surgery, Beijing Institute of Otorhinolaryngology, Beijing 100730, China
| |
Collapse
|
11
|
Guo L, Xiao K, Xie Y, Yang Z, Lei J, Cai L. Overexpression of HSPB6 inhibits osteosarcoma progress through the ERK signaling pathway. Clin Exp Med 2023; 23:5389-5398. [PMID: 37861934 PMCID: PMC10725330 DOI: 10.1007/s10238-023-01216-9] [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: 05/15/2023] [Accepted: 10/05/2023] [Indexed: 10/21/2023]
Abstract
Heat shock protein B6 (HSPB6) plays a certain role in the formation of several cancers, whereas its effect on osteosarcoma remains unclear. In this study, the effect of HSPB6 on osteosarcoma was validated through numerous experiments. HSPB6 was down-regulated in osteosarcoma. As indicated by the result of CCK-8 and colony formation assays, HSPB6 overexpression was likely to inhibit the osteosarcoma cells proliferation, whereas the flow cytometry analysis suggested that apoptosis of osteosarcoma cells was increased after HSPB6 overexpression. Furthermore, transwell and wound healing assays suggested that when HSPB6 was overexpressed, osteosarcoma cells migration and invasion were declined. Moreover, the western blotting assay suggested that the protein level of p-ERK1/2 was down-regulated in osteosarcoma when HSPB6 was overexpressed. Besides, the effect of HSPB6 on osteosarcoma in vivo was examined. As indicated by the result, HSPB6 overexpression was likely to prevent osteosarcoma growth and lung metastasis in vivo. As revealed by the findings of this study, HSPB6 overexpression exerted anticancer effects in osteosarcoma through the ERK signaling pathway and HSPB6 may be suitable target for osteosarcoma molecular therapies.
Collapse
Affiliation(s)
- Liangyu Guo
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Kangwen Xiao
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Yuanlong Xie
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Zhiqiang Yang
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China
| | - Jun Lei
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China.
| | - Lin Cai
- Department of Spine Surgery and Musculoskeletal Tumor, Zhongnan Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
12
|
Liu F, Li W, Jin Z, Ye J. METTL3-mediated m6A modification of circRNF220 modulates miR-330-5p/survivin axis to promote osteosarcoma progression. J Cancer Res Clin Oncol 2023; 149:17347-17360. [PMID: 37838643 PMCID: PMC10657300 DOI: 10.1007/s00432-023-05455-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2023] [Accepted: 10/01/2023] [Indexed: 10/16/2023]
Abstract
BACKGROUND Circular RNAs (circRNAs) play a crucial role in regulating various physiological processes. However, the precise regulatory mechanisms of circRNF220s in osteosarcoma (OS) are not well understood. METHODS The abundances of circRNF220, miR-330-5p, and survivin were determined using qRT-PCR. To assess the m6A accumulation in circRNF220, a methylated RNA immunoprecipitation (Me-RIP) assay was conducted. Cellular multiplication, motility, and invasion were examined using the cell Counting Kit-8 (CCK-8), EdU, colony formation, Transwell, and wound-healing assays. The binding relationships were measured through RNA immunoprecipitation (RIP) and luciferase reporter assays. In vivo functionality was assessed using xenograft models. RESULTS CircRNF220 was identified as being overexpressed in both OS cells and tissues. In vitro experiments demonstrated that silencing circRNF220 impeded the proliferation, invasion, and motility of OS cells. Similarly, in vivo studies confirmed that downregulating circRNF220 inhibited the growth of OS. Further mechanistic investigations unveiled that METTL3-modulated circRNF220 regulated the progression of OS by upregulating survivin expression through acting as a sponge for miR-330-5p. CONCLUSION The modulation of METTL3-regulated circRNF220 has been found to promote the progression of OS by modulating the miR-330-5p/survivin axis. This novel finding suggests a potentially unique approach to managing OS.
Collapse
Affiliation(s)
- Feng Liu
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Wen Li
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Zhihui Jin
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jia Ye
- Department of Orthopedics, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|
13
|
Zhang X, Liu Q, Zhang J, Song C, Han Z, Wang J, Shu L, Liu W, He J, Wang P. The emerging role of lncRNAs in osteoarthritis development and potential therapy. Front Genet 2023; 14:1273933. [PMID: 37779916 PMCID: PMC10538550 DOI: 10.3389/fgene.2023.1273933] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 09/04/2023] [Indexed: 10/03/2023] Open
Abstract
Osteoarthritis impairs the functions of various joints, such as knees, hips, hands and spine, which causes pain, swelling, stiffness and reduced mobility in joints. Multiple factors, including age, joint injuries, obesity, and mechanical stress, could contribute to osteoarthritis development and progression. Evidence has demonstrated that genetics and epigenetics play a critical role in osteoarthritis initiation and progression. Noncoding RNAs (ncRNAs) have been revealed to participate in osteoarthritis development. In this review, we describe the pivotal functions and molecular mechanisms of numerous lncRNAs in osteoarthritis progression. We mention that long noncoding RNAs (lncRNAs) could be biomarkers for osteoarthritis diagnosis, prognosis and therapeutic targets. Moreover, we highlight the several compounds that alleviate osteoarthritis progression in part via targeting lncRNAs. Furthermore, we provide the future perspectives regarding the potential application of lncRNAs in diagnosis, treatment and prognosis of osteoarthritis.
Collapse
Affiliation(s)
- Xiaofeng Zhang
- Department of Traumatology, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, Zhejiang, China
| | - Qishun Liu
- Department of Orthopedics, Zhejiang Medical & Health Group Hangzhou Hospital, Hang Gang Hospital, Hangzhou, China
| | - Jiandong Zhang
- Department of Orthopedics and Traumatology, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, Zhejiang, China
| | - Caiyuan Song
- Department of Traumatology, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, Zhejiang, China
| | - Zongxiao Han
- Department of Traumatology, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, Zhejiang, China
| | - Jinjie Wang
- Department of Traumatology, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, Zhejiang, China
| | - Lilu Shu
- Zhejiang Zhongwei Medical Research Center, Department of Medicine, Hangzhou, Zhejiang, China
| | - Wenjun Liu
- Zhejiang Zhongwei Medical Research Center, Department of Medicine, Hangzhou, Zhejiang, China
| | - Jinlin He
- Department of Traumatology, Hangzhou Fuyang Hospital of TCM Orthopedics and Traumatology, Hangzhou, Zhejiang, China
| | - Peter Wang
- Zhejiang Zhongwei Medical Research Center, Department of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Xu J, Hu S, Chen Q, Shu L, Wang P, Wang J. Integrated bioinformatics analysis of noncoding RNAs with tumor immune microenvironment in gastric cancer. Sci Rep 2023; 13:15006. [PMID: 37696973 PMCID: PMC10495442 DOI: 10.1038/s41598-023-41444-3] [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: 05/29/2023] [Accepted: 08/26/2023] [Indexed: 09/13/2023] Open
Abstract
In recent years, molecular and genetic research hotspots of gastric cancer have been investigated, including microRNAs, long noncoding RNAs (lncRNAs) and messenger RNA (mRNAs). The study on the role of lncRNAs may help to develop personalized treatment and identify potential prognostic biomarkers in gastric cancer. The RNA-seq and miRNA-seq data of gastric cancer were downloaded from the TCGA database. Differential analysis of RNA expression between gastric cancer samples and normal samples was performed using the edgeR package. The ceRNA regulatory network was visualized using Cytoscape. KEGG pathway analysis of mRNAs in the ceRNA network was performed using the clusterProfiler package. CIBERSORT was used to distinguish 22 immune cell types and the prognosis-related genes and immune cells were determined using Kaplan-Meier and Cox proportional hazard analyses. To estimate these nomograms, we used receiver operating characteristic and calibration curve studies. The ceRNA regulation network of gastric cancer was built in this study, and the genes in the network were analyzed for prognosis. A total of 980 lncRNAs were differentially expressed, of which 774 were upregulated and 206 were downregulated. A survival study identified 15 genes associated with gastric cancer prognosis, including VCAN-AS1, SERPINE1, AL139002.1, LINC00326, AC018781.1, C15orf54, hsa-miR-145. Monocytes and Neutrophils were associated with the survival rate of gastric cancer. Our research uncovers new ceRNA network for the detection, treatment, and monitoring of gastric cancer.
Collapse
Affiliation(s)
- Jun Xu
- First People's Hospital of Hangzhou Lin'an District, Affiliated Lin'an People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Shengnan Hu
- First People's Hospital of Hangzhou Lin'an District, Affiliated Lin'an People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Qiuli Chen
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China
| | - Lilu Shu
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China
| | - Peter Wang
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China.
| | - Jianjiang Wang
- First People's Hospital of Hangzhou Lin'an District, Affiliated Lin'an People's Hospital, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
15
|
Wu Y, Liang L, Li Q, Shu L, Wang P, Huang S. The role of pyroptosis-related lncRNA risk signature in ovarian cancer prognosis and immune system. Discov Oncol 2023; 14:149. [PMID: 37597098 PMCID: PMC10439870 DOI: 10.1007/s12672-023-00767-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/01/2023] [Accepted: 08/10/2023] [Indexed: 08/21/2023] Open
Abstract
Ovarian cancer is a leading cause of death in females with gynecologic cancers. Pyroptosis is a relatively new discovered programmed cell death that is believed to be associated with inflammation. However, studies on pyroptosis-related lncRNAs in ovarian cancer are limited. In this study, we identified 29 pyroptosis-related genes and screened out 72 pyroptosis-related lncRNAs. Furthermore, the 72 lncRNAs were eliminated to 2 survival-related lncRNAs using Cox regression and Lasso regression to build an ovarian cancer prognostic prediction signature and were further validated on the test set. We adopted a riskscore from the two-gene signature, and the survival in low-risk group was higher than the high-risk group. Functional enrichment analysis indicated that the differentially expressed genes (DEGs) between two risk groups were associated with tumor immunity. This study implies that pyroptosis-related genes are closely related to tumor immunity and could be potential therapeutic factors for ovarian cancer treatment.
Collapse
Affiliation(s)
- Yanling Wu
- Department of Gynecology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518067, Guangdong, China
| | - Lei Liang
- Department of Gynecology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518067, Guangdong, China
| | - Qin Li
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China
| | - Lilu Shu
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China
| | - Peter Wang
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, 310018, Zhejiang, China.
| | - Shufeng Huang
- Department of Gynecology, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, 518067, Guangdong, China.
| |
Collapse
|
16
|
Wang X, Zhang Y, Wu Y, Cheng H, Wang X. The role of E3 ubiquitin ligases and deubiquitinases in bladder cancer development and immunotherapy. Front Immunol 2023; 14:1202633. [PMID: 37215134 PMCID: PMC10196180 DOI: 10.3389/fimmu.2023.1202633] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Accepted: 04/21/2023] [Indexed: 05/24/2023] Open
Abstract
Bladder cancer is one of the common malignant urothelial tumors. Post-translational modification (PTMs), including ubiquitination, acetylation, methylation, and phosphorylation, have been revealed to participate in bladder cancer initiation and progression. Ubiquitination is the common PTM, which is conducted by E1 ubiquitin-activating enzyme, E2 ubiquitin-conjugating enzyme and E3 ubiquitin-protein ligase. E3 ubiquitin ligases play a key role in bladder oncogenesis and progression and drug resistance in bladder cancer. Therefore, in this review, we summarize current knowledge regarding the functions of E3 ubiquitin ligases in bladder cancer development. Moreover, we provide the evidence of E3 ubiquitin ligases in regulation of immunotherapy in bladder cancer. Furthermore, we mention the multiple compounds that target E3 ubiquitin ligases to improve the therapy efficacy of bladder cancer. We hope our review can stimulate researchers and clinicians to investigate whether and how targeting E3 ubiquitin ligases acts a novel strategy for bladder cancer therapy.
Collapse
|
17
|
Leitner N, Ertl R, Gabner S, Fuchs-Baumgartinger A, Walter I, Hlavaty J. Isolation and Characterization of Novel Canine Osteosarcoma Cell Lines from Chemotherapy-Naïve Patients. Cells 2023; 12:cells12071026. [PMID: 37048099 PMCID: PMC10093184 DOI: 10.3390/cells12071026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/18/2023] [Accepted: 03/20/2023] [Indexed: 03/30/2023] Open
Abstract
The present study aimed to establish novel canine osteosarcoma cell lines (COS3600, COS3600B, COS4074) and characterize the recently described COS4288 cells. The established D-17 cell line served as a reference. Analyzed cell lines differed notably in their biological characteristics. Calculated doubling times were between 22 h for COS3600B and 426 h for COS4074 cells. COS3600B and COS4288 cells produced visible colonies after anchorage-independent growth in soft agar. COS4288 cells were identified as cells with the highest migratory capacity. All cells displayed the ability to invade through an artificial basement membrane matrix. Immunohistochemical analyses revealed the mesenchymal origin of all COS cell lines as well as positive staining for the osteosarcoma-relevant proteins alkaline phosphatase and karyopherin α2. Expression of p53 was confirmed in all tested cell lines. Gene expression analyses of selected genes linked to cellular immune checkpoints (CD270, CD274, CD276), kinase activity (MET, ERBB2), and metastatic potential (MMP-2, MMP-9) as well as selected long non-coding RNA (MALAT1) and microRNAs (miR-9, miR-34a, miR-93) are provided. All tested cell lines were able to grow as multicellular spheroids. In all spheroids except COS4288, calcium deposition was detected by von Kossa staining. We believe that these new cell lines serve as useful biological models for future studies.
Collapse
Affiliation(s)
- Natascha Leitner
- Institute of Morphology, Working Group Histology, University of Veterinary Medicine, Veterinaerplatz 1, A-1210 Vienna, Austria
| | - Reinhard Ertl
- VetCore Facility for Research, University of Veterinary Medicine, Veterinaerplatz 1, A-1210 Vienna, Austria
| | - Simone Gabner
- Institute of Morphology, Working Group Histology, University of Veterinary Medicine, Veterinaerplatz 1, A-1210 Vienna, Austria
| | | | - Ingrid Walter
- Institute of Morphology, Working Group Histology, University of Veterinary Medicine, Veterinaerplatz 1, A-1210 Vienna, Austria
- VetCore Facility for Research, University of Veterinary Medicine, Veterinaerplatz 1, A-1210 Vienna, Austria
| | - Juraj Hlavaty
- Institute of Morphology, Working Group Histology, University of Veterinary Medicine, Veterinaerplatz 1, A-1210 Vienna, Austria
- Correspondence: ; Tel.: +431-250-77-3402; Fax: +431-250-77-3490
| |
Collapse
|
18
|
Lian B, Chen X, Shen K. Inhibition of histone deacetylases attenuates tumor progression and improves immunotherapy in breast cancer. Front Immunol 2023; 14:1164514. [PMID: 36969235 PMCID: PMC10034161 DOI: 10.3389/fimmu.2023.1164514] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 02/27/2023] [Indexed: 03/11/2023] Open
Abstract
Breast cancer is one of the common malignancies with poor prognosis worldwide. The treatment of breast cancer patients includes surgery, radiation, hormone therapy, chemotherapy, targeted drug therapy and immunotherapy. In recent years, immunotherapy has potentiated the survival of certain breast cancer patients; however, primary resistance or acquired resistance attenuate the therapeutic outcomes. Histone acetyltransferases induce histone acetylation on lysine residues, which can be reversed by histone deacetylases (HDACs). Dysregulation of HDACs via mutation and abnormal expression contributes to tumorigenesis and tumor progression. Numerous HDAC inhibitors have been developed and exhibited the potent anti-tumor activity in a variety of cancers, including breast cancer. HDAC inhibitors ameliorated immunotherapeutic efficacy in cancer patients. In this review, we discuss the anti-tumor activity of HDAC inhibitors in breast cancer, including dacinostat, belinostat, abexinostat, mocetinotat, panobinostat, romidepsin, entinostat, vorinostat, pracinostat, tubastatin A, trichostatin A, and tucidinostat. Moreover, we uncover the mechanisms of HDAC inhibitors in improving immunotherapy in breast cancer. Furthermore, we highlight that HDAC inhibitors might be potent agents to potentiate immunotherapy in breast cancer.
Collapse
Affiliation(s)
| | | | - Kunwei Shen
- *Correspondence: Xiaosong Chen, ; Kunwei Shen,
| |
Collapse
|
19
|
Zhang X, Wang X, Hou L, Xu Z, Liu Y, Wang X. Nanoparticles overcome adaptive immune resistance and enhance immunotherapy via targeting tumor microenvironment in lung cancer. Front Pharmacol 2023; 14:1130937. [PMID: 37033636 PMCID: PMC10080031 DOI: 10.3389/fphar.2023.1130937] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Accepted: 03/16/2023] [Indexed: 04/11/2023] Open
Abstract
Lung cancer is one of the common malignant cancers worldwide. Immune checkpoint inhibitor (ICI) therapy has improved survival of lung cancer patients. However, ICI therapy leads to adaptive immune resistance and displays resistance to PD-1/PD-L1 blockade in lung cancer, leading to less immune response of lung cancer patients. Tumor microenvironment (TME) is an integral tumor microenvironment, which is involved in immunotherapy resistance. Nanomedicine has been used to enhance the immunotherapy in lung cancer. In this review article, we described the association between TME and immunotherapy in lung cancer. We also highlighted the importance of TME in immunotherapy in lung cancer. Moreover, we discussed how nanoparticles are involved in regulation of TME to improve the efficacy of immunotherapy, including Nanomedicine SGT-53, AZD1080, Nanomodulator NRF2, Cisplatin nanoparticles, Au@PG, DPAICP@ME, SPIO NP@M-P, NBTXR3 nanoparticles, ARAC nanoparticles, Nano-DOX, MS NPs, Nab-paclitaxel, GNPs-hPD-L1 siRNA. Furthermore, we concluded that targeting TME by nanoparticles could be helpful to overcome resistance to PD-1/PD-L1 blockade in lung cancer.
Collapse
Affiliation(s)
- Xin Zhang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Xuemei Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Lijian Hou
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Zheng Xu
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yu’e Liu
- School of Medicine, Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, Tongji University, Shanghai, China
| | - Xueju Wang
- Department of Pathology, China-Japan Union Hospital, Jilin University, Changchun, China
- *Correspondence: Xueju Wang,
| |
Collapse
|
20
|
Wang M, Zhu L, Yang X, Li J, Liu Y, Tang Y. Targeting immune cell types of tumor microenvironment to overcome resistance to PD-1/PD-L1 blockade in lung cancer. Front Pharmacol 2023; 14:1132158. [PMID: 36874015 PMCID: PMC9974851 DOI: 10.3389/fphar.2023.1132158] [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: 12/26/2022] [Accepted: 02/06/2023] [Indexed: 02/17/2023] Open
Abstract
Lung cancer is the common malignant tumor with the highest mortality rate. Lung cancer patients have achieved benefits from immunotherapy, including immune checkpoint inhibitors (ICIs) therapy. Unfortunately, cancer patients acquire adaptive immune resistance, leading to poor prognosis. Tumor microenvironment (TME) has been demonstrated to play a critical role in participating in acquired adaptive immune resistance. TME is associated with molecular heterogeneity of immunotherapy efficacy in lung cancer. In this article, we discuss how immune cell types of TME are correlated with immunotherapy in lung cancer. Moreover, we describe the efficacy of immunotherapy in driven gene mutations in lung cancer, including KRAS, TP53, EGFR, ALK, ROS1, KEAP1, ZFHX3, PTCH1, PAK7, UBE3A, TNF-α, NOTCH, LRP1B, FBXW7, and STK11. We also emphasize that modulation of immune cell types of TME could be a promising strategy for improving adaptive immune resistance in lung cancer.
Collapse
Affiliation(s)
- Man Wang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lijie Zhu
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xiaoxu Yang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiahui Li
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Ying Tang
- Department of Respiratory Medicine, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
21
|
The Role of Tumor Microenvironment in Regulating the Plasticity of Osteosarcoma Cells. Int J Mol Sci 2022; 23:ijms232416155. [PMID: 36555795 PMCID: PMC9788144 DOI: 10.3390/ijms232416155] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 12/07/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
Osteosarcoma (OS) is a malignancy that is becoming increasingly common in adolescents. OS stem cells (OSCs) form a dynamic subset of OS cells that are responsible for malignant progression and chemoradiotherapy resistance. The unique properties of OSCs, including self-renewal, multilineage differentiation and metastatic potential, 149 depend closely on their tumor microenvironment. In recent years, the likelihood of its dynamic plasticity has been extensively studied. Importantly, the tumor microenvironment appears to act as the main regulatory component of OS cell plasticity. For these reasons aforementioned, novel strategies for OS treatment focusing on modulating OS cell plasticity and the possibility of modulating the composition of the tumor microenvironment are currently being explored. In this paper, we review recent studies describing the phenomenon of OSCs and factors known to influence phenotypic plasticity. The microenvironment, which can regulate OSC plasticity, has great potential for clinical exploitation and provides different perspectives for drug and treatment design for OS.
Collapse
|
22
|
Zhou M, Liu L, Wang J, Liu W. The role of long noncoding RNAs in therapeutic resistance in cervical cancer. Front Cell Dev Biol 2022; 10:1060909. [PMID: 36438563 PMCID: PMC9682114 DOI: 10.3389/fcell.2022.1060909] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 10/20/2022] [Indexed: 11/10/2022] Open
Abstract
Cervical cancer is one of the common tumors and often causes cancer-related death in women. Chemotherapy is a common cancer therapy, which displays a pivotal clinical benefit for cancer patients. However, chemoresistance becomes a big obstacle for failure of treatment in cancer patients. Recently, long noncoding RNAs (lncRNAs) have been identified to regulate drug resistance in human cancers, including cervical cancer. In this review, we describe the role of lncRNAs in regulation of chemotherapeutic resistance in cervical cancer. We also discuss the molecular mechanisms of lncRNA-mediated drug resistance in cervical cancer. Moreover, we describe that targeting lncRNAs could reverse drug resistance in cervical cancer. Therefore, lncRNAs could become effective therapeutic targets and chemotherapeutic sensitizers for cervical cancer patients.
Collapse
|
23
|
Chen W, Chen F, Gong M, Jin Z, Shu L, Wang ZW, Wang J. Comprehensive analysis of lncRNA-mediated ceRNA networkfor hepatocellular carcinoma. Front Oncol 2022; 12:1042928. [PMID: 36338699 PMCID: PMC9634570 DOI: 10.3389/fonc.2022.1042928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2022] [Accepted: 09/30/2022] [Indexed: 11/25/2022] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a high-burden cancer. The molecular mechanism of HCC has not been fully elucidated. Notably, current research has revealed a significant function for long non-coding RNAs (lncRNAs) in the prognosis of patients with HCC. Here, this study aims to construct a regulated lncRNA-mediated ceRNA network and find biological targets for the treatment of HCC. Methods Based on the RNA expression patterns from the TCGA, we did an analysis to determine which genes were expressed differently between liver tumor tissues and noncancerous tissues. Then, using bioinformatic tools, we built a lncRNA-miRNA-mRNA ceRNA network and used GO and KEGG functional analyses on the DEmRNAs connected to ceRNA networks. The main lncRNAs in the subnetwork were chosen, and we next looked at the relationships between these lncRNAs and the clinical characteristics of patients with HCC. The prognosis-related genes and immune cells were identified using Kaplan-Meier and Cox proportional hazard analyses, and CIBERSORT was utilized to separate the 22 immune cell types. CCK8 assay was performed to measure cell viability in HCC cells after lncRNA HOTTIP modulation. Results Differentially expressed mRNA and lncRNAs in HCC and paracancerous tissues were identified. There are 245 lncRNAs, 126 miRNAs, and 1980 mRNAs that are expressed differently in liver tumour tissues than in noncancerous cells. Function analysis showed that mRNAs in ceRNA network were significantly enriched in G1/S transition of mototiv cell cycle, positive regulation of cell cycle process, hepatocellular carcinoma, and cancer related pathways. CD8 T cells and T follicular helper cells had a favourable link with a 0.65 correlation coefficient. Additionally, there was a strong correlation between Eosinophils, activated NK cells, and B memory cells. Strikingly, depletion of lncRNA HOTTIP inhibited viability of HCC cells. In addition, miR-205 upregulation suppressed viability of HCC cells, while miR-205 downregulation repressed viability of HCC cells. Notably, miR-205 depletion rescued HOTTIP depletion-mediated suppression of cell viability in HCC. Conclusion A ceRNA network was created by examining the lncRNA, miRNA, and mRNA expression profiles of liver tumours from the TCGA database. LncRNA HOTTIP promoted cell viability via inhibition of miR-205 in HCC cells.
Collapse
Affiliation(s)
- Weiqing Chen
- First People’s Hospital of Hangzhou Lin’an District, Affiliated Lin’an People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Feihua Chen
- First People’s Hospital of Hangzhou Lin’an District, Affiliated Lin’an People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Mouchun Gong
- First People’s Hospital of Hangzhou Lin’an District, Affiliated Lin’an People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Zhaoqing Jin
- First People’s Hospital of Hangzhou Lin’an District, Affiliated Lin’an People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Lilu Shu
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, China
| | - Zhi-wei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Jianjiang Wang, ; Zhi-wei Wang,
| | - Jianjiang Wang
- First People’s Hospital of Hangzhou Lin’an District, Affiliated Lin’an People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Jianjiang Wang, ; Zhi-wei Wang,
| |
Collapse
|
24
|
Yang C, Xiang H, Fu K, Jin L, Yuan F, Xue B, Wang Z, Wang L. Lycorine suppresses cell growth and invasion via down-regulation of NEDD4 ligase in bladder cancer. Am J Cancer Res 2022; 12:4708-4720. [PMID: 36381314 PMCID: PMC9641406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 09/13/2022] [Indexed: 06/16/2023] Open
Abstract
Recent studies have shown that lycorine, a natural alkaloid compound, plays its anti-cancer role in several human malignancies including bladder cancer. However, the molecular mechanism of lycorine-induced antitumor activity has not been sufficiently investigated. The E3 ubiquitin ligase neural precursor cell expressed developmentally downregulated protein 4 (NEDD4, also known as NEDD4-1) plays a crucial role in tumorigenesis and progression of human cancer. Therefore, depletion of NEDD4 could be a prospective therapeutic strategy for the treatment of cancer. In this study, we investigated whether lycorine restrains tumor by inhibiting the expression of NEDD4 in bladder cancer. We observed that lycorine blocked bladder cancer cell proliferation, colony formation, metastasis and invasion. Moreover, we found that overexpression of NEDD4 in bladder cancer cells significantly promoted cell proliferation and motility, whereas downregulating of the NEDD4 gene expression by lycorine or siRNA suppressed cell growth and movement. Notably, lycorine increased gemcitabine sensitivity in bladder cancer cells. Importantly, lycorine significantly reduced tumor growth, whereas overexpression of NEDD4 accelerated tumor growth and rescued lycorine-triggered tumor inhibition in xenograft mouse model. In conclusion, our study demonstrated that lycorine could exert its antineoplastic activity via suppressing NEDD4 pathway in vitro and in vivo. Therefore, inhibition of NEDD4 expression by lycorine might be a potential efficient strategy for bladder cancer.
Collapse
Affiliation(s)
- Chuanlai Yang
- Scientific Research Department, The Second Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Han Xiang
- Department of Urology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Kai Fu
- Department of Urology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Lu Jin
- Department of Urology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Feng Yuan
- Department of Urology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Boxin Xue
- Department of Urology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| | - Zhiwei Wang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical UniversityWenzhou 325027, Zhejiang, China
| | - Lixia Wang
- Department of Urology, The Second Affiliated Hospital of Soochow UniversitySuzhou 215006, Jiangsu, China
| |
Collapse
|
25
|
Cheng Y, Wu X, Xia Y, Liu W, Wang P. The role of lncRNAs in regulation of DKD and diabetes-related cancer. Front Oncol 2022; 12:1035487. [PMID: 36313695 PMCID: PMC9606714 DOI: 10.3389/fonc.2022.1035487] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Accepted: 09/19/2022] [Indexed: 11/23/2022] Open
Abstract
Diabetes mellitus often results in several complications, such as diabetic kidney disease (DKD) and end-stage renal diseases (ESRDs). Cancer patients often have the dysregulated glucose metabolism. Abnormal glucose metabolism can enhance the tumor malignant progression. Recently, lncRNAs have been reported to regulate the key proteins and signaling pathways in DKD development and progression and in cancer patients with diabetes. In this review article, we elaborate the evidence to support the function of lncRNAs in development of DKD and diabetes-associated cancer. Moreover, we envisage that lncRNAs could be diagnosis and prognosis biomarkers for DKD and cancer patients with diabetes. Furthermore, we delineated that targeting lncRNAs might be an alternative approach for treating DKD and cancer with dysregulated glucose metabolism.
Collapse
Affiliation(s)
- Yawei Cheng
- Department of Disease Prevention, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, China
- Hainan Clinical Research Center for Preventive Treatment of Diseases, Haikou, China
- *Correspondence: Yawei Cheng, ; Peter Wang,
| | - Xiaowen Wu
- Department of Disease Prevention, Hainan Province Hospital of Traditional Chinese Medicine, Haikou, China
| | - Yujie Xia
- Department of Food Science and Technology Centers, National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Wenjun Liu
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, China
| | - Peter Wang
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, China
- *Correspondence: Yawei Cheng, ; Peter Wang,
| |
Collapse
|
26
|
Liu Y, Liu H, Sheng B, Pan S, Wang ZW, Zhu X. The functions of lncRNAs in the HPV-negative cervical cancer compared with HPV-positive cervical cancer. Apoptosis 2022; 27:685-696. [PMID: 35980559 DOI: 10.1007/s10495-022-01761-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2022] [Indexed: 12/24/2022]
Abstract
Cervical cancer is one of the most common female malignancies. Human papillomaviruses (HPV) are the main causative agents of virtually all cervical carcinomas. Nevertheless, emerging evidence has demonstrated that a small proportion of cervical cancer patients are HPV negative. Long noncoding RNAs (lncRNAs) have been identified to play a crucial role in cervical cancer development. Here, this review describes the incidence and development of HPV-negative cervical cancer. Moreover, HPV-negative cervical cancers are more likely diagnosed at non-squamous type, older ages, more advanced stage and metastases, and associated with poorer prognosis as compared to HPV-positive cervical cancer. Furthermore, the significant role and functions of lncRNAs underlying HPV-negative cervical cancer is clarified.
Collapse
Affiliation(s)
- Yi Liu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Hejing Liu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Bo Sheng
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Shuya Pan
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Zhi-Wei Wang
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| | - Xueqiong Zhu
- Center for Uterine Cancer Diagnosis & Therapy Research of Zhejiang Province, Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, China.
| |
Collapse
|
27
|
Song Z, Wang X, Chen F, Chen Q, Liu W, Yang X, Zhu X, Liu X, Wang P. LncRNA MALAT1 regulates METTL3-mediated PD-L1 expression and immune infiltrates in pancreatic cancer. Front Oncol 2022; 12:1004212. [PMID: 36212476 PMCID: PMC9533337 DOI: 10.3389/fonc.2022.1004212] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Accepted: 08/15/2022] [Indexed: 12/04/2022] Open
Abstract
Pancreatic cancer is the fourth leading cause of cancer death in the United States. The main methods of treating pancreatic cancer are surgery and chemotherapy, but the treatment efficacy is low with a poor prognosis. Immunotherapy represented by PD-1/PD-L1 has brought a milestone progress in the treatment of pancreatic cancer. However, the unique tumor microenvironment of pancreatic cancer presents challenges for immunotherapy. In addition, m6A is a common RNA modification and a potential molecular target in tumor therapy. The expression pattern of m6A in pancreatic cancer is still unclear. LncRNAs also play an essential role in pancreatic cancer development and treatment. In this study, we found that some m6A regulators were significantly elevated in pancreatic cancer and associated with the expression of PD-1/PD-L1. Moreover, we observed that METTL3 can increase the expression of PD-L1. Notably, METTL3 positively regulates the expression of lncRNA MALAT1 in pancreatic cancer cells. Strikingly, lncRNA MALAT1 increased the expression of PD-L1 in pancreatic cancer cells. This finding indicated that METTL3 regulated the expression of PD-L1 possibly via targeting lncRNA MALAT1 in pancreatic cancer cells. Lastly, MALAT1 governed the viability of pancreatic cancer cells. Taken together, lncRNA MALAT1 is involved in METTL3-mediated promotion of PD-L1 expression in pancreatic cancer.
Collapse
Affiliation(s)
- Zhengwei Song
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaoguang Wang
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Fei Chen
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Qiuli Chen
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, China
| | - Wenjun Liu
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, China
| | - Xiaodan Yang
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xun Zhu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiaorong Liu
- Department of Surgery, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
- *Correspondence: Xiaorong Liu, ; Peter Wang,
| | - Peter Wang
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, China
- *Correspondence: Xiaorong Liu, ; Peter Wang,
| |
Collapse
|
28
|
Mir-29b in Breast Cancer: A Promising Target for Therapeutic Approaches. Diagnostics (Basel) 2022; 12:diagnostics12092139. [PMID: 36140539 PMCID: PMC9497770 DOI: 10.3390/diagnostics12092139] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/26/2022] [Accepted: 08/30/2022] [Indexed: 11/22/2022] Open
Abstract
The miR-29 family comprises miR-29a, miR-29b, and miR-29c, and these molecules play crucial and partially overlapped functions in solid tumors, in which the different isoforms are variously de-regulated and mainly correlated with tumor suppression. miR-29b is the most expressed family member in cancer, in which it is involved in regulating gene expression at both transcriptional and post-transcriptional levels. This review focuses on the role of miR-29b in breast cancer, in which it plays a controversial role as tumor suppressor or onco-miRNA. Here we have highlighted the dual effect of miR-29b on breast tumor features, which depend on the prevailing function of this miRNA, on the mature miR-29b evaluated, and on the breast tumor characteristics. Remarkably, the analyzed miR-29b form emerged as a crucial element in the results obtained by various research groups, as the most abundant miR-29b-3p and the less expressed miR-29b1-5p seem to play distinct roles in breast tumors with different phenotypes. Of particular interest are the data showing that miR-29b1-5p counteracts cell proliferation and migration and reduces stemness in breast tumor cells with a triple negative phenotype. Even if further studies are required to define exactly the role of each miR-29b, our review highlights its possible implication in phenotype-specific management of breast tumors.
Collapse
|
29
|
Cao T, Cui Y, Wang Y, Wu L, Yu K, Chen K, Xia J, Li Y, Wang ZP, Ma J. CACNA1C-AS2 inhibits cell proliferation and suppresses cell migration and invasion via targeting FBXO45 and PI3K/AKT/mTOR pathways in glioma. Apoptosis 2022; 27:979-991. [PMID: 36038736 DOI: 10.1007/s10495-022-01764-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2022] [Indexed: 11/30/2022]
Abstract
Glioma is the most common brain cancer with a poor prognosis, and its underlying molecular mechanisms still needs to be further explored. In the current study, we discovered that an antisense lncRNA, CACNA1C-AS2, suppressed growth, migration and invasion of glioma cells, suggesting that CACNA1C-AS2 functions as a tumor suppressor. Furthermore, we found that CACNA1C-AS2 negatively regulated Fbxo45 protein expression in glioma cells. Impressively, extensive experimental results revealed that Fbxo45 accelerated growth, migration and invasion of glioma cells. Clinically, increased Fbxo45 expression was observed in 75 human glioma tissue samples. Moreover, in vivo experiments also demonstrated that Fbxo45 overexpression enhanced tumor growth in mice. Especially, we further identified that Fbxo45 activated mTORC1 rather than mTORC2 through PI3K/AKT signaling to promote cell growth and motility in glioma cells. Rescue experiments also exhibited that CACNA1C-AS2 inhibited cell growth and motility partly through down-regulating Fbxo45 expression in glioma. Our results provide the novel insights into the critical role of CACNA1C-AS2/Fbxo45/mTOR axis involved in regulating glioma tumorigenesis and progression, and further indicate that CACNA1C-AS2 and Fbxo45 may be the potential biomarkers and therapeutic targets for glioma.
Collapse
Affiliation(s)
- Tong Cao
- Department of Clinical Laboratory, the First Affiliated Hospital of Bengbu Medical College, Bengbu, 233004, Anhui, China.,Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Yue Cui
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, 233030, Anhui, China.,Department of Clinical Laboratory, Fuyang People's Hospital, Fuyang, 236001, Anhui, China
| | - Yingying Wang
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Linhui Wu
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, 233030, Anhui, China.,Clinical Laboratory Center, Lu'an People's Hospital, The Lu'an Affiliated Hospital of Anhui Medical University, Lu'an, 237000, Anhui, China
| | - Ke Yu
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Kai Chen
- Bengbu Medical College Key Laboratory of Cancer Research and Clinical Laboratory Diagnosis, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Jun Xia
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, 2600 Donghai Street, Bengbu, 233030, Anhui, China
| | - Yuyun Li
- Department of Clinical Laboratory Diagnostics, School of Laboratory Medicine, Bengbu Medical College, Bengbu, 233030, Anhui, China
| | - Zhiwei Peter Wang
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, 2600 Donghai Street, Bengbu, 233030, Anhui, China. .,Center of Scientific Research, the Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325027, Zhejiang, China.
| | - Jia Ma
- Department of Biochemistry and Molecular Biology, School of Laboratory Medicine, Bengbu Medical College, 2600 Donghai Street, Bengbu, 233030, Anhui, China.
| |
Collapse
|
30
|
Chen L, Wang J, Liu Q. Long noncoding RNAs as therapeutic targets to overcome chemoresistance in ovarian cancer. Front Cell Dev Biol 2022; 10:999174. [PMID: 36105363 PMCID: PMC9464811 DOI: 10.3389/fcell.2022.999174] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2022] [Accepted: 08/08/2022] [Indexed: 12/15/2022] Open
Abstract
Long noncoding RNAs (lncRNAs) have been characterized to play an essential role in ovarian tumorigenesis via controlling a variety of cellular processes, such as cell proliferation, invasion, apoptotic death, metastasis, cell cycle, migration, metabolism, immune evasion, and chemoresistance. The one obstacle for the therapeutic efficacy is due to the development of drug resistance in ovarian cancer patients. Therefore, in this review article, we describe the role of lncRNAs in chemoresistance in ovarian cancer. Moreover, we discuss the molecular mechanism of lncRNAs-involved drug resistance in ovarian cancer. We conclude that lncRNAs could be useful targets to overcome chemoresistance and improve therapeutic outcome in ovarian cancer patients.
Collapse
|
31
|
Xu Z, Chen Q, Shu L, Zhang C, Liu W, Wang P. Expression profiles of m6A RNA methylation regulators, PD-L1 and immune infiltrates in gastric cancer. Front Oncol 2022; 12:970367. [PMID: 36003776 PMCID: PMC9393729 DOI: 10.3389/fonc.2022.970367] [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: 06/15/2022] [Accepted: 06/30/2022] [Indexed: 12/01/2022] Open
Abstract
Gastric cancer is the fourth most frequent cancer and has a high death rate. Immunotherapy represented by PD-1 has brought hope for the treatment of advanced gastric cancer. Methylation of the m6A genes is linked to the onset and progression of numerous cancers, but there are few studies on gastric cancer. The main purpose of this study aims to analyze the relationship between m6A RNA methylation regulators, PD-L1, prognosis and tumor immune microenvironment (TIME) in gastric cancer. The Cancer Genome Atlas (TCGA) and Genotype Tissue Expression (GTEx) databases were used to acquire transcriptomic data and clinical information from gastric cancer patients. The changes in m6A regulator expression levels in gastric cancer tissues and normal tissues were studied. Consensus clustering analysis was used to separate gastric cancer samples into two categories. We employed Least Absolute Shrinkage, Selection Operator (LASSO) Cox regression analysis, Gene Set Enrichment Analysis (GSEA), and cBioPortal to analyze the m6A regulators, PD-L1 and TIME in gastric cancer. In gastric cancer tissues, the majority of m6A regulatory factors are considerably overexpressed. Two gastric cancer subgroups (Cluster1/2) based on consensus clustering of 21 m6A regulators. PD-L1 and PD-1 expression levels were significantly higher in gastric cancer tissues, and they were significantly linked with METTL3, WTAP, HNRNPD, ZC3H7B, METTL14, FTO, PCIF1, HNRNPC, YTHDF1 and YTDHF2. Cluster1 showed a large increase in resting memory CD4+ T cells, regulatory T cells, naïve B cells, active NK cells, and resting Mast cells. Cluster1 and Cluster2 were shown to be involved in numerous critical signaling pathways, including base excision repair, cell cycle, nucleotide excision repair, RNA degradation, and spliceosome pathways. Gastric cancer RiskScores based on prognostic factors have been found as independent prognostic indicators. The amount of tumor-infiltrating immune cells is dynamically affected by changes in the copy number of m6A methylation regulators associated with TIME.
Collapse
Affiliation(s)
- Zhiyuan Xu
- Department of Gastric Surgery, the Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institutes of Basic Medicine and Cancer (IBMC), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou, China
- *Correspondence: Zhiyuan Xu, ; Peter Wang,
| | - Qiuli Chen
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, Zhejiang, China
| | - Lilu Shu
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, Zhejiang, China
| | - Chunye Zhang
- National University of Singapore (Suzhou) Research Institute, Suzhou, China
| | - Wenjun Liu
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, Zhejiang, China
| | - Peter Wang
- Department of Research and Development, Zhejiang Zhongwei Medical Research Center, Hangzhou, Zhejiang, China
- *Correspondence: Zhiyuan Xu, ; Peter Wang,
| |
Collapse
|
32
|
Li R, Chen Z, Zhou Y, Maimaitirexiati G, Yan Q, Li Y, Maimaitiyimin A, Zhou C, Ren J, Liu C, Mainike A, Zhou P, Ding L. LncRNA SCAMP1 disrupts the balance between miR-26a-5p and ZEB2 to promote osteosarcoma cell viability and invasion. Front Oncol 2022; 12:967000. [PMID: 35992869 PMCID: PMC9382636 DOI: 10.3389/fonc.2022.967000] [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: 06/12/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022] Open
Abstract
Osteosarcoma often occurs in children and adolescents and affects their health. The survival rate of osteosarcoma patients is unsatisfactory due to the lack of early detection and metastasis development and drug resistance. Hence, dissection of molecular insight into osteosarcoma initiation and progression is pivotal to provide the new therapeutic strategy. In recent years, long noncoding RNAs (lncRNAs) have burst into stage in osteosarcoma development and malignant behaviors. LncRNA SCAMP1 has been discovered to play an essential role in carcinogenesis and progression. However, the mechanisms of lncRNA SCAMP1-involved tumorigenesis have not been reported in human osteosarcoma. In this study, we utilized multiple cellular biological approaches to determine the function of lncRNA SCAMP1 in osteosarcoma cells. Moreover, we performed several molecular biological approaches to define the mechanism by which lncRNA SCAMP1 regulated cell viability and invasion in osteosarcoma. We dissected that lncRNA SCAMP1 promoted progression of osteosarcoma via modulation of miR-26a-5p/ZEB2 axis. In conclusion, targeting lncRNA SCAMP1 and its downstream targets, miR-26a-5p and ZEB2, might be a useful approach for osteosarcoma therapy.
Collapse
Affiliation(s)
- Rong Li
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Zhen Chen
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Yubo Zhou
- Department of Orthopedics, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, China
| | - Gulikezi Maimaitirexiati
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qi Yan
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Yuting Li
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | | | - Changhui Zhou
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Jingqin Ren
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Chengqing Liu
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Abasi Mainike
- Department of Orthopedics, Xinjiang Medical University Affiliated Fifth Hospital, Urumqi, China
| | - Peng Zhou
- Department of Orthopedics, Xinjiang Medical University Affiliated Fifth Hospital, Urumqi, China
| | - Lu Ding
- Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Postdoctoral Research Center on Public Health and Preventive Medicine, Xinjiang Medical University, Xinjiang, China
- *Correspondence: Lu Ding,
| |
Collapse
|
33
|
Xia L, Chen J, Huang M, Mei J, Lin M. The functions of long noncoding RNAs on regulation of F-box proteins in tumorigenesis and progression. Front Oncol 2022; 12:963617. [PMID: 35928868 PMCID: PMC9343830 DOI: 10.3389/fonc.2022.963617] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Accepted: 06/27/2022] [Indexed: 11/28/2022] Open
Abstract
Accumulated evidence has revealed that F-box protein, a subunit of SCF E3 ubiquitin ligase complexes, participates in carcinogenesis and tumor progression via targeting its substrates for ubiquitination and degradation. F-box proteins could be regulated by cellular signaling pathways and noncoding RNAs in tumorigenesis. Long noncoding RNA (lncRNA), one type of noncoding RNAs, has been identified to modulate the expression of F-box proteins and contribute to oncogenesis. In this review, we summarize the role and mechanisms of multiple lncRNAs in regulating F-box proteins in tumorigenesis, including lncRNAs SLC7A11-AS1, MT1JP, TUG1, FER1L4, TTN-AS1, CASC2, MALAT1, TINCR, PCGEM1, linc01436, linc00494, GATA6-AS1, and ODIR1. Moreover, we discuss that targeting these lncRNAs could be helpful for treating cancer via modulating F-box protein expression. We hope our review can stimulate the research on exploration of molecular insight into how F-box proteins are governed in carcinogenesis. Therefore, modulation of lncRNAs is a potential therapeutic strategy for cancer therapy via regulation of F-box proteins.
Collapse
|
34
|
Li S, Tang M, Zen N, Liang J, Xing X, Huang D, Liu F, Zhang X. LncRNA OIP5-AS1 Knockdown Targets miR-183-5p/GLUL Axis and Inhibits Cell Proliferation, Migration and Metastasis in Nasopharyngeal Carcinoma. Front Oncol 2022; 12:921929. [PMID: 35756672 PMCID: PMC9214031 DOI: 10.3389/fonc.2022.921929] [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: 04/16/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
Nasopharyngeal carcinoma (NPC) is often associated with the infection of Epstein-Barr virus in nasopharynx and is mainly happened in South China and Southeast Asia. Recently, noncoding RNAs have been reported to regulate NPC carcinogenesis. LncRNA OIP5-AS1 participates in tumorigenesis and progression; however, the inherent mechanism of OIP5-AS1-mediated progression of NPC is unclear. In the current study, we aimed to explore the role of OIP5-AS1 in NPC progression. We measured the cell viability, apoptosis, migration, and invasion in NPC cells after OIP5-AS1 modulation. Moreover, we determined whether OIP5-AS1 exerts its oncogenic functions via sponging miR-183-5p in NPC. Furthermore, we determined whether glutamate ammonia ligase (GLUL) was a downstream target of miR-183-5p. We found that OIP5-AS1 downregulation inhibited the viability, migration and invasion of NPC via targeting miR-183-5p. We also identified that GLUL might be a potential downstream target of miR-183-5p in NPC cells. Mechanistically, OIP5-AS1 promotes cell motility via regulating miR-183-5p and GLUL in NPC cells. We concluded that OIP5-AS1 performed its biological functions via targeting miR-183-5p and GLUL in NPC cells.
Collapse
Affiliation(s)
- Shuo Li
- Department of Otolaryngology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China.,Department of Otolaryngology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Mingxing Tang
- Department of Otolaryngology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Nan Zen
- Department of Otolaryngology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China.,Department of Otolaryngology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Junyi Liang
- Department of Otolaryngology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China.,Department of Otolaryngology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Xiao Xing
- Department of Otolaryngology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China.,Department of Otolaryngology, Affiliated Shenzhen Sixth Hospital of Guangdong Medical University, Shenzhen, China
| | - Danglin Huang
- Department of Otolaryngology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China.,Department of Otolaryngology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Fei Liu
- Department of Otolaryngology, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China.,Department of Otolaryngology, Huazhong University of Science and Technology Union Shenzhen Hospital, Shenzhen, China
| | - Xiaomeng Zhang
- Department of Otorhinolaryngology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
35
|
Man X, Li Q, Wang B, Zhang H, Zhang S, Li Z. DNMT3A and DNMT3B in Breast Tumorigenesis and Potential Therapy. Front Cell Dev Biol 2022; 10:916725. [PMID: 35620052 PMCID: PMC9127442 DOI: 10.3389/fcell.2022.916725] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2022] [Accepted: 04/21/2022] [Indexed: 01/15/2023] Open
Abstract
Breast cancer has become a leading cause of cancer-related deaths in women worldwide. DNA methylation has been revealed to play an enormously important role in the development and progression of breast cancer. DNA methylation is regulated by DNA methyltransferases (DNMTs), including DNMT1, DNMT2, and DNMT3. DNMT3 family has three members: DNMT3A, DNMT3B, and DNMT3L. The roles and functions of DNMT1 in breast cancer have been well reviewed. In this article, the roles of DNMT3A and DNMT3B in breast tumorigenesis and development are reviewed. We also discuss the SNP and mutations of DNMT3A and DNMT3B in breast cancer. In addition, we summarize how DNMT3A and DNMT3B are regulated by non-coding RNAs and signaling pathways in breast cancer, and targeting the expression levels of DNMT3A and DNMT3B may be a promising therapeutic approach for breast cancer. This review will provide reference for further studies on the biological functions and molecular mechanisms of DNMT3A and DNMT3B in breast cancer.
Collapse
Affiliation(s)
- Xiaxia Man
- Department of Oncologic Gynecology, the First Hospital of Jilin University, Jilin, China
| | - Qi Li
- State and Local Joint Engineering Laboratory for Animal Models of Human Diseases, Academy of Translational Medicine, the First Hospital of Jilin University, Jilin, China
| | - Baogang Wang
- Department of Cardiac Surgery, the First Hospital of Jilin University, Jilin, China
| | - He Zhang
- Department of Oncologic Gynecology, the First Hospital of Jilin University, Jilin, China
| | - Songling Zhang
- Department of Oncologic Gynecology, the First Hospital of Jilin University, Jilin, China
| | - Ziyi Li
- State and Local Joint Engineering Laboratory for Animal Models of Human Diseases, Academy of Translational Medicine, the First Hospital of Jilin University, Jilin, China
| |
Collapse
|
36
|
Chen T, Liu J, Zhang H, Li J, Shang G. Long Intergenic Noncoding RNA00265 Enhances Cell Viability and Metastasis via Targeting miR-485-5p/USP22 Axis in Osteosarcoma. Front Oncol 2022; 12:907472. [PMID: 35692754 PMCID: PMC9179024 DOI: 10.3389/fonc.2022.907472] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 04/19/2022] [Indexed: 01/09/2023] Open
Abstract
Osteosarcoma is one of the bone malignancies in children and adolescents. Long noncoding RNAs (lncRNAs) have been demonstrated to participate in osteosarcoma development and progression. Linc00265 has been shown to involve in osteosarcoma oncogenesis; however, the underlying mechanism is largely unclear. In this study, we investigated the function of linc00265 in osteosarcoma cells, including cell viability, migration and invasion. Moreover, we elucidated mechanistically the involvement of linc00265 in osteosarcoma. We found that linc00265 overexpression promoted viability, migration and invasion of osteosarcoma cells. Notably, linc00265 sponged miR-485-5p and increased the expression of USP22, one target of miR-485-5p, in osteosarcoma cells. Strikingly, linc00265 exerted its oncogenic function via regulating miR-485-5p and USP22 in osteosarcoma. Taken together, targeting linc00265 is a promising approach for treating osteosarcoma patients.
Collapse
|