1
|
Schaft N, Dörrie J. The Role of Non-coding RNAs in Tumorigenesis, Diagnosis/Prognosis, and Therapeutic Strategies for Cutaneous Melanoma. Methods Mol Biol 2025; 2883:79-107. [PMID: 39702705 DOI: 10.1007/978-1-0716-4290-0_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] [Indexed: 12/21/2024]
Abstract
RNA is a substance with various biological functions. It serves as blueprint for proteins and shuttles information from the genes to the protein factories of the cells. However, these factories-the ribosomes-are also composed mainly of RNA, whose purpose is not storing information but enzymatic action. In addition, there is a cornucopia of RNA molecules within our cells that form a complex regulatory network, connected with all aspects of cellular development and maintenance. These non-coding RNAs can be used for diagnostics and therapeutic strategies in cancer. In this chapter we give an overview of recent developments in non-coding RNA-based diagnostics and therapies for cutaneous melanoma. It is not meant to be comprehensive; however, it describes examples based on some of the most recent publications in this field.
Collapse
Affiliation(s)
- Niels Schaft
- Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, CCC WERA, Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Deutsches Zentrum Immuntherapie (DZI), Bavarian Cancer Research Center (BZKF), Erlangen, Germany.
| | - Jan Dörrie
- Department of Dermatology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen, CCC WERA, Comprehensive Cancer Center Erlangen European Metropolitan Area of Nuremberg (CCC ER-EMN), Deutsches Zentrum Immuntherapie (DZI), Bavarian Cancer Research Center (BZKF), Erlangen, Germany
| |
Collapse
|
2
|
Chen S, Wen L, Wu Y, Xiao S, Lai Y, Ou J, Shen Y. Circular RNA circ-CARD8 regulates alveolar macrophage pyroptosis through the miR-580-3p/CARD8 pathway in acute lung injury. PLoS One 2024; 19:e0314936. [PMID: 39705232 DOI: 10.1371/journal.pone.0314936] [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: 05/10/2024] [Accepted: 11/18/2024] [Indexed: 12/22/2024] Open
Abstract
Pyroptosis is linked to the development of acute lung injury (ALI), and circular RNAs (circRNAs) play a role in ALI-related inflammation. However, the mechanisms by which circRNAs contribute to macrophage pyroptosis in ALI remain unclear. This study constructed an in vitro ALI model by inducing THP-1 cells with phorbol 12-myristate 13-acetate (PMA) and lipopolysaccharide (LPS). The expression and potential mechanism of circ-CARD8 in macrophage pyroptosis were then investigated. The interaction between circ-CARD8, hsa-miR-580-3p, and caspase recruitment domain family member 8 (CARD8) was confirmed through luciferase reporter assays and RNA-binding protein immunoprecipitation. Our data showed that circ-CARD8 was expressed at low levels. Meanwhile, the pyroptotic proteins caspase-1 and GSDMD, along with the secretion of chemokine (C-C motif) ligand 18 and interleukin 1 beta, were upregulated in the ALI cell model. Overexpression of circ-CARD8 reversed macrophage pyroptosis, whereas inhibition of circ-CARD8 promoted it. Furthermore, the expression of miR-580-3p, a downstream microRNA that binds to circ-CARD8, was reduced upon circ-CARD8 overexpression and increased following its inhibition. Additionally, overexpression of miR-580-3p suppressed the expression of CARD8, a downstream target of miR-580-3p, thereby promoting macrophage pyroptosis. The inhibition of miR-580-3p reversed the effect of circ-CARD8 silencing on macrophage pyroptosis and CARD8 expression. Therefore, our study confirms that the low expression of circ-CARD8 reduces the sponge adsorption of miR-580-3p, increasing its expression, which in turn targets and inhibits CARD8, ultimately promoting macrophage pyroptosis induced by LPS in THP-1 cells.
Collapse
Affiliation(s)
- Sida Chen
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Ling Wen
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Yumei Wu
- Yadi Sancun Community Health Service Center, Shenzhen Pingle Orthopaedic Hospital (Shenzhen Pingshan Traditional Chinese Medicine Hospital), Shenzhen, China
| | - Shan Xiao
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Yuting Lai
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Jintao Ou
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| | - Yan Shen
- Respiratory Department, Longgang Central Hospital, Shenzhen, China
| |
Collapse
|
3
|
Lyu Z, Mahenderan A, Radhakrishnan AKGK, Chin YS, Yin C. Swimming upregulates APOL3 through regulating macrophage polarization to inhibit glycolysis and the development of melanoma. 3 Biotech 2024; 14:307. [PMID: 39583207 PMCID: PMC11582285 DOI: 10.1007/s13205-024-04150-z] [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: 08/29/2024] [Accepted: 11/01/2024] [Indexed: 11/26/2024] Open
Abstract
This study investigated the role of swimming exercise in regulating melanoma tumour growth and glycolysis in cancer cells, the specific mechanism involved was also studied. In our study, a murine melanoma tumour model was established to assess the impact of swimming on tumour growth. The mRNA and protein expressions were assessed using qRT-PCR, western blot, and IHC. The metabolic behavior of melanoma cells was examined through lactic acid level measurements and glucose consumption assessments. CCK-8 and colony formation assays were used to detect cell viability and proliferation. ELISA was employed to determine the levels of cytokines secreted by macrophages. The interaction between APOL3 and STAT3 was analyzed by dual luciferase reporter gene and ChIP assays. Our results demonstrated that swimming exercise suppressed melanoma growth in mice by suppressing glycolysis, which might be related to APOL3 upregulation. In addition, downregulation of APOL3 in melanoma was associated with poor prognosis, and APOL3 overexpression markedly suppressed melanoma cell proliferation by reducing glucose uptake and lactate production in vitro. Mechanistically, STAT3 directly down-regulated APOL3 transcription. Swimming upregulated APOL3 by inactivating the IL-6R-STAT3 signaling axis in melanoma cells by inhibiting the secretion of IL-6 by M2 macrophages. As expected, IL-6 secreted by M2 macrophages promoted glycolysis in melanoma cells by reducing APOL3 expression. In summary, swimming inactivated the IL-6R/STAT3 signaling axis in melanoma cells by inhibiting the secretion of IL-6 by M2 macrophages, which could suppress the growth of melanoma in the body by upregulating APOL3 to inhibit glycolysis.
Collapse
Affiliation(s)
- Zhenlei Lyu
- Hainan Vocational University of Science and Technology, College of Medicine, No.18 Qiongshan Avenue, Meilan District, Haikou, 571100 Hainan People’s Republic of China
- Sports Science Programme, Faculty of Sports Science and Recreation, Universiti Teknologi MARA, 40450 Selangor, Shah Alam Malaysia
| | - Appukutty Mahenderan
- Sports Science Programme, Faculty of Sports Science and Recreation, Universiti Teknologi MARA, 40450 Selangor, Shah Alam Malaysia
| | | | - Yit Siew Chin
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, UPM Serdang Selangor Malaysia, 43400 Seri Kembangan, Malaysia
| | - Chao Yin
- Hainan Vocational University of Science and Technology, College of Medicine, No.18 Qiongshan Avenue, Meilan District, Haikou, 571100 Hainan People’s Republic of China
| |
Collapse
|
4
|
Thongkumkoon P, Sangphukieo A, Tongjai S, Noisagul P, Sangkhathat S, Laochareonsuk W, Kamolphiwong R, Budprom P, Teeyakasem P, Yongpitakwattana P, Thepbundit V, Sirikaew N, Klangjorhor J, Settakorn J, Moonmuang S, Suksakit P, Pasena A, Chaijaruwanich J, Yathongkhum W, Dissook S, Pruksakorn D, Chaiyawat P. Establishment, characterization, and genetic profiling of patient-derived osteosarcoma cells from a patient with retinoblastoma. Sci Rep 2024; 14:11056. [PMID: 38744935 PMCID: PMC11094034 DOI: 10.1038/s41598-024-60628-z] [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: 05/22/2023] [Accepted: 04/25/2024] [Indexed: 05/16/2024] Open
Abstract
Osteosarcoma is the most common malignant bone cancer in pediatric patients. Patients who respond poorly to chemotherapy experience worse clinical outcomes with a high mortality rate. The major challenge is the lack of effective drugs for these patients. To introduce new drugs for clinical approval, preclinical studies based on in vitro models must demonstrate the potency of the tested drugs, enabling the drugs to enter phase 1 clinical trials. Patient-derived cell culture is a promising testing platform for in vitro studies, as they more accurately recapitulate cancer states and genetic profiles compared to cell lines. In the present study, we established patient-derived osteosarcoma cells (PDC) from a patient who had previously been diagnosed with retinoblastoma. We identified a new variant of a germline mutation in the RB1 gene in the tissue of the patient. The biological effects of this PDC were studied to observe whether the cryopreserved PDC retained a feature of fresh PDC. The cryopreserved PDC preserved the key biological effects, including cell growth, invasive capability, migration, and mineralization, that define the conserved phenotypes compared to fresh PDC. From whole genome sequencing analysis of osteosarcoma tissue and patient-derived cells, we found that cryopreserved PDC was a minor population in the origin tissue and was selectively grown under the culture conditions. The cryopreserved PDC has a high resistance to conventional chemotherapy. This study demonstrated that the established cryopreserved PDC has the aggressive characteristics of osteosarcoma, in particular the chemoresistance phenotype that might be used for further investigation in the chemoresistant mechanism of osteosarcoma. In conclusion, the approach we applied for primary cell culture might be a promising method to generate in vitro models for functional testing of osteosarcoma.
Collapse
Affiliation(s)
- Patcharawadee Thongkumkoon
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Apiwat Sangphukieo
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Siripong Tongjai
- Department of Microbiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pitiporn Noisagul
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Surasak Sangkhathat
- Division of Surgery, Faculty of Medicine, Prince of Songkla University, Hatyai, Songkhla, 90110, Thailand
- Translational Medicine Research Center, Prince of Songkla University, Hatyai, Songkhla, 90110, Thailand
| | - Wison Laochareonsuk
- Division of Surgery, Faculty of Medicine, Prince of Songkla University, Hatyai, Songkhla, 90110, Thailand
- Translational Medicine Research Center, Prince of Songkla University, Hatyai, Songkhla, 90110, Thailand
| | - Rawikant Kamolphiwong
- Department of Biomedical Sciences and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Hat Yai, Songkhla, 90110, Thailand
| | - Piyaporn Budprom
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pimpisa Teeyakasem
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Petlada Yongpitakwattana
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Viraporn Thepbundit
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 10 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Nutnicha Sirikaew
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 10 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Jeerawan Klangjorhor
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Jongkolnee Settakorn
- Department of Pathology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sutpirat Moonmuang
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
- Office of Research Administration, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Pathacha Suksakit
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Arnat Pasena
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand
| | - Jeerayut Chaijaruwanich
- Department of Computer Science, Faculty of Science, Data Science Research Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Wilawan Yathongkhum
- Department of Computer Science, Faculty of Science, Data Science Research Center, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sivamoke Dissook
- Department of Biochemistry, Faculty of Medicine, Chiang Mai University, 10 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand.
| | - Dumnoensun Pruksakorn
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand.
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Department of Orthopedics, Faculty of Medicine, Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand.
| | - Parunya Chaiyawat
- Faculty of Medicine, Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Chiang Mai University, 110 Intawaroros Road, Si Phum, Muang, Chiang Mai, 50200, Thailand.
- Faculty of Medicine, Musculoskeletal Science and Translational Research (MSTR) Center, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
5
|
Tan Z, Luan S, Wang X, Jiao W, Jiang P. Mechanism study of lncRNA RMRP regulating esophageal squamous cell carcinoma through miR-580-3p/ATP13A3 axis. Discov Oncol 2024; 15:150. [PMID: 38722543 PMCID: PMC11082096 DOI: 10.1007/s12672-024-00990-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 04/19/2024] [Indexed: 05/12/2024] Open
Abstract
OBJECTIVE It is well-known that lncRNAs regulate energy metabolism in tumors. This study focused on the action of RMRP on esophageal squamous cell carcinoma (ESCC) cell proliferation, apoptosis, and glycolysis. METHODS In the resected ESCC tissues and adjacent tissues from patients, RMRP/miR-580-3p/ATP13A3 expressions were evaluated. ESCC cell proliferation rates and apoptotic rates were measured by CCK-8 and flow cytometry, respectively. Apoptosis related markers were examined by Western blot. Moreover, glucose uptake, lactic acid, and ATP were measured by commercial kits, whereas HK2 and PKM2 were evaluated by Western blot to study ESCC cell glycolysis. Finally, the editing program of RMRP/miR-580-3p/ATP13A3 was translated by luciferase reporter assay and RIP analysis. RESULTS RMRP and ATP13A3 were induced, while miR-580-3p was reduced in their expression in ESCC tissues. Silencing RMRP reduced proliferation, glycolysis, and anti-apoptosis ability of ESCC cells. RMRP sequestered miR-580-3p to target ATP13A3. Silenced ATP13A3 or overexpressed miR-580-3p rescued overexpressed RMRP-mediated promotion of proliferation, glycolysis, and anti-apoptosis of ESCC cells. CONCLUSION RMRP accelerates ESCC progression through the miR-580-3p/ATP13A3 axis, renewing a reference for lncRNA-based therapies for tumors.
Collapse
Affiliation(s)
- ZiRui Tan
- The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Chang'an District, Shijiazhuang City, 050000, Hebei Province, China
| | - ShengJie Luan
- Department of Tumor Chemoradiotherapy, Central Hospital of Qinghe County, Xingtai City, 054800, Hebei Province, China
| | - XiaoPeng Wang
- Department of Tumor Chemoradiotherapy, Central Hospital of Qinghe County, Xingtai City, 054800, Hebei Province, China
| | - WenPeng Jiao
- The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Chang'an District, Shijiazhuang City, 050000, Hebei Province, China
| | - Pu Jiang
- The Fourth Hospital of Hebei Medical University, No. 12, Jiankang Road, Chang'an District, Shijiazhuang City, 050000, Hebei Province, China.
| |
Collapse
|
6
|
Liu S, Wang H, Yang B, Hou B, Sun L, Pang H, Wang H, Fan Y. Circ_0068087 knockdown attenuates high-glucose-induced human tubular epithelial cell injury in a microribonucleic acid/progestin and adipoQ receptor 3-dependent manner in diabetic nephropathy. J Diabetes Investig 2024; 15:159-171. [PMID: 37985406 PMCID: PMC10804928 DOI: 10.1111/jdi.14107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 09/28/2023] [Accepted: 10/25/2023] [Indexed: 11/22/2023] Open
Abstract
AIMS/INTRODUCTION Previous studies have shown that circular ribonucleic acid mediates the occurrence of diabetic nephropathy. This study aimed to analyze the effects of circ_0068087 on high-glucose (HG)-induced human kidney 2 (HK2) cell dysfunction. MATERIALS AND METHODS Circ_0068087, miR-580-3p, and progestin and adipoQ receptor 3 (PAQR3) expression were detected by quantitative reverse transcription polymerase chain reaction. Cell viability and proliferation were investigated by Cell Counting Kit-8 and EdU assays, respectively. The cell apoptotic rate was assessed by flow cytometry. Inflammatory response was assessed by enzyme-linked immunoassays. Oxidative stress was evaluated by a superoxide dismutase activity assay kit and lipid peroxidation malondialdehyde assay kit. Molecular interaction was identified by dual-luciferase reporter assay. RESULTS Circ_0068087 and PAQR3 expression were significantly upregulated in diabetic nephropathy patients. HG treatment inhibited HK2 cell proliferation, but induced cell apoptosis, inflammation, oxidative stress and epithelial-mesenchymal transition by regulating circ_0068087. Circ_0068087 acted as a microribonucleic acid-580-3p (miR-580-3p) sponge, and miR-580-3p targeted PAQR3. Furthermore, circ_0068087 depletion repressed PAQR3 expression through miR-580-3p. MiR-580-3p inhibitors or PAQR3 introduction attenuated circ_0068087 silencing mediated-effects in HG-treated HK2 cells. CONCLUSION Circ_0068087 promoted HG-induced HK2 cell injuries by the regulation of the miR-580-3p/PAQR3 pathway.
Collapse
Affiliation(s)
- Shu‐yan Liu
- Department of EndocrinologyThe First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital)JiaozuoChina
| | - Hong Wang
- Department of GynecologyThe First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital)JiaozuoChina
| | - Bo Yang
- Department of NeurologyThe First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital)JiaozuoChina
| | - Baohua Hou
- Medical College of Henan Polytechnic UniversityJiaozuoChina
| | - Li‐sha Sun
- Department of GynecologyThe First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital)JiaozuoChina
| | - Hui Pang
- Department of OncologyThe First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital)JiaozuoChina
| | - Hui‐hui Wang
- Department of EndocrinologyThe First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital)JiaozuoChina
| | - Yan‐ping Fan
- Department of EndocrinologyThe First Affiliated Hospital of Henan Polytechnic University (Jiaozuo Second People's Hospital)JiaozuoChina
| |
Collapse
|
7
|
Grafanaki K, Grammatikakis I, Ghosh A, Gopalan V, Olgun G, Liu H, Kyriakopoulos GC, Skeparnias I, Georgiou S, Stathopoulos C, Hannenhalli S, Merlino G, Marie KL, Day CP. Noncoding RNA circuitry in melanoma onset, plasticity, and therapeutic response. Pharmacol Ther 2023; 248:108466. [PMID: 37301330 PMCID: PMC10527631 DOI: 10.1016/j.pharmthera.2023.108466] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/24/2023] [Accepted: 05/31/2023] [Indexed: 06/12/2023]
Abstract
Melanoma, the cancer of the melanocyte, is the deadliest form of skin cancer with an aggressive nature, propensity to metastasize and tendency to resist therapeutic intervention. Studies have identified that the re-emergence of developmental pathways in melanoma contributes to melanoma onset, plasticity, and therapeutic response. Notably, it is well known that noncoding RNAs play a critical role in the development and stress response of tissues. In this review, we focus on the noncoding RNAs, including microRNAs, long non-coding RNAs, circular RNAs, and other small RNAs, for their functions in developmental mechanisms and plasticity, which drive onset, progression, therapeutic response and resistance in melanoma. Going forward, elucidation of noncoding RNA-mediated mechanisms may provide insights that accelerate development of novel melanoma therapies.
Collapse
Affiliation(s)
- Katerina Grafanaki
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA; Department of Dermatology, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Ioannis Grammatikakis
- Cancer Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Arin Ghosh
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Vishaka Gopalan
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Gulden Olgun
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Huaitian Liu
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - George C Kyriakopoulos
- Department of Biochemistry, School of Medicine, University of Patras, 26504 Patras, Greece
| | - Ilias Skeparnias
- Laboratory of Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD, USA
| | - Sophia Georgiou
- Department of Dermatology, School of Medicine, University of Patras, 26504 Patras, Greece
| | | | - Sridhar Hannenhalli
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Glenn Merlino
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kerrie L Marie
- Division of Molecular and Cellular Function, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK.
| | - Chi-Ping Day
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
8
|
MicroRNA-136-5p from Endothelial Progenitor Cells-released Extracellular Vesicles Mediates TXNIP to Promote the Dissolution of Deep Venous Thrombosis. Shock 2022; 57:714-721. [PMID: 35583913 DOI: 10.1097/shk.0000000000001920] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Endothelial progenitor cells-released extracellular vesicles (EPCs-EVs) have previously been reported to promote the dissolution of deep venous thrombosis (DVT) through delivery of microRNA (miR). Given that, this research was projected to search the relative action of EPCs-EVs transferring of miR-136-5p in DVT. METHODS From EPCs transfected with miR-136-5p agomir or antagomir, EVs were extracted and then injected into DVT mice. Meanwhile, based on the treatment with EPCs-EVs loading miR-136-5p antagomir, silenced thioredoxin-interacting protein (TXNIP) lentivirus was injected into DVT mice to perform the rescue experiments. Afterwards, the length and weight of venous thrombosis, EPC apoptosis and inflammatory factors, plasmin, fibrinogen, and thrombin-antithrombin were measured. miR-136-5p and TXNIP expression in DVT mice, and their targeting relationship were evaluated. RESULTS miR-136-5p expression was suppressed and TXNIP expression was elevated in DVT mice. EPCs-EV reduced the length and weight of venous thrombosis, suppressed cell apoptosis and inflammatory reaction, as well as elevated level of plasmin, and reduced levels of fibrinogen and thrombin-antithrombin in DVT mice. Restored miR-136-5p loaded by EPCs-EV further attenuated DVT but EPCs-EV transfer of depleted miR-136-5p resulted in the opposite consequences. miR-136-5p targeted TXNIP and silenced TXNIP rescued the effect of EPCs-EV transfer of depleted miR-136-5p on DVT. CONCLUSION miR-136-5p from EPCs-EV suppresses TXNIP expression to reduce the thrombus size in DVT, offering a promising treatment target for DVT.
Collapse
|
9
|
Ni N, Fang X, Mullens DA, Cai JJ, Ivanov I, Bartholin L, Li Q. Transcriptomic Profiling of Gene Expression Associated with Granulosa Cell Tumor Development in a Mouse Model. Cancers (Basel) 2022; 14:2184. [PMID: 35565312 PMCID: PMC9105549 DOI: 10.3390/cancers14092184] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/05/2022] [Accepted: 04/24/2022] [Indexed: 11/16/2022] Open
Abstract
Ovarian granulosa cell tumors (GCTs) are rare sex cord-stromal tumors, accounting for ~5% ovarian tumors. The etiology of GCTs remains poorly defined. Genetically engineered mouse models are potentially valuable for understanding the pathogenesis of GCTs. Mice harboring constitutively active TGFβ signaling (TGFBR1-CA) develop ovarian GCTs that phenocopy several hormonal and molecular characteristics of human GCTs. To determine molecular alterations in the ovary upon TGFβ signaling activation, we performed transcriptomic profiling of gene expression associated with GCT development using ovaries from 1-month-old TGFBR1-CA mice and age-matched controls. RNA-sequencing and bioinformatics analysis coupled with the validation of select target genes revealed dysregulations of multiple cellular events and signaling molecules/pathways. The differentially expressed genes are enriched not only for known GCT-related pathways and tumorigenic events but also for signaling events potentially mediated by neuroactive ligand-receptor interaction, relaxin signaling, insulin signaling, and complements in TGFBR1-CA ovaries. Additionally, a comparative analysis of our data in mice with genes dysregulated in human GCTs or granulosa cells overexpressing a mutant FOXL2, the genetic hallmark of adult GCTs, identified some common genes altered in both conditions. In summary, this study has revealed the molecular signature of ovarian GCTs in a mouse model that harbors the constitutive activation of TGFBR1. The findings may be further exploited to understand the pathogenesis of a class of poorly defined ovarian tumors.
Collapse
Affiliation(s)
- Nan Ni
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (N.N.); (X.F.); (J.J.C.)
| | - Xin Fang
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (N.N.); (X.F.); (J.J.C.)
| | - Destiny A. Mullens
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (D.A.M.); (I.I.)
| | - James J. Cai
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (N.N.); (X.F.); (J.J.C.)
| | - Ivan Ivanov
- Department of Veterinary Physiology and Pharmacology, Texas A&M University, College Station, TX 77843, USA; (D.A.M.); (I.I.)
| | - Laurent Bartholin
- INSERM U1052, CNRS UMR5286, Centre de Recherche en Cancérologie de Lyon, Université Lyon 1, F-69000 Lyon, France;
- Centre Léon Bérard, F-69008 Lyon, France
| | - Qinglei Li
- Department of Veterinary Integrative Biosciences, Texas A&M University, College Station, TX 77843, USA; (N.N.); (X.F.); (J.J.C.)
| |
Collapse
|
10
|
Zhou W, Li J. Integrated Analysis of Genes Associated With Immune Microenvironment and Distant Metastasis in Uveal Melanoma. Front Cell Dev Biol 2022; 10:874839. [PMID: 35433689 PMCID: PMC9006059 DOI: 10.3389/fcell.2022.874839] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Accepted: 03/16/2022] [Indexed: 12/21/2022] Open
Abstract
Inflammatory infiltration plays an essential role in the progression of tumor malignancy. The aim of this study was to identify genes associated with inflammatory microenvironment and clinical traits for survival prediction of uveal melanoma (UVM) patients. The datasets and clinical characteristics of UVM were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. We divided the UVM patients into low and high immune cell infiltration groups, identified differentially expressed genes (DEGs), constructed weighted gene co-expression network, and established prognostic prediction model and nomogram for UVM. Our analysis showed that DEGs were enriched in cytokine signaling in immune system, positive regulation of immune response and adaptive immune system. A total of fifteen candidate genes were extracted from DEGs and genes that were positively associated with tumor metastasis. Subsequently, five prognostic genes were selected to construct the final prognostic prediction model, including two up-regulated genes LHFPL3 antisense RNA 1 (LHFPL3-AS1) and LYN proto-oncogene (LYN), and three down-regulated genes SLCO4A1 antisense RNA 1 (SLCO4A1-AS1), Zinc-α2-glycoprotein 1 (AZGP1) and Deleted in Liver Cancer-1 (DLC1) in the high risk group. The model showed an Area Under Curve (AUC) value of 0.877. Our analysis highlighted the importance of immune-related genes in the progression of UVM and also provided potential targets for the immunotherapy of UVM.
Collapse
|
11
|
Zhang L, Zhao Y, Guan H, Zhang D. HnRNPU-AS1 inhibits the proliferation, migration and invasion of HCC cells and induces autophagy through miR-556-3p/ miR-580-3p/SOCS6 axis. Cancer Biomark 2022; 34:443-457. [PMID: 35275521 DOI: 10.3233/cbm-210261] [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] [Indexed: 11/15/2022]
Abstract
BACKGROUND Long non-coding RNAs have drawn increasing research interest in cancer biology. This study aims to investigate the function roles and the underlying mechanism of HnRNPU-AS1 in Hepatocellular carcinoma (HCC). METHODS qRT-PCR was performed to detect the expression levels of HnRNPU-AS1, miR-556-3p, miR-580-3p in HCC tissues and cell lines. Western blot was used to determine protein levels of LC3-II, LC3-I, Beclin-1, P62, and SOCS6. Functional assays including CCK8 assay, colony formation assay, wound healing assay, Transwell assay were performed to evaluate the role of HnRNPU-AS1 in regulating the malignant phenotype of HCC cells. Dual luciferase reporter assay and RNA pull-down experiment were used to examined the RNA-RNA interaction. RESULTS HnRNPU-AS1 expression was decreased in HCC tissues and cell lines, which was associated with poor prognosis in HCC patients. Overexpression of HnRNPU-AS1 could inhibit the proliferation, migration, invasion but promote autophagy in HCC cells. Two miRNAs (miR-556-3p and miR-580-3p) were identified as potential targets of HnRNPU-AS1 in lncBASE database, which were significantly upregulated in HCC tissues and cell lines. Cell experiments demonstrated the effects of HnRNPU-AS1 overexpression could be attenuated by miR-556-3p or miR-580-3p overexpression. We further revealed that SOX6 was the downstream target of HnRNPU-AS1/miR-556-3p or miR-580-3p axis. Xenograft mouse model validated the tumor-suppressor role of HnRNPU-AS1 overexpression in vivo. CONCLUSIONS This study demonstrated the tumor suppressor function of HnRNPU-AS1 in HCC and identified the downstream molecules underlying its tumor suppressor function. Our results suggest that HnRNPU-AS1 suppresses HCC by targeting miR-556-3p and miR-580-3p/SOXS6 axis.
Collapse
|
12
|
Wang W, Zhang Z, Li Y, Gu A, Wang Y, Cai Y, Yu Y, Deng X. Down-regulated long non-coding RNA LHFPL3 antisense RNA 1 inhibits the radiotherapy resistance of nasopharyngeal carcinoma via modulating microRNA-143-5p/homeobox A6 axis. Bioengineered 2022; 13:5421-5433. [PMID: 35176945 PMCID: PMC9208473 DOI: 10.1080/21655979.2021.2024386] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The function of long non-coding RNA LHFPL3 antisense RNA 1 (LHFPL3-AS1) in cancer progression has been studied, while its role in nasopharyngeal carcinoma (NPC) remains unclear. This study aims to unravel the effects of LHFPL3-AS1 on NPC progression via microRNA (miR)-143-5p/homeobox A6 (HOXA6) axis. NPC tissues were collected and NPC cells were cultured. NPC cells were subjected to radiation therapy to construct the radiation therapy resistance NPC cell line. The levels of LHFPL3-AS1, miR-143-5p and HOXA6 in NPC cells and tissues were examined. LHFPL3-AS1, miR-143-5p or HOXA6 expression was changed and then transfected into radiation-resistant NPC cells to detect cell proliferation, colony formation, migration, invasion and cell apoptosis in vitro. The tumorigenesis in nude mice in vivo was conducted to detect tumor growth. The targeting relations among LHFPL3-AS1, miR-143-5p and HOXA6 were validated. It was discovered that LHFPL3-AS1 and HOXA6 expression was elevated while the miR-143-5p level was depleted in radiation-resistant NPC cells and NPC tissues. The silenced LHFPL3-AS1 or augmented miR-143-5p repressed the proliferation, colony formation, migration and invasion of radiation-resistant NPC cells, while accelerated cell apoptosis in vitro. Silenced LHFPL3-AS1 hindered tumor growth in vivo. MiR-143-5p deletion reversed the effects of reduced LHFPL3-AS1; while HOXA6 upregulation reversed the effects of enriched miR-143-5p. LHFPL3-AS1 sponged miR-143-5p that targeted HOXA6. It is concluded that the down-regulated LHFPL3-AS1 retards the development of radiation-resistant NPC cells via sponging miR-143-5p to modulate HOXA6. This study reveals novel therapeutic targets for NPC treatment.
Collapse
Affiliation(s)
- Weifeng Wang
- Department of Radiotherapy, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Zhuo Zhang
- Department of Radiotherapy, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Yundong Li
- Department of Radiotherapy, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Anqi Gu
- Department of Radiotherapy, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Yingyin Wang
- Department of Radiotherapy, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Yizheng Cai
- Department of Radiotherapy, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Yajie Yu
- Department of Radiotherapy, Hainan Cancer Hospital, Haikou, Hainan, China
| | - Xiaocong Deng
- Department of Head and Neck Surgery, Hainan Cancer Hospital, Haikou, Hainan, China
| |
Collapse
|
13
|
Melixetian M, Pelicci PG, Lanfrancone L. Regulation of LncRNAs in Melanoma and Their Functional Roles in the Metastatic Process. Cells 2022; 11:577. [PMID: 35159386 PMCID: PMC8834033 DOI: 10.3390/cells11030577] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 02/02/2022] [Accepted: 02/05/2022] [Indexed: 02/06/2023] Open
Abstract
Long non-coding RNAs (lncRNAs) are key regulators of numerous intracellular processes leading to tumorigenesis. They are frequently deregulated in cancer, functioning as oncogenes or tumor suppressors. As they act through multiple mechanisms, it is not surprising that they may exert dual functions in the same tumor. In melanoma, a highly invasive and metastatic tumor with the propensity to rapidly develop drug resistance, lncRNAs play different roles in: (i) guiding the phenotype switch and leading to metastasis formation; (ii) predicting the response of melanoma patients to immunotherapy; (iii) triggering adaptive responses to therapy and acquisition of drug resistance phenotypes. In this review we summarize the most recent findings on the lncRNAs involved in melanoma growth and spreading to distant sites, focusing on their role as biomarkers for disease diagnosis and patient prognosis, or targets for novel therapeutic approaches.
Collapse
Affiliation(s)
- Marine Melixetian
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (M.M.); (P.G.P.)
| | - Pier Giuseppe Pelicci
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (M.M.); (P.G.P.)
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Luisa Lanfrancone
- Department of Experimental Oncology, IEO, European Institute of Oncology IRCCS, 20139 Milan, Italy; (M.M.); (P.G.P.)
| |
Collapse
|
14
|
Long Noncoding RNA RMRP Contributes to Paclitaxel Sensitivity of Ovarian Cancer by Regulating miR-580-3p/MICU1 Signaling. JOURNAL OF ONCOLOGY 2022; 2022:8301941. [PMID: 35132320 PMCID: PMC8817877 DOI: 10.1155/2022/8301941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/21/2021] [Accepted: 12/23/2021] [Indexed: 11/25/2022]
Abstract
Ovarian cancer is a prevalent female malignancy affecting the health and life of an increasing population of women around the world. Paclitaxel (PTX) resistance is a significant clinical problem in the treatment of ovarian cancer. However, the regulation mechanism of PTX resistance remains unclear. In this investigation, we reported an innovative function of the long noncoding RNA RMRP in promoting PTX resistance and glycolysis of ovarian cancer cells. We observed that RMRP was highly expressed in the ovarian cancer samples, in which the expression of RMRP was elevated in the PTX-resistant patients compared with the PTX-sensitive patients. Meanwhile, RMRP was upregulated in PTX-resistant ovarian cancer cell lines. Functionally, we found that the silencing of RMRP by siRNA significantly enhanced the PTX sensitivity of PTX-resistant ovarian cancer cells, in which the IC50 of PTX was reduced by RMRP depletion. The RMRP knockdown reduced cell viabilities and enhanced cell apoptosis of PTX-resistant ovarian cancer cells. Moreover, we observed that glucose uptake was enhanced in PTX-resistant ovarian cancer cells. The depletion of RMRP decreased glucose uptake, lactate product, and ATP production in PTX-resistant ovarian cancer cells. About the mechanism, we identified that RMRP was able to sponge miR-580-3p to enhance mitochondrial calcium uptake 1 (MICU1) expression in PTX-resistant ovarian cancer cells. MICU1 overexpression and miR-580-3p repression could reverse the RMRP-inhibited proliferation of PTX-resistant ovarian cancer cells in vitro. Thus, we concluded that RMRP contributes to PTX resistance and glycolysis of ovarian cancer by enhancing MICU1 expression through sponging miR-580-3p. Targeting RMRP may serve as a potential therapeutic strategy for the treatment of PTX-resistant ovarian cancer patients.
Collapse
|
15
|
Ma J, Shi Q, Guo S, Xu P, Yi X, Yang Y, Zhang W, Liu Y, Liu L, Yue Q, Zhao T, Gao T, Guo W, Li C. Long Non-Coding RNA CD27-AS1-208 Facilitates Melanoma Progression by Activating STAT3 Pathway. Front Oncol 2022; 11:818178. [PMID: 35096622 PMCID: PMC8791859 DOI: 10.3389/fonc.2021.818178] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 12/17/2021] [Indexed: 11/21/2022] Open
Abstract
Melanoma is the most lethal skin cancer that originates from epidermal melanocytes. Recently, long non-coding RNAs (lncRNAs) are emerging as critical regulators of cancer pathogenesis and potential therapeutic targets. However, the expression profile of lncRNAs and their role in melanoma progression have not been thoroughly investigated. Herein, we firstly obtained the expression profile of lncRNAs in primary melanomas using microarray analysis and unveiled the differentially-expressed lncRNAs compared with nevus. Subsequently, a series of bioinformatics analysis showed the great involvement of dysregulated lncRNAs in melanoma biology and immune response. Further, we identified lncRNA CD27-AS1-208 as a novel nuclear-localized factor with prominent facilitative role in melanoma cell proliferation, invasion and migration. Mechanistically, CD27-AS1-208 could directly interact with STAT3 and contribute to melanoma progression in a STAT3-dependent manner. Ultimately, the role of CD27-AS1-208 in melanoma progression in vivo was also investigated. Collectively, the present study offers us a new horizon to better understand the role of lncRNAs in melanoma pathogenesis and demonstrates that CD27-AS1-208 up-regulation contributes to melanoma progression by activating STAT3 pathway. Targeting CD27-AS1-208 in melanoma cells can be exploited as a potential therapeutic approach that needs forward validation in clinical trials in the future.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | - Weinan Guo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| | - Chunying Li
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi’an, China
| |
Collapse
|
16
|
Wozniak M, Czyz M. The Functional Role of Long Non-Coding RNAs in Melanoma. Cancers (Basel) 2021; 13:cancers13194848. [PMID: 34638331 PMCID: PMC8508152 DOI: 10.3390/cancers13194848] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/23/2021] [Accepted: 09/25/2021] [Indexed: 12/15/2022] Open
Abstract
Melanoma is the most lethal skin cancer, with increasing incidence worldwide. The molecular events that drive melanoma development and progression have been extensively studied, resulting in significant improvements in diagnostics and therapeutic approaches. However, a high drug resistance to targeted therapies and adverse effects of immunotherapies are still a major challenge in melanoma treatment. Therefore, the elucidation of molecular mechanisms of melanomagenesis and cancer response to treatment is of great importance. Recently, many studies have revealed the close association of long noncoding RNAs (lncRNAs) with the development of many cancers, including melanoma. These RNA molecules are able to regulate a plethora of crucial cellular processes including proliferation, differentiation, migration, invasion and apoptosis through diverse mechanisms, and even slight dysregulation of their expression may lead to tumorigenesis. lncRNAs are able to bind to protein complexes, DNA and RNAs, affecting their stability, activity, and localization. They can also regulate gene expression in the nucleus. Several functions of lncRNAs are context-dependent. This review summarizes current knowledge regarding the involvement of lncRNAs in melanoma. Their possible role as prognostic markers of melanoma response to treatment and in resistance to therapy is also discussed.
Collapse
|
17
|
Wong NK, Luo S, Chow EYD, Meng F, Adesanya A, Sun J, Ma HMH, Jin W, Li WC, Yip SP, Huang CL. The Tyrosine Kinase-Driven Networks of Novel Long Non-coding RNAs and Their Molecular Targets in Myeloproliferative Neoplasms. Front Cell Dev Biol 2021; 9:643043. [PMID: 34414175 PMCID: PMC8369571 DOI: 10.3389/fcell.2021.643043] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Accepted: 07/09/2021] [Indexed: 01/16/2023] Open
Abstract
Recent research has focused on the mechanisms by which long non-coding RNAs (lncRNAs) modulate diverse cellular processes such as tumorigenesis. However, the functional characteristics of these non-coding elements in the genome are poorly understood at present. In this study, we have explored several mechanisms that involve the novel lncRNA and microRNA (miRNA) axis participating in modulation of drug response and the tumor microenvironment of myeloproliferative neoplasms (MPNs). We identified novel lncRNAs via mRNA sequencing that was applied to leukemic cell lines derived from BCR-ABL1-positive and JAK2-mutant MPNs under treatment with therapeutic tyrosine kinase inhibitors (TKI). The expression and sequence of novel LNC000093 were further validated in both leukemic cells and normal primary and pluripotent cells isolated from human blood, including samples from patients with chronic myelogenous leukemia (CML). Downregulation of LNC000093 was validated in TKI-resistant CML while a converse expression pattern was observed in blood cells isolated from TKI-sensitive CML cases. In addition to BCR-ABL1-positive CML cells, the driver mutation JAK2-V617F-regulated lncRNA BANCR axis was further identified in BCR-ABL1-negative MPNs. Further genome-wide validation using MPN patient specimens identified 23 unique copy number variants including the 7 differentially expressed lncRNAs from our database. The newly identified LNC000093 served as a competitive endogenous RNA for miR-675-5p and reversed the imatinib resistance in CML cells through regulating RUNX1 expression. The extrinsic function of LNC000093 in exosomal H19/miR-675-induced modulation for the microenvironment was also determined with significant effect on VEGF expression.
Collapse
Affiliation(s)
- Nonthaphat Kent Wong
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Shumeng Luo
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Eudora Y D Chow
- Department of Pathology, United Christian Hospital, Kwun Tong, Hong Kong
| | - Fei Meng
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Adenike Adesanya
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Jiahong Sun
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong.,Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Herman M H Ma
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong.,Department of Pathology, United Christian Hospital, Kwun Tong, Hong Kong
| | - Wenfei Jin
- Department of Biology, Southern University of Science and Technology, Shenzhen, China
| | - Wan-Chun Li
- Institute of Oral Biology, College of Dentistry, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| | - Chien-Ling Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Kowloon, Hong Kong
| |
Collapse
|
18
|
Tang G, Liu L, Xiao Z, Wen S, Chen L, Yang P. CircRAB3IP upregulates twist family BHLH transcription factor (TWIST1) to promote osteosarcoma progression by sponging miR-580-3p. Bioengineered 2021; 12:3385-3397. [PMID: 34224315 PMCID: PMC8806556 DOI: 10.1080/21655979.2021.1948487] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Circular RNAs (circ RNAs) have been found to play an important role in cancer development. However, the role of circRAB3IP in osteosarcoma (OS) is unclear. In the present study, We found that circRAB3IP was highly expressed in OS tissues and OS cells. High levels of circRAB3IP was correlated with advanced TNM stage, distant metastasis. CircRAB3IP knockdown inhibited cell proliferation, migration, and invasion. Moreover, circRAB3IP directly binds to miR-580-3p. TWIST1 is directly targeted by miR-580-3p. We also demonstrated that circRAB3IP act as the sponge of miR-580-3p to promote TWIST1 expression. CircRAB3IP promotes OS cells proliferation, migration, and invasion through modulating miR-580-3p/TWIST1 axis. Moreover, circRAB3IP facilitated tumor formation in vivo. Our findings suggested that circRAB3IP acts as an oncogene in OS by regulating miR-580-3p/TWIST1 axis.
Collapse
Affiliation(s)
- Guojun Tang
- Department of Spine Surgery, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, P.R. China
| | - Linghua Liu
- Department of Nursing, Hubei College of Chinese Medicine, Jingzhou, Hubei, P.R. China
| | - Zhihong Xiao
- Department of Spine Surgery, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, P.R. China
| | - Shuo Wen
- Department of Breast and Thyroid Surgery, Union Hospital West Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| | - Liangyuan Chen
- Department of Spine Surgery, The Second Affiliated Hospital, University of South China, Hengyang, Hunan, P.R. China
| | - Peng Yang
- Department of Breast and Thyroid Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, P.R. China
| |
Collapse
|
19
|
Li J, Xu X, Zhang D, Lv H, Lei X. LncRNA LHFPL3-AS1 Promotes Oral Squamous Cell Carcinoma Growth and Cisplatin Resistance Through Targeting miR-362-5p/CHSY1 Pathway. Onco Targets Ther 2021; 14:2293-2300. [PMID: 33833527 PMCID: PMC8020056 DOI: 10.2147/ott.s298679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 03/08/2021] [Indexed: 12/16/2022] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) is a common oral cancer. The current study aims to elucidate the potential roles of long noncoding RNA (lncRNA) LHFPL3-AS1 in OSCC development. Methods Gene expression was measured by qRT-PCR in tumor tissues and cell lines. Loss-of-function assays were performed to analyze the effects of LHFPL3-AS1 on malignant behaviors. Bioinformatics analysis was conducted to explore the downstream signaling pathway of LHFPL3-AS1 in OSCC. Results LHFPL3-AS1 was highly expressed in OSCC tissues and cell lines. LHFPL3-AS1 was upregulated in cisplatin-resistant tumor cell lines. LHFPL3-AS1 level was correlated with survival rate. LHFPL3-AS1 knockdown suppressed OSCC proliferation, migration and invasion. LHFPL3-AS1 downregulation reduced cisplatin resistance of OSCC cells. LHFPL3-AS1 was the competing endogenous RNA (ceRNA) for miR-194-5p to enhance CHSY1 expression. Conclusion LHFPL3-AS1/miR-362-5p/CHSY1 signaling pathway plays essential roles in regulating OSCC development and cisplatin resistance.
Collapse
Affiliation(s)
- Jiandong Li
- Department of Stomatology, Shenzhen Longhua District Central Hospital, Shenzhen, 518000, People's Republic of China
| | - Xiaohu Xu
- Department of Stomatology, Shenzhen Longhua District Central Hospital, Shenzhen, 518000, People's Republic of China
| | - Dandan Zhang
- Department of Stomatology, Shenzhen Longhua District Central Hospital, Shenzhen, 518000, People's Republic of China
| | - Han Lv
- Department of Stomatology, Shenzhen Longhua District Central Hospital, Shenzhen, 518000, People's Republic of China
| | - Xin Lei
- Department of Stomatology, Shenzhen Longhua District Central Hospital, Shenzhen, 518000, People's Republic of China
| |
Collapse
|