1
|
Abusharieh E, Aslam N, Zihlif MA, Bustanji Y, Wehaibi S, Abuarqoub D, Shahin D, Saadeh H, Barham R, Awidi AS. In vitro investigation of epigenetic regulators related to chemo-resistance and stemness of CD133 +VE cells sorted from U87MG cell line. Gene 2025; 956:149432. [PMID: 40157620 DOI: 10.1016/j.gene.2025.149432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 03/11/2025] [Accepted: 03/18/2025] [Indexed: 04/01/2025]
Abstract
Glioblastoma (GBM) is the most common and malignant adult primary brain tumor with frequent relapse and resistance to therapies. Glioma stem cells, a rare population, is thought to be the reason behind the treatment's failure. It is imperative to investigate the disease mechanisms and identify the biomarkers by which glioma stem cells would contribute to treatment relapse and resistance to already available chemotherapeutic agents. The CD133+VE cells were isolated from U87MG cell line and characterized by morphological features, cell viability, self-renewal efficiency, migration potential and karyotyping. Doxorubicin Cisplatin, Irinotecan, Etoposide and Temozolomide were used to determine the anti-proliferative effect on CD133+VE cells. Confocal microcopy was used to localize the chemotherapeutic agents in the CD133+VE cells. In quest of epigenetic biomarkers, RNA sequencing was performed to find the role of lncRNAs in stemness and resistance to therapies. U87cell line and CD133-VE cells were kept as controls for all the experiments. It was found that CD133+VEcells were highly proliferative with increased migration potential, elevated IC50 values against chemotherapeutic agents and showed distinct karyotyping related to pluripotency. Chemotherapeutic agent such as Doxorubicin was localized outside the nucleus revealing the drug resistance as evident by confocal microscopy. RNA sequencing revealed 126 differentially expressed lncRNAs (DELs) in CD133+VEcells among which lncRNA LOXL1-AS1 was highly upregulated and lncRNA PAX8-AS1 was significantly downregulated. These lncRNAs has been reported to be related to drug resistance, migration and epithelial- to- mesenchymal transmission (EMT), self-renewal and stemness properties contributing to poor prognosis and disease relapse.
Collapse
Affiliation(s)
- Elham Abusharieh
- Department of Pharmaceutical Science, Faculty of Pharmacy, Al-zaytoonah University of Jordan, Amman 11733, Jordan; Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; Department of Clinical Pharmacy and Biopharmaceutics, Faculty of Pharmacy, The University of Jordan, Amman, Jordan.
| | - Nazneen Aslam
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Malek A Zihlif
- Faculty of Medicine, The University of Jordan, Amman 11942, Jordan
| | - Yasser Bustanji
- Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah 27272, United Arab Emirates; Department of Clinical Pharmacy and Biopharmaceutics, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| | - Suha Wehaibi
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Duaa Abuarqoub
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; Department of Pharmacology and Biomedical Sciences, Faculty of Pharmacy and Medical Sciences, University of Petra. Amman 11196, Jordan
| | - Diana Shahin
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Heba Saadeh
- Department of Computer Science, KASIT, The University of Jordan, Amman, 11942 Jordan
| | - Raghad Barham
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan
| | - Abdalla S Awidi
- Cell Therapy Center, The University of Jordan, Amman 11942, Jordan; Faculty of Medicine, The University of Jordan, Amman 11942, Jordan; Department of Hematology and Oncology, Jordan University Hospital, The University of Jordan, Amman 11942, Jordan.
| |
Collapse
|
2
|
Shao W, Feng Y, Huang J, Li T, Gao S, Yang Y, Li D, Yang Z, Yao Z. Interaction of ncRNAs and the PI3K/AKT/mTOR pathway: Implications for osteosarcoma. Open Life Sci 2024; 19:20220936. [PMID: 39119480 PMCID: PMC11306965 DOI: 10.1515/biol-2022-0936] [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: 05/18/2024] [Revised: 07/07/2024] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Osteosarcoma (OS) is the most common primary malignant bone tumor in children and adolescents, and is characterized by high heterogeneity, high malignancy, easy metastasis, and poor prognosis. Recurrence, metastasis, and multidrug resistance are the main problems that limit the therapeutic effect and prognosis of OS. PI3K/AKT/mTOR signaling pathway is often abnormally activated in OS tissues and cells, which promotes the rapid development, metastasis, and drug sensitivity of OS. Emerging evidence has revealed new insights into tumorigenesis through the interaction between the PI3K/AKT/mTOR pathway and non-coding RNAs (ncRNAs). Therefore, we reviewed the interactions between the PI3K/AKT/mTOR pathway and ncRNAs and their implication in OS. These interactions have the potential to serve as cancer biomarkers and therapeutic targets in clinical applications.
Collapse
Affiliation(s)
- Weilin Shao
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Yan Feng
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Jin Huang
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Tingyu Li
- Clinical Oncology Institute, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Shengguai Gao
- Clinical Oncology Institute, Kunming Medical University, Kunming, Yunnan, 650500, China
| | - Yihao Yang
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Dongqi Li
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Zuozhang Yang
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, Yunnan, 650118, China
| | - Zhihong Yao
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, 650118, China
- Department of Cancer Research Institute, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), No. 519 Kunzhou Road, Xishan District, Kunming, Yunnan, 650118, China
| |
Collapse
|
3
|
Yousefnia S. A comprehensive review on lncRNA LOXL1-AS1: molecular mechanistic pathways of lncRNA LOXL1-AS1 in tumorigenicity of cancer cells. Front Oncol 2024; 14:1384342. [PMID: 39136001 PMCID: PMC11317273 DOI: 10.3389/fonc.2024.1384342] [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: 02/09/2024] [Accepted: 07/15/2024] [Indexed: 08/15/2024] Open
Abstract
Long non-coding RNAs (lncRNAs) are versatile RNAs that regulate various cellular processes, such as gene regulation, by acting as signals, decoys, guides, and scaffolds. A novel recognized lncRNA, LOXL1-antisense RNA 1 (LOXL1-AS1), is dysregulated in some diseases, including cancer, and acts as an oncogenic lncRNA in many types of cancer cells. Upregulation of LOXL1-AS1 has been involved in proliferation, migration, metastasis, and EMT, as well as inhibiting apoptosis in cancer cells. Most importantly, the malignant promoting activity of LOXL1-AS1 can be mostly mediated by sequestering specific miRNAs and inhibiting their binding to the 3´UTR of their target mRNAs, thereby indirectly regulating gene expression. Additionally, LOXL1-AS1 can decoy transcription factors and proteins and prevent their binding to their regulatory regions, inhibiting their mechanistic activity on the regulation of gene expression and signaling pathways. This review presents the mechanistic pathways of the oncogenic role of LOXL1-AS1 by modulating its target miRNAs and proteins in various cancer cells. Having information about the molecular mechanisms regulated by LOXL1-AS1 in cancer cells can open ways to find out particular prognostic biomarkers, as well as discover novel therapeutic approaches for different types of cancer.
Collapse
Affiliation(s)
- Saghar Yousefnia
- Department of Cell and Molecular Biology, Semnan University, Semnan, Iran
| |
Collapse
|
4
|
Yan C, Dou Y, Xia R, Liu S, Fu J, Li D, Wang R, Tie F, Li L, Jin H, An F. Research progress on the role of lncRNA, circular RNA, and microRNA networks in regulating ferroptosis in osteosarcoma. Biomed Pharmacother 2024; 176:116924. [PMID: 38876052 DOI: 10.1016/j.biopha.2024.116924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 06/07/2024] [Accepted: 06/09/2024] [Indexed: 06/16/2024] Open
Abstract
Noncoding RNAs (ncRNAs) do not participate in protein-coding. Ferroptosis is a newly discovered form of cell death mediated by reactive oxygen species and lipid peroxidation. Recent studies have shown that ncRNAs such as microRNAs, long noncoding RNAs, circular RNAs, and ferroptosis are involved in the occurrence and development of osteosarcoma (OS). Studies have confirmed that ncRNAs participate in the development of OS by regulating the ferroptosis. However, systematic summary on this topic are still lacking. This review summarises the potential role of ncRNAs in the diagnosis, treatment, drug resistance, and prognosis of OS and the basis for diagnosing, preventing, and treating clinical OS and developing effective drugs. This review summarises the latest research progress on ncRNAs that regulate ferroptosis in OS, attempts to clarify the molecular mechanisms by which ncRNAs regulate ferroptosis in the pathogenesis of OS, and elaborates on the involvement of ferroptosis in OS from the perspective of ncRNAs.
Collapse
Affiliation(s)
- Chunlu Yan
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Yinnan Dou
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Ruoliu Xia
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Shiqing Liu
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Jianchao Fu
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Duo Li
- The First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Rong Wang
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Feng Tie
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Linxin Li
- School of Traditional Chinese and Western Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China
| | - Hua Jin
- Clinical College of Chinese Medicine, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China.
| | - Fangyu An
- Teaching Experiment Training Center, Gansu University of Chinese Medicine, Lanzhou, Gansu 730000, China.
| |
Collapse
|
5
|
Wang X, Liu R, Li J, Wang B, Lin Y, Zi J, Yu M, Pu Y, Xiong W. Involvement of long non-coding RNA LOXL1-AS1 in the tumourigenesis and development of malignant tumours: a narrative review. Transl Cancer Res 2024; 13:3142-3155. [PMID: 38988912 PMCID: PMC11231786 DOI: 10.21037/tcr-23-2282] [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: 12/12/2023] [Accepted: 04/24/2024] [Indexed: 07/12/2024]
Abstract
Background and Objective Long noncoding RNAs (lncRNAs) are involved in a wide variety of physiological and pathological processes in organisms. LncRNAs play a significant role as oncogenic or tumour-suppressing factors in various biological processes associated with malignant tumours and are closely linked to the occurrence and development of malignancies. Lysyl oxidase like 1 antisense RNA 1 (LOXL1-AS1) is a recently discovered lncRNA. It is upregulated in various malignant tumours and is associated with pathological characteristics such as tumour size, tumour node metastasis (TNM) staging, lymph node metastasis, and tumour prognosis. LOXL1-AS1 exerts its oncogenic role by competitively binding with multiple microRNAs (miRs), thereby regulating the expression of downstream target genes and controlling relevant signalling pathways. This article aims to explore the structure and the function of LOXL1-AS1, and the relationship between LOXL1-AS1 and the occurrence and development of human malignant tumours to provide a reference for further clinical research. Methods English literature on LOXL1-AS1 in the occurrence and development of various malignant tumours was searched in PubMed. The main search terms were "LOXL1-AS1", "tumour". Key Content and Findings This article mainly summarizes the biological processes in which LOXL1-AS1 is involved in various human malignant tumours and the ways in which this lncRNA affects malignant biological behaviours such as proliferation, metastasis, invasion, and apoptosis of tumour cells through different molecular regulatory mechanisms. This article also explores the potential clinical significance and application prospects of LOXL1-AS1, aiming to provide a theoretical basis and reference for the clinical diagnosis, treatment, and screening of prognostic markers for malignant tumours. Conclusions LOXL1-AS1 acts as a competing endogenous RNA (ceRNA), binding to miRs to regulate downstream target genes and exert its oncogenic effects. LOXL1-AS1 may become a novel molecular biomarker for cancer diagnosis and treatment in humans, and it may also serve as an independent prognostic indicator.
Collapse
Affiliation(s)
- Xinmeng Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University, Dali, China
- Key Laboratory of Clinical Biochemistry Testing in Universities of Yunnan Province, School of Basic Medical Sciences, Dali University, Dali, China
| | - Ruai Liu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University, Dali, China
- Key Laboratory of Clinical Biochemistry Testing in Universities of Yunnan Province, School of Basic Medical Sciences, Dali University, Dali, China
| | - Jinsong Li
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University, Dali, China
- Key Laboratory of Clinical Biochemistry Testing in Universities of Yunnan Province, School of Basic Medical Sciences, Dali University, Dali, China
| | - Boyong Wang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University, Dali, China
- Key Laboratory of Clinical Biochemistry Testing in Universities of Yunnan Province, School of Basic Medical Sciences, Dali University, Dali, China
| | - Yaru Lin
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University, Dali, China
- Key Laboratory of Clinical Biochemistry Testing in Universities of Yunnan Province, School of Basic Medical Sciences, Dali University, Dali, China
| | - Jiaji Zi
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University, Dali, China
- Key Laboratory of Clinical Biochemistry Testing in Universities of Yunnan Province, School of Basic Medical Sciences, Dali University, Dali, China
| | - Min Yu
- Laboratory of Biochemistry and Molecular Biology, School of Life Sciences, Yunnan University, Kunming, China
| | - Yuanqian Pu
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University, Dali, China
- Key Laboratory of Clinical Biochemistry Testing in Universities of Yunnan Province, School of Basic Medical Sciences, Dali University, Dali, China
| | - Wei Xiong
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Dali University, Dali, China
- Key Laboratory of Clinical Biochemistry Testing in Universities of Yunnan Province, School of Basic Medical Sciences, Dali University, Dali, China
| |
Collapse
|
6
|
Fu XP, Ji CY, Tang WQ, Yu TT, Luo L. Long non-coding RNA LOXL1-AS1: a potential biomarker and therapeutic target in human malignant tumors. Clin Exp Med 2024; 24:93. [PMID: 38693424 PMCID: PMC11062969 DOI: 10.1007/s10238-024-01355-7] [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: 03/26/2024] [Accepted: 04/15/2024] [Indexed: 05/03/2024]
Abstract
Long non-coding RNAs (lncRNAs) are transcripts that contain more than 200 nucleotides. Despite their inability to code proteins, multiple studies have identified their important role in human cancer through different mechanisms. LncRNA lysyl oxidase like 1 antisense RNA 1 (LOXL1-AS1), a newly discovered lncRNA located on human chromosome 15q24.1, has recently been shown to be involved in the occurrence and progression of various malignancies, such as colorectal cancer, gastric cancer, hepatocellular carcinoma, prostate cancer, non-small cell lung cancer, ovarian cancer, cervical cancer, breast cancer, glioma, thymic carcinoma, pancreatic carcinoma. LOXL1-AS1 acts as competitive endogenous RNA (ceRNA) and via sponging various miRNAs, including miR-374b-5p, miR-21, miR-423-5p, miR-589-5p, miR-28-5p, miR-324-3p, miR-708-5p, miR-143-3p, miR-18b-5p, miR-761, miR-525-5p, miR-541-3p, miR-let-7a-5p, miR-3128, miR-3614-5p, miR-377-3p and miR-1224-5p to promote tumor cell proliferation, invasion, migration, apoptosis, cell cycle, and epithelial-mesenchymal transformation (EMT). In addition, LOXL1-AS1 is involved in the regulation of P13K/AKT and MAPK signaling pathways. This article reviews the current understanding of the biological function and clinical significance of LOXL1-AS1 in human cancers. These findings suggest that LOXL1-AS1 may be both a reliable biomarker and a potential therapeutic target for cancers.
Collapse
Affiliation(s)
- Xiao-Ping Fu
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Hongshan District, 856 Luoyu Road, Wuhan, 430070, People's Republic of China
| | - Chun-Yan Ji
- Department of Gastroenterology, Hubei Provincial Hospital of Traditional Chinese and Western Medicine, Wuhan, 430015, People's Republic of China
| | - Wen-Qian Tang
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Hongshan District, 856 Luoyu Road, Wuhan, 430070, People's Republic of China
| | - Ting-Ting Yu
- School of Clinical Medical, Hubei University of Chinese Medicine, Wuhan, 443000, People's Republic of China
| | - Lei Luo
- Department of Health Management Center, Hubei Provincial Hospital of Traditional Chinese Medicine, Hongshan District, 856 Luoyu Road, Wuhan, 430070, People's Republic of China.
| |
Collapse
|
7
|
Tang M, Rong Y, Liu S, Wu Z, Ma G, Li X, Cai H. Potential role of lncRNA LOXL1-AS1 in human cancer development: a narrative review. Transl Cancer Res 2024; 13:1997-2011. [PMID: 38737681 PMCID: PMC11082674 DOI: 10.21037/tcr-23-1450] [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: 08/13/2023] [Accepted: 02/29/2024] [Indexed: 05/14/2024]
Abstract
Background and Objective Long non-coding RNAs (lncRNAs) are a group of non-coding RNAs consisting of more than 200 nucleotides that are widely involved in various physiological and pathobiological processes in the body. LncRNA plays a crucial role in tumorigenesis and development with its unique functions, such as playing a role in a variety of biological processes of malignant tumors as a cancer-promoting factor or a cancer-suppressor factor. Lysyl oxidase-like protein 1-antisense RNA1 (LOXL1-AS1) is a novel functional lncRNA recently reported. This article reviews the current findings on the role of LOXL1-AS1 in cancer, and discusses the potential clinical significance and application prospects, in order to provide a theoretical basis and reference for the clinical diagnosis, treatment and screening of prognostic markers for malignant tumors. Methods The PubMed and Embase databases were searched using the keywords "cancer" or "tumor" or "neoplasm" and "LOXL1-AS1" for publications from 2018 to the present. The English literature was searched, with a focus on relevant articles. These articles validated the role and mechanism of LOXL1-AS1 in different cancers. Key Content and Findings LOXL1-AS1 is a recently reported novel lncRNA, which is abnormally expressed and upregulated in more than ten cancers, and is positively correlated with adverse clinical features and poor prognosis in cancer patients. LOXL1-AS1 competently binds to a variety of microRNAs to regulate the expression of downstream target genes and regulate related signaling pathways, including proliferation, migration, invasion and inhibition of malignant biological behaviors such as apoptosis. Conclusions LOXL1-AS1 is expected to become a novel biomarker for cancer diagnosis and treatment, with great potential as an independent prognostic indicator.
Collapse
Affiliation(s)
- Mingzheng Tang
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, China
- National Health Council Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
| | - Yao Rong
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, China
- National Health Council Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
| | - Songhua Liu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, China
| | - Zhihang Wu
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Guorong Ma
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Xiaofeng Li
- The First Clinical Medical College of Gansu University of Chinese Medicine (Gansu Provincial Hospital), Lanzhou, China
| | - Hui Cai
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou, China
- National Health Council Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou, China
- The First Clinical Medical College of Lanzhou University, Lanzhou, China
| |
Collapse
|
8
|
Liao X, Wei R, Zhou J, Wu K, Li J. Emerging roles of long non-coding RNAs in osteosarcoma. Front Mol Biosci 2024; 11:1327459. [PMID: 38516191 PMCID: PMC10955361 DOI: 10.3389/fmolb.2024.1327459] [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: 10/25/2023] [Accepted: 02/12/2024] [Indexed: 03/23/2024] Open
Abstract
Osteosarcoma (OS) is a highly aggressive and lethal malignant bone tumor that primarily afflicts children, adolescents, and young adults. However, the molecular mechanisms underlying OS pathogenesis remain obscure. Mounting evidence implicates dysregulated long non-coding RNAs (lncRNAs) in tumorigenesis and progression. These lncRNAs play a pivotal role in modulating gene expression at diverse epigenetic, transcriptional, and post-transcriptional levels. Uncovering the roles of aberrant lncRNAs would provide new insights into OS pathogenesis and novel tools for its early diagnosis and treatment. In this review, we summarize the significance of lncRNAs in controlling signaling pathways implicated in OS development, including the Wnt/β-catenin, PI3K/AKT/mTOR, NF-κB, Notch, Hippo, and HIF-1α. Moreover, we discuss the multifaceted contributions of lncRNAs to drug resistance in OS, as well as their potential to serve as biomarkers and therapeutic targets. This review aims to encourage further research into lncRNA field and the development of more effective therapeutic strategies for patients with OS.
Collapse
Affiliation(s)
- Xun Liao
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, China
| | - Rong Wei
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Junxiu Zhou
- Frontiers Medical Center, Tianfu Jincheng Laboratory, Chengdu, Sichuan, China
| | - Ke Wu
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jiao Li
- Laboratory of Molecular Oncology, Frontiers Science Center for Disease-related Molecular Network, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
9
|
Jefferi NES, Shamhari A‘A, Noor Azhar NKZ, Shin JGY, Kharir NAM, Azhar MA, Hamid ZA, Budin SB, Taib IS. The Role of ERα and ERβ in Castration-Resistant Prostate Cancer and Current Therapeutic Approaches. Biomedicines 2023; 11:biomedicines11030826. [PMID: 36979805 PMCID: PMC10045750 DOI: 10.3390/biomedicines11030826] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 02/23/2023] [Accepted: 02/25/2023] [Indexed: 03/11/2023] Open
Abstract
Castration-resistant prostate cancer, or CRPC, is an aggressive stage of prostate cancer (PCa) in which PCa cells invade nearby or other parts of the body. When a patient with PCa goes through androgen deprivation therapy (ADT) and the cancer comes back or worsens, this is called CRPC. Instead of androgen-dependent signalling, recent studies show the involvement of the estrogen pathway through the regulation of estrogen receptor alpha (ERα) and estrogen receptor beta (ERβ) in CRPC development. Reduced levels of testosterone due to ADT lead to low ERβ functionality in inhibiting the proliferation of PCa cells. Additionally, ERα, which possesses androgen independence, continues to promote the proliferation of PCa cells. The functions of ERα and ERβ in controlling PCa progression have been studied, but further research is needed to elucidate their roles in promoting CRPC. Finding new ways to treat the disease and stop it from becoming worse will require a clear understanding of the molecular processes that can lead to CRPC. The current review summarizes the underlying processes involving ERα and ERβ in developing CRPC, including castration-resistant mechanisms after ADT and available medication modification in mitigating CRPC progression, with the goal of directing future research and treatment.
Collapse
Affiliation(s)
- Nur Erysha Sabrina Jefferi
- Center of Diagnostics, Therapeutics and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Asma’ ‘Afifah Shamhari
- Center of Diagnostics, Therapeutics and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Nur Khayrin Zulaikha Noor Azhar
- Biomedical Science Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Joyce Goh Yi Shin
- Biomedical Science Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Nur Annisa Mohd Kharir
- Biomedical Science Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Muhammad Afiq Azhar
- Biomedical Science Programme, Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Zariyantey Abd Hamid
- Center of Diagnostics, Therapeutics and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Siti Balkis Budin
- Center of Diagnostics, Therapeutics and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
| | - Izatus Shima Taib
- Center of Diagnostics, Therapeutics and Investigative Studies (CODTIS), Faculty of Health Sciences, Universiti Kebangsaan Malaysia, Jalan Raja Muda Abdul Aziz, Kuala Lumpur 50300, Malaysia
- Correspondence: ; Tel.: +0603-92897608
| |
Collapse
|
10
|
Wang X, Chen Z, Zhou H, Liu W, Luo J. LncRNA LOXL1-AS1 expression in cancer prognosis: A meta-analysis. Medicine (Baltimore) 2022; 101:e32436. [PMID: 36596047 PMCID: PMC9803452 DOI: 10.1097/md.0000000000032436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Several studies showed that LncRNA LOXL1 antisense RNA 1 (LOXL1-AS1) is overexpressed in a variety of cancers and plays a role as an oncogene in cancer. The present meta-analysis aims to elucidate the relationship between LOXL1-AS1 expression and prognosis and clinicopathological features among cancer patients. METHODS PubMed, Web of Science, Cochrane Library, and EMBASE database were comprehensively and systematically searched. Pooled odds ratios (ORs) and hazard ratios with a 95% confidence interval (CI) were employed to assess the relationship between LOXL1-AS1 expression and clinical outcomes and clinicopathological features in cancer patients. RESULTS The present study finally enrolled 8 studies which included 657 cancer patients. The combined results indicated that the overexpression of LOXL1-AS1 was significantly associated with shorter overall survival (pooled hazard ratio = 1.99, 95% CI 1.49-2.65, P < .00001). Meanwhile, regarding clinicopathology of cancer patients, the upregulation of LOXL1-AS1 expression was closely related to lymph node metastasis (yes vs no OR = 4.01, 95% CI: 2.02-7.96, P < .0001) and distant metastasis (yes vs no OR = 3.04, 95% CI: 1.82-5.06, P < .0001), respectively. CONCLUSION High expression of LOXL1-AS1 in some cancers predicts shorter overall survival, distant metastasis, and lymph node metastasis. LOXL1-AS1 shows great promise as a prognostic biomarker in cancer patients.
Collapse
Affiliation(s)
- Xuhua Wang
- Department of Spine Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Zhaoyuan Chen
- Department of Spine Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Huaqiang Zhou
- Department of Spine Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Wuyang Liu
- Department of Spine Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
| | - Jiaquan Luo
- Department of Spine Surgery, The First Affiliated Hospital of Gannan Medical University, Ganzhou, P.R. China
- * Correspondence: Jiaquan Luo Department of Spine Surgery, The First Affiliated Hospital of Gannan Medical University, No. 128, Jingling West Road, Ganzhou City, Jiangxi Province 341099, China (e-mail: )
| |
Collapse
|
11
|
Xu K, Jiang X, Ariston Gabriel AN, Li X, Wang Y, Xu S. Evolving Landscape of Long Non-coding RNAs in Cerebrospinal Fluid: A Key Role From Diagnosis to Therapy in Brain Tumors. Front Cell Dev Biol 2021; 9:737670. [PMID: 34692695 PMCID: PMC8529119 DOI: 10.3389/fcell.2021.737670] [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: 07/07/2021] [Accepted: 08/31/2021] [Indexed: 11/23/2022] Open
Abstract
Long non-coding RNAs (lncRNAs) are a type of non-coding RNAs that act as molecular fingerprints and modulators of many pathophysiological processes, particularly in cancer. Specifically, lncRNAs can be involved in the pathogenesis and progression of brain tumors, affecting stemness/differentiation, replication, invasion, survival, DNA damage response, and chromatin dynamics. Furthermore, the aberrations in the expressions of these transcripts can promote treatment resistance, leading to tumor recurrence. The development of next-generation sequencing technologies and the creation of lncRNA-specific microarrays have boosted the study of lncRNA etiology. Cerebrospinal fluid (CSF) directly mirrors the biological fluid of biochemical processes in the brain. It can be enriched for small molecules, peptides, or proteins released by the neurons of the central nervous system (CNS) or immune cells. Therefore, strategies that identify and target CSF lncRNAs may be attractive as early diagnostic and therapeutic options. In this review, we have reviewed the studies on CSF lncRNAs in the context of brain tumor pathogenesis and progression and discuss their potential as biomarkers and therapeutic targets.
Collapse
Affiliation(s)
- Kanghong Xu
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | - Xinquan Jiang
- School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Taian, China
| | | | - Xiaomeng Li
- Department of Hematology, Jining First People's Hospital, Jining, China
| | - Yunshan Wang
- Department of Clinical Laboratory, The Second Hospital of Shandong University, Jinan, China
| | - Shuo Xu
- Department of Neurosurgery, Qilu Hospital of Shandong University and Institute of Brain and Brain-Inspired Science, Shandong University, Jinan, China.,Key Laboratory of Brain Function Remodeling, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
12
|
Chen J, Qian C, Ren P, Yu H, Kong X, Huang C, Luo H, Chen G. Light-Responsive Micelles Loaded With Doxorubicin for Osteosarcoma Suppression. Front Pharmacol 2021; 12:679610. [PMID: 34220512 PMCID: PMC8249570 DOI: 10.3389/fphar.2021.679610] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/19/2021] [Indexed: 01/14/2023] Open
Abstract
The enhancement of tumor targeting and cellular uptake of drugs are significant factors in maximizing anticancer therapy and minimizing the side effects of chemotherapeutic drugs. A key challenge remains to explore stimulus-responsive polymeric nanoparticles to achieve efficient drug delivery. In this study, doxorubicin conjugated polymer (Poly-Dox) with light-responsiveness was synthesized, which can self-assemble to form polymeric micelles (Poly-Dox-M) in water. As an inert structure, the polyethylene glycol (PEG) can shield the adsorption of protein and avoid becoming a protein crown in the blood circulation, improving the tumor targeting of drugs and reducing the cardiotoxicity of doxorubicin (Dox). Besides, after ultraviolet irradiation, the amide bond connecting Dox with PEG can be broken, which induced the responsive detachment of PEG and enhanced cellular uptake of Dox. Notably, the results of immunohistochemistry in vivo showed that Poly-Dox-M had no significant damage to normal organs. Meanwhile, they showed efficient tumor-suppressive effects. This nano-delivery system with the light-responsive feature might hold great promises for the targeted therapy for osteosarcoma.
Collapse
Affiliation(s)
- Jiayi Chen
- Bengbu Medical College, Bengbu, China.,Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | | | - Peng Ren
- Bengbu Medical College, Bengbu, China.,Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Han Yu
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Xiangjia Kong
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Chenglong Huang
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Huanhuan Luo
- Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Gang Chen
- Bengbu Medical College, Bengbu, China.,Jiaxing Key Laboratory of Basic Research and Clinical Translation on Orthopedic Biomaterials, Department of Orthopaedics, The Second Affiliated Hospital of Jiaxing University, Jiaxing, China
| |
Collapse
|
13
|
Wang J, Huang H, Zhang X, Ma H. LOXL1‑AS1 promotes thymoma and thymic carcinoma progression by regulating miR‑525‑5p‑ HSPA9. Oncol Rep 2021; 45:117. [PMID: 33907842 PMCID: PMC8107651 DOI: 10.3892/or.2021.8068] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 02/12/2021] [Indexed: 12/14/2022] Open
Abstract
Due to the lack of specific symptoms in early thymic epithelial tumours (TETs), patients are mostly in the advanced stage at the time of presentation. The aim of the present study was to explore the mechanism by which the long noncoding RNA (lncRNA) LOXL1‑AS1 affects thymoma and thymic carcinoma progression by targeting the miR‑525‑5p‑HSPA9 axis. Bioinformatics was used to analyse the process of LOXL1‑AS1 targeting miR‑525‑5p‑HSPA9 and its expression characteristics in TET. The relationships between LOXL1‑AS1, miR‑525‑5p, HSPA9 and prognosis were analysed. The dual luciferase reporter assay was applied to verify targeting. The gene was knocked down or overexpressed by plasmid transfection. Cell counting kit 8 (CCK‑8) assay, flow cytometry and Transwell assay were used to detect cell viability, apoptosis and invasion ability, respectively. Proteins and RNAs were examined by western blot analysis and qPCR, respectively. A tumour‑burdened assay was used to perform in vivo verification. LOXL1‑AS1 and HSPA9 were overexpressed in thymoma and thymic carcinoma; high levels of LOXL1‑AS1 and HSPA9 were associated with poor prognosis, and there was a significant positive correlation between their levels. Downregulation of miR‑525‑5p expression was also associated with poor prognosis of patients. Clinical trials also demonstrated the same trends. miR‑525‑5p inhibited the expression of HSPA9 protein by targeting the 3'‑untranslated region (UTR) of HSPA9 mRNA. LOXL1‑AS1 promoted the expression of HSPA9 as a sponge targeting miR‑525‑5p. Animal experiment results also showed that knockdown of miR‑525‑5p promoted cancer by promoting the expression of HSPA9. In conclusion, LOXL1‑AS1 and HSPA9 are highly expressed in thymoma and thymic carcinoma; miR‑525‑5p is expressed at low levels in thymoma and thymic carcinoma; and downregulation of miR‑525‑5p is associated with poor prognosis. In summary, this study demonstrates that LOXL1‑AS1 acts as a sponge that targets miR‑525‑5p to promote HSPA9 expression, thereby promoting the growth and invasion and inhibiting apoptosis of thymoma and thymic carcinoma cells.
Collapse
Affiliation(s)
- Jin Wang
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai 200080, P.R. China
| | - Haihua Huang
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai 200080, P.R. China
| | - Xiaomiao Zhang
- Department of Thoracic Surgery, Shanghai General Hospital, Shanghai 200080, P.R. China
| | - Haitao Ma
- Department of Cardiothoracic Surgery, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, P.R. China
| |
Collapse
|
14
|
LOXL1-AS1/miR-515-5p/STAT3 Positive Feedback Loop Facilitates Cell Proliferation and Migration in Atherosclerosis. J Cardiovasc Pharmacol 2021; 76:151-158. [PMID: 32453072 DOI: 10.1097/fjc.0000000000000853] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Existing research has elucidated the critical role of long noncoding RNAs (lncRNAs) in the progression of multiple human cardiovascular diseases, including atherosclerosis (AS). Nonetheless, whether long noncoding RNA LOXL1 antisense RNA 1 (LOXL1-AS1) regulates the biological functions in AS is exceedingly limited. In this research, we detected through reverse transcription-quantitative polymerase chain reaction that LOXL1-AS1 expression was markedly upregulated in patients with AS. The role of LOXL1-AS1 in vascular smooth muscle cells (VSMCs) and human umbilical vein endothelial cells (HUVECs) was unmasked by functional assays. Moreover, knockdown of LOXL1-AS1 exerted suppressive effect on proliferation and migration whereas accelerated apoptosis in VSMCs and HUVECs. Molecular mechanism assays revealed that signal transducer and activator of transcription 3 (STAT3) functioned as a transcription activator of LOXL1-AS1 in VSMCs and HUVECs. In addition, miR-515-5p was manifested to bind with LOXL1-AS1 (or STAT3) in VSMCs and HUVECs. Furthermore, LOXL1-AS1 could elevate STAT3 expression by sponging miR-515-5p in VSMCs and HUVECs. More importantly, rescue assays delineated that inhibited expression of miR-515-5p or elevated expression of STAT3 could reverse the restraining effect of LOXL1-AS1 depletion on the progression of AS in HUVECs. All these findings revealed the role of a LOXL1-AS1/miR-515-5p/STAT3 positive feedback loop in AS.
Collapse
|
15
|
Ghafouri-Fard S, Shirvani-Farsani Z, Hussen BM, Taheri M. The critical roles of lncRNAs in the development of osteosarcoma. Biomed Pharmacother 2021; 135:111217. [PMID: 33433358 DOI: 10.1016/j.biopha.2021.111217] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 12/29/2020] [Accepted: 12/31/2020] [Indexed: 02/07/2023] Open
Abstract
Osteosarcoma is rare malignancy of childhood and adolescence, with high morbidity and mortality despite accomplishment of diverse therapeutic modalities. Identification of the underlying mechanism of osteosarcoma evolution would help in better management of this rare malignancy. Lots of investigations have described abnormal regulation of long non-coding RNAs (lncRNAs) in clinical specimens of osteosarcoma and the established cell lines. This malignancy has been associated with over-expression of TUG1, LOXL1-AS1, MIR100HG, NEAT1, HULC, ANRIL and a number of other lncRNAs, while under-expression of lots of lncRNAs including LncRNA-p21, FER1L4, GAS5, LncRNA NR_136400 and LINC-PINT. Expression amounts of LUCAT1, LINC00922, SNHG12, FOXC2-AS1 and OIP5-AS1 lncRNAs have been associated with response to a number of chemotherapeutic agents. Taken together, lncRNAs are possible targets for proposing novel advanced therapeutic modalities for osteosarcoma.
Collapse
Affiliation(s)
- Soudeh Ghafouri-Fard
- Urogenital Stem Cell Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zeinab Shirvani-Farsani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University G.C., Tehran, Iran
| | - Bashdar Mahmud Hussen
- Pharmacognosy Department, College of Pharmacy, Hawler Medical University, Erbil, Iraq
| | - Mohammad Taheri
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
16
|
Li H, Chu J, Jia J, Sheng J, Zhao X, Xing Y, He F. LncRNA LOXL1-AS1 promotes esophageal squamous cell carcinoma progression by targeting DESC1. J Cancer 2021; 12:530-538. [PMID: 33391449 PMCID: PMC7738998 DOI: 10.7150/jca.51136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Accepted: 10/30/2020] [Indexed: 12/13/2022] Open
Abstract
Recently, ample evidence indicated that numerous aberrantly expressed long non-coding RNAs (lncRNAs) participated in the development of multiple malignancies. However, the expression and function of lncRNA LOXL1-AS1 in mediating esophageal squamous cell carcinoma (ESCC) carcinogenesis remains largely elusive. Here we validated that LOXL1-AS1 was significantly upregulated in ESCC tissues compared with the corresponding adjacent non-neoplastic tissues, and LOXL1-AS1 expression was positively correlated with ESCC patients' lymph node metastasis. Besides, LOXL1-AS1 knockdown impaired ESCC cells proliferation, migration and invasion capabilities in vitro. Furthermore, inhibiting LOXL1-AS1 in ESCC cells increased the percentage of cells at the G1 phase, accompanied by reducing in S phase in contrast to scramble control, and silencing of LOXL1-AS1 evoked ESCC cell apoptosis. From high throughput RNA sequencing (RNA-seq) analysis, we identified that differentially expressed in squamous cell carcinoma 1 (DESC1) was a critical downstream target of LOXL1-AS1. Taken together, we demonstrated the function and mechanism of LOXL1-AS1 in contributing ESCC progression for the first time, and indicated LOXL1-AS1 may be a novel therapeutic biomarker of ESCC.
Collapse
Affiliation(s)
- Hongle Li
- Department of Molecular Pathology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, Henan 450008, China
| | - Jie Chu
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jinlin Jia
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Jinxiu Sheng
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Xue Zhao
- Institute of Medical and Pharmaceutical Science, Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Yurong Xing
- Department of Physical Examination, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| | - Fucheng He
- Department of Medical Laboratory, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan 450052, China
| |
Collapse
|
17
|
Zhang L, Wan Q, Zhou H. Targeted-regulating of miR-515-5p by LncRNA LOXL1-AS1 on the proliferation and migration of trophoblast cells. Exp Mol Pathol 2020; 118:104588. [PMID: 33278425 DOI: 10.1016/j.yexmp.2020.104588] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 11/15/2020] [Accepted: 11/30/2020] [Indexed: 10/22/2022]
Abstract
OBJECTIVE This study aims to elucidate the molecular mechanism of LOXL1-AS1 in proliferation and migration of trophoblast cells. METHODS We have used specific small interfering RNAs (si-RNA) and microRNA (mi-RNA) to knock down the target gene or m-RNA. In this regard we used following siRNAs: si-NC, si-LOXL1-AS1, pcDNA-NC, pcDNA-LOXL1-AS1, miR-NC, miR-515-5p. These si-RNA and miRNA were transfected into JeG-3 cells. Real-time quantitative PCR (RT-qPCR) was used to detect the expressions of LOXL1-AS1 and miR-515-5p. Western blot was used to detect Cyclin D1, matrix metalloproteinase 2 (MMP2), matrix metalloproteinases 9 (MMP9), phosphorylated p65 (p-p65), profilin of phosphorylated nuclear transcription factor κB (p-IκBα). MTT assay was used to detect cell survival rate, and the luciferase assay was used to detected the targeting relationship of LOXL1-AS1 and miR-515-5p. RESULTS Our results showed that human placental trophoblast cells had higher level of LOXL1-AS1 in comparison to human choriocarcinoma cells, however, human placental trophoblast cells had lower level of miR-515-5p. In addition, the expression of CyclinD1, MMP2, MMP9 were significantly decreased in JeG-3 cell lines. We observed lower cell survival rate and lower cell migration number in JeG-3 cell lines. Our results demonstrated that LOXL1-AS1 could target miR-515-5p, and subsequently reverse the inhibitory effect of LOXL1-AS1 on proliferation and migration in JEG-3 cells. Also, lower expressions of p-p65 and p-IкBα in JeG-3 cells showed that miR-515-5p could reversed the inhibitory effect of LOXL1-AS1 on NF-κB signaling pathway. CONCLUSIONS The low expression of LOXL1-AS1 inhibits the proliferation and migration of human choriocarcinoma cells, which might be related to miR-515-5p and NF-κB signaling pathways.
Collapse
Affiliation(s)
- Li Zhang
- Department of Clinical Medicine, Jiangxi Medical College, Shangrao 334000, China
| | - Qingfeng Wan
- Department of Basic Medicine, Jiangxi Medical College, Shangrao 334000, China.
| | - Huiyun Zhou
- Academic Affairs Office of Jiangxi Medical College, Shangrao 334000, China
| |
Collapse
|
18
|
He G, Yao W, Li L, Wu Y, Feng G, Chen L. LOXL1-AS1 contributes to the proliferation and migration of laryngocarcinoma cells through miR-589-5p/TRAF6 axis. Cancer Cell Int 2020; 20:504. [PMID: 33061856 PMCID: PMC7552551 DOI: 10.1186/s12935-020-01565-5] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/18/2020] [Indexed: 12/12/2022] Open
Abstract
Background LOXL1-AS1 is a long non-coding RNA (lncRNA) that plays crucial roles in various cancers. However, the functional role of LOXL1-AS1 in laryngocarcinoma remains unclear. Thus we planned to probe into the function and underlying mechanism of LOXL1-AS1 in laryngocarcinoma. Methods Gene expression was evaluated in laryngocarcinoma cells using RT-qPCR. The ability of cell proliferation and migration was assessed by CCK8, colony formation, wound healing and transwell assays. The interaction among LOXL1-AS1, miR-589-5p and TRAF6 was detected by Ago2-RIP, RNA pull down and luciferase reporter assays. Results LOXL1-AS1 was overexpressed in laryngocarcinoma cells. Silencing of LOXL1-AS1 suppressed cell proliferation, migration and EMT in laryngocarcinoma. Moreover, miR-589-5p, the downstream of LOXL1-AS1, directly targeted TRAF6 in laryngocarcinoma. Importantly, LOXL1-AS1 augmented TRAF6 expression in laryngocarcinoma cells by sequestering miR-589-5p. Besides, miR-589-5p worked as a tumor-inhibitor while TRAF6 functioned as a tumor-facilitator in laryngocarcinoma. Of note, rescue experiments both in vitro and in vivo validated that LOXL1-AS1 aggravated the malignancy in laryngocarcinoma by targeting miR-589-5p/TRAF6 pathway. Conclusions LOXL1-AS1 promotes the proliferation and migration of laryngocarcinoma cells through absorbing miR-589-5p to upregulate TRAF6 expression.
Collapse
Affiliation(s)
- Guijun He
- Department of Otolaryngology and Head and Neck Surgery, Lianyungang Second People's Hospital, Lianyungang, 222023 Jiangsu China
| | - Wenfeng Yao
- Department of Otolaryngology, The First People's Hospital of Xinxiang City, Xinxiang, 453000 Henan China
| | - Liang Li
- Department of Otolaryngology and Head and Neck Surgery, Lianyungang Second People's Hospital, Lianyungang, 222023 Jiangsu China
| | - Yang Wu
- Department of Otolaryngology and Head and Neck Surgery, Lianyungang Second People's Hospital, Lianyungang, 222023 Jiangsu China
| | - Guojian Feng
- Department of Otolaryngology and Head and Neck Surgery, Lianyungang Second People's Hospital, Lianyungang, 222023 Jiangsu China
| | - Li Chen
- Department of Otorhinolaryngology, Zaozhuang Municipal Hospital, No. 41, Longtou Middle Road, Shizhong District, Zaozhuang, 277100 Shandong China
| |
Collapse
|
19
|
Schmitt HM, Johnson WM, Aboobakar IF, Strickland S, Gomez-Caraballo M, Parker M, Finnegan L, Corcoran DL, Skiba NP, Allingham RR, Hauser MA, Stamer WD. Identification and activity of the functional complex between hnRNPL and the pseudoexfoliation syndrome-associated lncRNA, LOXL1-AS1. Hum Mol Genet 2020; 29:1986-1995. [PMID: 32037441 PMCID: PMC7390937 DOI: 10.1093/hmg/ddaa021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2019] [Revised: 01/09/2020] [Accepted: 02/03/2020] [Indexed: 12/28/2022] Open
Abstract
Individuals with pseudoexfoliation (PEX) syndrome exhibit various connective tissue pathologies associated with dysregulated extracellular matrix homeostasis. PEX glaucoma is a common, aggressive form of open-angle glaucoma resulting from the deposition of fibrillary material in the conventional outflow pathway. However, the molecular mechanisms that drive pathogenesis and genetic risk remain poorly understood. PEX glaucoma-associated single-nucleotide polymorphisms are located in and affect activity of the promoter of LOXL1-AS1, a long non-coding RNA (lncRNA). Nuclear and non-nuclear lncRNAs regulate a host of biological processes, and when dysregulated, contribute to disease. Here we report that LOXL1-AS1 localizes to the nucleus where it selectively binds to the mRNA processing protein, heterogeneous nuclear ribonucleoprotein-L (hnRNPL). Both components of this complex are critical for the regulation of global gene expression in ocular cells, making LOXL1-AS1 a prime target for investigation in PEX syndrome and glaucoma.
Collapse
Affiliation(s)
- Heather M Schmitt
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
| | - William M Johnson
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
| | - Inas F Aboobakar
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
| | - Shelby Strickland
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701 USA
| | - María Gomez-Caraballo
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
| | - Megan Parker
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
| | - Laura Finnegan
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
- School of Genetics and Microbiology, Department of Genetics, Smurfit Institute, Trinity College Dublin, Dublin 2, Ireland
| | - David L Corcoran
- Genomic Analysis and Bioinformatics Shared Resource, Duke University, Duke University CIEMAS, Durham, NC 27708, USA
| | - Nikolai P Skiba
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
| | - R Rand Allingham
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
| | - Michael A Hauser
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
- Duke Molecular Physiology Institute, Duke University, Durham, NC 27701 USA
- Department of Medicine, Duke University, Durham, NC 27710, USA
| | - W Daniel Stamer
- Department of Ophthalmology, Duke University, Duke Eye Center AERI Rm 4014, Durham, NC 27710, USA
| |
Collapse
|
20
|
Zhang Y, Pu Y, Wang J, Li Z, Wang H. Research progress regarding the role of long non-coding RNAs in osteosarcoma. Oncol Lett 2020; 20:2606-2612. [PMID: 32782578 PMCID: PMC7400499 DOI: 10.3892/ol.2020.11807] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 05/29/2020] [Indexed: 12/16/2022] Open
Abstract
Osteosarcoma is a malignant tumor that occurs in children and adolescents. Although treatments for osteosarcoma have improved, the likelihood of survival remains low for most patients with metastasis and recurrence. Elucidating the mechanism underlying the development of osteosarcoma and chemotherapy resistance will be important to improve diagnosis and treatment. Long non-coding RNAs (lncRNAs), which are longer than 200 nucleotides in length and do not encode for proteins, have been shown to play a regulatory role in the occurrence and development of osteosarcoma, and are expected to serve as biomarkers and molecular targets. This review discusses the progress in the study of the role of lncRNAs in osteosarcoma, and highlights the recent developments in this field.
Collapse
Affiliation(s)
- Yanli Zhang
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Yanchuan Pu
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Jin Wang
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Zicai Li
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| | - Hulin Wang
- Department of Orthopedics, Wuwei People's Hospital, Wuwei, Gansu 733000, P.R. China
| |
Collapse
|
21
|
Zhao L, Zhang X, Guo H, Liu M, Wang L. LOXL1-AS1 Contributes to Non-Small Cell Lung Cancer Progression by Regulating miR-3128/RHOXF2 Axis. Onco Targets Ther 2020; 13:6063-6071. [PMID: 32636639 PMCID: PMC7326695 DOI: 10.2147/ott.s247900] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Accepted: 04/23/2020] [Indexed: 12/20/2022] Open
Abstract
Purpose The purpose of this study was to investigate the molecular mechanism of LncRNA LOXL1-AS1 in non-small cell lung cancer (NSCLC). Methods Lung cancer cell lines (H1299, A549, H520 and H596) and human normal lung epithelial cell line (BEAS-2B) were used in this study. Gene expression was measured by qRT-PCR (quantitative real-time PCR). The bioinformatics databases (miRDB and TargetScan7) were used to predict target genes. Luciferase assay and pull-down assay were processed for verifying the binding sites. CCK8 assay was used for detecting proliferation, and transwell assay was undertaken for migration and invasion. Results LncRNA LOXL1-AS1 was higher expressed in lung cancer tissues and cells. Moreover, LOXL1-AS1 expression was upregulated in tumor tissues with advanced stages and metastasis. After knocking down LOXL1-AS1, proliferation, invasion and migration of H1299 and A549 cells were inhibited. Interestingly, miR-3128 was negatively regulated by LncRNA LOXL1-AS1, which inhibited the expression of RHOXF2. Rescue assay also confirmed that miR-3128 inhibitor and oeRHOXF2 could rescue the effect of down-regulated LOXL1-AS1 on proliferation, invasion and migration progression. Conclusion LOXL1-AS1 promotes the progression of NSCLC by regulating miR-3128/RHOXF2 axis, which might be a new potential target for the diagnosis and treatment of NSCLC.
Collapse
Affiliation(s)
- Limin Zhao
- Department of Oncology, General Hospital of Heilongjiang General Administration of Agriculture and Reclamation, Harbin 150088, People's Republic of China
| | - Xuefei Zhang
- Department of Thoracic Surgery, The Second Hospital of Dalian Medical University, Dalian 116023, People's Republic of China
| | - Huannan Guo
- Department of Oncology, General Hospital of Heilongjiang General Administration of Agriculture and Reclamation, Harbin 150088, People's Republic of China
| | - Mingyang Liu
- Department of Oncology, General Hospital of Heilongjiang General Administration of Agriculture and Reclamation, Harbin 150088, People's Republic of China
| | - Liming Wang
- Department of Thoracic Surgery, The First Affiliated Hospital of China Medical University, Shenyang 110001, People's Republic of China
| |
Collapse
|
22
|
Liu Y, Xi Y, Chen G, Wu X, He M. URG4 mediates cell proliferation and cell cycle in osteosarcoma via GSK3β/β-catenin/cyclin D1 signaling pathway. J Orthop Surg Res 2020; 15:226. [PMID: 32552851 PMCID: PMC7301506 DOI: 10.1186/s13018-020-01681-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/28/2020] [Indexed: 11/26/2022] Open
Abstract
BACKGROUND Osteosarcoma is one of the most common malignant bone tumors with the annual global incidence of approximately four per million. Upregulated gene 4 (URG4) expression in the osteosarcoma tissue is closely associated with recurrence, metastasis, and poor prognosis of osteosarcoma. However, the biological function and underlying mechanisms of URG4 in osteosarcoma have not been elucidated. This study aimed to explore the expression and underlying mechanism of URG4 in osteosarcoma. METHODS The expression level of URG4 in osteosarcoma and normal tissues was compared using immunohistochemistry (IHC). PCR and western blotting (WB) techniques are used to detect URG4 mRNA and protein levels. Wound healing and Transwell analysis to assess the effect of URG4 on osteosarcoma cell migration and invasion. Cell Counting Kit-8 assay and colony proliferation assay were performed to evaluate the effects of silencing URG4 on the inhibition of cell proliferation. The cell cycle distribution was detected by flow cytometry, and a xenograft mouse model was used to verify the function of URG4 in vivo. RESULTS URG4 was found to be highly expressed in osteosarcoma tissues and cells, and its high expression was correlated with advanced Enneking stage, large tumor size, and tumor metastasis in osteosarcoma patients. The proliferation in osteosarcoma cell lines and cell cycle in the S phase was suppressed when siRNA was used to downregulate URG4. URG4 promoted cell proliferation and tumorigenesis in vitro and in vivo. WB verified that URG4 promotes cell proliferation in osteosarcoma via pGSK3β/β-catenin/cyclinD1 signaling. CONCLUSION URG4, which is high-expressed in osteosarcoma, promotes cell cycle progression via GSK3β/β-catenin/cyclin D1 signaling pathway and may be a novel biomarker and potential target for the treatment of osteosarcoma.
Collapse
Affiliation(s)
- Yayun Liu
- Department of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
- Department of Orthopaedics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, No. 152 Aiguo Road, Nanchang, 330006, Jiangxi, China
| | - Yizhe Xi
- Department of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China
| | - Gang Chen
- Department of Orthopaedics, Jiangxi Provincial People's Hospital Affiliated to Nanchang University, No. 152 Aiguo Road, Nanchang, 330006, Jiangxi, China
| | - Xidong Wu
- Department of drug safety evaluation, Jiangxi Testing Center of Medical Device, No. 181 Nanjing East Road, Nanchang, 330000, Jiangxi, China
| | - Maolin He
- Department of Spinal Surgery, The First Affiliated Hospital of Guangxi Medical University, No. 6 Shuangyong Road, Nanning, 530021, Guangxi, China.
| |
Collapse
|
23
|
Eivers SB, Greene AG, Dervan E, O’Brien C, Wallace D. Prevalence of Pseudoexfoliation Glaucoma Risk-associated Variants Within Lysyl Oxidase-like 1 in an Irish Population. J Glaucoma 2020; 29:417-422. [PMID: 32102031 PMCID: PMC7265999 DOI: 10.1097/ijg.0000000000001475] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 02/15/2020] [Indexed: 11/26/2022]
Abstract
PRECIS High-risk alleles of risk-associated single-nucleotide polymorphisms (SNPs) within the lysyl oxidase-like 1 (LOXL1) gene are associated with pseudoexfoliation in patients recruited from an Irish population. PURPOSE SNPs within the LOXL1 gene have been identified as a major risk factor for pseudoexfoliation syndrome (PXF) and pseudoexfoliation glaucoma (PXFG), specifically SNPs within exon 1 and intron 1 regions of the gene. The common haplotype (G-G) of 2 SNPs within exon 1, rs1048661, and rs3825942, is the strongest associated risk factor for PXF in white populations, but is switched in some populations to act as protective or low risk. Herein, a study was undertaken to genotype an Irish population for PXF/PXFG risk-associated SNPs within LOXL1. MATERIALS AND METHODS Patient cohorts of PXFG, PXF, and controls were recruited and genotyped for risk-associated SNPs within exon 1 (rs1048661 and rs3825942), along with 3 SNPs within intron 1 (rs1550437, rs6495085, and rs6495086) of LOXL1. RESULTS The risk G alleles of rs1048661 and rs3825942 were most prevalent in PXFG patients, and a significant association was found between rs3825942 and pseudoexfoliation (P=0.04). Genotypes of several intron 1 SNPs were found to be present at higher frequencies within the pseudoexfoliation patient cohort (PXF/PXFG) compared with control patients, wherein rs6495085 showed statistical association (P=0.04). The G-G-G haplotype of rs1048661, rs3825942, and rs6495085 was the most prevalent in PXFG patients compared with control patients or patients with PXF alone. Patients with the G-G-G haplotype were more likely to need surgery, suggestive of a more severe form of disease. CONCLUSION Collectively, these results represent the first study to assess the association of LOXL1 SNPs with PXFG in an Irish population.
Collapse
Affiliation(s)
- Sarah B. Eivers
- UCD Clinical Research Centre, UCD School of Medicine, University College Dublin
| | - Alison G. Greene
- UCD Clinical Research Centre, UCD School of Medicine, University College Dublin
| | - Edward Dervan
- Department of Ophthalmology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Colm O’Brien
- UCD Clinical Research Centre, UCD School of Medicine, University College Dublin
- Department of Ophthalmology, Mater Misericordiae University Hospital, Dublin, Ireland
| | - Deborah Wallace
- UCD Clinical Research Centre, UCD School of Medicine, University College Dublin
| |
Collapse
|
24
|
Han J, Shen X. Long noncoding RNAs in osteosarcoma via various signaling pathways. J Clin Lab Anal 2020; 34:e23317. [PMID: 32249459 PMCID: PMC7307344 DOI: 10.1002/jcla.23317] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2019] [Revised: 03/03/2020] [Accepted: 03/04/2020] [Indexed: 12/14/2022] Open
Abstract
Osteosarcoma is one of the most commonly seen bone malignancies with high incidence rate in both children and adults. Although the regulatory network of osteosarcoma has been greatly concerned for years, the mechanisms regarding its oncogenesis and development are still not clear. Recent discoveries have revealed that long noncoding RNAs (lncRNAs) play a crucial role in the development, progression, and invasion of osteosarcoma. Deregulated expression of lncRNAs has been found to participate in the regulation of various signaling transduction pathways in osteosarcoma. This review summarized roles of lncRNAs in the pathogenesis, development, and potential therapeutic of osteosarcoma via different signaling pathways. For examples, MALAT1, CCAT2, FER1L4, LOXL1‐AS1, OIP5‐AS1, PVT1, DBH‐AS1, and AWPPH regulate PI3K/Akt signaling; AWPPH and BE503655 regulate Wnt/β‐catenin signaling; NKILA and XIST regulate NF‐κB signaling; MEG3 and SNHG12 regulate Notch signaling; FOXD2‐AS1 and LINK‐A regulate HIF‐1α signaling; GClnc1 and HOTAIR regulate P53 signaling; ZFAS1, H19, and MALAT1 regulate MAPK, Hedgehog and Rac1/JNK signaling, respectively.
Collapse
Affiliation(s)
| | - Xiaohan Shen
- Ningbo Diagnostic Pathology Center (Shanghai Cancer Center Ningbo Pathology Center), Ningbo, China.,Ningbo Medical Center Lihuili Hospital, Ningbo, China
| |
Collapse
|
25
|
Zhao D, Zhang H, Long J, Li M. LncRNA SNHG7 Functions as an Oncogene in Cervical Cancer by Sponging miR-485-5p to Modulate JUND Expression. Onco Targets Ther 2020; 13:1677-1689. [PMID: 32161467 PMCID: PMC7049860 DOI: 10.2147/ott.s237802] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Accepted: 02/03/2020] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND Long non-coding RNA (LncRNA) SNHG7 is involved in the development of multiple cancers. However, its role in cervical cancer (CC) has not been elucidated. This study aimed to explore the function of SNHG7 in CC progression and the underlying mechanisms. MATERIALS AND METHODS The expression levels of SNHG7 and miR-485-5p in CC tissues and cell lines were measured by qPCR. Functional experiments including CCK-8 assay, wound healing assay, transwell assay, flow cytometry, Western blot, luciferases reporter assay and immunoprecipitation (RIP) were performed to explore the SNHG7/miR-485-5p/JUND pathway. Additionally, in vivo study was carried out by establishing tumor xenograft models. RESULTS We found that SNHG7 was markedly enhanced in CC tissues and cell lines, and associated with poor clinical characteristics. In vitro, knockdown of SNHG7 inhibited CC cell proliferation, migration and invasion, as well as aggravated cell apoptosis. As to mechanism investigation, rescue experiments revealed that miR-485-5p inhibitor could partially reverse the effects on CC cells induced by SNHG7 knockdown. SNHG7 upregulated JUND expression via miR-485-5p. Moreover, tumor xenograft models were established to confirm the findings in vivo. CONCLUSION SNHG7 promoted CC progression through miR-485-5p/JUND axis. The SNHG7/miR-485-5p/JUND pathway might provide a novel therapeutic target for CC treatment.
Collapse
Affiliation(s)
- Danyang Zhao
- Department of Reproductive Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, People’s Republic of China
| | - Hui Zhang
- Department of Obstetrics, Chongzuo People’s Hospital, Chongzuo, Guangxi Province, People’s Republic of China
| | - Jianxiong Long
- School of Public Health of Guangxi Medical University, Nanning, Guangxi Province, People’s Republic of China
| | - Mujun Li
- Department of Reproductive Center, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, People’s Republic of China
| |
Collapse
|
26
|
Li M, Cai O, Tan S. LOXL1-AS1 Drives The Progression Of Gastric Cancer Via Regulating miR-142-5p/PIK3CA Axis. Onco Targets Ther 2019; 12:11345-11357. [PMID: 31908498 PMCID: PMC6929932 DOI: 10.2147/ott.s223702] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 09/28/2019] [Indexed: 12/24/2022] Open
Abstract
Background Gastric cancer (GC) is a deadly disease, and its incidence is especially high in East Asia including China. Recently, some long non-coding RNA (lncRNAs) have been identified as oncogenes or tumor suppressors. This study aimed to determine the function and mechanism of lncRNA LOXL1-AS1 on the progression of GC. Methods RT-PCR was done to measure the expression levels of LOXL1-AS1 and miR-142-5p in GC tissues. The association between pathological indexes and LOXL1-AS1 expression was also analyzed. Human GC cell lines AGS and BGC823 were used as cell models. CCK-8 and colony formation assays were conducted to assess the effect of LOXL1-AS1 on the proliferation of GC cell lines. Transwell assay was conducted to determine the influence of LOXL1-AS1 on cell migration and invasion. Furthermore, luciferase reporter assay was carried out to confirm the relationship of miR-142-5p with LOXL1-AS1. Additionally, Western blot was done to detect the regulatory function of LOXL1-AS1 on PIK3CA, a target of miR-142-5p. In vivo experiment was also performed to validate the roles and mechanism of LOXL1-AS1 on the growth and metastasis of GC cells. Results LOXL1-AS1 expression in GC samples was significantly increased, which was correlated with unfavorable pathological indexes. Highly expressed LOXL1-AS1 was closely linked to shorter overall survival time and post-progression survival time of the patients. LOXL1-AS1 markedly modulated the malignant phenotypes of GC cells. Additionally, overexpressed LOXL1-AS1 notably reduced the expression of miR-142-5p, but enhanced the expression level of PIK3CA. In vivo experiments further validated that knockdown of LOXL1-AS1 inhibited the growth and metastasis of GC cells via regulating miR-142-5p and PIK3CA. Conclusion LOXL1-AS1 was a sponge of tumor suppressor miR-142-5p in GC, enhanced the expression of PIK3CA indirectly and functioned as an oncogenic lncRNA.
Collapse
Affiliation(s)
- Ming Li
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430000, People's Republic of China
| | - Ou Cai
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430000, People's Republic of China
| | - Shiyun Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei 430000, People's Republic of China
| |
Collapse
|
27
|
Li W, Zhang B, Jia Y, Shi H, Wang H, Guo Q, Li H. LncRNA LOXL1-AS1 regulates the tumorigenesis and development of lung adenocarcinoma through sponging miR-423-5p and targeting MYBL2. Cancer Med 2019; 9:689-699. [PMID: 31758653 PMCID: PMC6970024 DOI: 10.1002/cam4.2641] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 09/16/2019] [Accepted: 10/10/2019] [Indexed: 12/27/2022] Open
Abstract
Lung adenocarcinoma (LUAD) is the most common form of malignant tumor and closely correlated with high risk of death worldwide. Accumulating researches have manifested that long noncoding RNAs (lncRNAs) are deeply involved in the progression of multiple cancers. LncRNA LOXL1 antisense RNA 1 (LOXL1‐AS1) was identified as an oncogene in several cancers, nonetheless, its biological effect and regulatory mechanism have not been explained in LUAD. Our present study suggested that LOXL1‐AS1 expression was considerably increased in LUAD tissues and cells. Moreover, LOXL1‐AS1 deficiency notably hampered cell proliferation and migration as well as dramatically facilitated cell apoptosis. Through molecular mechanism assays, LOXL1‐AS1 was identified as a cytoplasmic RNA and acted as a sponge of miR‐423‐5p. Furthermore, MYBL2 was targeted and negatively modified by miR‐423‐5p. Rescue experiments revealed that MYBL2 knockdown could counteract miR‐423‐5p repression‐mediated enhancement on the progression of LOXL1‐AS1 downregulated LUAD cells. More importantly, MYBL2 was discovered to interact with LOXL1‐AS1 promoter, indicating a positive feedback loop of LOXL1‐AS1/miR‐423‐5p/MYBL2 in LUAD. These findings manifested the carcinogenic role of LOXL1‐AS1 and LOXL1‐AS1/miR‐423‐5p/MYBL2 feedback loop in LUAD, which could be helpful to explore effective therapeutic strategy for LUAD patients.
Collapse
Affiliation(s)
- Wei Li
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Biao Zhang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Youchao Jia
- Department of Medical Oncology, Affiliated Hospital of Hebei University, Baoding, China
| | - Hongyun Shi
- Department of Radiation Oncology, Affiliated Hospital of Hebei University, Baoding, China
| | - Haibo Wang
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Qiang Guo
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, China
| | - Hefei Li
- Department of Thoracic Surgery, Affiliated Hospital of Hebei University, Baoding, China
| |
Collapse
|
28
|
Xie N, Fei X, Liu S, Liao J, Li Y. LncRNA LOXL1-AS1 promotes invasion and proliferation of non-small-cell lung cancer through targeting miR-324-3p. Am J Transl Res 2019; 11:6403-6412. [PMID: 31737192 PMCID: PMC6834493] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 08/16/2019] [Indexed: 06/10/2023]
Abstract
LncRNAs are played crucial roles in athogenesis of NSCLC. LOXL1-AS1 involved in development of several tumors. So far, there is no study about expression and function pattern of the LOXL1-AS1 in NSCLC. In this reference, we firstly proved that LOXL1-AS1 was overexpressed in NSCLC cell lines (H23, A549, H1299 and SPC-A1) compared to 16HBE cell. The expression of LOXL1-AS1 was overexpressed in NSCLC specimens than adjacent control specimens. We found that 29 of 40 cases showed higher LOXL1-AS1 expression in NSCLC samples as compared to adjacent control specimens. Ectopic expression of LOXL1-AS1 promoted H1299 cell and H23 cell proliferation. LOXL1-AS1 overexpression promoted ki-67 and cyclin D1 expression in the NSCLC cell. Overexpression of LOXL1-AS1 promoted cell invasion and induced N-cadherin and Vimentin expression and suppressed E-cadherin expression in the NSCLC cell. LOXL1-AS1 acts as one sponge for miR-324-3p in NSCLC cell. Moreover, the expression of miR-324-3p was lower in NSCLC specimens than adjacent control specimens. We found that 24 of 40 cases showed lower miR-324-3p expression in NSCLC samples as compared to adjacent control specimens. Further correlation assay indicated a negative association between miR-324-3p and LOXL1-AS1 expression. miR-324-3p restoration attenuates the function of LOXL1-AS1 overexpression on NSCLC cell. These results indicated that LOXL1-AS1 enhanced NSCLC cell proliferation and invasion via sponging miR-324-3p in NSCLC cell.
Collapse
Affiliation(s)
- Ning Xie
- Thoracic Surgery, Yantaishan HospitalYantai, Shandong, P. R. China
| | - Xiuqu Fei
- Thoracic Surgery, Yantaishan HospitalYantai, Shandong, P. R. China
| | - Shuliang Liu
- Thoracic Surgery, Yantaishan HospitalYantai, Shandong, P. R. China
| | - Jie Liao
- Thoracic Surgery, Yantaishan HospitalYantai, Shandong, P. R. China
| | - Youjie Li
- Department of Biochemistry and Molecular Biology, Key Laboratory of Tumor Molecular Biology, Binzhou Medical UniversityYantai, Shandong, P. R. China
| |
Collapse
|
29
|
Schlötzer-Schrehardt U, Zenkel M. The role of lysyl oxidase-like 1 (LOXL1) in exfoliation syndrome and glaucoma. Exp Eye Res 2019; 189:107818. [PMID: 31563608 DOI: 10.1016/j.exer.2019.107818] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/23/2019] [Accepted: 09/25/2019] [Indexed: 12/27/2022]
Abstract
Exfoliation syndrome (XFS) is an age-related systemic disease that affects the extracellular matrix. It increases the risk of glaucoma (exfoliation glaucoma, XFG) and susceptibility to diseases of elastin-rich connective tissues. LOXL1 (lysyl oxidase-like 1) is still recognized as the major genetic effect locus in XFS and XFG in all populations worldwide, although its genetic architecture is incompletely understood. LOXL1 is a key cross-linking enzyme in elastic fiber formation and remodeling, which is compatible with the pathogenetic concept of XFS as a specific type of elastosis. This review provides an overview on the current knowledge about the role of LOXL1 in the etiology and pathophysiology of XFS and XFG. It covers the known genetic associations at the LOXL1 locus, potential mechanisms of gene regulation, implications of LOXL1 in XFS-associated fibrosis and connective tissue homeostasis, its role in the development of glaucoma and associated systemic diseases, and the currently available LOXL1-based in vivo and in vitro models. Finally, it also identifies gaps in knowledge and suggests potential areas for future research.
Collapse
Affiliation(s)
| | - Matthias Zenkel
- Department of Ophthalmology, University of Erlangen-Nürnberg, Schwabachanlage 6, 91054, Erlangen, Germany
| |
Collapse
|
30
|
Sun Q, Li J, Li F, Li H, Bei S, Zhang X, Feng L. LncRNA LOXL1-AS1 facilitates the tumorigenesis and stemness of gastric carcinoma via regulation of miR-708-5p/USF1 pathway. Cell Prolif 2019; 52:e12687. [PMID: 31468594 PMCID: PMC6869681 DOI: 10.1111/cpr.12687] [Citation(s) in RCA: 62] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Revised: 06/28/2019] [Accepted: 07/15/2019] [Indexed: 12/24/2022] Open
Abstract
Objectives As one of the most life‐threatening malignancies, gastric cancer is the third contributor of cancer mortalities globally. Increasing studies have proven the regulatory roles of lncRNAs in the development of diverse malignant tumours. But little is known about its function and molecular mechanism in gastric carcinoma. Materials and methods RT‐qPCR was performed to measure the expression pattern of LOXL1‐AS1 in gastric cancer. To ascertain its definite role, CCK‐8, EdU, Western blot, transwell and sphere formation assays were adopted. RNA pull‐down, RIP, ChIP and luciferase reporter assays were carried out to investigate the molecular mechanism of LOXL1‐AS1 in gastric carcinoma. Results LOXL1‐AS1 was highly expressed in tissues and cells of gastric cancer. The upregulation of LOXL1‐AS1 predicted poor prognosis in gastric carcinoma. Our findings demonstrated that LOXL1‐AS1 accelerated the deterioration of gastric cancer by inducing cell proliferation, migration, EMT and stemness. Moreover, the expression of USF1 in gastric cancer was higher than in normal control and LOXL1‐AS1 negatively modulated USF1. Functionally, LOXL1‐AS1 acted as a ceRNA to upregulate USF1 via sponging miR‐708‐5p. Besides, we confirmed USF1 promoted the transcription of stemness marker SOX2. Rescue experiments testified the stimulative role of LOXL1‐AS1/miR‐708‐5p/USF1 pathway in gastric cancer progression. It was also validated that LOXL1‐AS1 facilitated cell growth of gastric carcinoma in vivo. Conclusions Our study unravelled that LOXL1‐AS1/miR‐708‐5p/USF1 pathway contributed to the development of gastric cancer.
Collapse
Affiliation(s)
- Qi Sun
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Jian Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Fan Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Huanqin Li
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Songhua Bei
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Xiaohong Zhang
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| | - Li Feng
- Endoscopy Center, Minhang Hospital, Fudan University, Shanghai, China
| |
Collapse
|