1
|
Li HM, Wang LJ, Wang YP, Li XM, Pan HF. Differences in the expression of long noncoding RNAs in peripheral blood mononuclear cells indicate potential biomarkers for rheumatoid arthritis. Int Immunopharmacol 2024; 134:112218. [PMID: 38733828 DOI: 10.1016/j.intimp.2024.112218] [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: 02/01/2024] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 05/13/2024]
Abstract
OBJECTIVE Long noncoding RNAs (lncRNAs) play an increasingly important role in various autoimmune diseases. We aimed to characterize the expression profiles of lncRNAs in peripheral blood mononuclear cells (PBMCs) from RA patients and to assess the potential of these lncRNAs as RA biomarkers. METHODS Whole-transcriptome sequencing was used to establish a lncRNA expression profile. A total of 155 RA patients, 145 healthy controls, 59 systemic lupus erythematosus (SLE) patients and 59 primary Sjögren's syndrome (pSS) patients were recruited for this study. Four candidate lncRNAs (linc00152, lnc-ADM-1, ITSN1-2, and lnc-FTH1-7) were validated via qRT-PCR in independent samples, and their expression, association with RA clinical features and value as RA biomarkers were evaluated. RESULTS Linc00152 and lnc-ADM-1 exhibited upregulated expression (p = 0.001, p = 0.014, respectively), while ITSN1-2 and lnc-FTH1-7 exhibited downregulated expression (both p < 0.001, respectively) in RA patients compared to controls. Lnc-ADM-1 and lnc-FTH1-7 expression correlated positively with the C4 level (p = 0.016 and p = 0.012, respectively). ITSN1-2 levels were negatively associated with CRP levels (p = 0.024). Linc00152, lnc-ADM-1, ITSN1-2, and lnc-FTH1-7 showed potential as RA biomarkers, with the four-lncRNA panel distinguishing RA patients from controls, SLE patients, or pSS patients (AUC = 0.886, 0.746, and 0.749, respectively). CONCLUSION The altered expression of linc00152, lnc-ADM-1, ITSN1-2 and lnc-FTH1-7 in RA patients suggested that these genes may serve as potential biomarkers for RA and could be involved in its pathogenesis.
Collapse
Affiliation(s)
- Hong-Miao Li
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China
| | - Li-Jun Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yi-Ping Wang
- Westmead Institute for Medical Research, University of Sydney, Westmead, 2145 NSW, Australia
| | - Xiao-Mei Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China.
| | - Hai-Feng Pan
- Department of Epidemiology and Biostatistics, School of Public Health, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
2
|
Ghorbani A, Hosseinie F, Khorshid Sokhangouy S, Islampanah M, Khojasteh-Leylakoohi F, Maftooh M, Nassiri M, Hassanian SM, Ghayour-Mobarhan M, Ferns GA, Khazaei M, Nazari E, Avan A. The prognostic, diagnostic, and therapeutic impact of Long noncoding RNAs in gastric cancer. Cancer Genet 2024; 282-283:14-26. [PMID: 38157692 DOI: 10.1016/j.cancergen.2023.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Revised: 11/27/2023] [Accepted: 12/24/2023] [Indexed: 01/03/2024]
Abstract
Gastric cancer (GC), ranking as the third deadliest cancer globally, faces challenges of late diagnosis and limited treatment efficacy. Long non-coding RNAs (lncRNAs) emerge as valuable treasured targets for cancer prognosis, diagnosis, and therapy, given their high specificity, convenient non-invasive detection in body fluids, and crucial roles in diverse physiological and pathological processes. Research indicates the significant involvement of lncRNAs in various aspects of GC pathogenesis, including initiation, metastasis, and recurrence, underscoring their potential as novel diagnostic and prognostic biomarkers, as well as therapeutic targets for GC. Despite existing challenges in the clinical application of lncRNAs in GC, the evolving landscape of lncRNA molecular biology holds promise for advancing the survival and treatment outcomes of gastric cancer patients. This review provides insights into recent studies on lncRNAs in gastric cancer, elucidating their molecular mechanisms and exploring the potential clinical applications in GC.
Collapse
Affiliation(s)
- Atousa Ghorbani
- Department of Biology, East Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Fatemeh Hosseinie
- Department of Nursing, Faculty of Nursing and Midwifery, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Saeideh Khorshid Sokhangouy
- Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Muhammad Islampanah
- Student Research Committee, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Mina Maftooh
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mohammadreza Nassiri
- Recombinant Proteins Research Group, The Research Institute of Biotechnology, Ferdowsi University of Mashhad, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Ghayour-Mobarhan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex BN1 9PH, UK
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elham Nazari
- Department of Health Information Technology and Management, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran; Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
3
|
Hashemi M, Nazdari N, Gholamiyan G, Paskeh MDA, Jafari AM, Nemati F, Khodaei E, Abyari G, Behdadfar N, Raei B, Raesi R, Nabavi N, Hu P, Rashidi M, Taheriazam A, Entezari M. EZH2 as a potential therapeutic target for gastrointestinal cancers. Pathol Res Pract 2024; 253:154988. [PMID: 38118215 DOI: 10.1016/j.prp.2023.154988] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 11/18/2023] [Accepted: 11/27/2023] [Indexed: 12/22/2023]
Abstract
Gastrointestinal (GI) cancers continue to be a major cause of mortality and morbidity globally. Understanding the molecular pathways associated with cancer progression and severity is essential for creating effective cancer treatments. In cancer research, there is a notable emphasis on Enhancer of zeste homolog 2 (EZH2), a key player in gene expression influenced by its irregular expression and capacity to attach to promoters and alter methylation status. This review explores the impact of EZH2 signaling on various GI cancers, such as colorectal, gastric, pancreatic, hepatocellular, esophageal, and cholangiocarcinoma. The primary function of EZH2 signaling is to facilitate the accelerated progression of cancer cells. Additionally, EZH2 has the capacity to modulate the reaction of GI cancers to chemotherapy and radiotherapy. Numerous pathways, including long non-coding RNAs and microRNAs, serve as upstream regulators of EZH2 in these types of cancer. EZH2's enzymatic activity enables it to attach to target gene promoters, resulting in methylation that modifies their expression. EZH2 could be considered as an independent prognostic factor, with increased expression correlating with a worse disease prognosis. Additionally, a range of gene therapies including small interfering RNA, and anti-tumor agents are being explored to target EZH2 for cancer treatment. This comprehensive review underscores the current insights into EZH2 signaling in gastrointestinal cancers and examines the prospect of therapies targeting EZH2 to enhance patient outcomes.
Collapse
Affiliation(s)
- Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Naghmeh Nazdari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ghazaleh Gholamiyan
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mahshid Deldar Abad Paskeh
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Ali Moghadas Jafari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Fateme Nemati
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Elaheh Khodaei
- Department of Dermatology, Faculty of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ghazal Abyari
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Nazanin Behdadfar
- Young Researchers and Elite Club, Buinzahra Branch, Islamic Azad University, Buinzahra, Iran
| | - Behnaz Raei
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical-Surgical Nursing, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada
| | - Peng Hu
- Department of Emergency, Shanghai Tenth People's Hospital, Tongji University, Shanghai 200072, China
| | - Mohsen Rashidi
- Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran; The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
4
|
Wang W, Yun B, Hoyle RG, Ma Z, Zaman SU, Xiong G, Yi C, Xie N, Zhang M, Liu X, Bandyopadhyay D, Li J, Wang C. CYTOR Facilitates Formation of FOSL1 Phase Separation and Super Enhancers to Drive Metastasis of Tumor Budding Cells in Head and Neck Squamous Cell Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305002. [PMID: 38032139 PMCID: PMC10811474 DOI: 10.1002/advs.202305002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 10/31/2023] [Indexed: 12/01/2023]
Abstract
Tumor budding (TB) is a small tumor cell cluster with highly aggressive behavior located ahead of the invasive tumor front. However, the molecular and biological characteristics of TB and the regulatory mechanisms governing TB phenotypes remain unclear. This study reveals that TB exhibits a particular dynamic gene signature with stemness and partial epithelial-mesenchymal transition (p-EMT). Importantly, nuclear expression of CYTOR is identified to be the key regulator governing stemness and the p-EMT phenotype of TB cells, and targeting CYTOR significantly inhibits TB formation, tumor growth and lymph node metastasis in head and neck squamous cell carcinoma (HNSCC). Mechanistically, CYTOR promotes tumorigenicity and metastasis of TB cells by facilitating the formation of FOSL1 phase-separated condensates to establish FOSL1-dependent super enhancers (SEs). Depletion of CYTOR leads to the disruption of FOSL1-dependent SEs, which results in the inactivation of cancer stemness and pro-metastatic genes. In turn, activation of FOSL1 promotes the transcription of CYTOR. These findings indicate that CYTOR is a super-lncRNA that controls the stemness and metastasis of TB cells through facilitating the formation of FOSL1 phase separation and SEs, which may be an attractive target for therapeutic interventions in HNSCC.
Collapse
Affiliation(s)
- Wenjin Wang
- Hospital of StomatologySun Yat‐sen UniversityGuangzhou510055China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510080China
- Guanghua School of StomatologySun Yat‐sen UniversityGuangzhou510055China
| | - Bokai Yun
- Hospital of StomatologySun Yat‐sen UniversityGuangzhou510055China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510080China
- Guanghua School of StomatologySun Yat‐sen UniversityGuangzhou510055China
| | - Rosalie G Hoyle
- Department of Medicinal ChemistrySchool of PharmacyVirginia Commonwealth UniversityRichmondVA23298‐0540USA
| | - Zhikun Ma
- Department of Medicinal ChemistrySchool of PharmacyVirginia Commonwealth UniversityRichmondVA23298‐0540USA
| | - Shadid Uz Zaman
- Department of Medicinal ChemistrySchool of PharmacyVirginia Commonwealth UniversityRichmondVA23298‐0540USA
| | - Gan Xiong
- Hospital of StomatologySun Yat‐sen UniversityGuangzhou510055China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510080China
- Guanghua School of StomatologySun Yat‐sen UniversityGuangzhou510055China
| | - Chen Yi
- Hospital of StomatologySun Yat‐sen UniversityGuangzhou510055China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510080China
- Guanghua School of StomatologySun Yat‐sen UniversityGuangzhou510055China
| | - Nan Xie
- Hospital of StomatologySun Yat‐sen UniversityGuangzhou510055China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510080China
- Guanghua School of StomatologySun Yat‐sen UniversityGuangzhou510055China
| | - Ming Zhang
- Hospital of StomatologySun Yat‐sen UniversityGuangzhou510055China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510080China
- Guanghua School of StomatologySun Yat‐sen UniversityGuangzhou510055China
| | - Xiqiang Liu
- Department of Oral and Maxillofacial SurgeryNanfang Hospital, Southern Medical UniversityGuangzhou510515China
| | - Dipankar Bandyopadhyay
- Department of BiostatisticsSchool of MedicineVirginia Commonwealth UniversityRichmondVA23298‐0540USA
- Massey Cancer CenterVirginia Commonwealth UniversityRichmondVA23298‐0540USA
| | - Jiong Li
- Department of Medicinal ChemistrySchool of PharmacyVirginia Commonwealth UniversityRichmondVA23298‐0540USA
- Massey Cancer CenterVirginia Commonwealth UniversityRichmondVA23298‐0540USA
- Department of Oral and Craniofacial Molecular BiologySchool of DentistryVirginia Commonwealth UniversityRichmondVA23298‐0540USA
- Philips Institute for Oral Health ResearchSchool of DentistryVirginia Commonwealth UniversityRichmondVA23298‐0540USA
| | - Cheng Wang
- Hospital of StomatologySun Yat‐sen UniversityGuangzhou510055China
- Guangdong Provincial Key Laboratory of StomatologyGuangzhou510080China
- Guanghua School of StomatologySun Yat‐sen UniversityGuangzhou510055China
| |
Collapse
|
5
|
Deng X, Wang Y, Guo H, Wang Q, Rao S, Wu H. Pan-Cancer Analysis and Experimental Validation of SOX4 as a Potential Diagnosis, Prognosis, and Immunotherapy Biomarker. Cancers (Basel) 2023; 15:5235. [PMID: 37958409 PMCID: PMC10649301 DOI: 10.3390/cancers15215235] [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: 09/02/2023] [Revised: 10/11/2023] [Accepted: 10/13/2023] [Indexed: 11/15/2023] Open
Abstract
INTRODUCTION SOX4 plays an important role in tumorigenesis and cancer progression. The role of SOX4 in pan-cancer and its underlying molecular mechanism in liver hepatocellular carcinoma (LIHC) are not fully understood. In this study, a comprehensive analysis and experimental validation were performed to explore the function of SOX4 across tumor types. METHODS Raw data in regard to SOX4 expression in malignant tumors were downloaded from the TCGA and GTEx databases. The expression levels, prognostic values, genetic mutation, and DNA promoter methylation of SOX4 across tumor types were explored via systematic bioinformatics analysis. The ceRNA regulatory network, immune characteristics, and prognostic models were analyzed in LIHC. Finally, we conducted in vitro experiments including Western blotting, cell proliferative assay, trypan blue staining, and fluorescence microscopy to further explore the function of SOX4 in LIHC. RESULTS SOX4 expression was significantly upregulated in 24 tumor types. SOX4 expression level was strongly associated with unfavorable prognoses, genetic mutations, and DNA methylation levels across different tumor types. Especially in LIHC, LINC00152/hsa-miR-139-3p/SOX4 was identified as a crucial ceRNA network. Moreover, this study also provides insight into the roles of SOX4 expression in immune cell infiltration, macrophage polarization, immune subtype, molecular subtype, and immunomodulators, as well as the tumor immune microenvironment (TIME)-related prognosis, in LIHC. The study established six favorable prognostic models to predict LIHC prognosis based on the SOX4-associated genes. Finally, lenvatinib treatment can increase the expression of SOX4 in hepatocellular carcinoma cells and lead to drug resistance. Silencing SOX4 can effectively eliminate the drug resistance caused by lenvatinib treatment and inhibit the proliferation of cancer cells. CONCLUSIONS This study highlights that SOX4 may serve as a promising therapeutic target for tumor treatment.
Collapse
Affiliation(s)
- Xinna Deng
- Departments of Oncology, Hebei General Hospital, Shijiazhuang 050057, China; (X.D.); (H.G.); (Q.W.); (S.R.)
| | - Yashu Wang
- Department of Pathology, Hebei Medical University, Shijiazhuang 050011, China;
| | - Hao Guo
- Departments of Oncology, Hebei General Hospital, Shijiazhuang 050057, China; (X.D.); (H.G.); (Q.W.); (S.R.)
| | - Qian Wang
- Departments of Oncology, Hebei General Hospital, Shijiazhuang 050057, China; (X.D.); (H.G.); (Q.W.); (S.R.)
| | - Shuting Rao
- Departments of Oncology, Hebei General Hospital, Shijiazhuang 050057, China; (X.D.); (H.G.); (Q.W.); (S.R.)
- Department of Internal Medicine, Hebei Medical University, Shijiazhuang 050011, China
| | - Haijiang Wu
- Department of Pathology, Hebei Medical University, Shijiazhuang 050011, China;
| |
Collapse
|
6
|
Li S, Yao W, Liu R, Gao L, Lu Y, Zhang H, Liang X. Long non-coding RNA LINC00152 in cancer: Roles, mechanisms, and chemotherapy and radiotherapy resistance. Front Oncol 2022; 12:960193. [PMID: 36033524 PMCID: PMC9399773 DOI: 10.3389/fonc.2022.960193] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2022] [Accepted: 07/20/2022] [Indexed: 11/13/2022] Open
Abstract
Long non-coding RNA LINC00152 (cytoskeleton regulator, or LINC00152) is an 828-bp lncRNA located on chromosome 2p11.2. LINC00152 was originally discovered during research on hepatocarcinogenesis and has since been regarded as a crucial oncogene that regulates gene expression in many cancer types. LINC00152 is aberrantly expressed in various cancers, including gastric, breast, ovarian, colorectal, hepatocellular, and lung cancer, and glioma. Several studies have indicated that LINC00152 is correlated with cell proliferation, apoptosis, migration, invasion, cell cycle, epithelial-mesenchymal transition (EMT), chemotherapy and radiotherapy resistance, and tumor growth and metastasis. High LINC00152 expression in most tumors is significantly associated with poor patient prognosis. Mechanistic analysis has demonstrated that LINC00152 can serve as a competing endogenous RNA (ceRNA) by sponging miRNA, regulating the abundance of the protein encoded by a particular gene, or modulating gene expression at the epigenetic level. LINC00152 can serve as a diagnostic or prognostic biomarker, as well as a therapeutic target for most cancer types. In the present review, we discuss the roles and mechanisms of LINC00152 in human cancer, focusing on its functions in chemotherapy and radiotherapy resistance.
Collapse
Affiliation(s)
- Shuang Li
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Jinzhou Medical University, Jinzhou, China
| | - Weiping Yao
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Bengbu Medical College, Bengbu, China
| | - Ruiqi Liu
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Bengbu Medical College, Bengbu, China
| | - Liang Gao
- Cancer Center, Department of Medical Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Yanwei Lu
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
| | - Haibo Zhang
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- *Correspondence: Xiaodong Liang, ; Haibo Zhang,
| | - Xiaodong Liang
- Cancer Center, Department of Affiliated People’ Radiation Oncology, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, China
- Graduate Department, Jinzhou Medical University, Jinzhou, China
- *Correspondence: Xiaodong Liang, ; Haibo Zhang,
| |
Collapse
|
7
|
Xia L, Lin H, Zhou Y, Lian J. ZNF750 facilitates carcinogenesis via promoting the expression of long non-coding RNA CYTOR and influences pharmacotherapy response in colon adenocarcinoma. J Zhejiang Univ Sci B 2022; 23:587-596. [PMID: 35794688 DOI: 10.1631/jzus.b2100939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
The epidermal cell differentiation regulator zinc finger protein 750 (ZNF750) is a transcription factor containing the Cys2His2 (C2H2) domain, the zinc finger structure of which is located at the N-terminal 25-46 amino acids of ZNF750. It can promote the expression of differentiation-related factors while inhibiting the expression of progenitor cell-related genes. ZNF750 is directly regulated by p63 (encoded by the TP63 gene, belonging to the TP53 superfamily). The Krüppel-like factor 4 (KLF4), repressor element-1 (RE-1)-silencing transcription factor (REST) corepressor 1 (RCOR1), lysine demethylase 1A (KDM1A), and C-terminal-binding protein 1/2 (CTBP1/2) chromatin regulators cooperate with ZNF750 to repress epidermal progenitor genes and activate the expression of epidermal terminal differentiation genes (Sen et al., 2012; Boxer et al., 2014). Besides, ZNF750 and the regulatory network composed of bone morphogenetic protein (BMP) signaling pathway, long non-coding RNAs (lncRNAs) (anti-differentiation non-coding RNA (ANCR) and tissue differentiation-inducing non-protein coding RNA (TINCR)), musculoaponeurotic fibrosarcoma oncogene (MAF)/MAF family B (MAFB), grainy head-like 3 (GRHL3), and positive regulatory domain zinc finger protein 1 (PRDM1) jointly promote epidermal cell differentiation (Sen et al., 2012).
Collapse
Affiliation(s)
- Lu Xia
- Xiamen Cell Therapy Research Center, the First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Hexin Lin
- Department of Colorectal Surgery, the First Affiliated Hospital of Fujian Medical University, Fuzhou 350004, China
| | - Yanming Zhou
- Department of Gastrointestinal Surgery, the First Affiliated Hospital of Xiamen University, Xiamen 361000, China
| | - Jiabian Lian
- Department of Clinical Laboratory, the First Affiliated Hospital of Xiamen University, Xiamen 361000, China.
| |
Collapse
|
8
|
Transcription Factor E2F1 Exacerbates Papillary Thyroid Carcinoma Cell Growth and Invasion via Upregulation of LINC00152. Anal Cell Pathol (Amst) 2022; 2022:7081611. [PMID: 35592867 PMCID: PMC9113902 DOI: 10.1155/2022/7081611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 03/07/2022] [Accepted: 03/29/2022] [Indexed: 11/17/2022] Open
Abstract
Background Papillary thyroid carcinoma (PTC) is the most common thyroid neoplasm, whereas transcription factor E2F1 has been previously implicated in PTC progression. The current study sought to elucidate the underlying mechanism of E2F1 in PTC cell biological activities via regulation of long intergenic noncoding RNA 152 (LINC00152). Methods Firstly, the expression patterns of LINC00152 and E2F1 in PTC were determined. Besides, TPC-1 and IHH-4 cells were adopted to carry out a series of experiments. Cell proliferation was detected by means of a cell counting kit-8 assay and colony formation assay, while cell migration and invasion abilities were assessed using a Transwell assay. Next, the interaction between E2F1 and LINC00152 was certified. Lastly, xenograft transplantation was carried out to validate the effects of E2F1 depletion on PTC. Results Both LINC00152 and E2F1 were highly expressed in PTC cells. Knockdown of LINC00152 led to reduced cell activity, while LINC00152 overexpression brought about the opposing trends. Likewise, E2F1 knockdown quenched cell proliferation, migration, and invasion. However, the combination of E2F1 knockdown and LINC00152 overexpression resulted in augmented cell growth. In addition, E2F1 induced LINC00152 overexpression, which accelerated cell proliferation, migration, and invasion by activating the PI3K/AKT axis, whereas the administration of LY294002, the inhibitor of PI3K, led to reversal of the same. Finally, xenograft transplantation validated that E2F1 inhibition could suppress LY294002, thereby discouraging tumor growth. Conclusion Our findings highlighted that E2F1 augmented PTC cell proliferation and invasion by upregulating LINC00152 and the PI3K/AKT axis. Our discovery provides therapeutic implications for PTC alleviation.
Collapse
|
9
|
Stage-Specific Non-Coding RNA Expression Patterns during In Vitro Human B Cell Differentiation into Antibody Secreting Plasma Cells. Noncoding RNA 2022; 8:ncrna8010015. [PMID: 35202088 PMCID: PMC8878715 DOI: 10.3390/ncrna8010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 01/31/2022] [Accepted: 02/03/2022] [Indexed: 11/17/2022] Open
Abstract
The differentiation of B cells into antibody secreting plasma cells (PCs) is governed by a strict regulatory network that results in expression of specific transcriptomes along the activation continuum. In vitro models yielding significant numbers of PCs phenotypically identical to the in vivo state enable investigation of pathways, metabolomes, and non-coding (ncRNAs) not previously identified. The objective of our study was to characterize ncRNA expression during human B cell activation and differentiation. To achieve this, we used an in vitro system and performed RNA-seq on resting and activated B cells and PCs. Characterization of coding gene transcripts, including immunoglobulin (Ig), validated our system and also demonstrated that memory B cells preferentially differentiated into PCs. Importantly, we identified more than 980 ncRNA transcripts that are differentially expressed across the stages of activation and differentiation, some of which are known to target transcription, proliferation, cytoskeletal, autophagy and proteasome pathways. Interestingly, ncRNAs located within Ig loci may be targeting both Ig and non-Ig-related transcripts. ncRNAs associated with B cell malignancies were also identified. Taken together, this system provides a platform to study the role of specific ncRNAs in B cell differentiation and altered expression of those ncRNAs involved in B cell malignancies.
Collapse
|
10
|
Ye S, Lu Y, Ru Y, Wu X, Zhao M, Chen J, Xu M, Huang Q, Wang Y, Shi S, Bu S, Xi Y. LncRNAs GACAT3 and LINC00152 regulate each other through miR-103 and are associated with clinicopathological characteristics in colorectal cancer. J Clin Lab Anal 2020; 34:e23378. [PMID: 32462718 PMCID: PMC7521261 DOI: 10.1002/jcla.23378] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 11/29/2019] [Accepted: 12/04/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Long non-coding RNAs (lncRNAs) perform pivotal regulatory roles in tumor development. Our previous work revealed that the lncRNA gastric cancer-associated transcript 3 (GACAT3) was significantly overexpressed and associated with tumor size and metastasis in gastric cancer. METHODS Total RNAs were extracted from colorectal cancer (CRC) and reverse transcribed, and then quantitative real-time PCR (qRT-PCR) was conducted. Cell counting was performed to assess the effect of GACAT3 on CRC cell line proliferation. Bioinformatics prediction, dual luciferase assay, miRNA mimics, siRNAs, and transfection experiments were applied to determine whether GACAT3 and LINC00152 are reciprocally regulated by miR-103. The relationship between their expression levels and clinicopathological factors of patients was explored. A receiver operating characteristic (ROC) curve was used to assess the potential diagnostic value of GACAT3 and LINC00152. RESULTS GACAT3 was identified to be highly expressed in CRC tissues and associated with cell proliferation. Furthermore, we demonstrated that GACAT3 acted as a competing endogenous RNA of LINC00152 and they were both regulated by miR-103. Moreover, analysis of clinicopathological characteristics revealed that GACAT3 and LINC00152 were positively correlated with the depth of invasion, TNM stage, lymph node metastasis, and CA19-9 level. Importantly, a combination of GACAT3 and LINC00152 showed a superior diagnostic capacity compared with the use of the two molecules alone. CONCLUSION Our work shows that GACAT3 and LINC00152 are both overexpressed in CRC and they act as a ceRNA network. Therefore, our data suggest that GACAT3 and LINC00152 may be a promising potential diagnostic biomarker for CRC.
Collapse
Affiliation(s)
- Shazhou Ye
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Yicong Lu
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Yuqing Ru
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Xiaoyue Wu
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Ming Zhao
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Jiayi Chen
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Mingjun Xu
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Qin Huang
- Department of Endocrinology, Changhai Hospital, Second Military Medical University, Shanghai, China
| | - Yibo Wang
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Shanping Shi
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Shizhong Bu
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| | - Yang Xi
- Diabetes Research Center, Zhejiang Provincial Key Laboratory of Pathophysiology, Institute of Biochemistry and Molecular Biology, School of Medicine, Ningbo University, Ningbo, China
| |
Collapse
|
11
|
Zheng X, Dong S, Sun L, Xu J, Liu J, Hao R. LncRNA LINC00152 Promotes Laryngeal Cancer Progression by Sponging MiR-613. Open Med (Wars) 2020; 15:240-248. [PMID: 32266320 PMCID: PMC7126196 DOI: 10.1515/med-2020-0035] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2019] [Accepted: 01/29/2020] [Indexed: 12/18/2022] Open
Abstract
Background Long noncoding RNA (lncRNA) LINC00152 (CYTOR) has been reported to be upregulated and to serve as a diagnostic biomarker in multiple types of cancers, including laryngeal squamous cell cancer (LSCC). However, the functional role and molecular mechanisms of LINC00152 in LSCC progression need to be further investigated. Methods LINC00152 levels in LSCC and adjacent normal tissues were measured by quantitative real-time polymerase chain reaction (qRT-PCR). Gene knockdown of LINC00152 was achieved in LSCC cells by use of small interfering RNA (siRNA). Cell proliferation, apoptosis, migration and invasion were examined by a series of methods. The micoRNA (miRNA) interaction with LINC00152 was screened by starBase v2.0 and confirmed by luciferase reporter activity. Results LINC00152 levels in LSCC tissues were significantly higher than those in adjacent normal tissue, and patients with lymph node metastasis or an advanced clinical stage displayed higher LINC00152 expression. Moreover, siRNA-mediated LINC00152 knockdown significantly inhibited the proliferation, migration and invasion of LSCC cells and induced apoptosis in those cells. Mechanistically, LINC00152 functioned as a competing endogenous RNA (ceRNA) sponging miR-613. The inhibitory effect of LINC00152 knockdown on malignant behavior was abrogated by inhibiting miR-613. Conclusion LINC00152 exerts an oncogenic effect on the tumorigenesis of LSCC by sponging miR-613 and may serve as a potential target for treating LSCC.
Collapse
Affiliation(s)
- Xuesong Zheng
- Department of Otolaryngology Head and Neck surgery, the Affiliated Hospital of Beihua University, Jilin 132001, P.R. China
| | - Su Dong
- Departments of Anesthesia, the First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Lele Sun
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Jialu Xu
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Jia Liu
- Department of Thyroid Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China
| | - Rui Hao
- Department of Infection, The Affiliated Hospital of Beihua University, Jilin 132001, P.R. China
| |
Collapse
|
12
|
Binder S, Zipfel I, Friedrich M, Riedel D, Ende S, Kämpf C, Wiedemann K, Buschmann T, Puppel SH, Reiche K, Stadler PF, Horn F. Master and servant: LINC00152 - a STAT3-induced long noncoding RNA regulates STAT3 in a positive feedback in human multiple myeloma. BMC Med Genomics 2020; 13:22. [PMID: 32041604 PMCID: PMC7011539 DOI: 10.1186/s12920-020-0692-3] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Accepted: 01/31/2020] [Indexed: 12/16/2022] Open
Abstract
Background The survival of INA-6 human multiple myeloma cells is strictly dependent upon the Interleukin-6-activated transcription factor STAT3. Although transcriptional analyses have revealed many genes regulated by STAT3, to date no protein-coding STAT3 target gene is known to mediate survival in INA-6 cells. Therefore, the aim here was to identify and analyze non-protein-coding STAT3 target genes. In addition to the oncogenic microRNA-21, we previously described five long noncoding RNAs (lncRNAs) induced by STAT3, named STAiRs. Here, we focus on STAT3-induced RNA 18 (STAiR18), an mRNA-like, long ncRNA that is duplicated in the human lineage. One STAiR18 locus is annotated as the already well described LINC00152/CYTOR, however, the other harbors the MIR4435-2HG gene and is, up to now, barely described. Methods CAPTURE-RNA-sequencing was used to analyze STAiR18 transcript architecture. To identify the STAiR18 and STAT3 phenotype, siRNA-based knockdowns were performed and microarrays were applied to identify their target genes. RNA-binding partners of STAiR18 were determined by Chromatin-Isolation-by-RNA-Purification (ChIRP) and subsequent sequencing. STAT3 expression in dependence of STAiR18 was investigated by immunoblots, chromatin- and RNA-immunoprecipitations. Results As identified by CAPTURE-RNA sequencing, a complex splice pattern originates from both STAiR18 loci, generating different transcripts. Knockdown of the most abundant STAiR18 isoforms dramatically decreased INA-6 cell vitality, suggesting a functional role in myeloma cells. Additionally, STAiR18 and STAT3 knockdowns yielded overlapping changes of transcription patterns in INA-6 cells, suggesting a close functional interplay between the two factors. Moreover, Chromatin isolation by RNA purification (ChIRP), followed by genome-wide RNA sequencing showed that STAiR18 associates specifically with the STAT3 primary transcript. Furthermore, the knockdown of STAiR18 reduced STAT3 levels on both the RNA and protein levels, suggesting a positive feedback between both molecules. Furthermore, STAiR18 knockdown changes the histone methylation status of the STAT3 locus, which explains the positive feedback and indicates that STAiR18 is an epigenetic modulator. Conclusion Hence, STAiR18 is an important regulator of myeloma cell survival and is strongly associated with the oncogenic function of STAT3. The close functional interplay between STAT3 and STAiR18 suggests a novel principle of regulatory interactions between long ncRNAs and signaling pathways.
Collapse
Affiliation(s)
- Stefanie Binder
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany. .,Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany.
| | - Ivonne Zipfel
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany.,Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Maik Friedrich
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany.,Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Diana Riedel
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany
| | - Stefanie Ende
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany.,Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Christoph Kämpf
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Karolin Wiedemann
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Tilo Buschmann
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Sven-Holger Puppel
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Kristin Reiche
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| | - Peter F Stadler
- Bioinformatics Group, Department of Computer Science, and Interdisciplinary Center for Bioinformatics, University of Leipzig, Leipzig, Germany.,German Centre for Integrative Biodiversity Research - iDiv, Halle-Jena-Leipzig, Germany.,Max Planck Institute for Mathematics in the Sciences, Leipzig, Germany.,Department of Theoretical Chemistry, University of Vienna, Wien, Austria.,Center for RNA in Technology and Health, University of Copenhagen, København, Denmark.,Santa Fe Institute, Santa Fe, USA
| | - Friedemann Horn
- Institute of Clinical Immunology, Faculty of Medicine, University of Leipzig, Leipzig, Germany.,Fraunhofer Institute for Cell Therapy and Immunology, Department of Diagnostics, Leipzig, Germany
| |
Collapse
|
13
|
Ren X. Genome-wide analysis reveals the emerging roles of long non-coding RNAs in cancer. Oncol Lett 2019; 19:588-594. [PMID: 31897174 DOI: 10.3892/ol.2019.11141] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2019] [Accepted: 10/23/2019] [Indexed: 12/11/2022] Open
Abstract
Cancer is the most intractable human disease that is primarily caused by genetic alterations. Recently, the general application of microarrays and high-throughput sequencing technology has revealed various important roles of long noncoding RNAs (lncRNAs) in cancer. This review summarizes the function, mechanism, diagnostic and treatment potential of lncRNAs identified through genome-wide analysis in cancer. Cell-, tissue- and development stage-specific expression patterns are major characteristics of cancer-associated lncRNAs, and various genetic alterations are also implicated. Microarray and sequencing analyses serve important roles in mechanistic studies of either nuclear or cytoplasmic lncRNAs. Collectively, genome-wide analysis is the inexorable trend of future studies or clinical applications of lncRNAs and offers a novel perspective regarding the prognosis and treatment of cancer.
Collapse
Affiliation(s)
- Xiaoxia Ren
- Center for Life Sciences, School of Life Sciences, Yunnan University, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
14
|
Xu J, Guo J, Jiang Y, Liu Y, Liao K, Fu Z, Xiong Z. Improved characterization of the relationship between long intergenic non-coding RNA Linc00152 and the occurrence and development of malignancies. Cancer Med 2019; 8:4722-4731. [PMID: 31270960 PMCID: PMC6712457 DOI: 10.1002/cam4.2245] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2019] [Revised: 04/28/2019] [Accepted: 04/30/2019] [Indexed: 12/15/2022] Open
Abstract
Linc00152, located on chromosome 2p11.2, is a long intergenic non-coding RNA molecule with 828 nucleotides that is highly expressed in many types of human tumor tissues, especially in malignant tumors of the digestive system. Linc00152 promotes the occurrence and development of tumors by increasing tumor cell proliferation, invasion, metastasis, and apoptosis. Additionally, linc00152 contributes to the carcinogenesis of several cancers, including gastric cancer, liver cancer, hepatocellular carcinoma, gallbladder cancer, clear cell renal cell carcinoma, and colorectal cancer, by disturbing various signaling pathways (eg PI3K/AKT, mTOR, IL-1, and NOTCH 1 signaling pathways). High linc00152 expression levels are associated with chemoresistance as well as poor prognosis and shorter survival. Continual advances made in the relevant research have indicated that linc00152 may be useful as a new tumor molecular biomarker, applicable for tumor diagnosis, targeted therapy, and prognosis assessment. This review summarizes the progress in the research into the relationship between linc00152 and the occurrence and development of malignancies based on molecular functions, regulatory mechanisms, and clinical applications.
Collapse
Affiliation(s)
- Jiasheng Xu
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Jingjing Guo
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yangkai Jiang
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yujun Liu
- Queen Mary College of Nanchang University, Nanchang, China
| | - Kaili Liao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhonghua Fu
- Department of Burns, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenfang Xiong
- Department of Pathology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| |
Collapse
|