1
|
Wang Y, Hang K, Wu X, Ying L, Wang Z, Ling Z, Hu H, Pan Z, Zou X. SLAMF8 regulates osteogenesis and adipogenesis of bone marrow mesenchymal stem cells via S100A6/Wnt/β-catenin signaling pathway. Stem Cell Res Ther 2024; 15:349. [PMID: 39380096 PMCID: PMC11462740 DOI: 10.1186/s13287-024-03964-1] [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: 01/30/2024] [Accepted: 09/29/2024] [Indexed: 10/10/2024] Open
Abstract
BACKGROUND The inflammatory microenvironment plays an essential role in bone healing after fracture. The signaling lymphocytic activation molecule family (SLAMF) members deeply participate in inflammatory response and make a vast difference. METHODS We identified SLAMF8 in GEO datasets (GSE129165 and GSE176086) and co-expression analyses were performed to define the relationships between SLAMF8 and osteogenesis relative genes (RUNX2 and COL1A1). In vitro, we established SLAMF8 knockdown and overexpression mouse bone marrow mesenchymal stem cells (mBMSCs) lines. qPCR, Western blot, ALP staining, ARS staining, Oil Red O staining and Immunofluorescence analyses were performed to investigate the effect of SLAMF8 in mBMSCs osteogenesis and adipogenesis. In vivo, mice femoral fracture model was performed to explore the function of SLAMF8. RESULTS SLAMF8 knockdown significantly suppressed the expression of osteogenesis relative genes (RUNX2, SP7 and COL1A1), ALP activity and mineral deposition, but increased the expression of adipogenesis relative genes (PPARγ and C/EBPα). Additionally, SLAMF8 overexpression had the opposite effects. The role SLAMF8 played in mBMSCs osteogenic and adipogenic differentiation were through S100A6 and Wnt/β-Catenin signaling pathway. Moreover, SLAMF8 overexpression mBMSCs promoted the healing of femoral fracture. CONCLUSIONS SLAMF8 promotes osteogenesis and inhibits adipogenesis of mBMSCs via S100A6 and Wnt/β-Catenin signaling pathway. SLAMF8 overexpression mBMSCs effectively accelerate the healing of femoral fracture in mice.
Collapse
Affiliation(s)
- Yibo Wang
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Kai Hang
- Department of Orthopaedics, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310052, China
| | - Xiaoyong Wu
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310009, China
| | - Li Ying
- Department of Orthopedic, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Linhai, 317000, China
| | - Zhongxiang Wang
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310009, China
| | - Zemin Ling
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
- Department of Orthopedics, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Hao Hu
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Zhijun Pan
- Department of Orthopedic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, 310009, China.
- Orthopedics Research Institute of Zhejiang University, Hangzhou, 310009, China.
| | - Xuenong Zou
- Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Department of Spinal Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China.
| |
Collapse
|
2
|
Mosca N, Alessio N, Di Paola A, Marrapodi MM, Galderisi U, Russo A, Rossi F, Potenza N. Osteosarcoma in a ceRNET perspective. J Biomed Sci 2024; 31:59. [PMID: 38835012 DOI: 10.1186/s12929-024-01049-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 05/24/2024] [Indexed: 06/06/2024] Open
Abstract
Osteosarcoma (OS) is the most prevalent and fatal type of bone tumor. It is characterized by great heterogeneity of genomic aberrations, mutated genes, and cell types contribution, making therapy and patients management particularly challenging. A unifying picture of molecular mechanisms underlying the disease could help to transform those challenges into opportunities.This review deeply explores the occurrence in OS of large-scale RNA regulatory networks, denominated "competing endogenous RNA network" (ceRNET), wherein different RNA biotypes, such as long non-coding RNAs, circular RNAs and mRNAs can functionally interact each other by competitively binding to shared microRNAs. Here, we discuss how the unbalancing of any network component can derail the entire circuit, driving OS onset and progression by impacting on cell proliferation, migration, invasion, tumor growth and metastasis, and even chemotherapeutic resistance, as distilled from many studies. Intriguingly, the aberrant expression of the networks components in OS cells can be triggered also by the surroundings, through cytokines and vesicles, with their bioactive cargo of proteins and non-coding RNAs, highlighting the relevance of tumor microenvironment. A comprehensive picture of RNA regulatory networks underlying OS could pave the way for the development of innovative RNA-targeted and RNA-based therapies and new diagnostic tools, also in the perspective of precision oncology.
Collapse
Affiliation(s)
- Nicola Mosca
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Nicola Alessio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Alessandra Di Paola
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Maria Maddalena Marrapodi
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Umberto Galderisi
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Aniello Russo
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy
| | - Francesca Rossi
- Department of Woman, Child and General and Specialist Surgery, University of Campania "Luigi Vanvitelli", Naples, Italy
| | - Nicoletta Potenza
- Department of Environmental, Biological and Pharmaceutical Sciences and Technologies, University of Campania "Luigi Vanvitelli", Caserta, Italy.
| |
Collapse
|
3
|
Zhang T, Ji C, Zhang Y, Yuan M, Gao H, Yin Q. LncRNA SNHG1 Accelerates Cell Proliferation, Migration, and Invasion of Hepatoblastoma Through Mediating miR-6838-5p/PIM3/RhoA Axis. Biochem Genet 2024; 62:59-76. [PMID: 37248373 DOI: 10.1007/s10528-023-10404-x] [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: 02/02/2023] [Accepted: 05/16/2023] [Indexed: 05/31/2023]
Abstract
Hepatoblastoma (HB) is a common primary liver malignant tumor in children. Long non-coding RNAs (lncRNAs) are closely engaged in HB progression. The role and regulatory molecule mechanism of lncRNA small nucleolar RNA host gene 1 (SNHG1) in HB remain unclear. Through qRT-PCR or western blot, we found that SNHG1 and proviral integration site for moloney murine leukemia virus 3 (PIM3) were elevated but miR-6838-5p was decreased in HB cells. Cell biology experiments revealed that SNHG1 depletion or miR-6838-5p upregulation suppressed cell proliferation, migration, and invasion of HB cells. Mechanistically, luciferase activity assay validated that miR-6838-5p could interact with SNHG1 or PIM3. SNHG1 up-regulated PIM3 expression via sponging miR-6838-5p. Moreover, miR-6838-5p inhibitor abolished SNHG1 depletion-mediated suppression of malignant behaviors in HB cells. PIM3 overexpression neutralized miR-6838-5p mimics-mediated repression of malignant phenotypes in HB cells. Furthermore, miR-6838-5p overexpression suppressed RhoA activation, which was restored by PIM3 upregulation. What's more, the results at the cellular level were further verified by nude mice tumor formation experiment. In conclusion, SNHG1 regulated miR-6838-5p/PIM3/RhoA axis to promote malignant phenotypes of HB, which might provide novel therapeutic target for HB treatment.
Collapse
Affiliation(s)
- Tian Zhang
- General Surgery, Hunan Children's Hospital, No. 86, ZiYuan Road, Yuhua District, Changsha, 410007, Hunan Province, People's Republic of China
| | - Chunyi Ji
- General Surgery, Hunan Children's Hospital, No. 86, ZiYuan Road, Yuhua District, Changsha, 410007, Hunan Province, People's Republic of China
| | - Yanbing Zhang
- General Surgery, Hunan Children's Hospital, No. 86, ZiYuan Road, Yuhua District, Changsha, 410007, Hunan Province, People's Republic of China
| | - Miaoxian Yuan
- General Surgery, Hunan Children's Hospital, No. 86, ZiYuan Road, Yuhua District, Changsha, 410007, Hunan Province, People's Republic of China
| | - Hongqiang Gao
- General Surgery, Hunan Children's Hospital, No. 86, ZiYuan Road, Yuhua District, Changsha, 410007, Hunan Province, People's Republic of China
| | - Qiang Yin
- General Surgery, Hunan Children's Hospital, No. 86, ZiYuan Road, Yuhua District, Changsha, 410007, Hunan Province, People's Republic of China.
| |
Collapse
|
4
|
Yang Z, Liu Z, Lu W, Guo H, Chen J, Zhang Y. LncRNA WAC-AS1 promotes osteosarcoma Metastasis and stemness by sponging miR-5047 to upregulate SOX2. Biol Direct 2023; 18:74. [PMID: 37957698 PMCID: PMC10644615 DOI: 10.1186/s13062-023-00433-2] [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: 07/17/2023] [Accepted: 11/02/2023] [Indexed: 11/15/2023] Open
Abstract
Cancer stemness and osteosarcoma (OS) malignant progression are closely associated. However, the molecular mechanisms underlying this association have not been fully demonstrated. Long noncoding RNAs (lncRNAs) are an intriguing class of widely prevalent endogenous RNAs involved in OS progression, the vast majority of which have not been characterized functionally. Here, we identified tumor promoter lncRNA WAC-AS1 to be highly expressed in OS tumors and associated with worse survival. Further analysis revealed that WAC-AS1 increased tumorsphere formation of OS cells and promoted metastasis, as confirmed by cell proliferation, transwell and wound healing assays. MiR-5047 was identified as a downstream target of WAC-AS1. Subsequently, based on bioinformatics analysis, RIP assay and luciferase reporter assay, SOX2 mRNA was verified as a target of miR-5047. WAC-AS1 enhanced OS cell proliferation and stemness via acting as a ceRNA by binding to miR-5047, thereby increasing SOX2 expression. In addition, SOX2 bound to the promoter region of WAC-AS1 and promoted its transcription, thereby forming a positive feedback loop to regulate OS malignancy. Taken together, our findings show WAC-AS1 is a tumor promoter and a key regulator of OS cell stemness and metastasis via a miR-5047/SOX2 axis.
Collapse
Affiliation(s)
- Zhining Yang
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, Guangdong, PR China
| | - Zhaoyong Liu
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, 515041, Guangdong, China
| | - Weiqing Lu
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, Guangdong, PR China
| | - Huancheng Guo
- Department of Orthopaedics, First Affiliated Hospital of Shantou University Medical College, No.57 Changping Road, Shantou, 515041, Guangdong, China
| | - Jianzhou Chen
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, Guangdong, PR China
| | - Ying Zhang
- Department of Radiotherapy, Cancer Hospital of Shantou University Medical College, No. 7 Raoping Road, Shantou, 515041, Guangdong, PR China.
| |
Collapse
|
5
|
Sadrkhanloo M, Paskeh MDA, Hashemi M, Raesi R, Bahonar A, Nakhaee Z, Entezari M, Beig Goharrizi MAS, Salimimoghadam S, Ren J, Nabavi N, Rashidi M, Dehkhoda F, Taheriazam A, Tan SC, Hushmandi K. New emerging targets in osteosarcoma therapy: PTEN and PI3K/Akt crosstalk in carcinogenesis. Pathol Res Pract 2023; 251:154902. [PMID: 37922723 DOI: 10.1016/j.prp.2023.154902] [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: 08/21/2023] [Revised: 10/14/2023] [Accepted: 10/19/2023] [Indexed: 11/07/2023]
Abstract
Osteosarcoma (OS) is a malignant bone carcinoma that affects people in childhood and adulthood. The heterogeneous nature and chromosomal instability represent certain characteristics of OS cells. These cancer cells grow and migrate abnormally, making the prognosis undesirable for patients. Conventional and current treatments fail to completely eradicate tumor cells, so new therapeutics targeting genes may be considered. PI3K/Akt is a regulator of events such as growth, cell death, migration, and differentiation, and its expression changes during cancer progression. PTEN reduces PI3K/Akt expression, and its mutations and depletions have been reported in various tumors. Experimental evidence shows that there is upregulation of PI3K/Akt and downregulation of PTEN in OS. Increasing PTEN expression may suppress PI3K/Akt to minimize tumorigenesis. In addition, PI3K/Akt shows a positive association with growth, metastasis, EMT and metabolism of OS cells and inhibits apoptosis. Importantly, overexpression of PI3K/Akt causes drug resistance and radio-resistance and its level can be modulated by miRNAs, lncRNAs and circRNAs. Silencing PI3K/Akt by compounds and drugs can suppress OS. Here, we review in detail the function of the PTEN/PI3K/Akt in OS, revealing its biological function, function in tumor progression, resistance to therapy, and pharmacological significance.
Collapse
Affiliation(s)
| | - 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
| | - 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
| | - 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
| | - Alireza Bahonar
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Zahra Nakhaee
- Medical School, Gonabad University of Medical Sciences, Gonabad, 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
| | | | - Shokooh Salimimoghadam
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Shahid Chamran University of Ahvaz, Ahvaz, Iran
| | - Jun Ren
- Shanghai Institute of Cardiovascular Diseases, Department of Cardiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Noushin Nabavi
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, V6H3Z6 Vancouver, BC, Canada
| | - 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.
| | - Farshid Dehkhoda
- Department of Orthopedics, Imam Hossein Hospital, Shahid Beheshti University of Medical Sciences, Tehran, 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.
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia
| | - Kiavash Hushmandi
- Department of Food Hygiene and Quality Control, Division of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran.
| |
Collapse
|
6
|
Zeng H, Zhou S, Cai W, Kang M, Zhang P. LncRNA SNHG1: role in tumorigenesis of multiple human cancers. Cancer Cell Int 2023; 23:198. [PMID: 37684619 PMCID: PMC10492323 DOI: 10.1186/s12935-023-03018-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Accepted: 08/06/2023] [Indexed: 09/10/2023] Open
Abstract
Small nucleolar RNA host gene 1 (SNHG1) is an important member of the SNHG family. This family is composed of a group of host genes that can be processed into small nucleolar RNAs and play important biological functions. In an oncogenic role, the SNHG1 expression is increased in various cancers, which has immense application prospects in the diagnosis, treatment, and prognosis of malignant tumors. In this review, we have summarized the role and molecular mechanism of SNHG1 in the development of various cancers. In addition, we have emphasized the clinical significance of SNHG1 in cancers in our article. This molecule is expected to be a new marker for potential usage in the diagnosis, prognosis, and treatment of cancer.
Collapse
Affiliation(s)
- Huang Zeng
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Shouang Zhou
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Weiqiang Cai
- School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Mingqiang Kang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China.
| | - Peipei Zhang
- Department of Thoracic Surgery, Fujian Medical University Union Hospital, 29 Xinquan Road, Gulou, Fuzhou, 350001, China.
| |
Collapse
|
7
|
Toshida K, Itoh S, Harada N, Morinaga A, Yugawa K, Tomiyama T, Kosai-Fujimoto Y, Tomino T, Kurihara T, Nagao Y, Morita K, Oda Y, Yoshizumi T. Cancer-associated fibroblasts promote tumor cell growth via miR-493-5p in intrahepatic cholangiocarcinoma. Cancer Sci 2023; 114:937-947. [PMID: 36369960 PMCID: PMC9986089 DOI: 10.1111/cas.15644] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 10/18/2022] [Accepted: 11/09/2022] [Indexed: 11/14/2022] Open
Abstract
The association between tumor microenvironment (TME) and cancer-associated fibroblasts (CAFs) in intrahepatic cholangiocarcinoma (ICC) progression is poorly understood. This study aimed to reveal whether specific microRNAs (miRNAs) in extracellular vesicles (EVs) derived from CAFs were involved in ICC progression. Conditioned medium (CM) and EVs in the CM of CAFs and normal fibroblasts (NFs) derived from ICC specimens were used to investigate the effects on tumor cell lines. miRNA microarray assay was used to examine the miRNAs of EVs derived from CAFs and NFs in ICC, and the effects of miR-493-5p on tumor cell lines were examined. Additionally, databases were used to identify miR-493-5p targets, and the relationship between prognosis of ICC patients and cocaine- and amphetamine-regulated transcript propeptide (CARTPT), one of the targets of miR-493-5p, expression in ICC tissues was retrospectively analyzed. Compared with NF-derived CM and EVs, CAF-derived CM and EVs promoted cell lines in proliferation, scratch, migration, and invasion assays. miRNA microarray analysis revealed that miR-493-5p was significantly increased in CAF-derived EVs compared to NF-derived EVs. Tumor cell lines transfected with miR-493-5p were promoted in proliferation and scratch assays. Immunohistochemical staining was performed on 76 ICC specimens; both overall and recurrence-free survival rates were significantly worse in the CARTPT-negative group. Univariate and multivariate analyses showed that low CARTPT expression was an independent poor prognostic factor for overall and recurrence-free survival. Overall, our data suggest that CAFs in the ICC TME suppress CARTPT in tumor cells and promote tumor cells via miR-493-5p in EVs.
Collapse
Affiliation(s)
- Katsuya Toshida
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Shinji Itoh
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Noboru Harada
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Akinari Morinaga
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Kyohei Yugawa
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan.,Department of Anatomic Pathology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Takahiro Tomiyama
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Yukiko Kosai-Fujimoto
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Takahiro Tomino
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Takeshi Kurihara
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Yoshihiro Nagao
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Kazutoyo Morita
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Yoshinao Oda
- Department of Anatomic Pathology, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| | - Tomoharu Yoshizumi
- Department of Surgery and Science, Graduate School of Medicine, Kyushu University, Fukuoka, Japan
| |
Collapse
|
8
|
S100A6 Protein-Expression and Function in Norm and Pathology. Int J Mol Sci 2023; 24:ijms24021341. [PMID: 36674873 PMCID: PMC9866648 DOI: 10.3390/ijms24021341] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 01/13/2023] Open
Abstract
S100A6, also known as calcyclin, is a calcium-binding protein belonging to the S100 protein family. It was first identified and purified more than 30 years ago. Initial structural studies, focused mostly on the mode and affinity of Ca2+ binding and resolution of the resultant conformational changes, were soon complemented by research on its expression, localization and identification of binding partners. With time, the use of biophysical methods helped to resolve the structure and versatility of S100A6 complexes with some of its ligands. Meanwhile, it became clear that S100A6 expression was altered in various pathological states and correlated with the stage/progression of many diseases, including cancers, indicative of its important, and possibly causative, role in some of these diseases. This, in turn, prompted researchers to look for the mechanism of S100A6 action and to identify the intermediary signaling pathways and effectors. After all these years, our knowledge on various aspects of S100A6 biology is robust but still incomplete. The list of S100A6 ligands is growing all the time, as is our understanding of the physiological importance of these interactions. The present review summarizes available data concerning S100A6 expression/localization, interaction with intracellular and extracellular targets, involvement in Ca2+-dependent cellular processes and association with various pathologies.
Collapse
|
9
|
Sharma U, Kaur Rana M, Singh K, Jain A. LINC00324 promotes cell proliferation and metastasis of esophageal squamous cell carcinoma through sponging miR-493-5p via MAPK signaling pathway. Biochem Pharmacol 2023; 207:115372. [PMID: 36493845 DOI: 10.1016/j.bcp.2022.115372] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 11/26/2022] [Accepted: 11/29/2022] [Indexed: 12/12/2022]
Abstract
Long non-coding RNAs have been demonstrated to promote proliferation and metastasis via regulating the miRNA/mRNA regulatory axis in various malignancies. Based on our preliminary study, we investigated the mechanism of LINC00324 through miR-493-5p/MAPK1 in esophageal squamous cell carcinoma (ESCC) pathogenesis. Herein, we confirmed that LINC00324 is significantly upregulated in ESCC primary cells and esophageal squamous cell carcinoma cell line KYSE-70. Silencing of LINC00324 modulates cell proliferation markers, p21, p27, c-Myc, and Cyclin D1 and epithelial-to-mesenchymal transition markers, slug, snail, ZEB1, vimentin, ZO-1, and E-cadherin protein expression in ESCC. Through bioinformatics and dual luciferase reporter assays, we identified miR-493-5p as the direct target molecule of LINC00324. We further revealed that LINC00324 negatively regulates miR-493-5p expression in ESCC. Moreover, our multiple gain-and loss-of-functional experiments proved that a combination of miR-493-5p and LINC00324 significantly rescued ESCC cell proliferation and metastatic phenotypes. Mechanistically, LINC00324 promotes ESCC pathogenesis by acting as a competing endogenous RNA and sponges miR-493-5p activity thereby activating MAPK1 during ESCC progression. We believe that targeting LINC00324 /miR-493-5p/MAPK1 axis may provide new therapeutic avenues for ESCC.
Collapse
Affiliation(s)
- Uttam Sharma
- Central University of Punjab, Village-Ghudda, Bathinda, Punjab 151401, India
| | - Manjit Kaur Rana
- Department of Pathology/Lab Medicine, All India Institute of Medical Sciences, Bathinda 151001, Punjab, India
| | - Karuna Singh
- Department of Radiotherapy, Advanced Cancer Institute, Bathinda affiliated with Baba Farid University of Health Sciences, Faridkot 151203, Punjab, India
| | - Aklank Jain
- Central University of Punjab, Village-Ghudda, Bathinda, Punjab 151401, India.
| |
Collapse
|
10
|
The Cuproptosis-Related Long Noncoding RNA Signature Predicts Prognosis and Tumour Immune Analysis in Osteosarcoma. COMPUTATIONAL AND MATHEMATICAL METHODS IN MEDICINE 2022; 2022:6314182. [PMID: 36388161 PMCID: PMC9646308 DOI: 10.1155/2022/6314182] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 08/25/2022] [Accepted: 09/23/2022] [Indexed: 01/24/2023]
Abstract
Background Cuprotopsis is a type of programmed cell death discovered in recent years. Long noncoding RNAs (lncRNAs) play an important regulatory role in programmed cell death. The effect of cuproptosis-related lncRNAs on osteosarcoma is unknown. Our work, based on cuproptosis-related lncRNAs, proposes a gene signature to assess the prognosis of patients with osteosarcoma. Methods Osteosarcoma gene expression data from The Cancer Genome Atlas (TCGA), clinical features of osteosarcoma and RNA sequencing data of normal adipose tissue were obtained from the UCSC Xena database. A cuproptosis-related lncRNA risk model was established to calculate the risk score. At the same time, cluster analysis, clinicopathological analysis, functional enrichment analysis, and prediction of compounds with potential therapeutic value were evaluated. We analyzed whether there was a correlation between the risk score and tumour immunity. RT-qPCR was used to verify the expression level of lncRNA. Results Nine lncRNAs (AC124798.1, AC006033.2, AL450344.2, AL512625.2, LINC01060, LINC00837, AC004943.2, AC064836.3, and AC100821.2) were identified to create a risk model and indicate the prognosis of patients with osteosarcoma. The high-risk group had a worse prognosis than the low-risk group. Analysis of clinicopathological features, principal component analysis, receiver operating characteristic curve, c-index curve, and comparative analysis of models proved that the model is reliable. Functional enrichment analysis suggests that the risk score may correlate with cell energy metabolism and tumour-related biological function. Three potentially therapeutic compounds have been predicted. These analyses may be beneficial to the treatment of osteosarcoma in the future. RT-qPCR verified the expression level of three lncRNA (LINC01060, NKILA, and SNHG8). Conclusions Cuproptosis-related lncRNAs have a strong relationship with osteosarcoma patients. Nine lncRNA models can effectively forecast the prognosis of osteosarcoma and may play a significant role in the individualized treatment of osteosarcoma patients in the future.
Collapse
|
11
|
Xu N, Zhou X, Shi W, Ye M, Cao X, Chen S, Xu C. Integrative analysis of circulating microRNAs and the placental transcriptome in recurrent pregnancy loss. Front Physiol 2022; 13:893744. [PMID: 35991164 PMCID: PMC9390878 DOI: 10.3389/fphys.2022.893744] [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: 03/23/2022] [Accepted: 07/08/2022] [Indexed: 11/13/2022] Open
Abstract
Recurrent pregnancy loss (RPL) is a major type of pathological pregnancy that still lacks reliable early diagnosis and effective treatment. The placenta is critical to fetal development and pregnancy success because it participates in critical processes such as early embryo implantation, vascular remodeling, and immunological tolerance. RPL is associated with abnormalities in the biological behavior of placental villous trophoblasts, resulting in aberrant placental function. MicroRNAs (miRNAs) are increasingly being recognized as essential regulators of placental development, as well as potential biomarkers. In this study, plasma miRNAs and placental messenger RNAs (mRNAs) from RPL patients and normal pregnant (NP) controls were sequenced and analyzed. Compared to those in NP controls, 108 circulating miRNAs and 1199 placental mRNAs were differentially expressed in RPL samples. A total of 140 overlapping genes (overlapping between plasma miRNA target genes and actual placental disorder genes) were identified, and functional enrichment analysis showed that these genes were mainly related to cell proliferation, angiogenesis, and cell migration. The regulatory network among miRNAs, overlapping genes, and downstream biological processes was analyzed by protein–protein interactions and Cytoscape. Moreover, enriched mRNAs, which were predictive targets of the differentially expressed plasma miRNAs miR-766-5p, miR-1285-3p, and miR-520a-3p, were accordingly altered in the placenta. These results suggest that circulating miRNAs may be involved in the pathogenesis of RPL and are potential noninvasive biomarkers for RPL.
Collapse
Affiliation(s)
- Naixin Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xuanyou Zhou
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Weihui Shi
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Mujin Ye
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
| | - Xianling Cao
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
| | - Songchang Chen
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- *Correspondence: Songchang Chen, ; Chenming Xu,
| | - Chenming Xu
- International Peace Maternity and Child Health Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
- Shanghai Key Laboratory of Embryo Original Diseases, Shanghai, China
- Obstetrics and Gynecology Hospital, Institute of Reproduction and Development, Fudan University, Shanghai, China
- *Correspondence: Songchang Chen, ; Chenming Xu,
| |
Collapse
|
12
|
Li R, Chen Z, Zhou Y, Maimaitirexiati G, Yan Q, Li Y, Maimaitiyimin A, Zhou C, Ren J, Liu C, Mainike A, Zhou P, Ding L. LncRNA SCAMP1 disrupts the balance between miR-26a-5p and ZEB2 to promote osteosarcoma cell viability and invasion. Front Oncol 2022; 12:967000. [PMID: 35992869 PMCID: PMC9382636 DOI: 10.3389/fonc.2022.967000] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Accepted: 07/04/2022] [Indexed: 11/30/2022] Open
Abstract
Osteosarcoma often occurs in children and adolescents and affects their health. The survival rate of osteosarcoma patients is unsatisfactory due to the lack of early detection and metastasis development and drug resistance. Hence, dissection of molecular insight into osteosarcoma initiation and progression is pivotal to provide the new therapeutic strategy. In recent years, long noncoding RNAs (lncRNAs) have burst into stage in osteosarcoma development and malignant behaviors. LncRNA SCAMP1 has been discovered to play an essential role in carcinogenesis and progression. However, the mechanisms of lncRNA SCAMP1-involved tumorigenesis have not been reported in human osteosarcoma. In this study, we utilized multiple cellular biological approaches to determine the function of lncRNA SCAMP1 in osteosarcoma cells. Moreover, we performed several molecular biological approaches to define the mechanism by which lncRNA SCAMP1 regulated cell viability and invasion in osteosarcoma. We dissected that lncRNA SCAMP1 promoted progression of osteosarcoma via modulation of miR-26a-5p/ZEB2 axis. In conclusion, targeting lncRNA SCAMP1 and its downstream targets, miR-26a-5p and ZEB2, might be a useful approach for osteosarcoma therapy.
Collapse
Affiliation(s)
- Rong Li
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Zhen Chen
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Yubo Zhou
- Department of Orthopedics, Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Urumqi, China
| | - Gulikezi Maimaitirexiati
- CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Qi Yan
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Yuting Li
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | | | - Changhui Zhou
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Jingqin Ren
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Chengqing Liu
- College of Public Health, Xinjiang Medical University, Urumqi, China
| | - Abasi Mainike
- Department of Orthopedics, Xinjiang Medical University Affiliated Fifth Hospital, Urumqi, China
| | - Peng Zhou
- Department of Orthopedics, Xinjiang Medical University Affiliated Fifth Hospital, Urumqi, China
| | - Lu Ding
- Traditional Chinese Medicine Hospital Affiliated to Xinjiang Medical University, Postdoctoral Research Center on Public Health and Preventive Medicine, Xinjiang Medical University, Xinjiang, China
- *Correspondence: Lu Ding,
| |
Collapse
|
13
|
Wang G, Zhang X, Feng W, Wang J. Prediction of Prognosis and Immunotherapy of Osteosarcoma Based on Necroptosis-Related lncRNAs. Front Genet 2022; 13:917935. [PMID: 35692813 PMCID: PMC9178207 DOI: 10.3389/fgene.2022.917935] [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: 04/11/2022] [Accepted: 04/25/2022] [Indexed: 11/13/2022] Open
Abstract
Background: Osteosarcoma (OS) is the most common primary tumor of bone in adolescents, and its survival rate is generally less than 20% when metastases occur. Necroptosis, a novel form of programmed necrotic cell death distinct from apoptosis, has been increasingly recognized as a promising therapeutic strategy. This study sought to identify long non-coding RNAs (lncRNAs) associated with necrotizing apoptosis to predict prognosis and target drug use to improve patient survival. Methods: Transcriptomic data and clinical data from 85 OS patients with survival time data and expression profiles from 85 random normal adipose tissue samples were extracted from the UCSC Xena website (http://xena.ucsc.edu/). Nine necroptosis-associated differential prognostic lncRNAs were then identified by analysis of variance, correlation analysis, univariate Cox (uni-Cox) regression, and Kaplan–Meier analysis. Then, patients were randomized into training or testing groups. According to uni-Cox, we obtained prognostic lncRNAs in the training group and intersected them with the abovementioned nine lncRNAs to obtain the final necrotizing apoptosis–related differential prognostic lncRNAs (NRlncRNAs). Next, we performed the least absolute shrinkage and selection operator (LASSO) to construct a risk model of NRlncRNAs. Kaplan–Meier analysis, ROC curves, nomograms, calibration curves, and PCA were used to validate and evaluate the models and grouping. We also analyzed the differences in tumor immunity and drugs between risk groups. Results: We constructed a model containing three NRlncRNAs (AL391121.1, AL354919.2, and AP000851.2) and validated its prognostic predictive power. The value of the AUC curve of 1-, 3-, and 5-year survival probability was 0.806, 0.728, and 0.731, respectively. Moreover, we found that the overall survival time of patients in the high-risk group was shorter than that in the low-risk group. GSEA and ssGSEA showed that immune-related pathways were mainly abundant in the low-risk group. We also validated the differential prediction of immune checkpoint expression, tumor immunity, and therapeutic compounds in the two risk groups. Conclusion: Overall, NRlncRNAs have important functions in OS, and these three NRlncRNAs can predict the prognosis of OS and provide guidance for immunotherapy in OS.
Collapse
|