1
|
Su KJ, Qiu C, Greenbaum J, Zhang X, Liu A, Liu Y, Luo Z, Mungasavalli Gnanesh SS, Tian Q, Zhao LJ, Shen H, Deng HW. Genomic structural variations link multiple genes to bone mineral density in a multi-ethnic cohort study: Louisiana osteoporosis study. J Bone Miner Res 2024; 39:1474-1485. [PMID: 39167757 PMCID: PMC11425707 DOI: 10.1093/jbmr/zjae133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 07/11/2024] [Accepted: 08/07/2024] [Indexed: 08/23/2024]
Abstract
Osteoporosis, characterized by low BMD, is a highly heritable metabolic bone disorder. Although single nucleotide variations (SNVs) have been extensively studied, they explain only a fraction of BMD heritability. Although genomic structural variations (SVs) are large-scale genomic alterations that contribute to genetic diversity in shaping phenotypic variations, the role of SVs in osteoporosis susceptibility remains poorly understood. This study aims to identify and prioritize genes that harbor BMD-related SVs. We performed whole genome sequencing on 4982 subjects from the Louisiana Osteoporosis Study. To obtain high-confidence SVs, the detection of SVs was performed using an ensemble approach. The SVs were tested for association with BMD variation at the hip (HIP), femoral neck (FNK), and lumbar spine (SPN), respectively. Additionally, we conducted co-occurrence analysis using multi-omics approaches to prioritize the identified genes based on their functional importance. Stratification was employed to explore the sex- and ethnicity-specific effects. We identified significant SV-BMD associations: 125 for FNK-BMD, 99 for SPN-BMD, and 83 for HIP-BMD. We observed SVs that were commonly associated with both FNK and HIP BMDs in our combined and stratified analyses. These SVs explain 13.3% to 19.1% of BMD variation. Novel bone-related genes emerged, including LINC02370, ZNF family genes, and ZDHHC family genes. Additionally, FMN2, carrying BMD-related deletions, showed associations with FNK or HIP BMDs, with sex-specific effects. The co-occurrence analysis prioritized an RNA gene LINC00494 and ZNF family genes positively associated with BMDs at different skeletal sites. Two potential causal genes, IBSP and SPP1, for osteoporosis were also identified. Our study uncovers new insights into genetic factors influencing BMD through SV analysis. We highlight BMD-related SVs, revealing a mix of shared and specific genetic influences across skeletal sites and gender or ethnicity. These findings suggest potential roles in osteoporosis pathophysiology, opening avenues for further research and therapeutic targets.
Collapse
Affiliation(s)
- Kuan-Jui Su
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Chuan Qiu
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Jonathan Greenbaum
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Xiao Zhang
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Anqi Liu
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Yong Liu
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Zhe Luo
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Shashank Sajjan Mungasavalli Gnanesh
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Qing Tian
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Lan-Juan Zhao
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Hui Shen
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| | - Hong-Wen Deng
- Deming Department of Medicine, School of Medicine, Tulane Center for Biomedical Informatics and Genomics, Tulane University, New Orleans, LA 70112, United States
| |
Collapse
|
2
|
Liu D, Zhao X, Zhang Q, Zhou F, Tong X. Bone marrow mesenchymal stem cell-derived exosomes promote osteoblast proliferation, migration and inhibit apoptosis by regulating KLF3-AS1/miR-338-3p. BMC Musculoskelet Disord 2024; 25:122. [PMID: 38336637 PMCID: PMC10854165 DOI: 10.1186/s12891-024-07236-0] [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: 11/01/2023] [Accepted: 01/28/2024] [Indexed: 02/12/2024] Open
Abstract
AIM This study aimed to investigate the effect and mechanism of bone marrow mesenchymal stem cell-derived exosomes on osteoblast function. METHODS The expression of KLF3-AS1 and miR-338-3p in serum of fracture patients was detected by qRT-PCR. Exosomes from BMSCs were isolated by ultrafast centrifugation. MC3T3-E1 cells were cultured in vitro as experimental cells. Intracellular gene expression was regulated by transfection of si-KLF3-AS1 or miR-338-3p inhibitors. MTT assay, Transwell assay and flow cytometry were used to evaluate cell viability, migration, and apoptosis. The luciferase reporter gene was used to verify the targeting relationship between KLF3-AS1 and miR-338-3p. Bioinformatics analysis was used to identify the basic functions and possible enrichment pathways of miR-338-3p target genes. RESULTS The expressions of KLF3-AS1 and miR-338-3p in the serum of fracture patients were down-regulated and up-regulated, respectively. The expression of KLF3-AS1 was increased in MC3T3-E1 cells cultured with BMSCs-Exo, while the viability and migration ability of MC3T3-E1 cells were enhanced, and the apoptosis ability was weakened. Further analysis revealed miR-338-3p was the target gene of KLF3-AS1. The expression of miR-338-3p was downregulated in MC3T3-E1 cells cultured with BMSCs-Exo. Inhibition of miR-338-3p in MC3T3-E1 cells enhanced the viability and migration ability of MC3T3-E1 cells when cultured with BMSCs-Exo, while suppressing apoptosis. Bioinformatics analysis demonstrated that the target genes of miR-338-3p were predominantly localized at the axon's initiation site, involved in biological processes such as development and growth regulation, and mainly enriched in MAPK and ErbB signaling pathways. CONCLUSION In vitro, BMSCs-Exo exhibits the capacity to enhance proliferation and migration while inhibiting apoptosis of MC3T3-E1 cells, potentially achieved through modulation of KLF3-AS1 and miR-338-3p expression in MC3T3-E1 cells.
Collapse
Affiliation(s)
- Dacheng Liu
- Department of Orthopedics, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, 269 University Road, Tongshan District, Xuzhou, 221100, Jiangsu, China
| | - Xuechao Zhao
- Department of Orthopedics, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, 269 University Road, Tongshan District, Xuzhou, 221100, Jiangsu, China
| | - Qiang Zhang
- Department of Orthopedics, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, 269 University Road, Tongshan District, Xuzhou, 221100, Jiangsu, China
| | - Fei Zhou
- Operating Room, Xuzhou Central Hospital, Xuzhou, 221006, China
| | - Xiangyang Tong
- Department of Orthopedics, Xuzhou Municipal Hospital Affiliated to Xuzhou Medical University, 269 University Road, Tongshan District, Xuzhou, 221100, Jiangsu, China.
| |
Collapse
|
3
|
Vrščaj LA, Marc J, Ostanek B. Interactome of PTH-Regulated miRNAs and Their Predicted Target Genes for Investigating the Epigenetic Effects of PTH (1-34) in Bone Metabolism. Genes (Basel) 2022; 13:genes13081443. [PMID: 36011354 PMCID: PMC9407897 DOI: 10.3390/genes13081443] [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] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/08/2022] [Accepted: 08/09/2022] [Indexed: 11/16/2022] Open
Abstract
Osteoporosis is a metabolic bone disease that mostly affects the elderly. A lot of drugs are available, mostly with an antiresorptive effect but just a few with an osteoanabolic effect, meaning they promote bone building. PTH (1-34) or teriparatide is an osteoanabolic drug, but its efficacy varies between individuals. We performed a literature review and extracted a dataset of 62 microRNAs (miRNAs) from 10 different studies; predicted miRNA target interactions (MTIs) were obtained with the help of four software tools: DIANA, miRWalk, miRDB and TargetScan. With the construction of an interactome of PTH-regulated miRNAs and their predicted target genes, we elucidated miR-146a-5p, miR-551b-5p, miR-205-3p, miR-33a-3p, miR-338-5p as miRNAs with the most interactions and miR-410-3p as the miRNA targeting bone-related pathways with the highest significance. These miRNAs could help in further understanding the mechanism of action of PTH on bone metabolism and osteoporosis. They also have the potential for novel network-based biomarkers for osteoporosis treatment efficacy and safety and as new therapeutic targets.
Collapse
|
4
|
Li M, Zhang S, Ma Y, Yang Y, An R. Role of hsa‑miR‑105 during the pathogenesis of paclitaxel resistance and its clinical implication in ovarian cancer. Oncol Rep 2021; 45:84. [PMID: 33846814 PMCID: PMC8025119 DOI: 10.3892/or.2021.8035] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 01/21/2021] [Indexed: 12/17/2022] Open
Abstract
More than 70% of patients with epithelial ovarian cancer (EOC), one of the leading cause of gynecological cancer-related deaths worldwide, are diagnosed at an advanced stage of the disease. Currently, the mainstay for treatment of advanced EOC is tumor debulking surgery followed by combined platinum- and paclitaxel (PTX)-based chemotherapy. However, most patients eventually develop chemoresistance, which remains a major obstacle to successful treatment. Herein, by using clinical specimens and experimentally induced cell models, we found that the expression levels of hsa-miR-105 were significantly decreased in PTX-resistant EOC tissues and cell lines. Follow-up functional experiments demonstrated that repression of hsa-miR-105 conferred resistance to paclitaxel in EOC cells, whereas restoration of hsa-miR-105 expression in situ via intratumoral injection of hsa-miR-105 micrON™ agomir potentiated in vivo sensitivity to PTX and thereafter significantly inhibited tumor growth in a PTX-challenged xenograft model. Mechanistically, hsa-miR-105 exerted its tumor suppressor function by directly inhibiting the zinc and ring finger 2 (ZNRF2) signaling pathway. Importantly, aberrant expression of hsa-miR-105 in both tumor and circulating samples predicted a poor post-chemotherapy prognosis in EOC patients. These findings collectively suggest that hsa-miR-105 may act as a potent tumor suppressor miRNA during the progression of EOC, likely affecting cell proliferation, invasiveness and chemosensitivity to PTX, and functioning at least in part via inhibition of ZNRF2 signaling. The stability and availability and ease in measurement of circulating hsa-miR-105 make it a valuable diagnostic/prognostic biomarker candidate for chemotherapy of EOC.
Collapse
Affiliation(s)
- Mao Li
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| | - Shun Zhang
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yuan Ma
- Department of Obstetrics and Gynecology, Reproductive Medicine Center, Tangdu Hospital, Air Force Military Medical University, Xi'an, Shaanxi 710038, P.R. China
| | - Yang Yang
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Xi'an Medical University, Xi'an, Shaanxi 710082, P.R. China
| | - Ruifang An
- Department of Obstetrics and Gynecology, First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, P.R. China
| |
Collapse
|
5
|
Pala E, Denkçeken T. Differentially expressed circulating miRNAs in postmenopausal osteoporosis: a meta-analysis. Biosci Rep 2019; 39:BSR20190667. [PMID: 31023966 PMCID: PMC6522747 DOI: 10.1042/bsr20190667] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 04/22/2019] [Accepted: 04/23/2019] [Indexed: 02/06/2023] Open
Abstract
MicroRNAs (miRNAs) have been proven to play a crucial role in postmenopausal osteoporosis (PMO), and studies on their diagnostic value have been increasing. In our study, we aim to identify the key miRNAs in the PMO that might be potential biomarkers. A comprehensive systematic literature search was conducted by searching PubMed, Web of Science, Embase and Cochrane Library databases. In the total of 16 independent miRNA expression studies which contained 327 PMO patients and 328 postmenopausal (PM) healthy control samples, miRNAs were evaluated by using robust rank aggregation (RRA) method. A statistically significant meta-signature of up-regulated hsa-miR-133a-3p (P = 1.38e-03) was determined. Then bioinformatics analysis to recruit putative target genes prediction of hsa-miR-133a-3p and pathway enrichment analysis to reveal what biological processes this miRNA may affect were conducted. It was indicated that pathways were commonly associated with adrenergic signaling in cardiomyocytes, adherens junction, PI3K-Akt signaling pathway and AMPK signaling pathway. Furthermore, STRING and Cytoscape tools were used to visualize the interactions between target genes of hsa-miR-133a-3p. Six genes were detected as hub genes among 576 targets which were CDC42, RHOA, EGFR, VAMP2, PIK3R2 and FN1. After Kyoto Encyclopedia of Genes and Genomes pathway analysis, it was detected that these hub genes were mostly enriched in signaling pathways and cancer. In this meta-analysis, it is stated that circulating hsa-miR-133a-3p may serve as a potential non-invasive biomarker and therapeutic target in PMO.
Collapse
Affiliation(s)
- Elif Pala
- Department of Medical Biology, Faculty of Medicine, SANKO University, Gaziantep, Turkey
| | - Tuba Denkçeken
- Department of Biophysics, Faculty of Medicine, SANKO University, Gaziantep, Turkey
| |
Collapse
|