1
|
Cunha WR, Martin de la Vega M, Rodrigues de Barros P, Espinosa-Diez C. lncRNAs in vascular senescence and microvascular remodeling. Am J Physiol Heart Circ Physiol 2025; 328:H1238-H1252. [PMID: 40251747 DOI: 10.1152/ajpheart.00750.2024] [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: 10/31/2024] [Revised: 11/19/2024] [Accepted: 04/14/2025] [Indexed: 04/21/2025]
Abstract
Long noncoding RNAs (lncRNAs) have emerged as critical regulators of vascular senescence and microvascular remodeling, processes that significantly contribute to the development of age-related diseases in organs such as the kidneys, heart, and lungs. Through mechanisms like chromatin remodeling, transcriptional regulation, and posttranscriptional modifications, lncRNAs modulate gene expression, thereby influencing cellular processes such as apoptosis, inflammation, fibrosis, and angiogenesis. In chronic kidney disease, cardiovascular disease, and pulmonary disorders, lncRNAs play a central role in promoting vascular dysfunction, endothelial cell aging, and fibrosis. This review focuses on how lncRNAs contribute to endothelial dysfunction, fibrosis, and vascular aging, emphasizing their roles in disease progression within the kidneys, heart, and lungs, where lncRNA-mediated vascular changes play a significant role in disease progression. Understanding the interactions between lncRNAs, vascular senescence, and microvascular remodeling offers promising avenues for developing targeted therapeutic strategies to mitigate the impact of aging on vascular health.
Collapse
Affiliation(s)
- Warlley Rosa Cunha
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Maria Martin de la Vega
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States
| | - Paula Rodrigues de Barros
- Department of Pharmacology, Institute of Biomedical Sciences, University of Sao Paulo, Sao Paulo, Brazil
| | - Cristina Espinosa-Diez
- Center for Molecular Medicine & Genetics, Wayne State University School of Medicine, Detroit, Michigan, United States
- Department of Physiology, Wayne State University School of Medicine, Detroit, Michigan, United States
| |
Collapse
|
2
|
Wang Y, Chen G, Ji X, Wong SK, Ekeuku SO, Chin KY. Ejiao as a preventive agent for osteoporosis - a scoping review of current evidence. J Orthop Surg Res 2025; 20:445. [PMID: 40325487 PMCID: PMC12054324 DOI: 10.1186/s13018-025-05759-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 03/26/2025] [Indexed: 05/07/2025] Open
Abstract
Ejiao, a traditional Chinese medicinal product derived from donkey's hide, has long been used to promote blood formation and treat various ailments. Recently, growing pharmacological evidence has suggested that Ejiao and its formulations may also possess bone-protecting properties, making it a potential candidate for preventing and treating osteoporosis. This scoping review aims to summarise the current scientific evidence on the anti-osteoporosis potential of Ejiao and its formulations in osteoporosis prevention. A comprehensive literature search was conducted using PubMed, Web of Science, Scopus, and China National Knowledge Infrastructure up to October 2024. Primary studies published in English or Mandarin, regardless of study design, that investigated the effects of Ejiao on bone in vivo or bone cells in vitro were included. A total of 22 studies were included, comprising five studies on Ejiao alone and 17 studies on Ejiao-based formulations. The findings indicated that Ejiao alone enhanced osteoblast differentiation by increasing alkaline phosphatase synthesis and reducing bone remodelling markers in ovariectomised rats. However, its direct effect on bone mineralisation and density remains uncertain due to the absence of an exogenous mineral source. In contrast, Ejiao-based formulations, such as calcium-Ejiao oral liquid and Donkey-hide glue reinforcing bone oral solution, demonstrated more pronounced bone-protective effects, including improving bone density, enhancing bone repair, and supporting vitamin D metabolism in both animal models and clinical studies. These findings suggest that while Ejiao alone may promote osteoblast activity, its role in osteoporosis management may be more effective when combined with essential minerals. Further long-term studies and human clinical trials are needed to clarify its therapeutic potential and underlying molecular mechanisms.
Collapse
Affiliation(s)
- Yuanzhong Wang
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, 56000, Malaysia
- Chongqing Chemical Industry Vocational College, Changshou, Chongqing, 401220, China
| | - Guiju Chen
- Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, Hubei, 441100, China
| | - Xia Ji
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, 56000, Malaysia
- Hospital of Inner Mongolia Baotou Steel (The Third Affiliated Hospital of Inner Mongolia Medical University), Baotou, Inner Mongolia, 014010, China
- Department of Obstetrics & Gynaecology, Tianjin Medical University General Hospital, Heping, Tianjin, 300070, China
| | - Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, 56000, Malaysia
| | - Sophia Ogechi Ekeuku
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, 56000, Malaysia.
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Level 17, Jalan Yaacob Lahtif, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia.
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Cheras, Kuala Lumpur, 56000, Malaysia.
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Level 17, Jalan Yaacob Lahtif, Bandar Tun Razak, Cheras, Kuala Lumpur, 56000, Malaysia.
| |
Collapse
|
3
|
Dalle Carbonare L, Cominacini M, Trabetti E, Bombieri C, Pessoa J, Romanelli MG, Valenti MT. The bone microenvironment: new insights into the role of stem cells and cell communication in bone regeneration. Stem Cell Res Ther 2025; 16:169. [PMID: 40221779 PMCID: PMC11993959 DOI: 10.1186/s13287-025-04288-4] [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: 11/22/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025] Open
Abstract
Mesenchymal stem cells (MSCs) play a crucial role in bone formation and remodeling. Intrinsic genetic factors and extrinsic environmental cues regulate their differentiation into osteoblasts. Within the bone microenvironment, a complex network of biochemical and biomechanical signals orchestrates bone homeostasis and regeneration. In addition, the crosstalk among MSCs, immune cells, and neighboring cells-mediated by extracellular vesicles and non-coding RNAs (such as circular RNAs and micro RNAs) -profoundly influences osteogenic differentiation and bone remodeling. Recent studies have explored specific signaling pathways that contribute to effective bone regeneration, highlighting the potential of manipulating the bone microenvironment to enhance MSC functionality. The integration of advanced biomaterials, gene editing techniques, and controlled delivery systems is paving the way for more targeted and efficient regenerative therapies. Furthermore, artificial intelligence could improve bone tissue engineering, optimize biomaterial design, and enable personalized treatment strategies. This review explores the latest advancements in bone regeneration, emphasizing the intricate interplay among stem cells, immune cells, and signaling molecules. By providing a comprehensive overview of these mechanisms and their clinical implications, we aim to shed light on future research directions in this rapidly evolving field.
Collapse
Affiliation(s)
- L Dalle Carbonare
- Department of Engineering for the Innovation Medicine, University of Verona, 37100, Verona, Italy
| | - M Cominacini
- Department of Engineering for the Innovation Medicine, University of Verona, 37100, Verona, Italy
| | - E Trabetti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy
| | - C Bombieri
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy
| | - J Pessoa
- Department of Medical Sciences and Institute of Biomedicine-Ibimed, University of Aveiro, 3810 - 193, Aveiro, Portugal
| | - M G Romanelli
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy
| | - M T Valenti
- Department of Neurosciences, Biomedicine and Movement Sciences, University of Verona, 37100, Verona, Italy.
| |
Collapse
|
4
|
Wang C, Mu X, Sun J. Research progress of cycloartane triterpenoids and pharmacological activities. Arch Pharm (Weinheim) 2025; 358:e2400923. [PMID: 40071692 DOI: 10.1002/ardp.202400923] [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: 12/06/2024] [Revised: 01/20/2025] [Accepted: 02/18/2025] [Indexed: 05/13/2025]
Abstract
Cycloartane triterpenoids are widely distributed in the plant kingdom, and there have been reports of hundreds of families containing cycloartane triterpenoids. But the types and content of cycloartane are different among various plants. In recent years, a large amount of cycloartane triterpenoids have been extracted and studied from different plants, and some types of cycloartane triterpenoids exhibit great pharmacological activities in terms of antiaging, antioxidant, anti-inflammatory, anticancer, antiarrhythmic effects, and so on. Herein, we have systematically reviewed these research on the structure of naturally occurring, synthetic, and semisynthetic cycloartane triterpenoids, with particular emphasis on their pharmacological activities.
Collapse
Affiliation(s)
- Chen Wang
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan, China
| | - Xiaodong Mu
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan, China
| | - Jingyong Sun
- School of Pharmaceutical Sciences & Institute of Materia Medica, Shandong First Medical University & Shandong Academy of Medical Sciences, NHC Key Laboratory of Biotechnology Drugs (Shandong Academy of Medical Sciences), Key Laboratory for Rare & Uncommon Diseases of Shandong Province, Jinan, China
| |
Collapse
|
5
|
Li C, Yuan Y, Jia Y, Zhou Q, Wang Q, Jiang X. Cellular senescence: from homeostasis to pathological implications and therapeutic strategies. Front Immunol 2025; 16:1534263. [PMID: 39963130 PMCID: PMC11830604 DOI: 10.3389/fimmu.2025.1534263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 01/15/2025] [Indexed: 02/20/2025] Open
Abstract
Cellular aging is a multifactorial and intricately regulated physiological process with profound implications. The interaction between cellular senescence and cancer is complex and multifaceted, senescence can both promote and inhibit tumor progression through various mechanisms. M6A methylation modification regulates the aging process of cells and tissues by modulating senescence-related genes. In this review, we comprehensively discuss the characteristics of cellular senescence, the signaling pathways regulating senescence, the biomarkers of senescence, and the mechanisms of anti-senescence drugs. Notably, this review also delves into the complex interactions between senescence and cancer, emphasizing the dual role of the senescent microenvironment in tumor initiation, progression, and treatment. Finally, we thoroughly explore the function and mechanism of m6A methylation modification in cellular senescence, revealing its critical role in regulating gene expression and maintaining cellular homeostasis. In conclusion, this review provides a comprehensive perspective on the molecular mechanisms and biological significance of cellular senescence and offers new insights for the development of anti-senescence strategies.
Collapse
Affiliation(s)
- Chunhong Li
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Yixiao Yuan
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| | - YingDong Jia
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Zhou
- Department of Oncology, Suining Central Hospital, Suining, Sichuan, China
| | - Qiang Wang
- Gastrointestinal Surgical Unit, Suining Central Hospital, Suining, Sichuan, China
| | - Xiulin Jiang
- Department of Medicine, Health Cancer Center, University of Florida, Gainesville, FL, United States
| |
Collapse
|
6
|
Zhou Q, Liu J, Qi Y, Hu Y, Li Y, Cong C, Chen Y. Jianpi qingre tongluo prescription alleviates the senescence-associated secretory phenotype with osteoarthritis by regulating STAG1/TP53/P21 signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 337:118953. [PMID: 39423944 DOI: 10.1016/j.jep.2024.118953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/01/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Jianpi Qingre Chubi prescription primarily consists of a compound formula, also known as Huangqin Qingre Chubi Capsules (HQC), which strengthens the spleen and resolves dampness, clear heat, and collaterals. Long-term clinical use has shown that HQC improves joint swelling and pain in patients with osteoarthritis. Mechanistically, we demonstrated that HQC inhibits inflammatory responses, extracellular matrix degradation, and delays chondrocyte senescence. AIM To determine the bioactivity and mechanism of action of Jianpi Qingre Tongluo prescription (HQC) on osteoarthritis (OA). MATERIALS AND METHODS First, the chondroprotective effects of HQC were assessed using histopathology, immunohistochemical staining and protein blotting in an OA rat model. Additionally, we identified key targets for crucial targets of HQC in OA using the Network Pharmacology and Gene Expression Omnibus (GEO) dataset (GSE98918 and GSE152805). In vitro conditions, IL-1β-treated chondrocytes served to study the impact of HQC on OA development and the senescence-associated secretory phenotype (SASP). This was evaluated using a series of approaches, such as flow cytometry assays, and immunofluorescence staining, and then verified by rescue experiments. RESULTS Therapy with HQC attenuated the severity of osteoarthritis (demonstrated by histopathology, OARSI grading scores, and Mankin scores) and SASP factors (as indicated by IL-1β, IL-6, IL-4, IL-37, MMP13, ADAMTS5, COL2A1, and ACAN levels, and apoptotic cell death). HQC might treat osteoarthritis via four important targets (STAG1, TP53, P21, and P16), with the p53 signalling pathway representing one of the main pathways. The HQC acts primarily on chondrocyte clusters. In vitro experiments indicated that STAG1 overexpression accelerates chondrocyte apoptosis, promotes SASP factor expression and extracellular matrix (ECM) degradation, and facilitates OA progression. HQC-containing serum suppressed the expression of the STAG1/TP53/P21 pathway, regulated SASP factors, and restored ECM balance. CONCLUSION Jianpi Qingre Tongluo prescription modulated SASP factors by regulating the STAG1/TP53/P21 signal transduction axis and decelerating cartilage senescence and degradation in patients with OA. Jianpi Qingre Tongluo may be an effective drug candidate.
Collapse
Affiliation(s)
- Qiao Zhou
- Department of Geriatrics, The Second Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China; The First Clinical School of Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Jian Liu
- Department of Rheumatism Immunity, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Yajun Qi
- The First Clinical School of Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Yuedi Hu
- The First Clinical School of Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Yang Li
- The First Clinical School of Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Chengzhi Cong
- The First Clinical School of Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| | - Yiming Chen
- The First Clinical School of Medicine, Anhui University of Chinese Medicine, Hefei, Anhui, China.
| |
Collapse
|
7
|
Alavimanesh S, Nayerain Jazi N, Choubani M, Saeidi F, Afkhami H, Yarahmadi A, Ronaghi H, Khani P, Modarressi MH. Cellular senescence in the tumor with a bone niche microenvironment: friend or foe? Clin Exp Med 2025; 25:44. [PMID: 39849183 PMCID: PMC11759293 DOI: 10.1007/s10238-025-01564-8] [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: 10/30/2024] [Accepted: 01/08/2025] [Indexed: 01/25/2025]
Abstract
Cellular senescence is understood to be a biological process that is defined as irreversible growth arrest and was originally recognized as a tumor-suppressive mechanism that prevents further propagation of damaged cells. More recently, cellular senescence has been shown to have a dual role in prevention and tumor promotion. Senescent cells carry a senescence-associated secretory phenotype (SASP), which is altered by secretory factors including pro-inflammatory cytokines, chemokines, and other proteases, leading to the alteration of the tissue microenvironment. Though senescence would eventually halt the growth of cancerous potential cells, SASP contributes to the tumor environment by promoting inflammation, matrix remodeling, and tumor cell invasion. The paradox of tumor prevention/promotion is particularly relevant to the bone niche tumor microenvironment, where longer-lasting, chronic inflammation promotes tumor formation. Insights into a mechanistic understanding of cellular senescence and SASP provide the basis for targeted therapies, such as senolytics, which aim to eliminate senescent cells, or SASP inhibitors, which would eliminate the tumor-promoting effects of senescence. These therapeutic interventions offer significant clinical implications for treating cancer and healthy aging.
Collapse
Affiliation(s)
- Sajad Alavimanesh
- Student Research Committee, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Negar Nayerain Jazi
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Bacteriology and Virology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Maedeh Choubani
- School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farzane Saeidi
- Department of Medical Genetics, School of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| | - Aref Yarahmadi
- Department of Biology, Khorramabad Branch, Islamic Azad University, Khorramabad, Iran
| | - Hossein Ronaghi
- Department of Orthopedic, Faculty of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Pouria Khani
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| | - Mohammad Hossein Modarressi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
8
|
Liu H, Dong H, Zhou Y, Jin M, Hao H, Yuan Y, Jia H. Nonlinear relationship between cardiometabolic index and bone mineral density in U.S. adults: the mediating role of percent body fat. Sci Rep 2024; 14:22449. [PMID: 39342025 PMCID: PMC11439067 DOI: 10.1038/s41598-024-73427-3] [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: 06/30/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Both lipid metabolism and obesity are crucial factors in osteoporosis, influencing the relevance of the cardiometabolic index (CMI), a new body fat index incorporating obesity and lipid metrics. Our study aims to explore the relationship between CMI and lumbar spine bone mineral density (BMD) and the mediating role of body fat percentage. METHODS Utilizing the National Health and Nutrition Examination Survey (NHANES) data from 2011 to 2018, we conducted a cross-sectional analysis. We employed multiple linear regression models, subgroup analyses, generalized additive models (GAM), smooth curve fitting, and mediation analysis to evaluate the linear and nonlinear relationships between CMI and lumbar spine BMD. RESULTS The study involved 5,124 participants with an average lumbar spine BMD of 1.03 ± 0.15 g/cm2. We identified a negative correlation between CMI and lumbar spine BMD (β = -0.015; 95% CI: -0.023, -0.008). Nonlinear associations were evident, with inflection points at CMI values of 1.12 and 2.86. Subgroup analyses showed consistent negative correlations across all categories without significant differences (p for interaction > 0.05). Moreover, body fat percentage negatively correlated with BMD (β = -0.005; 95% CI: -0.006, -0.004) and mediated 9.41% of the relationship between CMI and BMD. CONCLUSION Increased CMI levels are associated with lower lumbar spine BMD, with body fat percentage significantly mediating this relationship. This underscores the importance of managing body composition in the context of bone health, highlighting CMI's potential utility in osteoporosis risk assessment.
Collapse
Affiliation(s)
- Heng Liu
- Department of Urology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Huqiang Dong
- School of Public Health, Ningxia Medical University, Yinchuan, 750004, Ningxia, China
| | - Yu Zhou
- Department of Urology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Mingchu Jin
- Department of Urology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Haidong Hao
- Department of Urology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Yutang Yuan
- Department of Urology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China
| | - Hongtao Jia
- Department of Urology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, People's Republic of China.
| |
Collapse
|
9
|
Wu C, Wang C, Xiao B, Li S, Sheng Y, Wang Q, Tao J, Zhang Y, Jiang X. Integration analysis of lncRNA and mRNA expression data identifies DOCK4 as a potential biomarker for elderly osteoporosis. BMC Med Genomics 2024; 17:70. [PMID: 38443923 PMCID: PMC10916189 DOI: 10.1186/s12920-024-01837-3] [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: 01/22/2024] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND We aimed to identify some potential biomarkers for elderly osteoporosis (OP) by integral analysis of lncRNA and mRNA expression data. METHODS A total of 8 OP cases and 5 healthy participants were included in the study. Fasting peripheral venous blood samples were collected from individuals, and total RNA was extracted. RNA-seq library was prepared and sequenced on the Illumina HiSeq platform. Differential gene expression analysis was performed using "DESeq2" package in R language. Functional enrichment analysis was conducted using the "clusterProfiler" package, and the cis- and trans-regulatory relationships between lncRNA and target mRNA were analyzed by the lncTar software. A protein-protein interaction (PPI) network was constructed using the STRING database, and hub genes were identified through the MCODE plugin in Cytoscape. RESULTS We identified 897 differentially expressed lncRNAs (DELs) and 1366 differentially expressed genes (DEGs) between normal and OP samples. After co-expression network analysis and cis-trans regulatory genes analysis, we identified 69 candidate genes regulated by lncRNAs. Then we further screened 7 genes after PPI analysis. The target gene DOCK4, trans-regulated by two lncRNAs, was found to be significantly upregulated in OP samples. Additionally, 4 miRNAs were identified as potential regulators of DOCK4. The potential diagnostic value of DOCK4 and its two trans-regulatory lncRNAs was supported by ROC analysis, indicating their potential as biomarkers for OP. CONCLUSION DOCK4 is a potential biomarker for elderly osteoporosis diagnostic. It is identified to be regulated by two lncRNAs and four miRNAs.
Collapse
Affiliation(s)
- Chengai Wu
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Chao Wang
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Bin Xiao
- Department of Spine Surgery, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Shan Li
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yueyang Sheng
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Qianqian Wang
- Department of Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China
| | - Jianfeng Tao
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Yanzhuo Zhang
- Department of Molecular Orthopaedics, National Center for Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, China
| | - Xu Jiang
- Department of Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, No. 31, Xinjiekou East Street, Xicheng District, Beijing, 100035, China.
| |
Collapse
|