1
|
Xu X, Gong C, Wang Y, Yin Z, Wang X, Wu X, Fang Z, Wei S. FOXF1 promotes ovarian cancer metastasis by facilitating HMGA2-mediated USP30-dependent S100A6 deubiquitination. Biochim Biophys Acta Mol Basis Dis 2024; 1871:167633. [PMID: 39694080 DOI: 10.1016/j.bbadis.2024.167633] [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: 02/29/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
Ovarian cancer is the most common type of gynecological malignant tumor, with the highest mortality rate among female genital malignant tumors. In this study, we initially identified forkhead box F1 (FOXF1) as a potential prognostic biomarker of ovarian cancer through bioinformatics analysis. FOXF1 expression was higher in ovarian cancer tissue samples and served as an unfavorable prognostic factor. In vitro and in vivo experiments demonstrated that FOXF1 enhanced ovarian cancer cell migration and tumor dissemination. Chromatin immunoprecipitation-polymerase chain reaction and luciferase assays revealed that FOXF1 bound directly to the high-mobility group AT-hook 2 (HMGA2) promoter and significantly induced its transcriptional activity. Subsequent co-immunoprecipitation and mass spectrometry analyses demonstrated that HMGA2 stabilized S100 calcium-binding protein A6 (S100A6) protein through recruitment of the deubiquitinase, ubiquitin-specific peptidase 30 (USP30), thereby inhibiting S100A6 degradation. Rescue experiments further illustrated that FOXF1 induced ovarian cancer cell mobility in an HMGA2/S100A6-dependent manner. Additionally, FOXF1, HMGA2, USP30, and S100A6 were clinically relevant in patients with ovarian cancer. This is the first study to reveal the molecular mechanisms underlying FOXF1-mediated ovarian cancer metastasis and demonstrate that FOXF1 represents a potential therapeutic target in patients with metastatic ovarian cancer.
Collapse
Affiliation(s)
- Xi Xu
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Chaoju Gong
- Central Laboratory, The Municipal Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China.
| | - Yunfeng Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, China.
| | - Zhidong Yin
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Xiaogang Wang
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| | - Xuebiao Wu
- Center for Molecular Pathology, Department of Pathophysiology, Gannan Medical University, Ganzhou 341000, China.
| | - Zejun Fang
- Central Laboratory, Sanmen People's Hospital, Sanmen 317100, China.
| | - Shumei Wei
- Department of Pathology, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China.
| |
Collapse
|
2
|
Lu X, Zhao Y, Peng X, Lu C, Wu Z, Xu H, Qin Y, Xu Y, Wang Q, Hao Y, Geng D. Comprehensive Overview of Interface Strategies in Implant Osseointegration. ADVANCED FUNCTIONAL MATERIALS 2024. [DOI: 10.1002/adfm.202418849] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Indexed: 01/05/2025]
Abstract
AbstractWith the improvement of implant design and the expansion of application scenarios, orthopedic implants have become a common surgical option for treating fractures and end‐stage osteoarthritis. Their common goal is rapidly forming and long‐term stable osseointegration. However, this fixation effect is limited by implant surface characteristics and peri‐implant bone tissue activity. Therefore, this review summarizes the strategies of interface engineering (osteogenic peptides, growth factors, and metal ions) and treatment methods (porous nanotubes, hydrogel embedding, and other load‐release systems) through research on its biological mechanism, paving the way to achieve the adaptation of both and coordination between different strategies. With the transition of the osseointegration stage, interface engineering strategies have demonstrated varying therapeutic effects. Especially, the activity of osteoblasts runs almost through the entire process of osseointegration, and their physiological activities play a dominant role in bone formation. Furthermore, diseases impacting bone metabolism exacerbate the difficulty of achieving osseointegration. This review aims to assist future research on osseointegration engineering strategies to improve implant‐bone fixation, promote fracture healing, and enhance post‐implantation recovery.
Collapse
Affiliation(s)
- Xiaoheng Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuhu Zhao
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Xiaole Peng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
- Department of Orthopedics The First Affiliated Hospital of Chongqing Medical University 1 Youyi Street Chongqing 400016 China
| | - Chengyao Lu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Zebin Wu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Hao Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yi Qin
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yaozeng Xu
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Qing Wang
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| | - Yuefeng Hao
- Orthopedics and Sports Medicine Center The Affiliated Suzhou Hospital of Nanjing Medical University 242 Guangji Street Suzhou Jiangsu 215006 China
| | - Dechun Geng
- Department of Orthopedics The First Affiliated Hospital of Soochow University 188 Shizi Street Suzhou Jiangsu 215006 China
| |
Collapse
|
3
|
Wang J, Zhen C, Zhang G, Yang Z, Shang P. A 0.2 T-0.4 T Static Magnetic Field Improves the Bone Quality of Mice Subjected to Hindlimb Unloading and Reloading Through the Dual Regulation of BMSCs via Iron Metabolism. Int J Mol Sci 2024; 25:13136. [PMID: 39684847 DOI: 10.3390/ijms252313136] [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: 09/21/2024] [Revised: 11/07/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Osteoporosis is the most prevalent metabolic bone disease, especially when aggravated by aging and long-term bed rest of various causes and also when coupled with astronauts' longer missions in space. Research on the use of static magnetic fields (SMFs) has been progressing as a noninvasive method for osteoporosis due to the complexity of the disease, the inconsistency of the effects of SMFs, and the ambiguity of the mechanism. This paper studied the effects of mice subjected to hindlimb unloading (UL, HLU) and reloading by the 0.2 T-0.4 T static magnetic field (MMF). Primary bone marrow mesenchymal stem cells (BMSCs) were extracted to explore the mechanism. Eight-week-old male C57BL/6 mice were used as an osteoporosis model by HLU for four weeks. The HLU recovery period (reloading, RL) was carried out on all FVEs and recovered in the geomagnetic field (45-64 μT, GMF) and MMF, respectively, for 12 h/d for another 4 weeks. The tibia and femur of mice were taken; also, the primary BMSCs were extracted. MMF promoted the recovery of mechanical properties after HLU, increased the number of osteoblasts, and decreased the number of adipocytes in the bone marrow. MMF decreased the total iron content and promoted the total calcium content in the tibia. In vitro experiments showed that MMF promoted the osteogenic differentiation of BMSCs and inhibited adipogenic differentiation, which is related to iron metabolism, the Wnt/β-catenin pathway, and the PPARγ pathway. MMF accelerated the improvement in bone metabolism and iron metabolism in RL mice to a certain extent, which improved the bone quality of mice. MMF mainly promoted osteogenic differentiation and reduced the adipogenic differentiation of BMSCs, which provides a reliable research direction and transformation basis for the osteoporosis of elderly, bedridden patients and astronauts.
Collapse
Affiliation(s)
- Jianping Wang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Chenxiao Zhen
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Gejing Zhang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhouqi Yang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
| | - Peng Shang
- School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Key Laboratory for Space Bioscience and Biotechnology, Institute of Special Environmental Biophysics, School of Life Sciences, Northwestern Polytechnical University, Xi'an 710072, China
- Research & Development Institute, Northwestern Polytechnical University, Shenzhen 518057, China
| |
Collapse
|
4
|
Shi K, Wei J, Chen J. MiR-223-3p Promotes Osteoporosis Progression by Repressing Osteogenic Differentiation via Targeting FHL1/Wnt/β-catenin Signaling. Cell Biochem Biophys 2024:10.1007/s12013-024-01579-0. [PMID: 39613991 DOI: 10.1007/s12013-024-01579-0] [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] [Accepted: 09/22/2024] [Indexed: 12/01/2024]
Abstract
The aim of this research was to unveil the potential along with potential mechanism of miR-223-3p in osteoporosis. RT-qPCR together with western blot was implemented to examine miR-223-3p, FHL1, along with osteogenic markers levels during bone marrow mesenchymal stem cells (BMSCs) differentiation. The ALP activity staining along with alizarin red staining (ARS) were implemented to assess ALP activity as well as the mineralization ability of BMSCs. Binding sequences for miR-223-3p and FHL1 from starBase website were validated through dual-luciferase reporter gene assay. MiR-223-3p was down-regulated in BMSCs during osteoblasts differentiation, and miR-223-3p elevation hindered BMSCs' osteogenic differentiation. FHL1 belonged to the target mRNA of miR-223-3p. FHL1 presented up-regulation in BMSCs during osteoblasts differentiation. More importantly, FHL1 expression was negative modulated by miR-223-3p in BMSCs during osteoblasts differentiation, and FHL1 elevation could inverse the inhibited BMSCs' osteogenic differentiation modulated by miR-223-3p elevation. Furthermore, miR-223-3p elevation repressed the Wnt/β-catenin pathway activity in lithium chloride-treated BMSCs, and FHL1 overexpression counteracted the inhibitory effect of the Wnt/β-catenin pathway caused by miR-223-3p up-regulation. Collectively, miR-223-3p accelerates osteoporosis progression by repressing osteogenic differentiation through targeting FHL1/Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Kairi Shi
- Department of Orthopedic Surgery, Ningbo No.6 Hospital, Ningbo, Zhejiang, 315040, China
| | - Junyu Wei
- Department of Orthopedic Surgery, Ningbo No.6 Hospital, Ningbo, Zhejiang, 315040, China
| | - Jianming Chen
- Department of Orthopedic Surgery, Ningbo No.6 Hospital, Ningbo, Zhejiang, 315040, China.
| |
Collapse
|
5
|
Fan L, Zhang L, Zhang X, Wei W, Liu Z. Long Noncoding RNA EMX2-AS Facilitates Osteoblast Differentiation and Bone Formation by Inhibiting EMX2 Protein Translation and Activating Wnt/ β-Catenin Pathway. Stem Cells Int 2024; 2024:4397807. [PMID: 39628661 PMCID: PMC11614513 DOI: 10.1155/sci/4397807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 06/24/2024] [Accepted: 11/11/2024] [Indexed: 12/06/2024] Open
Abstract
Long noncoding RNAs (lncRNAs), as a potentially new and crucial element of biological regulation, have gained widespread attention in recent years. Our previous work identified lncRNA empty spiracles homeobox 2 antisence (EMX2-AS) was significantly increased during the osteoblast differentiation of mesenchymal stem cells (MSCs). Overexpression of lncRNA EMX2-AS promoted osteogenesis in vitro and enhanced heterotopic bone formation in vivo, whereas lncRNA EMX2-AS knockdown had the opposite effect. EMX2 could negatively regulate the osteoblast differentiation of MSCs. lncRNA EMX2-AS was 80% expressed in the cytoplasm during osteoblast differentiation in MSCs. Mechanistic analysis revealed that lncRNA EMX2-AS acts as a positive regulator of osteogenic differentiation through interaction with EMX2 and suppression of its expression at the translational level and Wnt/β-catenin pathway is involved in lncRNA EMX2-AS/EMX2 regulated osteogenic differentiation. Our findings not only provide new targets for the treatment of diseases related to osteoblast differentiation disruption but also enrich the understanding of the regulation mechanisms of lncRNA during stem cell differentiation.
Collapse
Affiliation(s)
- Linyuan Fan
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| | - Li Zhang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| | - Xin Zhang
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| | - Wei Wei
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| | - Zhaohui Liu
- Department of Gynecology, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Maternal and Child Health Care Hospital Beijing, Beijing 100026, China
| |
Collapse
|
6
|
Xu J, Huang Z, Shi S, Xia J, Chen G, Zhou K, Zhang Y, Bian C, Shen Y, Yin X, Lu L, Gu H. Glial maturation factor-β deficiency prevents oestrogen deficiency-induced bone loss by remodelling the actin network to suppress adipogenesis of bone marrow mesenchymal stem cells. Cell Death Dis 2024; 15:829. [PMID: 39543090 PMCID: PMC11564563 DOI: 10.1038/s41419-024-07234-z] [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: 07/27/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
An imbalance between the adipogenesis and osteogenesis of bone marrow mesenchymal stem cells (BMSCs) is considered the basic pathogenesis of osteoporosis. Although actin cytoskeleton remodelling plays a crucial role in the differentiation of BMSCs, the role of actin cytoskeleton remodelling in the adipogenesis of BMSCs and postmenopausal osteoporosis (PMOP) has remained elusive. Glia maturation factor-beta (GMFB) has a unique role in remodelling the polymerization/depolymerization cycles of actin. We observed that GMFB expression was increased in bone tissue from both ovariectomized (OVX) rats and PMOP patients. GMFB knockout inhibited the accumulation of bone marrow adipocytes and increased bone mass in the OVX rat model. The inhibition of adipocyte differentiation in GMFB knockout BMSCs was mediated via actin cytoskeleton remodelling and the Ca2+-calcineurin-NFATc2 axis. Furthermore, we found that GMFB shRNA treatment in vivo had favourable effects on osteoporosis induced by OVX. Together, these findings suggest a pathological association of the GMFB with PMOP and highlight the potential of the GMFB as a therapeutic target for osteoporosis patients.
Collapse
Affiliation(s)
- Jun Xu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Zhongyue Huang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Si Shi
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of medicine, Shanghai, PR China
| | - Jiangni Xia
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Guangnan Chen
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Kaifeng Zhou
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Yiming Zhang
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Chong Bian
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China
| | - Yuqin Shen
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of medicine, Shanghai, PR China
| | - Xiaofan Yin
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China.
| | - Lixia Lu
- Department of Rehabilitation, Tongji Hospital Affiliated to Tongji University, Tongji University School of medicine, Shanghai, PR China.
- Department of Biochemistry and Molecular Biology, Tongji University School of medicine, Shanghai, PR China.
| | - Huijie Gu
- Department of Orthopedics, Minhang Hospital, Fudan University, Shanghai, PR China.
| |
Collapse
|
7
|
Coppola U, Saha B, Kenney J, Waxman JS. A Foxf1-Wnt-Nr2f1 cascade promotes atrial cardiomyocyte differentiation in zebrafish. PLoS Genet 2024; 20:e1011222. [PMID: 39495809 PMCID: PMC11563408 DOI: 10.1371/journal.pgen.1011222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 11/14/2024] [Accepted: 10/21/2024] [Indexed: 11/06/2024] Open
Abstract
Nr2f transcription factors (TFs) are conserved regulators of vertebrate atrial cardiomyocyte (AC) differentiation. However, little is known about the mechanisms directing Nr2f expression in ACs. Here, we identified a conserved enhancer 3' to the nr2f1a locus, which we call 3'reg1-nr2f1a (3'reg1), that can promote Nr2f1a expression in ACs. Sequence analysis of the enhancer identified putative Lef/Tcf and Foxf TF binding sites. Mutation of the Lef/Tcf sites within the 3'reg1 reporter, knockdown of Tcf7l1a, and manipulation of canonical Wnt signaling support that Tcf7l1a is derepressed via Wnt signaling to activate the transgenic enhancer and promote AC differentiation. Similarly, mutation of the Foxf binding sites in the 3'reg1 reporter, coupled with gain- and loss-of-function analysis supported that Foxf1 promotes expression of the enhancer and AC differentiation. Functionally, we find that Wnt signaling acts downstream of Foxf1 to promote expression of the 3'reg1 reporter within ACs and, importantly, both Foxf1 and Wnt signaling require Nr2f1a to promote a surplus of differentiated ACs. CRISPR-mediated deletion of the endogenous 3'reg1 abrogates the ability of Foxf1 and Wnt signaling to produce surplus ACs in zebrafish embryos. Together, our data support that downstream members of a conserved regulatory network involving Wnt signaling and Foxf1 function on a nr2f1a enhancer to promote AC differentiation in the zebrafish heart.
Collapse
Affiliation(s)
- Ugo Coppola
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Bitan Saha
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Jennifer Kenney
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
| | - Joshua S. Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Developmental Biology Division, Cincinnati Children’s Hospital Medical Center, Cincinnati, Ohio, United States of America
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, Ohio, United States of America
| |
Collapse
|
8
|
Peng K, Xia RP, Zhao F, Xiao Y, Ma TD, Li M, Feng Y, Zhou CG. ALKBH5 facilitates the progression of infantile hemangioma by increasing FOXF1 expression in a m 6A-YTHDF2 dependent manner to activate HK-2 signaling. Mol Cell Biochem 2024; 479:3153-3166. [PMID: 38306011 DOI: 10.1007/s11010-024-04936-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 01/08/2024] [Indexed: 02/03/2024]
Abstract
Alkylation repair homolog protein 5 (ALKBH5) is reported to participate in infantile hemangioma (IH) progression. However, the underlying mechanism of ALKBH5 in IH remains unclear. Using qRT-PCR and Western blotting, ALKBH5, forkhead box F1 (FOXF1) and hexokinase 2 (HK-2) expressions in IH tissues and IH-derived endothelial cells XPTS-1 were assessed. The Me-RIP assay was used to analyze FOXF1 m6A level. CCK8, colony formation, flow cytometry and transwell assays were employed to determine IH cell viability, proliferation, apoptosis, migration and invasion. The interactions between YTH (YT521-B homology) domain 2 (YTHDF2), FOXF1 and HK-2 were analyzed by RIP, dual luciferase reporter gene assay and/or ChIP assay. The in vivo IH growth was evaluated in immunocompromised mice. FOXF1 was overexpressed in IH tissues, and its silencing inhibited IH cell proliferation, migration and invasion whereas promoting cell apoptosis in vitro. ALKBH5 upregulation facilitated FOXF1 mRNA stability and expression in IH cells in a m6A-YTHDF2-dependent manner. FOXF1 downregulation reversed the impact of ALKBH5 upregulation on IH cellular phenotypes. It also turned out that FOXF1 positively regulated HK-2 expression in IH cells through interacting with the HK-2 promoter. HK-2 upregulation abolished FOXF1 knockdown's inhibition on IH cell aggressive behaviors. ALKBH5 or FOXF1 silencing suppressed IH tumor development via HK-2 signaling in immunocompromised mice. ALKBH5 promoted FOXF1 expression m6A-YTHDF2 dependently, which in turn elevated HK-2 expression, thereby accelerating IH development.
Collapse
Affiliation(s)
- Kun Peng
- Department of Fetal and Neonatal Surgery, Hunan Children's Hospital, No.86, Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, People's Republic of China
| | - Ren-Peng Xia
- Department of Fetal and Neonatal Surgery, Hunan Children's Hospital, No.86, Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, People's Republic of China
| | - Fan Zhao
- Department of Fetal and Neonatal Surgery, Hunan Children's Hospital, No.86, Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, People's Republic of China
| | - Yong Xiao
- Department of Fetal and Neonatal Surgery, Hunan Children's Hospital, No.86, Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, People's Republic of China
| | - Ti-Dong Ma
- Department of Fetal and Neonatal Surgery, Hunan Children's Hospital, No.86, Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, People's Republic of China
| | - Ming Li
- Department of Fetal and Neonatal Surgery, Hunan Children's Hospital, No.86, Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, People's Republic of China
| | - Yong Feng
- Department of Fetal and Neonatal Surgery, Hunan Children's Hospital, No.86, Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, People's Republic of China
| | - Chong-Gao Zhou
- Department of Fetal and Neonatal Surgery, Hunan Children's Hospital, No.86, Ziyuan Road, Yuhua District, Changsha, 410007, Hunan, People's Republic of China.
| |
Collapse
|
9
|
Qu Z, Zhao S, Zhang Y, Wang X, Yan L. Natural Compounds for Bone Remodeling: Targeting osteoblasts and relevant signaling pathways. Biomed Pharmacother 2024; 180:117490. [PMID: 39332184 DOI: 10.1016/j.biopha.2024.117490] [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: 06/23/2024] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 09/29/2024] Open
Abstract
In the process of bone metabolism and bone remodeling, bone marrow mesenchymal stem cells (BM-MSCs) differentiate into osteoblasts (OBs) under certain conditions to enable the formation of new bone, and normal bone reconstruction and pathological bone alteration are closely related to the differentiation and proliferation functions of OBs. Osteogenic differentiation of BM-MSCs involves multiple signaling pathways, which function individually but interconnect intricately to form a complex signaling regulatory network. Natural compounds have fewer adverse effects than chemically synthesized drugs, optimize bone health, and are more suitable for long-term use. In this paper, we focus on OBs, summarize the current research progress of signaling pathways related to OBs differentiation, and review the molecular mechanisms by which chemically synthesized drugs with potential anti-osteoporosis properties regulate OBs-mediated bone formation.
Collapse
Affiliation(s)
- Zechao Qu
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Songchuan Zhao
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yong Zhang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Xiaohao Wang
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Liang Yan
- Department of Spinal Surgery, Honghui Hospital of Xi'an Jiaotong University, Xi'an, China.
| |
Collapse
|
10
|
Li D, Cao C, Li Z, Chang Z, Cai P, Zhou C, Liu J, Li K, Du B. Icariside II protects from marrow adipose tissue (MAT) expansion in estrogen-deficient mice by targeting S100A16. J Mol Endocrinol 2024; 73:e240020. [PMID: 39101576 PMCID: PMC11466200 DOI: 10.1530/jme-24-0020] [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: 03/11/2024] [Accepted: 08/05/2024] [Indexed: 08/06/2024]
Abstract
Icariside II, a flavonoid glycoside, is the main component found invivo after the administration of Herba epimedii and has shown some pharmacological effects, such as prevention of osteoporosis and enhancement of immunity. Increased levels of marrow adipose tissue are associated with osteoporosis. S100 calcium-binding protein A16 (S100A16) promotes the differentiation of bone marrow mesenchymal stem cells (BMSCs) into adipocytes. This study aimed to confirm the anti-lipidogenesis effect of Icariside II in the bone marrow by inhibiting S100A16 expression. We used ovariectomy (OVX) and BMSC models. The results showed that Icariside II reduced bone marrow fat content and inhibited BMSCs adipogenic differentiation and S100A16 expression, which correlated with lipogenesis. Overexpression of S100A16 eliminated the inhibitory effect of Icariside II on lipid formation. β-catenin participated in the regulation adipogenesis mediated by Icariside II/S100A16 in the bone. In conclusion, Icariside II protects against OVX-induced bone marrow adipogenesis by downregulating S100A16, in which β-catenin might also be involved.
Collapse
Affiliation(s)
- Dong Li
- Department of Orthopedics, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chenhao Cao
- Department of Orthopedics, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhuofan Li
- Department of Orthopedics, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Zhiyong Chang
- Department of Orthopedics, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Ping Cai
- Department of Orthopedics, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Chenxi Zhou
- Department of Orthopedics, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Jun Liu
- Department of Orthopedics, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Kaihua Li
- Department of Orthopedics, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| | - Bin Du
- Department of Orthopedics, The Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing, Jiangsu, China
| |
Collapse
|
11
|
Liu L, Luo S, Li Q, Huang K, Jiang Y, Zeng L, Lan X, Li Q, Xiao J. Role of Wnt5a in modulation of osteoporotic adipose-derived stem cells and osteogenesis. Cell Prolif 2024:e13747. [PMID: 39288944 DOI: 10.1111/cpr.13747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 08/28/2024] [Accepted: 08/30/2024] [Indexed: 09/19/2024] Open
Abstract
Osteoporosis, a condition marked by the deterioration of bone microarchitecture and increased facture risk, arises from a disruption in bone metabolism, with osteoclasts surpassing osteoblasts in bone resorption versus formation. The Wnt signalling pathway, a key regulator of bone maintenance, remains partially understood in osteoporosis. Our research delves into the role of Wnt-related molecules in this disease. In osteoporotic adipose-derived stem cells (OP-ASCs), we detected a significant decrease in Ctnnb1 and Frizzled-6 (Fzd6), contrasted by an increase in Gsk-3β and Wnt5a. Activation of the Wnt pathway by LiCl resulted in elevated Ctnnb1 and Fzd6, but decreased Gsk-3β and Wnt5a levels, promoting OP-ASCs' bone-formation capacity. In contrast, inhibition of this pathway by DKK-1 led to diminished Ctnnb1 and Fzd6, and increased Gsk-3β and Wnt5a, adversely affecting osteogenesis. Furthermore, our findings show that overexpressing Wnt5a impedes, while silencing it enhances the bone-forming capability of OP-ASCs. In a cranial bone defect model, the implantation of Wnt5a-silenced OP-ASCs with biphasic calcium phosphate scaffolds significantly promoted new bone formation. These observations indicated a repression of the canonical Wnt pathway and a stimulation of the non-canonical pathway in OP-ASCs. Silencing Wnt5a increased the osteogenic and regenerative abilities of OP-ASCs. Our study suggests targeting Wnt5a could be a promising strategy for enhancing bone regeneration in post-menopausal osteoporosis.
Collapse
Affiliation(s)
- Lin Liu
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
| | - Shihong Luo
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
| | - Qiumei Li
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
| | - Kui Huang
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
| | - Yuan Jiang
- Medical Service Center of Sichuan Province, Chengdu, China
| | - Lu Zeng
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
| | - Xiaorong Lan
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
| | - Qing Li
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
| | - Jingang Xiao
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- Luzhou Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Luzhou, China
- Department of Oral Implantology, The Affiliated Stomatological Hospital, Southwest Medical University, Luzhou, China
- Department of Oral and Maxillofacial Surgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
12
|
Lai M, Chen X, Feng J, Ruan Z, Lin J. Morinda officinalis polysaccharide boosts osteogenic differentiation of bone marrow mesenchymal stem cells by Wnt/β-catenin signaling. Am J Transl Res 2024; 16:4492-4503. [PMID: 39398614 PMCID: PMC11470318 DOI: 10.62347/wmli2601] [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: 04/28/2024] [Accepted: 08/04/2024] [Indexed: 10/15/2024]
Abstract
OBJECTIVES To investigate the role of Morinda officinalis polysaccharide (MOP) in the protein expression of the Wnt/β-catenin signaling cascade during the osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs), and to elucidate the mechanisms by which MOP enhances osteogenic differentiation at the cellular level. METHODS BMSCs were isolated and cultured using the whole bone marrow adherence method, followed by flow cytometry for the detection of BMSC marker antigens. Two groups were prepared: a low-dose MOP (L-MOP, 10 µg/mL) group and a high-dose MOP (H-MOP, 40 µg/mL) group. MTT assays and cell clone formation assays were performed to evaluate the effects of different MOP doses on BMSC proliferation. Alizarin red staining (ARS) and alkaline phosphatase (ALP) staining were conducted to assess the impact of varying MOP doses on nodule calcification and ALP activity in BMSCs. Additionally, western blot assays were carried out to determine the effects of different MOP concentrations on the expression levels of osteogenesis-related factors and Wnt/β-catenin pathway proteins in BMSCs. RESULTS Highly purified BMSCs were successfully extracted. Subsequent assays demonstrated that BMSCs exhibited enhanced proliferation at all MOP doses, particularly at the H-MOP dose, compared to the control group. Both L-MOP and H-MOP increased calcium content and ALP activity in BMSCs, as well as elevated the expression of osteogenic factors and Wnt/β-catenin pathway proteins compared to the blank control group. However, the addition of Dickkopf-1 (DKK1) significantly reduced BMSC proliferation and osteogenic differentiation compared to the H-MOP group. CONCLUSIONS MOP can enhance BMSC proliferation and osteogenic differentiation by activating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Manxiang Lai
- Department of Pharmacy, Guangdong Food and Drug Vocational CollegeGuangzhou 510520, Guangdong, China
| | - Xia Chen
- Department of Nursing, Guangdong Food and Drug Vocational CollegeGuangzhou 510520, Guangdong, China
| | - Juan Feng
- Department of Health Management and Biotechnology, Guangdong Food and Drug Vocational CollegeGuangzhou 510520, Guangdong, China
| | - Zhiyan Ruan
- Department of Pharmacy, Guangdong Food and Drug Vocational CollegeGuangzhou 510520, Guangdong, China
| | - Jiwei Lin
- Prevention and Treatment Center, Shenzhen Hospital of Traditional Chinese MedicineShenzhen 518000, Guangdong, China
| |
Collapse
|
13
|
Wu Y, Lyu Z, Hu F, Yang L, Yang K, Chen M, Wang Y. A chondroitin sulphate hydrogel with sustained release of SDF-1α for extensive cartilage defect repair through induction of cell homing and promotion of chondrogenesis. J Mater Chem B 2024; 12:8672-8687. [PMID: 39115288 DOI: 10.1039/d4tb00624k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Articular cartilage damage represents a prevalent clinical disease in orthopedics, with its regeneration and repair constituting a central focus in ongoing research endeavors. While hydrogel technology has achieved notable progress in the field of cartilage regeneration, addressing the repair of larger cartilage defects remains a significant and formidable challenge. In pursuit of achieving the repair of extensive cartilage defects, this study designed a polydopamine-modified chondroitin sulfate hydrogel loaded with SDF-1α (P-SCMA). This hydrogel, capable of directly providing glycosaminoglycans (GAGs), served as a platform for carrying growth factors and attracting mesenchymal stem cells for the in situ reconstruction of extensive cartilage defects. The results indicate that the P-SCMA hydrogel is capable of not only directly providing GAGs but also sustainably releasing SDF-1α. In the early stages, it promotes cell adhesion and proliferation and induces cell homing, while in the later stages, it further induces chondrogenesis by inhibiting the Wnt/β-catenin pathway. This bioactive hydrogel, which possesses the functions of providing GAGs, promoting cell proliferation, inducing cell homing and chondrogenesis, is capable of promoting cartilage repair in multiple ways, providing new perspectives for the repair of extensive cartilage defects.
Collapse
Affiliation(s)
- Yuezhou Wu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Zhuocheng Lyu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Fei Hu
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Linjun Yang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Ke Yang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - Mo Chen
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| | - You Wang
- Department of Bone and Joint Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, 145 Middle Shandong Road, Shanghai, 200001, China.
| |
Collapse
|
14
|
He G, Ke Y, Yuan J, Zhang B, Dai L, Liu J, Zhang X. NSD2-mediated H3K36me2 exacerbates osteoporosis via activation of hoxa2 in bone marrow mesenchymal stem cells. Cell Signal 2024; 121:111294. [PMID: 38996954 DOI: 10.1016/j.cellsig.2024.111294] [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: 04/11/2024] [Revised: 06/26/2024] [Accepted: 07/07/2024] [Indexed: 07/14/2024]
Abstract
BACKGROUND Osteoporosis (OP) is a prevalent disease associated with age, and one of the primary pathologies is the defect of osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). This study aimed to elucidate whether Nuclear Receptor Binding SET Domain Protein 2 (NSD2) transcriptionally regulates osteogenic differentiation of BMSCs in osteoporosis. METHODS Identification of human BMSCs (hBMSCs) in vitro was measured by flow cytometry. Osteogenesis of hBMSCs in vitro was measured by Alizarin Red and Alkaline Phosphatase staining. The protein levels of H3K36me1/2/3, NSD2, and Hoxa2 were measured by western blotting. The mRNA levels of NSD2, Runx2, and BSP were measured by qPCR. The role of NSD2 in the osteogenic differentiation of BMSCs was further identified by silencing NSD2 via shRNA or overexpression of NSD2 via lentivirus transfection. The interactions of NSD2, H3K36me2 and Hoxa2 were identified via chromatin immunoprecipitation (ChIP). Luciferase reporting analysis was employed to confirm that NSD2 regulated the transcriptional activity of Hoxa2. Ovariectomized (OVX) was performed on mice to construct osteoporosis (OP) model. Subsequently, the bone mass was assessed by micro computed tomography (micro-CT) scan. RESULTS During the osteogenesis of OP-derived hBMSCs, the levels of NSD2 and H3K36me2 significantly increased in 14 days of osteogenic induction. Inhibition of NSD2 via shRNA increased the RUNX2 and BSP expression of hBMSCs, while overexpression of NSD2 decreased RUNX2 and BSP expression of hBMSCs. ChIP analysis indicated NSD2-mediated H3K36me2 reduced the osteogenic differentiation of hBMSCs by regulating the osteogenic inhibitor Hoxa2. Accordingly, inhibition of NSD2 in vivo via tail vein injection of LV-shNSD2 lentivirus greatly alleviated OVX-induced osteoporosis in mice. CONCLUSION We demonstrated that NSD2 inhibited the osteogenic differentiation in hBMSCs by transcriptionally downregulating Hoxa2 via H3K36me2 dimethylation. Inhibition of NSD2 effectively attenuated bone loss in murine osteoporosis and NSD2 is a promising target for clinical treatment of osteoporosis.
Collapse
Affiliation(s)
- Guanghui He
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi, Taiyuan, PR China
| | - Yanqin Ke
- School of Medical Sciences, Shanxi Medical University, Shanxi, Taiyuan, PR China
| | - Jie Yuan
- Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi, Taiyuan, PR China
| | - Bingjun Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Facility for Translational Medicine, Shanghai, PR China
| | - Liming Dai
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; National Facility for Translational Medicine, Shanghai, PR China.
| | - Jinlong Liu
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Xiaoling Zhang
- Department of Orthopedic Surgery, Xin Hua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, PR China; Department of Orthopedics, Second Hospital of Shanxi Medical University, Shanxi, Taiyuan, PR China; School of Medical Sciences, Shanxi Medical University, Shanxi, Taiyuan, PR China; National Facility for Translational Medicine, Shanghai, PR China.
| |
Collapse
|
15
|
Chen JH, Ye L, Zhu SL, Yang Y, Xu N. DNMT1-Mediated the Downregulation of FOXF1 Promotes High Glucose-induced Podocyte Damage by Regulating the miR-342-3p/E2F1 Axis. Cell Biochem Biophys 2024; 82:2957-2975. [PMID: 39014186 DOI: 10.1007/s12013-024-01409-3] [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] [Accepted: 07/03/2024] [Indexed: 07/18/2024]
Abstract
Podocyte damage plays a crucial role in the occurrence and development of diabetic nephropathy (DN). Accumulating evidence suggests that dysregulation of transcription factors plays a crucial role in podocyte damage in DN. However, the biological functions and underlying mechanisms of most transcription factors in hyperglycemia-induced podocytes damage remain largely unknown. Through integrated analysis of data mining, bioinformatics, and RT-qPCR validation, we identified a critical transcription factor forkhead box F1 (FOXF1) implicated in DN progression. Moreover, we discovered that FOXF1 was extensively down-regulated in renal tissue and serum from DN patients as well as in high glucose (HG)-induced podocyte damage. Meanwhile, our findings showed that FOXF1 might be a viable diagnostic marker for DN patients. Functional experiments demonstrated that overexpression of FOXF1 strikingly enhanced proliferation, outstandingly suppressed apoptosis, and dramatically reduced inflammation and fibrosis in HG-induced podocytes damage. Mechanistically, we found that the downregulation of FOXF1 in HG-induced podocyte damage was caused by DNMT1 directly binding to FOXF1 promoter and mediating DNA hypermethylation to block FOXF1 transcriptional activity. Furthermore, we found that FOXF1 inhibited the transcriptional expression of miR-342-3p by binding to the promoter of miR-342, resulting in reduced sponge adsorption of miR-342-3p to E2F1, promoting the expression of E2F1, and thereby inhibiting HG-induced podocytes damage. In conclusion, our findings showed that blocking the FOXF1/miR-342-3p/E2F1 axis greatly alleviated HG-induced podocyte damage, which provided a fresh perspective on the pathogenesis and therapeutic strategies for DN patients.
Collapse
Affiliation(s)
- Jie-Hui Chen
- Department of Nephrology, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 510082, China.
| | - Ling Ye
- Department of Nephrology, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 510082, China
| | - Sheng-Lang Zhu
- Department of Nephrology, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 510082, China
| | - Yun Yang
- Department of Nephrology, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 510082, China
| | - Ning Xu
- Department of Nephrology, Shenzhen Nanshan People's Hospital and The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, 510082, China
| |
Collapse
|
16
|
Xiao H, Li W, Qin Y, Lin Z, Qian C, Wu M, Xia Y, Bai J, Geng D. Crosstalk between Lipid Metabolism and Bone Homeostasis: Exploring Intricate Signaling Relationships. RESEARCH (WASHINGTON, D.C.) 2024; 7:0447. [PMID: 39165638 PMCID: PMC11334918 DOI: 10.34133/research.0447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 07/17/2024] [Indexed: 08/22/2024]
Abstract
Bone is a dynamic tissue reshaped by constant bone formation and bone resorption to maintain its function. The skeletal system accounts for approximately 70% of the total volume of the body, and continuous bone remodeling requires quantities of energy and material consumption. Adipose tissue is the main energy storehouse of the body and has a strong adaptive capacity to participate in the regulation of various physiological processes. Considering that obesity and metabolic syndrome have become major public health challenges, while osteoporosis and osteoporotic fractures have become other major health problems in the aging population, it would be interesting to explore these 2 diseases together. Currently, an increasing number of researchers are focusing on the interactions between multiple tissue systems, i.e., multiple organs and tissues that are functionally coordinated together and pathologically pathologically interact with each other in the body. However, there is lack of detailed reviews summarizing the effects of lipid metabolism on bone homeostasis and the interactions between adipose tissue and bone tissue. This review provides a detailed summary of recent advances in understanding how lipid molecules and adipose-derived hormones affect bone homeostasis, how bone tissue, as a metabolic organ, affects lipid metabolism, and how lipid metabolism is regulated by bone-derived cytokines.
Collapse
Affiliation(s)
- Haixiang Xiao
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
- Department of Orthopedics, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine,
University of Science and Technology of China, Hefei 230022, China
| | - Wenming Li
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yi Qin
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Zhixiang Lin
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Chen Qian
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Mingzhou Wu
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Yu Xia
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jiaxiang Bai
- Department of Orthopedics, Jingjiang People’s Hospital Affiliated to Yangzhou University, Jingjiang 214500, Jiangsu Province, China
| | - Dechun Geng
- Department of Orthopedics,
The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| |
Collapse
|
17
|
He H, Chen M, Long F, Ma C, Wang H, Qin J, Chen L. The "toxic window" of amoxicillin exposure during pregnancy on long bone development in fetal mice. Life Sci 2024; 350:122759. [PMID: 38815897 DOI: 10.1016/j.lfs.2024.122759] [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: 02/26/2024] [Revised: 05/19/2024] [Accepted: 05/27/2024] [Indexed: 06/01/2024]
Abstract
AIMS Amoxicillin is a broad-spectrum beta-lactam antibiotic used to treat infectious diseases in pregnant women. Studies have shown that prenatal amoxicillin exposure (PAmE) has developmental toxicity on fetal development. However, the effect of PAmE on long bone development has not been reported. This study aimed to investigate the "toxic window" of PAmE on long bone development and explore its possible mechanism in fetal mice. MATERIALS AND METHODS Pregnant mice were administered amoxicillin by gavage at different stages (gestational day (GD)10-12 and GD16-18), different doses (150 and 300 mg/kg·d) and different courses (single and multiple courses). Fetal femurs were collected at GD18 and bone development related indicators were detected. KEY FINDINGS The results showed that PAmE significantly reduced the length of the femur and primary ossification center of fetal mice, and inhibited the development of fetal growth plate. Meanwhile, PAmE inhibited the development of bone marrow mesenchymal stem cells, osteoclasts and endothelial cells in fetal long bone. Further, we found the fetal long bone developmental toxicity induced by PAmE was most significant at late-pregnancy (GD16-18), high dose (300 mg/kg·d) and multiple-course group. Besides, PAmE inhibited the expression of Wnt/β-catenin signaling pathway in fetal long bone. The β-catenin mRNA expression was significantly positively correlated with the development indexes of fetal long bone. SIGNIFICANCE PAmE has toxic effects on long bone development, and there was an obvious "toxic window" of PAmE on the long bone development in fetal mice. The Wnt/β-catenin signaling pathway may mediate PAmE-induced fetal long bone development inhibition.
Collapse
Affiliation(s)
- Hangyuan He
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Ming Chen
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Fei Long
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Chi Ma
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Hui Wang
- Department of Pharmacology, Basic Medical School of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China
| | - Jun Qin
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| | - Liaobin Chen
- Department of Joint Surgery and Sports Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China; Hubei Provincial Key Laboratory of Developmentally Originated Disease, Wuhan 430071, China.
| |
Collapse
|
18
|
Huang Q, Wang J. CBP-mediated FOXO4 acetylation facilitates postmenopausal osteoporosis (PMO) progression through the inhibition of the Wnt/β-catenin signaling pathway. Histol Histopathol 2024; 39:1017-1024. [PMID: 38037460 DOI: 10.14670/hh-18-680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2023]
Abstract
FOXO4 was previously identified as a potential biomarker and therapeutic target for postmenopausal osteoporosis (PMO) using bioinformatic analysis, but its specific function and molecular mechanism in the progression of osteoporosis was not reported. The current study was designed to investigate the biological function and underlying mechanism of FOXO4 in PMO. Our results showed that FOXO4 expression was significantly upregulated in the serum samples of PMO patients, which was also negatively correlated with the expression of osteogenesis genes (OCN and ALP). In addition, FOXO4 depletion alleviated osteoporosis by facilitating osteogenic differentiation and inhibiting adipogenic differentiation in human bone marrow mesenchymal stem cells (hBMSCs). Overexpression of FOXO4 exerted the opposite effects on the osteogenic/adipogenic differentiation in hBMSCs. Moreover, FOXO4 knockdown activated the Wnt/β-catenin signaling whereas the inhibition of Wnt/β-catenin signaling overturned the effects of FOXO4 deficiency on osteoporosis. Furthermore, FOXO4 upregulation in PMO was caused by CBP-induced acetylation. In summary, our data demonstrated that FOXO4 was a potent biomarker for PMO and mediated the balance between osteogenesis and adipogenesis in hBMSCs by regulating Wnt/β-catenin signaling.
Collapse
Affiliation(s)
- Qiubo Huang
- Department of Orthopedics, Liyang People's Hospital, Liyang City, Jiangsu Province, PR China
| | - Jiang Wang
- Department of Orthopedics, Liyang People's Hospital, Liyang City, Jiangsu Province, PR China.
| |
Collapse
|
19
|
Yuan Y, Chen L, Yang J, Zhou S, Fang Y, Zhang Q, Zhang N, Li Y, Yuan L, Jia F, Ni S, Xiang C. Enhanced homing of mesenchymal stem cells for in situ niche remodeling and bone regeneration. NANO RESEARCH 2024; 17:7449-7460. [DOI: 10.1007/s12274-024-6715-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/20/2024] [Accepted: 04/22/2024] [Indexed: 09/09/2024]
|
20
|
Lu Y, Wang W, Yang B, Cao G, Du Y, Liu J. Screening and Analysis of Core Genes for Osteoporosis Based on Bioinformatics Analysis and Machine Learning Algorithms. Indian J Orthop 2024; 58:944-954. [PMID: 38948379 PMCID: PMC11208356 DOI: 10.1007/s43465-024-01152-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Accepted: 04/08/2024] [Indexed: 07/02/2024]
Abstract
Objective This study aimed to identify osteoporosis-related core genes using bioinformatics analysis and machine learning algorithms. Methods mRNA expression profiles of osteoporosis patients were obtained from the Gene Expression Profiles (GEO) database, with GEO35958 and GEO84500 used as training sets, and GEO35957 and GSE56116 as validation sets. Differential gene expression analysis was performed using the R software "limma" package. A weighted gene co-expression network analysis (WGCNA) was conducted to identify key modules and modular genes of osteoporosis. Kyoto Gene and Genome Encyclopedia (KEGG), Gene Ontology (GO), and gene set enrichment analysis (GSEA) were performed on the differentially expressed genes. LASSO, SVM-RFE, and RF machine learning algorithms were used to screen for core genes, which were subsequently validated in the validation set. Predicted microRNAs (miRNAs) from the core genes were also analyzed, and differential miRNAs were validated using quantitative real-time PCR (qPCR) experiments. Results A total of 1280 differentially expressed genes were identified. A disease key module and 215 module key genes were identified by WGCNA. Three core genes (ADAMTS5, COL10A1, KIAA0040) were screened by machine learning algorithms, and COL10A1 had high diagnostic value for osteoporosis. Four core miRNAs (has-miR-148a-3p, has-miR-195-3p, has-miR-148b-3p, has-miR-4531) were found by intersecting predicted miRNAs with differential miRNAs from the dataset (GSE64433, GSE74209). The qPCR experiments validated that the expression of has-miR-195-3p, has-miR-148b-3p, and has-miR-4531 was significantly increased in osteoporosis patients. Conclusion This study demonstrated the utility of bioinformatics analysis and machine learning algorithms in identifying core genes associated with osteoporosis.
Collapse
Affiliation(s)
- Yongxia Lu
- Department of Endocrinology and Metabolism, Chengdu Seventh People’s Hospital, Chengdu, China
| | - Wei Wang
- Department of Endocrinology and Metabolism, Chengdu Seventh People’s Hospital, Chengdu, China
| | - Baiyuan Yang
- Department of Neurology, Chengdu Seventh People’s Hospital, Chengdu, China
| | - Gui Cao
- Department of Endocrinology and Metabolism, Chengdu Seventh People’s Hospital, Chengdu, China
| | - Yue Du
- Department of Endocrinology and Metabolism, Chengdu Seventh People’s Hospital, Chengdu, China
| | - JingYu Liu
- Department of Neurology, Chengdu Seventh People’s Hospital, Chengdu, China
| |
Collapse
|
21
|
Zhang F, Zhang W. Research progress in Alzheimer's disease and bone-brain axis. Ageing Res Rev 2024; 98:102341. [PMID: 38759893 DOI: 10.1016/j.arr.2024.102341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 05/06/2024] [Accepted: 05/11/2024] [Indexed: 05/19/2024]
Abstract
Alzheimer's disease (AD) is the most common type of cognitive impairment. AD is closely related to orthopedic diseases, such as osteoporosis and osteoarthritis, in terms of epidemiology and pathogenesis. Brain and bone tissues can regulate each other in different manners through bone-brain axis. This article reviews the research progress of the relationship between AD and orthopedic diseases, bone-brain axis mechanisms of AD, and AD therapy by targeting bone-brain axis, in order to deepen the understanding of bone-brain communication, promote early diagnosis and explore new therapy for AD patients.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Wei Zhang
- Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China; Center for Cognitive Neurology, Department of Neurology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.
| |
Collapse
|
22
|
Wang B, Hang H, Wang H, Li D, Jiang Z, Zhang X. Preparation of Puerarin Long Circulating Liposomes and its Effect on Osteoporosis in Castrated Rats. J Pharm Sci 2024; 113:1823-1835. [PMID: 38608726 DOI: 10.1016/j.xphs.2024.04.005] [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: 02/03/2024] [Revised: 04/07/2024] [Accepted: 04/08/2024] [Indexed: 04/14/2024]
Abstract
Osteoporosis is a disease that causes low bone mass and deterioration of bone microarchitecture. Puerarin is a natural isoflavone compound that has been shown to possess anti-inflammatory, antioxidant and ameliorative effects on osteoporosis with less adverse reactions. However, its fast metabolism and low oral bioavailability limit its application. This study aimed to prepare d-α-tocopherol polyethylene glycol 1000 succinate (TPGS)- modified Puerarin Long Circulating Liposomes (TPGS-Puerarin-liposomes), in order to improve the oral bioavailability of puerarin, before evaluation of its pharmacological activity in vitro and in vivo. We employed film dispersion method to develop TPGS-Puerarin-liposomes before appropriate characterizations. Afterwards, we utilized in vivo imaging, pharmacokinetic analysis and in vitro drug release testing to further evaluate the in vivo and in vitro delivery efficiency. In addition, we established a castrated osteoporosis rat model to observe the changes in femur tissue structure and bone micromorphology via hematoxylin-eosin (HE) staining and Micro Computed Tomography (Micro CT). Besides, levels of oxidative stress and inflammatory indicators, as well as expression of wnt/β-catenin pathway-related proteins were detected. In terms of physiochemical properties, the respective mean particle size (PS) and zeta potential (ZP) of TPGS-Puerarin-liposomes were 76.63±0.59 nm and -25.54±0.11 mV. The liposomal formulation exhibited encapsulation efficiency (EE) of 95.08±0.25% and drug loading (DL) of 7.84±0.07%, along with excellent storage stability. Compared with free drugs, the TPGS-Puerarin-liposomes demonstrated a sustained release effect and could increase blood concentration of puerarin in rats, thereby significantly improving its bioavailability. Also, in vivo studies have confirmed potential of the liposomes to promote bone tissue targeting and accumulation of puerarin, coupled with significant improvement of the osteoporotic status. Besides, the liposomes could also reduce levels of oxidative stress and inflammatory factors in serum and bone tissue. Additionally, we discovered that TPGS-Puerarin-liposomes increased Wnt, β-catenin and T-cell factor (TCF) expressions at protein level in the wnt/β-catenin signaling pathway. This study has demonstrated the potential of TPGS-Puerarin-liposomes for treatment of osteoporosis.
Collapse
Affiliation(s)
- Baojun Wang
- Department of spinal surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou 225200, Jiangsu, China
| | - Haifeng Hang
- Department of spinal surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou 225200, Jiangsu, China
| | - Hang Wang
- Department of spinal surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou 225200, Jiangsu, China
| | - Dongdong Li
- Department of spinal surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou 225200, Jiangsu, China
| | - Zhiyu Jiang
- Department of spinal surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou 225200, Jiangsu, China
| | - Xing Zhang
- Department of spinal surgery, Jiangdu People's Hospital Affiliated to Yangzhou University, Yangzhou 225200, Jiangsu, China.
| |
Collapse
|
23
|
Li F, Wang Y, Cao J, Chen Q, Gao Y, Li R, Yuan L. Integrated analysis of genes shared between type 2 diabetes mellitus and osteoporosis. Front Pharmacol 2024; 15:1388205. [PMID: 38966541 PMCID: PMC11222565 DOI: 10.3389/fphar.2024.1388205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/16/2024] [Indexed: 07/06/2024] Open
Abstract
Background The relationship between type 2 diabetes mellitus (T2DM) and osteoporosis (OP) has been widely recognized in recent years, but the mechanism of interaction remains unknown. The aim of this study was to investigate the genetic features and signaling pathways that are shared between T2DM and OP. Methods We analyzed the GSE76894 and GSE76895 datasets for T2DM and GSE56815 and GSE7429 for OP from the Gene Expression Omnibus (GEO) database to identify shared genes in T2DM and OP, and we constructed coexpression networks based on weighted gene coexpression network analysis (WGCNA). Shared genes were then further analyzed for functional pathway enrichment. We selected the best common biomarkers using the least absolute shrinkage and selection operator (LASSO) algorithm and validated the common biomarkers, followed by RT-PCR, immunofluorescence, Western blotting, and enzyme-linked immunosorbent assay (ELISA) to validate the expression of these hub genes in T2DM and OP mouse models and patients. Results We found 8,506 and 2,030 DEGs in T2DM and OP, respectively. Four modules were identified as significant for T2DM and OP using WGCNA. A total of 19 genes overlapped with the strongest positive and negative modules of T2DM and OP. Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed these genes may be involved in pantothenate and CoA biosynthesis and the glycosaminoglycan biosynthesis-chondroitin sulfate/dermatan sulfate and renin-angiotensin system signaling pathway. The LASSO algorithm calculates the six optimal common biomarkers. RT-PCR results show that LTB, TPBG, and VNN1 were upregulated in T2DM and OP. Immunofluorescence and Western blot show that VNN1 is upregulated in the pancreas and bones of T2DM model mice and osteoporosis model mice. Similarly, the level of VNN1 in the sera of patients with T2DM, OP, and T2DM and OP was higher than that in the healthy group. Conclusion Based on the WGCNA and LASSO algorithms, we identified genes and pathways that were shared between T2DM and OP. Both pantothenate and CoA biosynthesis and the glycosaminoglycan biosynthesis-chondroitin sulfate/dermatan sulfate and renin-angiotensin systems may be associated with the pathogenesis of T2DM and OP. Moreover, VNN1 may be a potential diagnostic marker for patients with T2DM complicated by OP. This study provides a new perspective for the systematic study of possible mechanisms of combined OP and T2DM.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Li Yuan
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
24
|
Feng S, Feng Z, Wei Y, Zheng X, Deng Z, Liao Z, Jin Y, Chen R, Zhao L. EEF1B2 regulates bone marrow-derived mesenchymal stem cells bone-fat balance via Wnt/β-catenin signaling. Cell Mol Life Sci 2024; 81:260. [PMID: 38878096 PMCID: PMC11335296 DOI: 10.1007/s00018-024-05297-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: 08/02/2023] [Revised: 02/25/2024] [Accepted: 05/26/2024] [Indexed: 06/29/2024]
Abstract
The pathological advancement of osteoporosis is caused by the uneven development of bone marrow-derived mesenchymal stem cells (BMSCs) in terms of osteogenesis and adipogenesis. While the role of EEF1B2 in intellectual disability and tumorigenesis is well established, its function in the bone-fat switch of BMSCs is still largely unexplored. During the process of osteogenic differentiation, we observed an increase in the expression of EEF1B2, while a decrease in its expression was noted during adipogenesis. Suppression of EEF1B2 hindered the process of osteogenic differentiation and mineralization while promoting adipogenic differentiation. On the contrary, overexpression of EEF1B2 enhanced osteogenesis and strongly inhibited adipogenesis. Furthermore, the excessive expression of EEF1B2 in the tibias has the potential to mitigate bone loss and decrease marrow adiposity in mice with osteoporosis. In terms of mechanism, the suppression of β-catenin activity occurred when EEF1B2 function was suppressed during osteogenesis. Our collective findings indicate that EEF1B2 functions as a regulator, influencing the differentiation of BMSCs and maintaining a balance between bone and fat. Our finding highlights its potential as a therapeutic target for diseases related to bone metabolism.
Collapse
Affiliation(s)
- Shuhao Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Zihang Feng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Yiran Wei
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Xiaoyong Zheng
- Orthopaedic Department, The 4th medical center of Chinese PLA General Hospital, Beijing, 100089, China
| | - Zhonghao Deng
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Zheting Liao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Yangchen Jin
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Ruge Chen
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China
| | - Liang Zhao
- Department of Orthopedics, Nanfang Hospital, Southern Medical University, No. 1838, North Guangzhou Avenue, Baiyun District, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
25
|
Li SY, Xue ST, Li ZR. Osteoporosis: Emerging targets on the classical signaling pathways of bone formation. Eur J Pharmacol 2024; 973:176574. [PMID: 38642670 DOI: 10.1016/j.ejphar.2024.176574] [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: 12/06/2023] [Revised: 03/30/2024] [Accepted: 04/10/2024] [Indexed: 04/22/2024]
Abstract
Osteoporosis is a multifaceted skeletal disorder characterized by reduced bone mass and structural deterioration, posing a significant public health challenge, particularly in the elderly population. Treatment strategies for osteoporosis primarily focus on inhibiting bone resorption and promoting bone formation. However, the effectiveness and limitations of current therapeutic approaches underscore the need for innovative methods. This review explores emerging molecular targets within crucial signaling pathways, including wingless/integrated (WNT), bone morphogenetic protein (BMP), hedgehog (HH), and Notch signaling pathway, to understand their roles in osteogenesis regulation. The identification of crosstalk targets between these pathways further enhances our comprehension of the intricate bone metabolism cycle. In summary, unraveling the molecular complexity of osteoporosis provides insights into potential therapeutic targets beyond conventional methods, offering a promising avenue for the development of new anabolic drugs.
Collapse
Affiliation(s)
- Si-Yan Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Si-Tu Xue
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| | - Zhuo-Rong Li
- Institute of Medicinal Biotechnology, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, 100050, China.
| |
Collapse
|
26
|
Yadalam PK, Ramadoss R, Suresh R. Weighted Gene Co-expression Network Analysis of the Inflammatory Wnt Signaling Reveals Biomarkers Related to Bone Formation. Cureus 2024; 16:e63510. [PMID: 39081453 PMCID: PMC11288288 DOI: 10.7759/cureus.63510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 06/29/2024] [Indexed: 08/02/2024] Open
Abstract
Background and aim Osteocytes regulate bone metabolism and balance through various mechanisms, including the Wnt (Wingless-related integration site signal transduction) signaling pathway. Weighted gene co-expression network analysis (WGCNA) is a computational method to identify functionally related genes based on expression patterns, especially in the Wnt-beta-catenin and osteo-regenerative pathways. This study aims to analyze gene modules of the Wnt signaling pathway from WGCNA analysis. Methods The study used a microarray dataset from the GEO (GSE228306) to analyze differential gene expression in human primary monocytes. The study standardized datasets using Robust Multi-Array Average (RMA) expression measure and Integrated Differential Expression and Pathway (IDEP) analysis tool, building a co-expression network for group-specific component (GC) genes. Results The study uses WGCNA to identify co-expression modules with dysregulated mRNAs, revealing enrichment in Wnt-associated pathways and top hub-enriched genes like colony-stimulating factor 3 (CSF3), interleukin-6 (IL-6), IL-23 subunit alpha (IL23A), suppressor of cytokine signaling 1 (SOCS1), and C-C motif chemokine ligand 19 (CCL19). Conclusion WGCNA analysis of the Wnt signaling pathway will involve functional annotation, network visualization, validation, integration with other omics data, and addressing method limitations for better understanding.
Collapse
Affiliation(s)
- Pradeep Kumar Yadalam
- Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Ramya Ramadoss
- Oral Pathology and Oral Biology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Ramya Suresh
- Oral Biology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| |
Collapse
|
27
|
Mao Y, Xie X, Sun G, Yu S, Ma M, Chao R, Wan T, Xu W, Chen X, Sun L, Zhang S. Multifunctional Prosthesis Surface: Modification of Titanium with Cinnamaldehyde-Loaded Hierarchical Titanium Dioxide Nanotubes. Adv Healthc Mater 2024; 13:e2303374. [PMID: 38366905 DOI: 10.1002/adhm.202303374] [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: 10/04/2023] [Revised: 02/14/2024] [Indexed: 02/18/2024]
Abstract
Orthopedic prostheses are the ultimate therapeutic solution for various end-stage orthopedic conditions. However, aseptic loosening and pyogenic infections remain as primary complications associated with these devices. In this study, a hierarchical titanium dioxide (TiO2) nanotube drug delivery system loaded with cinnamaldehyde for the surface modification of titanium implants, is constructed. These specially designed dual-layer TiO2 nanotubes enhance material reactivity and provide an extensive drug-loading platform within a short time. The introduction of cinnamaldehyde enhances the bone integration performance of the scaffold (simultaneously promoting bone formation and inhibiting bone resorption), anti-inflammatory capacity, and antibacterial properties. In vitro experiments have demonstrated that this system promoted osteogenesis by upregulating both Wnt/β-catenin and MAPK signaling pathways. Furthermore, it inhibits osteoclast formation, suppresses macrophage-mediated inflammatory responses, and impedes the proliferation of Staphylococcus aureus and Escherichia coli. In vivo experiments shows that this material enhances bone integration in a rat model of femoral defects. In addition, it effectively enhances the antibacterial and anti-inflammatory properties in a subcutaneous implant in a rat model. This study provides a straightforward and highly effective surface modification strategy for orthopedic Ti implants.
Collapse
Affiliation(s)
- Yi Mao
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xinru Xie
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Guangxin Sun
- Department of Oral and Maxillofacial Surgery, China Medical University School and Hospital of Stomatology, Shenyang, Liaoning, 110002, China
| | - Shiqi Yu
- Department of Nursing, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Mingqi Ma
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Rui Chao
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Tianhao Wan
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Weifeng Xu
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Xuzhuo Chen
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| | - Lei Sun
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
- Department of Stomatology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230601, China
| | - Shanyong Zhang
- Department of Oral Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Shanghai, 200011, China
| |
Collapse
|
28
|
Liao J, Lu L, Chu X, Xiong Y, Zhou W, Cao F, Cheng P, Shahbazi MA, Liu G, Mi B. Cell membrane coated nanoparticles: cutting-edge drug delivery systems for osteoporosis therapy. NANOSCALE 2024; 16:8236-8255. [PMID: 38584466 DOI: 10.1039/d3nr06264c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
Osteoporosis, characterized by a reduction in bone mineral density, represents a prevalent skeletal disorder with substantial global health implications. Conventional therapeutic strategies, exemplified by bisphosphonates and hormone replacement regimens, though effective, encounter inherent limitations and challenges. Recent years have witnessed the surge of cell-membrane-coated nanoparticles (CMNPs) as a promising intervention for osteoporosis, leveraging their distinct attributes including refined biocompatibility, heightened pharmaceutical payload capacity, as well as targeted drug release kinetics. However, a comprehensive review consolidating the application of CMNPs-based therapy for osteoporosis remains absent within the existing literature. In this review, we provide a concise overview of the distinctive pathogenesis associated with osteoporosis, alongside an in-depth exploration of the physicochemical attributes intrinsic to CMNPs derived from varied cellular sources. Subsequently, we explore the potential utility of CMNPs, elucidating emerging trends in their deployment for osteoporosis treatment through multifaceted therapeutic approaches. By linking the notable attributes of CMNPs with their roles in mitigating osteoporosis, this review serves as a catalyst for further advances in the design of advanced CMNPs tailored for osteoporosis management. Ultimately, such progress is promising for enhancing outcomes in anti-bone loss interventions, paving the way for clinical translation in the near future.
Collapse
Affiliation(s)
- Jiewen Liao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Li Lu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Xiangyu Chu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| | - Wu Zhou
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Faqi Cao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Peng Cheng
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan 430022, China
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 21 Nanyang Link, Singapore 637371, Singapore
| |
Collapse
|
29
|
Bian F, Goda C, Wang G, Lan YW, Deng Z, Gao W, Acharya A, Reza AA, Gomez-Arroyo J, Merjaneh N, Ren X, Goveia J, Carmeliet P, Kalinichenko VV, Kalin TV. FOXF1 promotes tumor vessel normalization and prevents lung cancer progression through FZD4. EMBO Mol Med 2024; 16:1063-1090. [PMID: 38589650 PMCID: PMC11099127 DOI: 10.1038/s44321-024-00064-8] [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: 07/24/2023] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 04/10/2024] Open
Abstract
Cancer cells re-program normal lung endothelial cells (EC) into tumor-associated endothelial cells (TEC) that form leaky vessels supporting carcinogenesis. Transcriptional regulators that control the reprogramming of EC into TEC are poorly understood. We identified Forkhead box F1 (FOXF1) as a critical regulator of EC-to-TEC transition. FOXF1 was highly expressed in normal lung vasculature but was decreased in TEC within non-small cell lung cancers (NSCLC). Low FOXF1 correlated with poor overall survival of NSCLC patients. In mice, endothelial-specific deletion of FOXF1 decreased pericyte coverage, increased vessel permeability and hypoxia, and promoted lung tumor growth and metastasis. Endothelial-specific overexpression of FOXF1 normalized tumor vessels and inhibited the progression of lung cancer. FOXF1 deficiency decreased Wnt/β-catenin signaling in TECs through direct transcriptional activation of Fzd4. Restoring FZD4 expression in FOXF1-deficient TECs through endothelial-specific nanoparticle delivery of Fzd4 cDNA rescued Wnt/β-catenin signaling in TECs, normalized tumor vessels and inhibited the progression of lung cancer. Altogether, FOXF1 increases tumor vessel stability, and inhibits lung cancer progression by stimulating FZD4/Wnt/β-catenin signaling in TECs. Nanoparticle delivery of FZD4 cDNA has promise for future therapies in NSCLC.
Collapse
Affiliation(s)
- Fenghua Bian
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Chinmayee Goda
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Guolun Wang
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Ying-Wei Lan
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Zicheng Deng
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Wen Gao
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
| | - Anusha Acharya
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Abid A Reza
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Jose Gomez-Arroyo
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Nawal Merjaneh
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, 1919 E Thomas Rd., Phoenix, AZ, 85016, USA
| | - Xiaomeng Ren
- Division of Asthma Research of Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA
| | - Jermaine Goveia
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, 3000, Belgium
| | - Peter Carmeliet
- Laboratory of Angiogenesis and Vascular Metabolism, Department of Oncology and Leuven Cancer Institute (LKI), KU Leuven, VIB Center for Cancer Biology, Leuven, 3000, Belgium
- Center for Biotechnology, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Vladimir V Kalinichenko
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA
- Division of Neonatology, Phoenix Children's Hospital, 1919 E Thomas Rd., Phoenix, AZ, 85016, USA
| | - Tanya V Kalin
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, 3333 Burnet Ave., Cincinnati, OH, 45229, USA.
- Department of Child Health, Phoenix Children's Research Institute, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA.
- Center for Cancer and Blood Disorders, Phoenix Children's Hospital, 1919 E Thomas Rd., Phoenix, AZ, 85016, USA.
- Department of Internal Medicine, Division of Pulmonary and Critical Care, University of Arizona College of Medicine-Phoenix, 475 N 5th Street, Phoenix, AZ, 85004, USA.
| |
Collapse
|
30
|
Thangavelu M, Kim PY, Cho H, Song JE, Park S, Bucciarelli A, Khang G. A Gellan Gum, Polyethylene Glycol, Hydroxyapatite Composite Scaffold with the Addition of Ginseng Derived Compound K with Possible Applications in Bone Regeneration. Gels 2024; 10:257. [PMID: 38667676 PMCID: PMC11049517 DOI: 10.3390/gels10040257] [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: 03/08/2024] [Revised: 04/07/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024] Open
Abstract
Engineered bone scaffolds should mimic the natural material to promote cell adhesion and regeneration. For this reason, natural biopolymers are becoming a gold standard in scaffold production. In this study, we proposed a hybrid scaffold produced using gellan gum, hydroxyapatite, and Poly (ethylene glycol) within the addition of the ginseng compound K (CK) as a candidate for bone regeneration. The fabricated scaffold was physiochemically characterized. The morphology studied by scanning electron microscopy (SEM) and image analysis revealed a pore distribution suitable for cells growth. The addition of CK further improved the biological activity of the hybrid scaffold as demonstrated by the MTT assay. The addition of CK influenced the scaffold morphology, decreasing the mean pore diameter. These findings can potentially help the development of a new generation of hybrid scaffolds to best mimic the natural tissue.
Collapse
Affiliation(s)
| | - Pil-Yun Kim
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea; (P.-Y.K.); (H.C.); (J.-E.S.)
| | - Hunhwi Cho
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea; (P.-Y.K.); (H.C.); (J.-E.S.)
| | - Jeong-Eun Song
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea; (P.-Y.K.); (H.C.); (J.-E.S.)
| | - Sunjae Park
- Department of Polymer Nano Science & Technology and Polymer Materials Fusion Research Center, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea;
| | - Alessio Bucciarelli
- Laboratorio RAMSES, IRCCS Istituto Ortopedico Rizzoli, Via di Barbiano 1/10, 40136 Bologna, Italy
| | - Gilson Khang
- Department of Bionanotechnology and Bio-Convergence Engineering, Jeonbuk National University, 567 Baekje-daero, Deokjin-gu, Jeonju-si 54896, Jeonbuk, Republic of Korea; (P.-Y.K.); (H.C.); (J.-E.S.)
| |
Collapse
|
31
|
Jiang H, Wang Y, Tang Z, Peng X, Li C, Dang Y, Ma R. Calycosin alleviates titanium particle-induced osteolysis by modulating macrophage polarization and subsequent osteogenic differentiation. J Cell Mol Med 2024; 28:e18157. [PMID: 38494857 PMCID: PMC10945085 DOI: 10.1111/jcmm.18157] [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: 08/08/2023] [Revised: 01/01/2024] [Accepted: 01/12/2024] [Indexed: 03/19/2024] Open
Abstract
Periprosthetic osteolysis (PPO) caused by wear particles is one of the leading causes of implant failure after arthroplasty. Macrophage polarization imbalance and subsequent osteogenic inhibition play a crucial role in PPO. Calycosin (CA) is a compound with anti-inflammatory and osteoprotective properties. This study aimed to evaluate the effects of CA on titanium (Ti) particle-induced osteolysis, Ti particle-induced macrophage polarization and subsequent osteogenic deficits, and explore the associated signalling pathways in a Ti particle-stimulated calvarial osteolysis mouse model using micro-CT, ELISA, qRT-PCR, immunofluorescence and western blot techniques. The results showed that CA alleviated inflammation, osteogenic inhibition and osteolysis in the Ti particle-induced calvarial osteolysis mouse model in vivo. In vitro experiments showed that CA suppressed Ti-induced M1 macrophage polarization, promoted M2 macrophage polarization and ultimately enhanced osteogenic differentiation of MC3T3-E1 cells. In addition, CA alleviated osteogenic deficits by regulating macrophage polarization homeostasis via the NF-κB signalling pathway both in vivo and in vitro. All these findings suggest that CA may prove to be an effective therapeutic agent for wear particle-induced osteolysis.
Collapse
Affiliation(s)
- Hui Jiang
- Department of OrthopedicsThe Affiliated Jinling Hospital of Nanjing Medical UniversityNanjingChina
| | - Yang Wang
- Department of OrthopedicsThe Affiliated Jinling Hospital of Nanjing Medical UniversityNanjingChina
| | - Zhao Tang
- Department of OrthopedicsThe Affiliated Jinling Hospital of Nanjing Medical UniversityNanjingChina
| | - Xianjiang Peng
- Department of Anesthesiology, Xi'an Children's HospitalAffiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Chan Li
- Department of Anesthesiology, Xi'an Children's HospitalAffiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Yangjie Dang
- Department of Anesthesiology, Xi'an Children's HospitalAffiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| | - Rui Ma
- Department of Anesthesiology, Xi'an Children's HospitalAffiliated Children's Hospital of Xi'an Jiaotong UniversityXi'anChina
| |
Collapse
|
32
|
Coppola U, Kenney J, Waxman JS. A Foxf1-Wnt-Nr2f1 cascade promotes atrial cardiomyocyte differentiation in zebrafish. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.03.13.584759. [PMID: 38558972 PMCID: PMC10980076 DOI: 10.1101/2024.03.13.584759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/04/2024]
Abstract
Nr2f transcription factors (TFs) are conserved regulators of vertebrate atrial cardiomyocyte (AC) differentiation. However, little is known about the mechanisms directing Nr2f expression in ACs. Here, we identified a conserved enhancer 3' to the nr2f1a locus, which we call 3'reg1-nr2f1a (3'reg1), that can promote Nr2f1a expression in ACs. Sequence analysis of the enhancer identified putative Lef/Tcf and Foxf TF binding sites. Mutation of the Lef/Tcf sites within the 3'reg1 reporter, knockdown of Tcf7l1a, and manipulation of canonical Wnt signaling support that Tcf7l1a is derepressed via Wnt signaling to activate the transgenic enhancer and promote AC differentiation. Similarly, mutation of the Foxf binding sites in the 3'reg1 reporter, coupled with gain- and loss-of-function analysis supported that Foxf1 promotes expression of the enhancer and AC differentiation. Functionally, we find that Wnt signaling acts downstream of Foxf1 to promote expression of the 3'reg1 reporter within ACs and, importantly, both Foxf1 and Wnt signaling require Nr2f1a to promote a surplus of differentiated ACs. CRISPR-mediated deletion of the endogenous 3'reg1 abrogates the ability of Foxf1 and Wnt signaling to produce surplus ACs in zebrafish embryos. Together, our data support that downstream members of a conserved regulatory network involving Wnt signaling and Foxf1 function on a nr2f1a enhancer to promote AC differentiation in the zebrafish heart.
Collapse
Affiliation(s)
- Ugo Coppola
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Jennifer Kenney
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Joshua S. Waxman
- Molecular Cardiovascular Biology Division and Heart Institute, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA
- Developmental Biology Division, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
- Department of Pediatrics, University of Cincinnati, College of Medicine, Cincinnati, OH 45229, USA
| |
Collapse
|
33
|
Zhao Y, Guo J, Mu Q, Liu R, Liu H, Xu Y, Li Y. Exploring quality evaluation markers of Fructus Psoraleae based on chemometric analysis integrated with network pharmacology. PHYTOCHEMICAL ANALYSIS : PCA 2024; 35:321-335. [PMID: 37816590 DOI: 10.1002/pca.3290] [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: 07/29/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 10/12/2023]
Abstract
INTRODUCTION Fructus Psoraleae (FP) is a well-known traditional Chinese medicine for the treatment of osteoporosis. However, major quality differences were witnessed owing to its various origins, thus influencing its safety and efficacy. OBJECTIVES The study aimed to evaluate the quality of FP from different origins and predict its quality evaluation markers. METHODS Ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight tandem mass spectrometry was employed for tentative characterisation of the constituents in 10 batches of FP, followed by the utilisation of multivariate statistical analysis methods including principal component analysis and orthogonal partial least squares discriminant analysis for quality evaluation. Network pharmacology approaches were utilised to explore the underlying mechanism of the screened chemotaxonomic markers in treating osteoporosis. RESULTS Forty-one components in FP including, chalcones, coumarins, coumestans, flavonoids, iso-flavonoids, and phenolics, were characterised based on their fragmentation pathways. Ten batches of FP were basically divided into three categories, and eight chemotaxonomic markers including isopsoralen, calamenene, bakuchiol, psoralen, bavachinin, isoneobavaisoflavone, corylifol C, and neobavaisoflavone were screened. Network pharmacology revealed that the chemotaxonomic markers can act on targets such as AKT1, HSP90AA1, and EGFR and possess effects mainly through glycolysis and wnt/β-catenin signalling to alleviate osteoporosis. Molecular docking and molecular dynamic simulation confirmed the good binding affinity and stability between proteins and selected markers. So, eight chemotaxonomic markers were all preferentially recommended as quality evaluation markers. CONCLUSION The study not only provides a reference for the improvement of quality control of FP but also offers a theoretical basis for its further in-depth research in osteoporosis.
Collapse
Affiliation(s)
- Yuting Zhao
- School of Chinese Material Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
| | - Junfeng Guo
- College of Clinical Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Qixuan Mu
- China Pharmaceutical University, Nanjing, Jiangsu, China
| | - Ruojin Liu
- School of Chinese Material Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hui Liu
- School of Chinese Material Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanyan Xu
- School of Chinese Material Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yubo Li
- School of Chinese Material Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
34
|
Guan X, Liang J, Xiang Y, Li T, Zhong X. BARX1 repressed FOXF1 expression and activated Wnt/β-catenin signaling pathway to drive lung adenocarcinoma. Int J Biol Macromol 2024; 261:129717. [PMID: 38290639 DOI: 10.1016/j.ijbiomac.2024.129717] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 01/01/2024] [Accepted: 01/17/2024] [Indexed: 02/01/2024]
Abstract
BACKGROUND Underlying molecular mechanisms of BARX homeobox 1 (BARX1) in lung adenocarcinoma (LUAD) remain elusive. METHODS Abnormally expressed genes in LUAD tissues were analyzed by RNA-sequencing. CCK-8, colony formation, transwell, and wound healing assays examined proliferation, colony formation, invasion, and migration of LUAD cells, respectively. Electrophoretic mobility shift assay and chromatin immunoprecipitation assay examined the interaction between BARX1 and Forkhead Box F1 (FOXF1). Xenograft mouse model of LUAD was constructed to monitor the growth and metastasis of tumor. RESULTS BARX1 was upregulated, FOXF1 was downregulated in LUAD tissues and cells. There was a negative correlation between BARX1 and FOXF1 expression. BARX1 deficiency limited malignant phenotypes of LUAD cells, including proliferation, invasion, migration and EMT. In vivo, BARX1 knockdown suppressed tumor growth and metastasis in A549-drove xenograft mouse model. BARX1 interacted with FOXF1 promoter and repressed FOXF1 expression. Upregulation of BARX1 promoted the expression of Wnt5a, β-catenin, and phosphorylated-glycogen synthase kinase-3 beta (p-GSK3β), whereas inhibited FOXF1, p-β-catenin, and GSK3β in LUAD cells. BARX1 knockdown caused an opposite result. Rescue assays uncovered that FOXF1 reversed the impact of BARX1 on malignant phenotypes and Wnt/β-catenin of LUAD cells. CONCLUSION BARX1 repressed FOXF1 expression and activated Wnt/β-catenin signaling pathway to drive lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiaojiao Guan
- Department of Pathology, Shengjing Hospital of China Medical University, Shenyang 110004, China
| | - Jie Liang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Yifan Xiang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China
| | - Tian Li
- School of Basic Medicine, Fourth Military Medical University, Xi'an 710032, China.
| | - Xinwen Zhong
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China.
| |
Collapse
|
35
|
Li JY, Wang TT, Ma L, Zhang Y, Zhu D. Silencing of Jumonji domain-containing 1C inhibits the osteogenic differentiation of bone marrow mesenchymal stem cells via nuclear factor-κB signaling. World J Stem Cells 2024; 16:151-162. [PMID: 38455099 PMCID: PMC10915961 DOI: 10.4252/wjsc.v16.i2.151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 12/20/2023] [Accepted: 01/17/2024] [Indexed: 02/26/2024] Open
Abstract
BACKGROUND Osteoporosis is a common metabolic bone disorder induced by an imbalance between osteoclastic activity and osteogenic activity. During osteoporosis, bone mesenchymal stem cells (BMSCs) exhibit an increased ability to differentiate into adipocytes and a decreased ability to differentiate into osteoblasts, resulting in bone loss. Jumonji domain-containing 1C (JMJD1C) has been demonstrated to suppress osteoclastogenesis. AIM To examine the effect of JMJD1C on the osteogenesis of BMSCs and the potential underlying mechanism. METHODS BMSCs were isolated from mouse bone marrow tissues. Oil Red O staining, Alizarin red staining, alkaline phosphatase staining and the expression of adipogenic and osteogenic-associated genes were assessed to determine the differentiation of BMSCs. Bone marrow-derived macrophages (BMMs) were incubated with receptor activator of nuclear factor-kappa Β ligand to induce osteoclast differentiation, and osteoclast differentiation was confirmed by tartrate-resistant acid phosphatase staining. Other related genes were measured via reverse transcription coupled to the quantitative polymerase chain reaction and western blotting. Enzyme-linked immunosorbent assays were used to measure the levels of inflammatory cytokines, including tumor necrosis factor alpha, interleukin-6 and interleukin-1 beta. RESULTS The osteogenic and adipogenic differentiation potential of BMSCs isolated from mouse bone marrow samples was evaluated. JMJD1C mRNA and protein expression was upregulated in BMSCs after osteoblast induction, while p-nuclear factor-κB (NF-κB) and inflammatory cytokines were not significantly altered. Knockdown of JMJD1C repressed osteogenic differentiation and enhanced NF-κB activation and inflammatory cytokine release in BMSCs. Moreover, JMJD1C expression decreased during BMM osteoclast differentiation. CONCLUSION The JMJD1C/NF-κB signaling pathway is potentially involved in BMSC osteogenic differentiation and may play vital roles in the pathogenesis of osteoporosis.
Collapse
Affiliation(s)
- Jing-Yi Li
- Department of Medical Cosmetology, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China
| | - Ting-Ting Wang
- Department of General Gerontology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou 450052, Henan Province, China
| | - Li Ma
- Department of Plastic Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yu Zhang
- Senior Department of Hematology, The Fifth Medical Centre, General Hospital of Chinese People's Liberation Army, Beijing 100071, China
| | - Di Zhu
- Department of Orthopaedic Surgery, Beijing Tiantan Hospital, Capital Medical University, Beijing 100070, China.
| |
Collapse
|
36
|
Wang H, Zhang H, Zhang Y, Wang P. Icariin promotes osteogenic differentiation of human bone marrow mesenchymal stem cells by regulating USP47/SIRT1/Wnt/β-catenin. Chem Biol Drug Des 2024; 103:e14431. [PMID: 38373741 DOI: 10.1111/cbdd.14431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2023] [Revised: 11/30/2023] [Accepted: 12/18/2023] [Indexed: 02/21/2024]
Abstract
Icariin has been shown to promote osteogenic differentiation of bone marrow mesenchymal stem cells (BMSCs). However, the underlying molecular mechanism by which Icariin regulates osteogenic differentiation needs to be further revealed. The viability of BMSCs was assessed by cell counting kit 8 assay. BMSC osteogenic differentiation ability was evaluated by detecting alkaline phosphatase activity and performing alizarin red S staining. The protein levels of osteogenic differentiation-related markers, sirtuin 1 (SIRT1), ubiquitin-specific protease 47 (USP47), and Wnt/β-catenin-related markers were determined using western blot. SIRT1 mRNA level was measured using quantitative real-time PCR. The regulation of USP47 on SIRT1 was confirmed by ubiquitination detection and co-immunoprecipitation analysis. Icariin could promote BMSC osteogenic differentiation. SIRT1 expression was enhanced by Icariin, and its knockdown suppressed Icariin-induced BMSC osteogenic differentiation. Moreover, deubiquitinating enzyme USP47 could stabilize SIRT1 protein expression. Besides, SIRT1 overexpression reversed the inhibiting effect of USP47 knockdown on BMSC osteogenic differentiation, and USP47 knockdown also restrained Icariin-induced BMSC osteogenic differentiation. Additionally, Icariin enhanced the activity of the Wnt/β-catenin pathway by upregulating SIRT1. Icariin facilitated BMSC osteogenic differentiation via the USP47/SIRT1/Wnt/β-catenin pathway.
Collapse
Affiliation(s)
- Hongrui Wang
- Department of Orthopedics, First Affiliated Hospital of Naval Medical University, Shanghai, China
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongyue Zhang
- Department of Orthopedics, First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yuntong Zhang
- Department of Orthopedics, First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Panfeng Wang
- Department of Orthopedics, First Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
37
|
Zhao D, Saiding Q, Li Y, Tang Y, Cui W. Bone Organoids: Recent Advances and Future Challenges. Adv Healthc Mater 2024; 13:e2302088. [PMID: 38079529 DOI: 10.1002/adhm.202302088] [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: 07/04/2023] [Revised: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Bone defects stemming from tumorous growths, traumatic events, and diverse conditions present a profound conundrum in clinical practice and research. While bone has the inherent ability to regenerate, substantial bone anomalies require bone regeneration techniques. Bone organoids represent a new concept in this field, involving the 3D self-assembly of bone-associated stem cells guided in vitro with or without extracellular matrix material, resulting in a tissue that mimics the structural, functional, and genetic properties of native bone tissue. Within the scientific panorama, bone organoids ascend to an esteemed status, securing significant experimental endorsement. Through a synthesis of current literature and pioneering studies, this review offers a comprehensive survey of the bone organoid paradigm, delves into the quintessential architecture and ontogeny of bone, and highlights the latest progress in bone organoid fabrication. Further, existing challenges and prospective directions for future research are identified, advocating for interdisciplinary collaboration to fully harness the potential of this burgeoning domain. Conclusively, as bone organoid technology continues to mature, its implications for both clinical and research landscapes are poised to be profound.
Collapse
Affiliation(s)
- Ding Zhao
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Qimanguli Saiding
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yihan Li
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Yunkai Tang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| | - Wenguo Cui
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, P. R. China
| |
Collapse
|
38
|
Li RX, Xu N, Guo YN, Wang Y, Liang YW, Zhou XL, Jiang WT, Wei JX, Zhang XY, Zhou LN, Zhu L, Zhou YM, Xu J. Hemoglobin is associated with BMDs and risk of the 10-year probability of fractures in patients with type 2 diabetes mellitus. Front Endocrinol (Lausanne) 2024; 15:1305713. [PMID: 38323109 PMCID: PMC10846305 DOI: 10.3389/fendo.2024.1305713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024] Open
Abstract
Purpose This study aimed to investigate the associations between hemoglobin (HGB) levels and bone mineral density (BMD) and fracture risk in type 2 diabetes mellitus(T2DM) population of different ages. Method This cross-sectional study included 641 patients with T2DM (57.9% males). BMD of the femoral neck (FN), total hip (TH), and lumbar spine (LS) were measured using dual-energy X-ray absorptiometry. The 10-year probability of fracture was assessed using a fracture risk assessment tool (FRAX). HGB and other biochemical indices were measured in a certified laboratory at our hospital. Statistical analysis was performed using SPSS 26.0 and R language (R version 4.1.0). Generalized additive models (GAMs) were used to identify the associations between HGB and BMD and fracture risk. Results Patients with osteoporosis have lower HGB levels than the non-osteoporotic population and lower FN BMD in patients with anemia than in the non-anemic population. In patients with T2DM, there was sex- and age-related variability in the correlation between HGB levels and BMDs and fracture risk. In older men, HGB level was an independent determinant of BMD and was positively correlated with FN and TH BMD. In non-older women, HGB level was an independent determinant of BMD and fracture risk, positively associated with BMDs and negatively associated with 10-year probability of fracture risk. GAMs revealed a positive linear association between HGB level and BMDs in non-older female patients but not in older male patients. Conclusion Our study provides a new perspective on the association of HGB level and BMDs with fracture risk. Relatively high HGB levels are a protective factor for bone quality in patients with T2DM. However, the bone-protective effect of HGB is influenced by age and sex and persists only in older men and non-older women with T2DM.
Collapse
Affiliation(s)
- Ren-xuan Li
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Na Xu
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Yu-ning Guo
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Yan Wang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Yan-wei Liang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Xiao-lian Zhou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Wen-tong Jiang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Jian-xia Wei
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Xin-yuan Zhang
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| | - Li-na Zhou
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Lei Zhu
- Department of Endocrinology, Shandong Provincial Third Hospital, Jinan, Shandong, China
| | - Yan-man Zhou
- Department of Nephrology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jin Xu
- Department of Endocrinology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong, China
- Key Laboratory of Endocrine Glucose & Lipids Metabolism and Brain Aging, Ministry of Education; Department of Endocrinology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- Department of Endocrinology, Shandong Clinical Research Center of Diabetes and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Institute of Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, “Chuangxin China” Innovation Base of Stem Cell and Gene Therapy for Endocrine Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Laboratory of Prevention and Control for Endocrine and Metabolic Diseases, Jinan, Shandong, China
- Department of Endocrinology, Shandong Engineering Research Center of Stem Cell and Gene Therapy for Endocrine and Metabolic Diseases, Jinan, Shandong, China
| |
Collapse
|
39
|
Huang C, Li Y, Li B, Liu X, Luo D, Liu Y, Wei M, Yang Z, Xu Y. Identifying potential ferroptosis key genes for diagnosis and treatment of postmenopausal osteoporosis through competitive endogenous RNA network analysis. Heliyon 2024; 10:e23672. [PMID: 38226266 PMCID: PMC10788451 DOI: 10.1016/j.heliyon.2023.e23672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 11/24/2023] [Accepted: 12/09/2023] [Indexed: 01/17/2024] Open
Abstract
Objective Postmenopausal osteoporosis (PMOP) is a common systemic metabolic bone disorder that is owing to the reduced estrogen secretion and imbalance of bone absorption and bone formation in postmenopausal women. Ferroptosis has been identified as a novel modulatory mechanism of osteoporosis. Nevertheless, the particular modulatory mechanism between ferroptosis and PMOP is still unclear. The objective of the current investigation was to detect potential biomarkers connected to ferroptosis in PMOP and discover its probable mechanism through bioinformatics. Methods We downloaded PMOP-related microarray datasets from the database of Gene Expression Omnibus (GEO) and obtained the differentially expressed genes (DEGs). Utilizing bioinformatics analysis, the DEGs were intersected with the ferroptosis dataset to obtain ferroptosis-connected mRNAs. Enrichment analysis employing KOBAS 3.0 was conducted to comprehend the biological functions and enrichment pathways of the DEGs. The generation of the protein-protein interaction (PPI) network was conducted with the aim of identifying central genes. Lastly, the coexpression and competitive endogenous RNA (ceRNA) networks were built using Cytoscape. With the help of external datasets GSE56815 to verify the reliability of the hub genes by plotting ROC curves. Results We identified 178 DE microRNAs (miRNAs), 138 DE circular RNAs (circRNAs), and 86 ferroptosis-related mRNAs. Enrichment analysis exhibited that mRNAs were primarily connected with the signaling pathways of PI3K/Akt, metabolism, mTOR, FoxO, HIF-1, AMPK, MAPK, ferroptosis, VEGF, and NOD-like receptors. Generation of the PPI network detected eight hub genes. The circRNA/miR-23b-3p/PTEN axis may relieve PMOP by inhibiting ferroptosis through targeting the pathway of PI3K/Akt signaling, which is a vital modulatory pathway for PMOP progression. Moreover, the ROC curves ultimately indicates that the four hub genes have greater diagnostic importance in PMOP samples in contrast to the normal group samples, which may be possible markers for PMOP diagnosis. Conclusions Bioinformatics analysis identified four hub genes, namely, PTEN, SIRT1, VEGFA, and KRAS, as potential biomarkers for PMOP diagnosis and management. Moreover, the circRNA/miR-23b-3p/PTEN axis may relieve PMOP by suppressing ferroptosis through targeting the pathway of PI3K/Akt signaling, providing a new avenue to explore the pathogenesis of PMOP.
Collapse
Affiliation(s)
- Chengcheng Huang
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
- Department of Endocrinology and Metabology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - Yang Li
- Department of Orthopedic, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - Bo Li
- Department of Orthopedic, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - Xiujuan Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
- Department of Endocrinology and Metabology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - Dan Luo
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
- Department of Endocrinology and Metabology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - Yuan Liu
- First Clinical Medical College, Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - Mengjuan Wei
- Department of Endocrinology and Metabology, The Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - ZhenGuo Yang
- Department of Orthopedic, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| | - Yunsheng Xu
- Department of Orthopedic, The Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, Shandong 250000, China
| |
Collapse
|
40
|
Shang X, Hao X, Hou W, Liu J, Chi R, Deng X, Pan C, Xu T. Exercise-induced modulation of myokine irisin on muscle-bone unit in the rat model of post-traumatic osteoarthritis. J Orthop Surg Res 2024; 19:49. [PMID: 38195597 PMCID: PMC10777589 DOI: 10.1186/s13018-024-04532-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Accepted: 01/03/2024] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND AND AIM Post-traumatic osteoarthritis (PTOA) is a subtype of osteoarthritis (OA). Exercise may produce and release the myokine irisin through muscle fiber contraction. However, the effect of exercise-promoted irisin production on the internal interactions of the muscle-bone unit in PTOA studies remains unclear. METHODS Eighteen 8-week-old Sprague-Dawley (SD) rats were randomly divided into three groups: Sham/sedentary (Sham/Sed), PTOA/sedentary (PTOA/Sed), and PTOA/treadmill-walking (PTOA/TW). The PTOA model was established by transection of anterior cruciate ligament (ACLT) and destabilization of medial meniscus (DMM). After 4 weeks of modeling, the PTOA/TW group underwent treadmill exercise (15 m/min, 30 min/d, 5 d/ week, 8 weeks), and the other two groups were free to move in the cage. Evaluation and correlation analysis of muscle, cartilage, subchondral bone and serological indexes were performed after euthanasia. RESULTS Eight weeks of treadmill exercise effectively alleviated the trauma-induced OA phenotype, thereby maintaining cartilage and subchondral bone integrity in PTOA, and reducing quadriceps atrophy and myofibril degradation. Exercise reversed the down-regulated expression of peroxisome proliferator-activated receptor-gamma coactivator-1α (PGC-1α) and fibronectin type III structural domain protein 5 (FNDC5) in muscle tissue of PTOA rats, and increased the blood irisin level, and the irisin level was positively correlated with the expression of PGC-1α and FNDC5. In addition, correlation analysis showed that irisin metabolism level was strongly negatively correlated with Osteoarthritis Research Society International (OARSI) and subchondral bone loss, indicating that irisin may be involved in cartilage biology and PTOA-related changes in cartilage and subchondral bone. Moreover, the metabolic level of irisin was strongly negatively correlated with muscle fiber cross-sectional area (CSA), Atrogin-1 and muscle ring-finger protein-1(MuRF-1) expression, suggesting that irisin may alleviate muscle atrophy through autocrine action. CONCLUSION Treadmill exercise can alleviate the atrophy and degeneration of muscle fibers in PTOA rats, reduce the degradation of muscle fibrin, promote the expression of serum irisin, and alleviate the degeneration of articular cartilage and subchondral bone loss in PTOA rats. These results indicate that treadmill exercise can affect the process of PTOA by promoting the expression of myokine irisin in rat muscle-bone unit.
Collapse
Affiliation(s)
- Xingru Shang
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Xiaoxia Hao
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Wenjie Hou
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Jiawei Liu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Ruimin Chi
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Xiaofeng Deng
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Chunran Pan
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China
| | - Tao Xu
- Department of Rehabilitation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095#, Jie-Fang Avenue, Qiaokou District, Wuhan, 430030, Hubei, China.
| |
Collapse
|
41
|
Li QL, Wu YX, Zhang YX, Mao J, Zhang ZX. Enhancing osteogenic differentiation of MC3T3-E1 cells during inflammation using UPPE/β-TCP/TTC composites via the Wnt/β-catenin pathway. RSC Adv 2024; 14:1527-1537. [PMID: 38179095 PMCID: PMC10763654 DOI: 10.1039/d3ra05529a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024] Open
Abstract
Periodontitis can lead to defects in the alveolar bone, thus increasing the demand for dependable biomaterials to repair these defects. This study aims to examine the pro-osteogenic and anti-bacterial properties of UPPE/β-TCP/TTC composites (composed of unsaturated polyphosphoester [UPPE], β-tricalcium phosphate [β-TCP], and tetracycline [TTC]) under an inflammatory condition. The morphology of MC3T3-E1 cells on the composite was examined using scanning electron microscopy. The toxicity of the composite to MC3T3-E1 cells was assessed using the Alamar-blue assay. The pro-osteogenic potential of the composite was assessed through ALP staining, ARS staining, RT-PCR, and WB. The antimicrobial properties of the composite were assessed using the zone inhibition assay. The results suggest that: (1) MC3T3-E1 cells exhibited stable adhesion to the surfaces of all four composite groups; (2) the UPPE/β-TCP/TTC composite demonstrated significantly lower toxicity to MC3T3-E1 cells; and (3) the UPPE/β-TCP/TTC composite had the most pronounced pro-osteogenic effect on MC3T3-E1 cells by activating the WNT/β-catenin pathway and displaying superior antibacterial properties. UPPE/β-TCP/TTC, as a biocomposite, has been shown to possess antibacterial properties and exhibit excellent potential in facilitating osteogenic differentiation of MC3T3-E1 cells.
Collapse
Affiliation(s)
- Qi-Lin Li
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan 430022 China
| | - Ya-Xin Wu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan 430022 China
| | - Yu-Xiao Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan 430022 China
| | - Jing Mao
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan 430022 China
| | - Zhi-Xing Zhang
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology Wuhan 430030 China
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration Wuhan 430022 China
| |
Collapse
|
42
|
Huang H, Qian Y, Feng Y, Wang Y, Qian P, Xu F, Wang Q. Erxian Decoction-induced serum exosomes slowed bone marrow mesenchymal stem cell senescence through mitophagy. J Gene Med 2024; 26:e3617. [PMID: 37935422 DOI: 10.1002/jgm.3617] [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: 08/25/2023] [Revised: 09/17/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
OBJECTIVE Erxian Decoction (EXD) is traditionally employed in the treatment of menopausal syndromes, although its underlying mechanisms remain largely undefined. Given that the senescence of bone marrow mesenchymal stem cells (BMSCs) is intertwined with organismal aging and associated diseases, this study endeavored to elucidate the influence of EXD on aging BMSCs and uncover the mechanisms through which EXD impedes BMSC senescence. METHODS Initially, we probed the anti-senescent mechanisms of EXD on BMSCs via network pharmacology. We subsequently isolated and identified exosomes from the serum of EXD-fed rats (EXD-Exos) and administered these to H2 O2 -induced aging BMSC. Assays were conducted to assess BMSC senescence indicators and markers pertinent to mitochondrial autophagy. Treatments with mitophagy inhibitors and activators were then employed to substantiate our findings. RESULTS Protein-protein interaction (PPI) network analyses spotlighted AKT1, TP53, TNF, JUN, VEGFA, IL6, CASP3 and EGFR as focal targets. Gene Ontology and Kyoto Encylcopedia of Genes and Genomes pathway analyses underscored oxidative stress, mitophagy and cell proliferation as pivotal processes. Our cellular assays ascertained that EXD-Exos mitigated H2 O2 -induced senescence phenotypes in BMSCs. Moreover, EXD-Exos ameliorated disrupted mitophagy in BMSCs, as evidenced by enhanced cellular membrane potential and diminished reactive oxygen species levels. Intriguingly, EXD-Exos also preserved the osteogenic differentiation potential of BMSCs while curtailing their adipogenic propensity. CONCLUSION Our findings compellingly suggest that EXD counteracts BMSC senescence by fostering mitophagy.
Collapse
Affiliation(s)
- Haoqiang Huang
- Department of Orthopaedics, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China
| | - Yinhua Qian
- Department of Orthopaedics, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China
| | - Ye Feng
- School of Stomatology, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Yitao Wang
- Department of Orthopaedics, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China
| | - Pingkang Qian
- Department of Orthopaedics, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China
| | - Feng Xu
- Department of Orthopaedics, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China
| | - Qing Wang
- Department of Orthopaedics, Kunshan Affiliated Hospital of Nanjing University of Chinese Medicine, Kunshan, Jiangsu, China
| |
Collapse
|
43
|
Zhang F, Lv M, Wang S, Li M, Wang Y, Hu C, Hu W, Wang X, Wang X, Liu Z, Fan Z, Du J, Sun Y. Ultrasound-triggered biomimetic ultrashort peptide nanofiber hydrogels promote bone regeneration by modulating macrophage and the osteogenic immune microenvironment. Bioact Mater 2024; 31:231-246. [PMID: 37637084 PMCID: PMC10450354 DOI: 10.1016/j.bioactmat.2023.08.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 07/28/2023] [Accepted: 08/08/2023] [Indexed: 08/29/2023] Open
Abstract
The immune microenvironment plays a vital role in bone defect repair. To create an immune microenvironment that promotes osteogenesis, researchers are exploring ways to enhance the differentiation of M2-type macrophages. Functional peptides have been discovered to effectively improve this process, but they are limited by low efficiency and rapid degradation in vivo. To overcome these issues, peptide with both M2 regulatory and self-assembly modules was designed as a building block to construct an ultrasound-responsive nanofiber hydrogel. These nanofibers can be released from hydrogel in a time-dependent manner upon ultrasound stimulation, activating mitochondrial glycolytic metabolism and the tricarboxylic acid cycle, inhibiting reactive oxygen species production and enhancing M2 macrophage polarization. The hydrogel exhibits advanced therapeutic potential for bone regeneration by triggering M2 macrophages to secrete BMP-2 and IGF-I, accelerating the differentiation of bone marrow mesenchymal stem cells (BMSCs) into osteoblasts. Thus, modularly designed biomimetic ultrashort peptide nanofiber hydrogels provide a novel strategy to rebuild osteogenic immune microenvironments for bone repair.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 200072, China
| | - Mingchen Lv
- Department of Gynaecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 201804, China
| | - Siyuan Wang
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 200072, China
| | - Mengyao Li
- Department of Immunology and Microbiology, The Minister of Education Key Laboratory of Cell Death and Differentiation, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, China
| | - Yu Wang
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 200072, China
| | - Congjiao Hu
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 200072, China
| | - Wei Hu
- Department of Gynaecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 201804, China
| | - Xuekui Wang
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 200072, China
| | - Xiaogang Wang
- Key Laboratory of Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, China
| | - Zhiduo Liu
- Department of Immunology and Microbiology, The Minister of Education Key Laboratory of Cell Death and Differentiation, Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, China
| | - Zhen Fan
- Department of Gynaecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 201804, China
| | - Jianzhong Du
- Department of Gynaecology and Obstetrics, Shanghai Fourth People's Hospital, School of Medicine, Tongji University, Shanghai, 200434, China
- Department of Polymeric Materials, School of Materials Science and Engineering, Tongji University, 201804, China
| | - Yao Sun
- Department of Oral Implantology, Stomatological Hospital and Dental School of Tongji University, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, 200072, China
| |
Collapse
|
44
|
Luo W, Zhang N, Wang Z, Chen H, Sun J, Yao C, Zhang Y. LncRNA USP2-AS1 facilitates the osteogenic differentiation of bone marrow mesenchymal stem cells by targeting KDM3A/ETS1/USP2 to activate the Wnt/β-catenin signaling pathway. RNA Biol 2024; 21:1-13. [PMID: 38131611 PMCID: PMC10761055 DOI: 10.1080/15476286.2023.2290771] [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] [Revised: 08/06/2023] [Accepted: 08/10/2023] [Indexed: 12/23/2023] Open
Abstract
Human bone marrow mesenchymal stem cells (HBMSCs) can promote new bone formation. Previous studies have proven the ability of long non-coding RNAs (lncRNAs) to modulate the osteogenic differentiation of mesenchymal stem cells. However, the molecular mechanism modulated by lncRNAs in affecting the osteogenic differentiation of HBMSCs remains largely unknown. Thus, this study aims to reveal the role of lncRNA ubiquitin-specific peptidase 2 antisense RNA 1 (USP2-AS1) in regulating the osteogenic differentiation of HBMSCs and investigate its regulatory mechanism. Through bioinformatics analysis and RT-qPCR, we confirmed that USP2-AS1 expression was increased in HBMSCs after culturing in osteogenic differentiation medium (OM-HBMSCs). Moreover, we uncovered that knockdown of USP2-AS1 inhibited the osteogenic differentiation of HBMSCs. Further exploration indicated that USP2-AS1 positively regulated the expression of its nearby gene USP2. Mechanistically, USP2-AS1 recruited lysine demethylase 3A (KDM3A) to stabilize ETS proto-oncogene 1 (ETS1), transcription factor that transcriptionally activated USP2. Additionally, USP2-induced Wnt/β-catenin signalling pathway activation via deubiquitination of β-catenin protein. In summary, our study proved that lncRNA USP2-AS1 facilitates the osteogenic differentiation of HBMSCs by targeting KDM3A/ETS1/USP2 axis to activate the Wnt/β-catenin signalling pathway.
Collapse
Affiliation(s)
- Wanxin Luo
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Na Zhang
- Department of Endocrinology, Jiangxi Provincial People’s Hospital, Nanchang, Jiangxi, China
- Medical College of Nanchang University, Nanchang, Jiangxi, China
| | - Ziping Wang
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Hao Chen
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Jie Sun
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Chen Yao
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Yafeng Zhang
- Department of Orthopaedics, the Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| |
Collapse
|
45
|
Zhao D, He J, Zhao X, Sheng X, Feng Z, Wang X, Zhang C, Wang S, Geng B, Xia Y. A novel lncRNA GM15416 regulates osteoblast apoptosis and differentiation through the c-Fos/Fas axis and mitigates osteoporosis. Int J Biol Macromol 2024; 254:127824. [PMID: 37924900 DOI: 10.1016/j.ijbiomac.2023.127824] [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: 07/24/2023] [Revised: 10/20/2023] [Accepted: 10/26/2023] [Indexed: 11/06/2023]
Abstract
Osteoporosis (OP) is a common systemic bone disorder, and the programmed cell death of osteoblasts is closely linked to the development of osteoporosis. Previous studies have shown that c-fos can cause osteoblast apoptosis. Furthermore, it has been demonstrated that long non-coding RNA (lncRNA) plays a pervasive role in regulating the biology of osteoblasts. Nevertheless, the precise role and mechanism of long non-coding RNA (lncRNA) in relation to c-Fos at the transcriptional level in osteoblast cell death remain uncertain. Compared with normal osteoblasts, serum deprivation resulted in significant upregulation of the transcription factor c-Fos and apoptosis-related Fas proteins in osteoblasts. In addition, the expression of lncRNA GM15416 related to c-Fos was significantly increased. The results showed that overexpression of c-Fos leads to an increase in downstream Fas protein, which subsequently leads to osteoblast apoptosis and hinders osteogenesis. On the contrary, a decrease in lncRNA GM15416 expression leads to a decrease in c-Fos/Fas expression, which hinders osteoblast apoptosis and promotes osteogenesis. Our results suggest that lncRNA GM15416 exerts inhibitory effects on osteoblast apoptosis and acts as a preventive factor against osteoporosis. As a result, GM15416 emerges as an important lncRNA associated with osteoporosis and holds potential as a future therapeutic target.
Collapse
Affiliation(s)
- Dacheng Zhao
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Pain Department of the Second Hospital of Lanzhou University, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, Pr China
| | - Jinwen He
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Xiaobing Zhao
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Xiaoyun Sheng
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Zhiwei Feng
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Xingwen Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Chengjun Zhang
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Shenghong Wang
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China
| | - Bin Geng
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China.
| | - Yayi Xia
- Department of Orthopaedics, Lanzhou University Second Hospital, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Gansu Province Clinical Medical Research Center for Orthopedics, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China; Key Laboratory of Bone and joint Disease research of Gansu Province, No. 82 Cuiying Gate, Chengguan District, Lanzhou 730030, Gansu, PR China.
| |
Collapse
|
46
|
Zheng K, Deng M, Yu Y, Zhou J, Hou Y, Chen L, Ma Y, Chen Y, Chen H, Guo X, Luo R, Liao J, Meng S, Zhang J, Yan P, Zhang Y, Hu L, Qian A, Yin C. Basic knowledge and research methods. BONE CELL BIOMECHANICS, MECHANOBIOLOGY AND BONE DISEASES 2024:3-29. [DOI: 10.1016/b978-0-323-96123-3.00001-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
47
|
Tian B, Bai J, Sheng L, Chen H, Chang W, Zhang Y, Yao C, Zhou C, Wang X, Shan H, Dong Q, Wang C, Zhou X. P7C3 Ameliorates Bone Loss by Inhibiting Osteoclast Differentiation and Promoting Osteogenesis. JBMR Plus 2023; 7:e10811. [PMID: 38130773 PMCID: PMC10731119 DOI: 10.1002/jbm4.10811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Accepted: 08/13/2023] [Indexed: 12/23/2023] Open
Abstract
Bone homeostasis, the equilibrium between bone resorption and formation, is essential for maintaining healthy bone tissue in adult humans. Disruptions of this process can lead to pathological conditions such as osteoporosis. Dual-targeted agents, capable of inhibiting excessive bone resorption and stimulating bone formation, are being explored as a promising strategy for developing new treatments to address osteoporosis. In this study, we investigated the effects of P7C3 on bone remodeling and its potential therapeutic role in osteoporosis treatment in mice. Specifically, P7C3 can remarkably suppress receptor activator of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclast differentiation in bone marrow macrophages via the Akt-NF-κB-NFATc1 signaling pathway. Additionally, RNA sequencing (RNAseq) analysis revealed that P7C3 promoted osteoblast differentiation and function through the Wnt/β-catenin signaling pathway, thereby enhancing bone formation. Furthermore, μCT analysis and histological examination of bone tissues from P7C3-treated mice showed attenuation of both Ti-induced bone erosion and ovariectomy (OVX)-induced bone loss. These findings suggest that P7C3 may have a novel function in bone remodeling and may be a promising therapeutic agent for the treatment of osteoporosis. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Bo Tian
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Jinyu Bai
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Lei Sheng
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Hao Chen
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Wenju Chang
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Yue Zhang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Chenlu Yao
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Chenmeng Zhou
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Xiaoyu Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Huajian Shan
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Qirong Dong
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| | - Chao Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & SoftMaterials (FUNSOM)Soochow UniversitySuzhouChina
| | - Xiaozhong Zhou
- Department of OrthopedicsThe Second Affiliated Hospital of Soochow UniversitySuzhouChina
| |
Collapse
|
48
|
Wen R, Huang R, Xu K, Cheng Y, Yi X. Beneficial effects of Apelin-13 on metabolic diseases and exercise. Front Endocrinol (Lausanne) 2023; 14:1285788. [PMID: 38089606 PMCID: PMC10714012 DOI: 10.3389/fendo.2023.1285788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/13/2023] [Indexed: 12/18/2023] Open
Abstract
Apelin, a novel endogenous ligand of the G-protein-coupled receptor APJ, is encoded by the APLN gene and can be hydrolyzed into multiple subtypes, with Apelin-13 being one of the most active subtypes of the Apelin family. Recent studies have revealed that Apelin-13 functions as an adipokine that participates in the regulation of different biological processes, such as oxidative stress, inflammation, apoptosis, and energy metabolism, thereby playing an important role in the prevention and treatment of various metabolic diseases. However, the results of recent studies on the association between Apelin-13 and various metabolic states remain controversial. Furthermore, Apelin-13 is regulated or influenced by various forms of exercise and could therefore be categorized as a new type of exercise-sensitive factor that attenuates metabolic diseases. Thus, in this review, our purpose was to focus on the relationship between Apelin-13 and related metabolic diseases and the regulation of response movements, with particular reference to the establishment of a theoretical basis for improving and treating metabolic diseases.
Collapse
Affiliation(s)
- Ruiming Wen
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Ruiqi Huang
- School of Physical Education, Liaoning Normal University, Dalian, Liaoning, China
| | - Ke Xu
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Yang Cheng
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| | - Xuejie Yi
- School of Sports Health, Shenyang Sport University, Shenyang, Liaoning, China
| |
Collapse
|
49
|
Chen Q, Yang Z, Sun X, Long R, Shen J, Wang Z. Inokosterone activates the BMP2 to promote the osteogenic differentiation of bone marrow mesenchymal stem cells and improve bone loss in ovariectomized rats. Biochem Biophys Res Commun 2023; 682:349-358. [PMID: 37839103 DOI: 10.1016/j.bbrc.2023.10.032] [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: 07/21/2023] [Revised: 09/21/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023]
Abstract
Evidence suggests that enhancing the osteogenic ability of bone marrow-derived mesenchymal stem cells (BMSCs) may be beneficial in the fight against osteoporosis (OP) effects. Inokosterone (IS) is a major active constituent of Achyranthis bidentatae radix (ABR), which stimulates osteogenic differentiation of mouse embryonic osteoblasts. This study aims to investigate effect of IS on OP using osteogenic differentiated BMSCs and ovariectomy (OVX)-induced OP rats. The BMSCs were treated with 50, 100, or 200 mg/L IS and OP rats were given 2 or 4 mg/kg of IS by gavage. Cell viability, the osteogenic differentiation marker protein expression level, and mineralization were observed. This study proved that IS improved cell viability, osteogenic differentiation, and cellular mineralization in BMSCs and raised expression levels of bone morphogenetic protein-2 (BMP2), Smad1, runt-related transcription factor 2 (RUNX2), collagen I, ALP, and OCN. By BMP2 knockdown/overexpression, this study also proved the BMP2 signaling pathway activation is a potential biological mechanism of IS to improve osteogenic differentiation and mineralization in osteogenic differentiated BMSCs. In OVX-induced OP rats, IS was observed to antagonize bone loss, improve osteogenic differentiation marker protein expression levels, and activate BMP-2, smad1, and RUNX2. These findings provide scientific support for further investigation of the biological mechanisms of IS in ameliorating OP.
Collapse
Affiliation(s)
- Qiang Chen
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Zhihua Yang
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Xiangyi Sun
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Ruchao Long
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Jianwei Shen
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China
| | - Zhen Wang
- Department of Orthopedics, Affiliated Xiaoshan Hospital, Hangzhou Normal University, Hangzhou, 311200, China.
| |
Collapse
|
50
|
Xue J, Liu L, Liu H, Li Z. LncRNA SNHG14 activates autophagy via regulating miR-493-5p/Mef2c axis to alleviate osteoporosis progression. Commun Biol 2023; 6:1120. [PMID: 37925525 PMCID: PMC10625635 DOI: 10.1038/s42003-023-05493-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 10/19/2023] [Indexed: 11/06/2023] Open
Abstract
Osteoporosis is a progressive bone disease caused by impaired function of endogenous bone marrow-derived mesenchymal stem cells (BMSCs). Herein, we investigated the mechanism of lncRNA SNHG14 in osteoporosis progression. BMSCs were isolated from BALB/c mice. The osteogenic ability of BMSCs was assessed by Alkaline phosphatase (ALP) and Alizarin Red S Staining (ARS) staining. The interaction between miR-493-5p and SNHG14 or myocyte enhancer factor 2 C (Mef2c) was confirmed by dual-luciferase reporter assay. Bone histomorphometry changes were evaluated to analyze SNHG14'roles in osteoporosis in vivo. Our results illustrated SNHG14 and Mef2c levels were increased in a time-dependent manner in BMSCs, and miR-493-5p expression was decreased. SNHG14 knockdown inhibited osteogenic differentiation of BMSCs, and SNHG14 upregulation had the opposite effect. SNHG14 overexpression elevated bone mineral density and bone trabecular number, and alleviated osteoporosis progression in vivo. Mechanically, miR-493-5p was a target of SNHG14, and miR-493-5p targeted the Mef2c gene directly. SNHG14 overexpression reversed the inhibition of miR-493-5p on the osteogenic ability of BMSCs, and miR-493-5p silencing accelerated BMSCs osteogenesis by activating Mef2c-mediated autophagy to accelerate BMSCs osteogenesis. In short, SNHG14 activated autophagy via regulating miR-493-5p/Mef2c axis to alleviate osteoporosis progression, which might provide a new molecular target for osteoporosis treatment.
Collapse
Affiliation(s)
- Jingbo Xue
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China.
| | - Lulu Liu
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| | - Hao Liu
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| | - Zepeng Li
- The First Affiliated Hospital, Department of Spine Surgery, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan Province, PR China
| |
Collapse
|