1
|
Wang Y, Xiao Y, Yang X, He F, Hu J, Yang G, Wang W. Bone marrow mesenchymal stem cells overexpressing stromal cell- derived factor 1 aid in bone formation in osteoporotic mice. BMC Musculoskelet Disord 2024; 25:878. [PMID: 39497150 PMCID: PMC11536944 DOI: 10.1186/s12891-024-07957-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 10/14/2024] [Indexed: 11/06/2024] Open
Abstract
BACKGROUND Osteoporosis is characterized by low systemic bone mineral content and destruction of bone microarchitecture. Promoting bone regeneration and reversing its loss by infusion of exogenous bone marrow mesenchymal stem cells (BMSCs) is a potentially effective treatment for osteoporosis. However, their limited migration to target organs reduces the therapeutic effect of the cells. Stromal cell-derived factor 1 (SDF1) is a chemokine that induces targeted cell migration through the SDF1/CXCR4 (C-X-C chemokine receptor 4) axis and can induce migration of exogenous mesenchymal stem cells to sites of high SDF1 concentration. There are no studies on BMSCs overexpressing SDF1 (SDF1-BMSCs) in osteoporotic mice in vivo. We aimed to investigate if the increased SDF1 concentration facilitated cell migration to the bone. METHODS We used lentivirus to construct BMSCs overexpressing SDF1 or knocking down CXCR4. We verified the proliferation ability of the cells in vitro using Cell Counting Kit-8 (CCK8) and 5-Bromodeoxyuridinc (BrdU), the migration ability of the cells using Transwell, and the osteogenic and lipogenic ability of the cells using osteogenic and lipogenic induction solutions. In in vivo experiments, we induced osteoporosis in 72 female mice by ovariectomy and injected different groups of cells via the tail vein. Femoral tissue samples were collected for a fixed time, and the osteogenic and homing abilities of the cells were verified by MicroCT and tissue section staining. RESULTS We successfully demonstrated that high expression of SDF1 promoted cell proliferation and migration in vitro, without affecting their cell differentiation ability. In an ovariectomized mouse model, SDF1-BMSCs were more likely to be home to the femur than the BMSCs, had a better pro-osteogenic ability, and had higher expression of Wnt-1. Blocking the SDF1/CXCR4 axis reduced the homing of exogenous mesenchymal stem cells (MSCs) to the femur and their osteogenic capacity. CONCLUSIONS SDF1-BMSCs can further promote bone formation by increasing the number of cells homing to the femur in osteoporotic mice. Our study shows that stem cells can promote their proliferation and home to the femur via the SDF1/CXCR4 axis and further help bone formation via Wnt-1 signaling.
Collapse
Affiliation(s)
- Yanghao Wang
- Department of Pathology, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Ya Xiao
- First Clinical College, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - XinYu Yang
- Clinical Oncology College, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China
| | - Fei He
- Department of Orthopedic, Qujing Affiliated Hospital of Kunming Medical University, Qujing, Yunnan, China
| | - Jun Hu
- Department of Orthopedic, The First People's Hospital of Kunming, Kunming, Yunnan, China
| | - Guang Yang
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| | - Weizhou Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
2
|
He P, Yang Z, Li H, Zhou E, Hou Z, Sang H. miR-18a-5p promotes osteogenic differentiation of BMSC by inhibiting Notch2. Bone 2024; 188:117224. [PMID: 39117162 DOI: 10.1016/j.bone.2024.117224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 07/17/2024] [Accepted: 08/03/2024] [Indexed: 08/10/2024]
Abstract
Postmenopausal osteoporosis (PMOP) is a metabolic disorder characterized by the loss of bone density, which increases the risk of developing complications such as fractures. A pivotal factor contributing to the onset of PMOP is the diminished osteogenic differentiation capacity of bone marrow mesenchymal stem cells (BMSCs). MicroRNAs (miRNAs) play a substantial role in this process; however, their specific impact on regulating BMSCs osteogenesis remains unclear. Studies have evidenced a reduced expression of miR-18a-5p in PMOP, and concomitantly, our observations indicate an augmented expression of miR-18a-5p during the osteogenic differentiation of BMSCs. This investigation seeks to elucidate the regulatory influence of miR-18a-5p on BMSC osteogenic differentiation and the underlying mechanisms. In vitro experiments demonstrated that the overexpression of miR-18a-5p facilitated the osteogenic differentiation of BMSCs, while the downregulation of miR-18a-5p yielded converse outcomes. Mechanistically, We employed bioinformatics techniques to screen out the target gene Notch2 of miR-18a-5p. Subsequently, dual-luciferase reporter gene assays and rescue experiments substantiated that miR-18a-5p promotes BMSC osteogenic differentiation by suppressing Notch2. Finally, miR-18a-5p was overexpressed via adenovirus injection into the femoral bone marrow cavity, with results demonstrating its capability to enhance osteogenic differentiation and alleviate PMOP symptoms. Our findings disclose that miR-18a-5p fosters osteogenic differentiation of BMSC by inhibiting Notch2, thereby offering novel targets and strategies for PMOP treatment.
Collapse
Affiliation(s)
- Peipei He
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zefeng Yang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Hetong Li
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Enhui Zhou
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China; The Third School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Zuoxu Hou
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China
| | - Hongxun Sang
- Department of Orthopedics, Shenzhen Hospital, Southern Medical University, Shenzhen, China.
| |
Collapse
|
3
|
Wang H, Zhao Y, Liu H, Zhang X, Lv S, Zhou T, Cui H, Zhao J, Li X. Untargeted metabolomics revealed the mechanism of aucubin on glucocorticoid-induced osteoporosis in mice through modulating arachidonic acid metabolism. J Pharm Biomed Anal 2024; 248:116273. [PMID: 38878451 DOI: 10.1016/j.jpba.2024.116273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 05/28/2024] [Accepted: 06/02/2024] [Indexed: 07/20/2024]
Abstract
Glucocorticoid-induced osteoporosis (GIOP) represents the most prevalent form of secondary osteoporosis. Aucubin (AU), a principal active component found in traditional herbal medicines such as Eucommia ulmoides, has been demonstrated to enhance osteoblast differentiation. Nonetheless, the precise therapeutic effects of AU on GIOP and the complex underlying regulatory mechanisms warrant further investigation. We first established a GIOP model in female mice and then assessed the therapeutic effects of AU using micro-CT analysis, biomechanical testing, measurements of serum calcium (Ca) and phosphorus (P) levels, and histological analyses using Hematoxylin and Eosin (HE) and Masson staining. Subsequently, non-targeted metabolomics was employed in order to study the effects of AU on serum metabolites in GIOP mice. The levels of the factors related to these metabolites were quantified using real-time quantitative reverse transcription polymerase chain reaction (RT-qPCR) and western blot analyses. Finally, the effects of AU on osteoblastic and osteoclastic differentiation were examined. We found that AU significantly ameliorated bone microarchitecture and strength in GIOP mice. It mitigated pathological damages such as impairment of trabecular bone structure and reduction in collagen fibers, while concurrently elevating serum levels of Ca and P. Non-targeted metabolomics revealed that Arachidonic acid (AA) metabolism serves as a common pathway between the control and GIOP groups, as well as between the high-dose AU (AUH) and GIOP groups. AU notably upregulates prostaglandin-endoperoxide synthase 2 (PTGS2) and microsomal prostaglandin-E synthase 1 (PTGES) expression and downregulates prostaglandin-H2 D-isomerase (PTGDS) expression. Furthermore, AU treatment increased the expression of runt-related transcription factor 2 (Runx2) and transcription factor Sp7 (Osterix), enhanced serum alkaline phosphatase (ALP) activity, and reduced osteoclast expression. These results indicate that AU is a potential drug for treating GIOP, and its mechanism is related to regulating AA metabolism and promoting osteoblast differentiation. However, the key targets of AU in treating GIOP still need further exploration.
Collapse
Affiliation(s)
- Hengjun Wang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Yunchao Zhao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Huan Liu
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Xuelei Zhang
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Shuquan Lv
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine in Osteoarthrosis Research, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Diabetes, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Tingting Zhou
- Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine in Osteoarthrosis Research, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Hebei Province Integrated Traditional Chinese and Western Medicine 3D Printing Technology Innovation Center, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China
| | - Huantian Cui
- The First School of Clinical Medicine, Yunnan University of Chinese Medicine, Kunming, Yunnan Province 650500, China
| | - Jianyong Zhao
- Graduate School, Hebei University of Chinese Medicine, Shijiazhuang, Hebei Province 050091, China; Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China.
| | - Xiaoming Li
- Department of Orthopedics, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Hebei Key Laboratory of Integrated Traditional Chinese and Western Medicine in Osteoarthrosis Research, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China; Department of Hebei Province Integrated Traditional Chinese and Western Medicine 3D Printing Technology Innovation Center, Cangzhou Hospital of Integrated Traditional Chinese Medicine and Western Medicine of Hebei Province Affiliated to Hebei University of Chinese Medicine, Cangzhou, Hebei Province 061013, China.
| |
Collapse
|
4
|
Yin M, Zhou D, Jia F, Su X, Li X, Sun R, Li J. Metabolomics analysis of the potential mechanism of Yi-Guan-Jian decoction to reverse bone loss in glucocorticoid-induced osteoporosis. J Orthop Surg Res 2023; 18:409. [PMID: 37277810 DOI: 10.1186/s13018-023-03778-6] [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: 02/22/2023] [Accepted: 04/02/2023] [Indexed: 06/07/2023] Open
Abstract
BACKGROUND Glucocorticoid-induced osteoporosis (GIOP) is a disease in which long-term use of glucocorticoid causes bone loss, deterioration of bone microstructure and fracture. Currently, clinical drugs targeting this disease have certain side effects. There is still a need to find effective drugs with fewer side effects. The theory of traditional Chinese medicine suggests that YGJ has therapeutic effect on GIOP, but it has not been explained. Therefore, this study aims to explore the protective effect of YGJ on GIOP mouse models and elucidate the underlying mechanism through LC-MS-based metabolomics analysis. METHODS The general condition of 8 week age male C57BL/6J mice was recorded after 8 weeks of treatment with dexamethasone (DEX) and YGJ. Bone-related parameters and bone morphology were determined by Micro-CT. HE staining was used to observe the pathological changes of bone tissue. Serum levels of bone metabolism markers were detected by ELISA. Liver metabolomics analysis was conducted to search for the significant markers of anti-GIOP of YGJ and the metabolic pathway affecting it. RESULTS After treatment, YGJ significantly reversed the weight loss caused by DEX; increase the number of bone trabecular in ROI region, significantly improve the bone-related parameters of GIOP mice, and increase the levels of alkaline phosphatase and osteocalcin. In the study of metabolic mechanism, YGJ reversed 24 potential markers in GIOP mice. These included cortisol, 3-hydroxybutyric acid, taurine, esculin and uric acid, which are closely associated with osteoporosis. Topological analysis results showed that YGJ had the most significant effect on taurine and hypotaurine metabolism, with - log10 (P) > 2.0 and Impact > 0.4. CONCLUSIONS Yi-Guan-Jian decoction can increase bone density and improve bone microstructure by regulating the levels of alkaline phosphatase and osteocalcin and reverse bone loss in GIOP mouse model. The underlying metabolic mechanism may be related to taurine and hypotaurine metabolic pathway.
Collapse
Affiliation(s)
- Mengxing Yin
- Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Dezhi Zhou
- Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Fu Jia
- Department of Orthopedics, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China.
| | - Xiaosan Su
- Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Xiufang Li
- West Yunnan University of Applied Sciences, Dali, Yunnan, China
| | - Ruifen Sun
- Yunnan University of Chinese Medicine, Kunming, Yunnan, China
| | - Junmin Li
- Department of Orthopedics, Yan'an Hospital Affiliated to Kunming Medical University, Kunming, Yunnan, China.
| |
Collapse
|
5
|
Wang H, Luo Y, Wang H, Li F, Yu F, Ye L. Mechanistic advances in osteoporosis and anti-osteoporosis therapies. MedComm (Beijing) 2023; 4:e244. [PMID: 37188325 PMCID: PMC10175743 DOI: 10.1002/mco2.244] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/15/2023] [Accepted: 03/06/2023] [Indexed: 05/17/2023] Open
Abstract
Osteoporosis is a type of bone loss disease characterized by a reduction in bone mass and microarchitectural deterioration of bone tissue. With the intensification of global aging, this disease is now regarded as one of the major public health problems that often leads to unbearable pain, risk of bone fractures, and even death, causing an enormous burden at both the human and socioeconomic layers. Classic anti-osteoporosis pharmacological options include anti-resorptive and anabolic agents, whose ability to improve bone mineral density and resist bone fracture is being gradually confirmed. However, long-term or high-frequency use of these drugs may bring some side effects and adverse reactions. Therefore, an increasing number of studies are devoted to finding new pathogenesis or potential therapeutic targets of osteoporosis, and it is of great importance to comprehensively recognize osteoporosis and develop viable and efficient therapeutic approaches. In this study, we systematically reviewed literatures and clinical evidences to both mechanistically and clinically demonstrate the state-of-art advances in osteoporosis. This work will endow readers with the mechanistical advances and clinical knowledge of osteoporosis and furthermore present the most updated anti-osteoporosis therapies.
Collapse
Affiliation(s)
- Haiwei Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- Department of EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Yuchuan Luo
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- Department of EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Haisheng Wang
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Feifei Li
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Fanyuan Yu
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- Department of EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| | - Ling Ye
- State Key Laboratory of Oral Diseases & National Clinical Research Center for Oral DiseasesWest China Hospital of StomatologySichuan UniversityChengduChina
- Department of EndodonticsWest China Hospital of StomatologySichuan UniversityChengduChina
| |
Collapse
|
6
|
Xu K, Zhang L, Yu N, Ren Z, Wang T, Zhang Y, Zhao X, Yu T. Effects of advanced glycation end products (AGEs) on the differentiation potential of primary stem cells: a systematic review. Stem Cell Res Ther 2023; 14:74. [PMID: 37038234 PMCID: PMC10088298 DOI: 10.1186/s13287-023-03324-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 03/27/2023] [Indexed: 04/12/2023] Open
Abstract
The formation and accumulation of advanced glycation end products (AGEs) have been associated with aging and the development, or worsening, of many degenerative diseases, such as atherosclerosis, chronic kidney disease, and diabetes. AGEs can accumulate in a variety of cells and tissues, and organs in the body, which in turn induces oxidative stress and inflammatory responses and adversely affects human health. In addition, under abnormal pathological conditions, AGEs create conditions that are not conducive to stem cell differentiation. Moreover, an accumulation of AGEs can affect the differentiation of stem cells. This, in turn, leads to impaired tissue repair and further aggravation of diabetic complications. Therefore, this systematic review clearly outlines the effects of AGEs on cell differentiation of various types of primary isolated stem cells and summarizes the possible regulatory mechanisms and interventions. Our study is expected to reveal the mechanism of tissue damage caused by the diabetic microenvironment from a cellular and molecular point of view and provide new ideas for treating complications caused by diabetes.
Collapse
Affiliation(s)
- Kuishuai Xu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Liang Zhang
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Ning Yu
- Department of Abdominal Ultrasound, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Zhongkai Ren
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Tianrui Wang
- Department of Traumatology, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Yingze Zhang
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xia Zhao
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| | - Tengbo Yu
- Department of Sports Medicine, The Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China.
| |
Collapse
|
7
|
Hou JL, Yang WY, Zhang Q, Feng H, Wang XB, Li H, Zhou S, Xiao SM. Integration of Metabolomics and Transcriptomics to Reveal the Metabolic Characteristics of Exercise-Improved Bone Mass. Nutrients 2023; 15:nu15071694. [PMID: 37049535 PMCID: PMC10097349 DOI: 10.3390/nu15071694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 03/25/2023] [Accepted: 03/28/2023] [Indexed: 04/03/2023] Open
Abstract
(1) Background: Exercise is effective in promoting and maintaining bone mass. The aim of this study was to detect the exercise-induced metabolic changes in bone tissue of zebrafish. (2) Methods: Thirty-eight zebrafish (Danio rerio, six months old) were analyzed. The exercise group (n = 19) received 8 weeks of counter-current swimming training. The control group (n = 19) was not subjected to exercise. Mineralization was quantified, and alkaline phosphatase (Alp) and anti-tartrate acid phosphatase (Trap) activities were estimated (n = 12). The metabolomics (n = 12) and transcriptomics (n = 14) data of bone tissue were used for the integration analyses. (3) Results: The results showed that the exercise training improved the bone mineralization of zebrafish, e.g., the exercise group (5.74 × 104 ± 7.63 × 103) had a higher mean optical density than the control group (5.26 × 104 ± 8.56 × 103, p = 0.046) for the caudal vertebrae. The amount of mineralized matrix in scales of the exercised zebrafish was also higher (0.156 ± 0.012 vs. 0.102 ± 0.003, p = 0.005). Both histological staining and biochemical analysis revealed increased Alp activity (0.81 ± 0.26 vs. 0.76 ± 0.01, p = 0.002) and decreased Trap activity (1.34 ± 0.01 vs. 1.36 ± 0.01, p = 0.005) in the exercise group. A total of 103 different metabolites (DMs, VIP ≥ 1, fold change (FC) ≥ 1.20 or ≤0.83, p < 0.050) were identified. Alanine, aspartate and glutamate metabolism, β-alanine metabolism, pyrimidine metabolism, and pantothenate and CoA biosynthesis were the significantly enriched metabolic pathways (p < 0.050). A total of 35 genes (q ≤ 0.050 (BH), |Log2FC| ≥ 0.5) were coenriched with the 103 DMs in the four identified pathways. Protein–protein interaction network analysis of the 35 genes showed that entpd3, entpd1, and cmpk2 were the core genes. (4) Conclusions: The results of this study suggest that alanine, aspartate and glutamate metabolism, β-alanine metabolism, pyrimidine metabolism, and pantothenate and CoA biosynthesis contributed to exercise-induced improvements in bone mass.
Collapse
Affiliation(s)
- Jin-Li Hou
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Wan-Yu Yang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Qiong Zhang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Hao Feng
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Xiao-Bao Wang
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Hui Li
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
| | - Sheng Zhou
- College of Marine Sciences, South China Agricultural University, Guangzhou 510642, China
- Correspondence: (S.Z.); (S.-M.X.); Tel.: +86-20-8757-7692 (S.Z.); +86-20-8733-0151 (S.-M.X.)
| | - Su-Mei Xiao
- Department of Epidemiology, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, School of Public Health, Sun Yat-sen University, Guangzhou 510080, China
- Correspondence: (S.Z.); (S.-M.X.); Tel.: +86-20-8757-7692 (S.Z.); +86-20-8733-0151 (S.-M.X.)
| |
Collapse
|
8
|
He C, Lv Q, Liu Z, Long S, Li H, Xiao Y, Yang X, Liu Y, Liu C, Wang Z. Random and aligned electrostatically spun PLLA nanofibrous membranes enhance bone repair in mouse femur midshaft defects. J Biomater Appl 2023; 37:1582-1592. [PMID: 36662630 DOI: 10.1177/08853282221144220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Long-segment bone defects are a common clinical challenge and abstract biomaterials are a promising therapy. Poly-L-lactic acid (PLLA) nanofibrous membranes prepared by electrostatic spinning have a good bone repair potential. However, there are random and aligned surface morphologies of electrostatic spun PLLA nanofibrous membranes, which can affect the migration, proliferation, and differentiation ability of cells. The role of surface morphology in the repair of long bone defects in vivo is currently unknown. In this study, random and aligned electrostatically spun PLLA nanofibrous membranes were prepared, characterised, and implanted into a femur midshaft defect mouse model. The ability of electrostatically spun PLLA nanofibrous membranes to enhance bone repair was tested using X-ray photography, high-resolution micro-computed tomography (micro-CT), and pathological section specimens. The results showed that both random and aligned electrostatically spun PLLA nanofibrous membranes enhanced bone regeneration at bone defects, but the aligned ones exhibited superior results. These results provide a theoretical basis for engineering the surface morphology of bone repair materials.
Collapse
Affiliation(s)
- Chengkai He
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,The Basic Medical School of Kunming Medical University, Kunming, China
| | - Qiong Lv
- Outpatient Department, The Second Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhui Liu
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Shengyu Long
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Haohan Li
- The First Clinical College of Kunming Medical University, Kunming, China
| | - Ya Xiao
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Xingyu Yang
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Yuhang Liu
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Cai Liu
- The Basic Medical School of Kunming Medical University, Kunming, China
| | - Zhihua Wang
- Trauma Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China.,Yunnan Provincial Clinical Medical for Bone and Joint Diseases, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
9
|
Li D, Wu Y, Shi K, Shao M, Duan Y, Yu M, Feng C. Untargeted metabolomics reveals the effect of rearing systems on bone quality parameters in chickens. Front Genet 2023; 13:1071562. [PMID: 36685899 PMCID: PMC9846032 DOI: 10.3389/fgene.2022.1071562] [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: 10/16/2022] [Accepted: 12/15/2022] [Indexed: 01/05/2023] Open
Abstract
The objective of this study was to investigate the effects of rearing systems on the bone quality parameters in chickens using a metabolomics strategy. A total of 419 male one-day-old chicks were randomly allocated to two groups, a floor rearing group (FRG, n = 173) and a cage rearing group (CRG, n = 246). At 6, 8, 10, and 12 weeks of age, all chickens were radiographed by a digital X-ray machine, and body weight was recorded. At 12 weeks of age, 12 birds were selected from each group to obtain tibia and femur, and bone quality parameters of bone mineral density (BMD), mineral content (BMC), breaking strength (BBS), stiffness, Young's modulus (YM), ash content, calcium content, and phosphorus content were determined. An untargeted metabolomics assay was performed to identify changes in the serum metabolic profile (n = 8 birds/group). The results showed that cage-reared chickens had wider tibiae and greater body weight compared with floor-reared chickens. There were no significant differences in BMC or BBS between the two groups (p > 0.05), but BMD, ash content, calcium content, and phosphorus content of the tibia and femur of FRG were significantly higher than those of CRG (p < 0.05). Greater stiffness and YM of the femur were also observed in birds raised in the FRG compared with those raised in the CRG (p < 0.05). Taken together, the results suggest that rearing systems affected bone quality parameters. Furthermore, 148 and 149 differential metabolites were identified in positive and negative ion modes by LC-MS/MS analysis, among which 257 metabolites were significantly correlated with 16 bone quality parameters, including leucine, myristoleic acid, glycocholic acid, and N-phenylacetamide. KEGG analysis indicated that 15 metabolic pathways, including six pathways of amino acid metabolism, two pathways of lipid metabolism, and two pathways of carbohydrate metabolism, were responsible for bone quality. Overall, the present study demonstrated the effect of rearing systems on bone quality parameters, and identified several metabolites and metabolic pathways associated with bone quality parameters.
Collapse
|
10
|
Zhao Z, Cai Z, Chen A, Cai M, Yang K. Application of metabolomics in osteoporosis research. Front Endocrinol (Lausanne) 2022; 13:993253. [PMID: 36452325 PMCID: PMC9702081 DOI: 10.3389/fendo.2022.993253] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Accepted: 10/28/2022] [Indexed: 11/15/2022] Open
Abstract
Osteoporosis (OP) is a systemic disease characterized by bone metabolism imbalance and bone microstructure destruction, which causes serious social and economic burden. At present, the diagnosis and treatment of OP mainly rely on imaging combined with drugs. However, the existing pathogenic mechanisms, diagnosis and treatment strategies for OP are not clear and effective enough, and the disease progression that cannot reflect OP further restricts its effective treatment. The application of metabolomics has facilitated the study of OP, further exploring the mechanism and behavior of bone cells, prevention, and treatment of the disease from various metabolic perspectives, finally realizing the possibility of a holistic approach. In this review, we focus on the application of metabolomics in OP research, especially the newer systematic application of metabolomics and treatment with herbal medicine and their extracts. In addition, the prospects of clinical transformation in related fields are also discussed. The aim of this study is to highlight the use of metabolomics in OP research, especially in exploring the pathogenesis of OP and the therapeutic mechanisms of natural herbal medicine, for the benefit of interdisciplinary researchers including clinicians, biologists, and materials engineers.
Collapse
Affiliation(s)
- Zhenyu Zhao
- Department of Orthopaedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- 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, Shanghai, China
| | - Zhengwei Cai
- 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, Shanghai, China
| | - Aopan Chen
- Department of Orthopaedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Ming Cai
- Department of Orthopaedics, Shanghai Tenth People’s Hospital, School of Medicine, Tongji University, Shanghai, China
- *Correspondence: Ming Cai, ; Kai Yang,
| | - Kai Yang
- 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, Shanghai, China
- *Correspondence: Ming Cai, ; Kai Yang,
| |
Collapse
|