1
|
Lee GL, Hu CC, Chen MF, Chang Y, Lin YC, Wu YY, Hsu YH. Inosine from purine metabolism enhances fracture healing by coupling fibrinolysis and angiogenesis of type H vessels. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167818. [PMID: 40157654 DOI: 10.1016/j.bbadis.2025.167818] [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: 12/03/2024] [Revised: 02/24/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025]
Abstract
Fibrinolysis-angiogenesis coupling is crucial for successful fracture healing, in which type H vessels play an indispensable role. However, the metabolic control of fibrinolysis-type H angiogenesis coupling in fracture healing remains unclear. We used a metabolomics approach to gain metabolic insights from mouse fracture models (0.3 mm and 1.0 mm femur defects). Furthermore, human umbilical vein endothelial cells (HUVECs) and MC3T3-E1 cells were employed as in vitro models to examine the effects of identified metabolites on endothelial events and osteogenesis, respectively. CD31 and endomucin (Emcn) were used for detecting type H markers, and tissue-type plasminogen activator (tPA) and plasminogen activator inhibitor-1 (PAI-1) for fibrinolysis. A femur defect of 1.5 mm in mice was included to validate the therapeutic potential of identified metabolites by bone imaging and micro-CT. Purine metabolism is the most significant pathway in fracture healing; three purinergic metabolites, adenosine, adenine, and inosine, are regulated in mouse serum. According to the in vivo results, CD31HiEmcnHi type H vessels are upregulated near the defect site in mouse femur and are associated with tPA expression, but not PAI-1. Additionally, in vitro experiments examining the endothelial functions of HUVECs demonstrated that these three metabolites promote cell migration and tube formation rather than proliferation. Fibrinolytic activity and type H phenotype in HUVECs were induced only by inosine through activation of the adenosine A2A receptor. Inosine, regulated during fracture healing, has the capacity to synchronously induce fibrinolysis and type H phenotype in line with osteogenesis, indicating its role in enhancing fracture healing.
Collapse
Affiliation(s)
- Guan-Lin Lee
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Chih-Chien Hu
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Mei-Feng Chen
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yuhan Chang
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Yu-Chih Lin
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
| | - Ying-Yu Wu
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan
| | - Yung-Heng Hsu
- Bone and Joint Research Center, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; Department of Orthopedic Surgery, Chang Gung Memorial Hospital, Taoyuan 33305, Taiwan; College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan.
| |
Collapse
|
2
|
Du Y, Xiao X, Liu F, Zhu W, Mo J, Liu Z. Causal effects of metabolites on malignant neoplasm of bone and articular cartilage: a mendelian randomization study. Front Genet 2025; 16:1366743. [PMID: 40098980 PMCID: PMC11911353 DOI: 10.3389/fgene.2025.1366743] [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: 01/07/2024] [Accepted: 02/07/2025] [Indexed: 03/19/2025] Open
Abstract
Objective Previous research has demonstrated that metabolites play a significant role in modulating disease phenotypes; nevertheless, the causal association between metabolites and malignant malignancies of bones and joint cartilage (MNBAC)has not been fully elucidated. Methods This study used two-sample Mendelian randomization (MR) to explore the causal correlation between 1,400 metabolites and MNBAC. Data from recent genome-wide association studies (GWAS) involving 8,299 individuals were summarized. The GWAS summary data for metabolites were acquired from the IEU Open GWAS database, while those for MNBAC were contributed by the Finnish Consortium. We employed eight distinct MR methodologies: simple mode, maximum likelihood estimator, MR robust adjusted profile score, MR-Egger, weighted mode, weighted median, MR-PRESSO and inverse variance weighted to scrutinize the causal association between metabolites engendered by each gene and MNBAC. Consequently, we evaluated outliers, horizontal pleiotropy, heterogeneity, the impact of single nucleotide polymorphisms (SNPs), and adherence to the normal distribution assumption in the MR analysis. Results Our findings suggested a plausible causative relationship between N-Formylmethionine (FMet) levels, lignoceroylcarnitine (C24) levels, and MNBAC. We observed a nearly significant causal association between FMet levels and MNBAC within the cohort of 1,400 metabolites (P = 0.024, odds ratio (OR) = 3.22; 95% CI [1.16-8.92]). Moreover, we ascertained a significant causal link between levels of C24 and MNBAC (P = 0.0009; OR = 0.420; 95%CI [0.25-0.70]). These results indicate a potential causative relationship between FMet, C24 level and MNBAC. Conclusion The occurrence of MNBAC may be causally related to metabolites. This might unveil new possibilities for investigating early detection and treatment of MNBAC.
Collapse
Affiliation(s)
- Yongwei Du
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiqiu Xiao
- Department of Orthopedics, 8th People Hospital of Nankang, Ganzhou, China
| | - Fuping Liu
- Department of Emergency, Shangyou Hospital of Traditional Chinese Medicine, Ganzhou, China
| | - Wenqing Zhu
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jianwen Mo
- Department of Orthopedics, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Zhen Liu
- Department of Rehabilitation, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
3
|
Yu H, Yang S, Jiang T, Li T, Duan H, Li M. Repair mechanisms of bone system tissues based on comprehensive perspective of multi-omics. Cell Biol Toxicol 2025; 41:45. [PMID: 39966216 PMCID: PMC11836151 DOI: 10.1007/s10565-025-09995-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 01/28/2025] [Indexed: 02/20/2025]
Abstract
Bone disorders affect more than half of the adult population worldwide who may have a poor quality of life and physical independence worldwide. Multi-omic techniques are increasingly adopted and applied to determine the molecular mechanisms of bone tissue repair, providing perspective towards personalized medical intervention. Data from genomics, epigenomics, transcriptomics, proteomics, glycomics, and lipidomics were combined to elucidate dynamic processes in bone repair. In this narrative review, the key role of genetic and epigenetic factors in regulating injured cellular responses is highlighted, and changes in RNA and protein expression during the healing phase, as well as glucolipid metabolism adaptation, are described in detail how the repair process is affected. In a word, the integration of multi-omic techniques in this review not only benefits the comprehensive identification of new biomarkers, but also facilitates the development of personalized treatment strategies of bone disorders to revolutionize regenerative medicine.
Collapse
Affiliation(s)
- Honghao Yu
- Departments of Spine Surgery, Shengjing Hospital of China Medical University, Shengyang, China
| | - Shize Yang
- Department of Thoracic Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Tianlong Jiang
- Department of Orthopedic Surgery, First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Tian Li
- Tianjin Key Laboratory of Acute Abdomen Disease-Associated Organ Injury and ITCWM Repair, Institute of Integrative Medicine of Acute Abdominal Diseases, Tianjin Nankai Hospital, Tianjin Medical University, 8 Changjiang Avenue, Tianjin, 300100, China.
| | - Hongmei Duan
- Department of Rheumatology and Immunology, First Affiliated Hospital of China Medical University, Shenyang, 110001, China.
| | - Minglei Li
- Department of Pediatric Orthopaedics, Shengjing Hospital of China Medical University, 36 Sanhao St, Shenyang, 110004, China.
| |
Collapse
|
4
|
Casarrubios L, Cicuéndez M, Polo-Montalvo A, Feito MJ, Martínez-Del-Pozo Á, Arcos D, Duarte IF, Portolés MT. Metabolomic characterization of MC3T3-E1pre-osteoblast differentiation induced by ipriflavone-loaded mesoporous nanospheres. BIOMATERIALS ADVANCES 2025; 166:214085. [PMID: 39490191 DOI: 10.1016/j.bioadv.2024.214085] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024]
Abstract
This study reports on the metabolic changes accompanying the differentiation of MC3T3-E1 osteoprogenitor cells induced by mesoporous bioactive glass nanospheres (nMBG) loaded with ipriflavone (nMBG-IP). Ipriflavone (IP) is a synthetic isoflavone known for inhibiting bone resorption, maintaining bone density, and preventing osteoporosis. Delivering IP intracellularly is a promising strategy to modulate bone remodeling at significantly lower doses compared to free drug administration. Our results demonstrate that nMBG are efficiently internalized by pre-osteoblasts and, when loaded with IP, induce their differentiation. This differentiation process is accompanied by pronounced metabolic alterations, as monitored by NMR analysis of medium supernatants and cell extracts (exo- and endo-metabolomics, respectively). The main effects include an early-stage intensification of glycolysis and changes in several metabolic pathways, such as nucleobase metabolism, osmoregulatory and antioxidant pathways, and lipid metabolism. Notably, the metabolic impacts of nMBG-IP and free IP were very similar, whereas nMBG alone induced only mild changes in the intracellular metabolic profile without affecting the cells' consumption/secretion patterns or lipid composition. This finding indicates that the observed effects are primarily related to IP-induced differentiation and that nMBG nanospheres serve as convenient carriers with both efficient internalization and minimal metabolic impact. Furthermore, the observed link between pre-osteoblast differentiation and metabolism underscores the potential of utilizing metabolites and metabolic reprogramming as strategies to modulate the osteogenic process, for instance, in the context of osteoporosis and other bone diseases.
Collapse
Affiliation(s)
- Laura Casarrubios
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Mónica Cicuéndez
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain; Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Alberto Polo-Montalvo
- Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain; Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - María José Feito
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain
| | - Álvaro Martínez-Del-Pozo
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain
| | - Daniel Arcos
- Departamento de Química en Ciencias Farmacéuticas, Facultad de Farmacia, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria Hospital 12 de Octubre i + 12, Plaza Ramón y Cajal s/n, Madrid 28040, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, ISCIII, Madrid 28040, Spain
| | - Iola F Duarte
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro 3810-193, Portugal
| | - María Teresa Portolés
- Departamento de Bioquímica y Biología Molecular, Facultad de Ciencias Químicas, Universidad Complutense de Madrid, Madrid 28040, Spain; Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid 28040, Spain; CIBER de Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, ISCIII, Madrid 28040, Spain.
| |
Collapse
|
5
|
Li Q, Wang J, Zhao C. From Genomics to Metabolomics: Molecular Insights into Osteoporosis for Enhanced Diagnostic and Therapeutic Approaches. Biomedicines 2024; 12:2389. [PMID: 39457701 PMCID: PMC11505085 DOI: 10.3390/biomedicines12102389] [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] [Received: 09/16/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 10/28/2024] Open
Abstract
Osteoporosis (OP) is a prevalent skeletal disorder characterized by decreased bone mineral density (BMD) and increased fracture risk. The advancements in omics technologies-genomics, transcriptomics, proteomics, and metabolomics-have provided significant insights into the molecular mechanisms driving OP. These technologies offer critical perspectives on genetic predispositions, gene expression regulation, protein signatures, and metabolic alterations, enabling the identification of novel biomarkers for diagnosis and therapeutic targets. This review underscores the potential of these multi-omics approaches to bridge the gap between basic research and clinical applications, paving the way for precision medicine in OP management. By integrating these technologies, researchers can contribute to improved diagnostics, preventative strategies, and treatments for patients suffering from OP and related conditions.
Collapse
Affiliation(s)
- Qingmei Li
- Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| | - Jihan Wang
- Institute of Medical Research, Northwestern Polytechnical University, Xi’an 710072, China
| | - Congzhe Zhao
- Honghui Hospital, Xi’an Jiaotong University, Xi’an 710054, China
| |
Collapse
|
6
|
Lv H, Zhang G, Hu ZM, Chu QS, Wang JX, Jiang T. Dissecting Causal Relationships Between Plasma Metabolites and Osteoporosis: A Bidirectional Mendelian Randomization Study. CHINESE MEDICAL SCIENCES JOURNAL = CHUNG-KUO I HSUEH K'O HSUEH TSA CHIH 2024; 39:182-188. [PMID: 39275811 DOI: 10.24920/004356] [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] [Indexed: 09/16/2024]
Abstract
OBJECTIVES To investigate the causal relationships between plasma metabolites and osteoporosis via Mendelian randomization (MR) analysis. METHODS Bidirectional MR was used to analyze pooled data from different genome-wide association studies (GWAS). The causal effect of plasma metabolites on osteoporosis was estimated using the inverse variance weighted method, intersections of statistically significant metabolites obtained from different sources of osteoporosis-related GWAS aggregated data was determined, and then sensitivity analysis was performed on these metabolites. Heterogeneity between single nucleotide polymorphisms was evaluated by Cochran's Q test. Horizontal pleiotropy was assessed through the application of the MR-Egger intercept method and the MR-PRESSO method. The causal effect of osteoporosis on plasma metabolites was also evaluated using the inverse variance weighted method. Additionally, pathway analysis was conducted to identify potential metabolic pathways involved in the regulation of osteoporosis. RESULTS Primary analysis and sensitivity analysis showed that 77 and 61 plasma metabolites had a causal relationship with osteoporosis from the GWAS data in the GCST90038656 and GCST90044600 datasets, respectively. Five common metabolites were identified via intersection. X-13684 levels and the glucose-to-maltose ratio were negatively associated with osteoporosis, whereas glycoursodeoxycholate levels and arachidoylcarnitine (C20) levels were positively associated with osteoporosis (all P < 0.05). The relationship between X-11299 levels and osteoporosis showed contradictory results (all P < 0.05). Pathway analysis indicated that glycine, serine, and threonine metabolism, valine, leucine, and isoleucine biosynthesis, galactose metabolism, arginine biosynthesis, and starch and sucrose metabolism pathways were participated in the development of osteoporosis. CONCLUSIONS We found a causal relationship between plasma metabolites and osteoporosis. These results offer novel perspectives with important implications for targeted metabolite-focused interventions in the management of osteoporosis.
Collapse
Affiliation(s)
- Hao Lv
- Department of Orthopaedics, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China
- Graduate School of Anhui University of Chinese Medicine, Hefei 230012, China
| | - Ge Zhang
- Department of Geriatrics, The Third People's Hospital of Hefei, Hefei 230041, China
| | - Zhi-Mu Hu
- Graduate School of Anhui University of Chinese Medicine, Hefei 230012, China
| | - Qing-Song Chu
- Graduate School of Anhui University of Chinese Medicine, Hefei 230012, China
| | - Jiu-Xiang Wang
- Experimental Center of Clinical Research, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China.
| | - Ting Jiang
- Department of Orthopaedics, The First Affiliated Hospital of Anhui University of Chinese Medicine, Hefei 230031, China.
| |
Collapse
|
7
|
Cheng X, Zhang L, Cao B, Zhao C. Correlation between serum hemoglobin levels and bone mineral density in adults: A cross-sectional study. Medicine (Baltimore) 2024; 103:e39612. [PMID: 39287248 PMCID: PMC11404974 DOI: 10.1097/md.0000000000039612] [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: 05/02/2024] [Revised: 06/14/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Serum hemoglobin plays an important role in bone metabolism. However, the association between serum hemoglobin levels and bone mineral density (BMD) remains unclear. Therefore, this study aimed to explore the relationship between serum hemoglobin levels and lumbar spine BMD in adults. We conducted a cross-sectional study by utilizing data from the National Health and Nutrition Examination Survey from 2011 to 2018. The serum hemoglobin level was examined as an independent variable, while the lumbar spine BMD was utilized as the dependent variable. Weighted multivariate linear regression models and stratified analysis by age, sex, and race/ethnicity were applied after controlling for confounding factors to assess the relationship between serum hemoglobin levels and the lumbar spine BMD. Additionally, smooth curve fitting and threshold effect analyses were utilized to depict the nonlinear relationship between the 2 variables. A total of 11,658 participants (6004 men and 5654 women) aged ≥ 18 years were included in this study. When the serum hemoglobin level was represented as a continuous variable and fully adjusted in the regression model, the hemoglobin level was significantly negatively correlated with the lumbar spine BMD (β = -0.0035, 95% confidence interval: -0.0065 to -0.0004, P = .024555); this significant negative correlation persisted when the serum hemoglobin level was transformed into a categorical variable, except in the Q2 group (β = -0.0046, 95% confidence interval: -0.0142 to -0.0050, P = .348413). When different confounding factors were used including sex, age, and race/ethnicity, the stratified subgroups exhibited a negative correlation between the serum hemoglobin level and the lumbar spine BMD. Additionally, smooth curve fitting and threshold effect analyses showed a negative correlation between the serum hemoglobin level and the lumbar spine BMD, with a saturation effect at 15 g/dL. Our findings demonstrated an association between hemoglobin levels and the lumbar spine BMD in adults, characterized by a nonlinear relationship. Thus, monitoring the serum hemoglobin level could aid in the early detection of risks associated with bone metabolic disorders such as osteoporosis.
Collapse
Affiliation(s)
- Xiaosong Cheng
- Medical School of Yan’an University, Yan’an, Shaanxi, China
| | - Liangliang Zhang
- Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Bo Cao
- Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| | - Chengjin Zhao
- Affiliated Hospital of Yan’an University, Yan’an, Shaanxi, China
| |
Collapse
|
8
|
Bonicelli A, Taylor G, Procopio N. Extraction and untargeted analysis of metabolome from undemineralised cortical bone matrix. Mol Omics 2024. [PMID: 39073399 DOI: 10.1039/d4mo00015c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Liquid chromatography coupled with tandem mass spectrometry (LC-MS/MS) untargeted metabolomics has become the gold standard for the profiling of low-molecular-weight compounds. Recently, this discipline has raised great interest in forensic sciences, especially in the field of toxicology and for post-mortem interval estimation. The current study aims at evaluating three extraction protocols and two LC-MS/MS assays run in both positive and negative modes, to identify the most suitable method to conduct post-mortem metabolomic profiling of bone tissue. A fragment of the anterior tibia of a 82 years-old male sampled from a human taphonomy facility was powdered via freeze-milling. The powdered sub-samples were extracted in five replicates per protocol. Methods tested were (I) a biphasic chloroform-methanol-water protocol, (II) a single phase methanol-water protocol, and (III) a single phase methanol-acetonitrile-water protocol. LC-MS/MS analyses were carried out via high performance liquid chromatography, either on hydrophilic interaction (HILIC) or on reversed-phase (C18) columns in both positive and negative ionisation modes, coupled with a Q-TOF mass spectrometer. Results suggest that the highest consistency between replicates and quality control samples was obtained with the single phase extractions (i.e., methanol-acetonitrile-water), whilst the ideal combination of instrumental set up HILIC chromatography in positive ionisation mode and of C18 chromatography in negative ionisation mode. For the purpose of forensic investigations, a combination of a single phase extraction and the two aforementioned chromatographic and mass spectrometry modes could represent an ideal set up for obtaining bone metabolomic profiles from taphonomically altered bones.
Collapse
Affiliation(s)
- Andrea Bonicelli
- School of Law and Policing, Research Centre for Field Archaeology and Forensic Taphonomy, University of Central Lancashire, Preston, UK.
| | - George Taylor
- Biological Mass Spectrometry (BioMS) Core Facility, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, M13 9PT, UK
| | - Noemi Procopio
- School of Law and Policing, Research Centre for Field Archaeology and Forensic Taphonomy, University of Central Lancashire, Preston, UK.
| |
Collapse
|
9
|
Malvandi AM, Halilaj E, Faraldi M, Mangiavini L, Cristoni S, Leoni V, Lombardi G. Enhanced molecular release from elderly bone samples using collagenase I: insights into fatty acid metabolism alterations. J Transl Med 2024; 22:143. [PMID: 38336738 PMCID: PMC10858523 DOI: 10.1186/s12967-024-04948-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 02/03/2024] [Indexed: 02/12/2024] Open
Abstract
BACKGROUND Bone is a metabolically active tissue containing different cell types acting as endocrine targets and effectors. Further, bone is a dynamic depot for calcium, phosphorous and other essential minerals. The tissue matrix is subjected to a constant turnover in response to mechanical/endocrine stimuli. Bone turnover demands high energy levels, making fatty acids a crucial source for the bone cells. However, the current understanding of bone cell metabolism is poor. This is partly due to bone matrix complexity and difficulty in small molecules extraction from bone samples. This study aimed to evaluate the effect of metabolite sequestering from a protein-dominated matrix to increase the quality and amount of metabolomics data in discovering small molecule patterns in pathological conditions. METHODS Human bone samples were collected from 65 to 85 years old (the elderly age span) patients who underwent hip replacement surgery. Separated cortical and trabecular bone powders were treated with decalcifying, enzymatic (collagenase I and proteinase K) and solvent-based metabolite extraction protocols. The extracted mixtures were analyzed with the high-resolution mass spectrometry (HRMS). Data analysis was performed with XCMS and MetaboAnalystR packages. RESULTS Fast enzymatic treatment of bone samples before solvent addition led to a significantly higher yield of metabolite extraction. Collagenase I and proteinase K rapid digestion showed more effectiveness in cortical and trabecular bone samples, with a significantly higher rate (2.2 folds) for collagenase I. Further analysis showed significant enrichment in pathways like de novo fatty acid biosynthesis, glycosphingolipid metabolism and fatty acid oxidation-peroxisome. CONCLUSION This work presents a novel approach for bone sample preparation for HRMS metabolomics. The disruption of bone matrix conformation at the molecular level helps the molecular release into the extracting solvent and, therefore, can lead to higher quality results and trustable biomarker discovery. Our results showed β-oxidation alteration in the aged bone sample. Future work covering more patients is worthy to identify the effective therapeutics to achieve healthy aging.
Collapse
Affiliation(s)
- Amir Mohammad Malvandi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy.
| | - Esra Halilaj
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Martina Faraldi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy
| | - Laura Mangiavini
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy
- Department of Biomedical Sciences for Health, University of Milan, Milan, Italy
| | | | - Valerio Leoni
- Department of Laboratory Medicine, University of Milano-Bicocca, Azienda Socio Sanitaria Territoriale Della Brianza, ASST-Brianza, Desio Hospital, Desio, Italy
| | - Giovanni Lombardi
- Laboratory of Experimental Biochemistry & Molecular Biology, IRCCS Istituto Ortopedico Galeazzi, Via Cristina Belgioioso 173, 20157, Milan, Italy
- Department of Athletics, Strength and Conditioning, Poznań University of Physical Education, Poznań, Poland
| |
Collapse
|
10
|
Ning T, Guo H, Ma M, Zha Z. BRD4 facilitates osteogenic differentiation of human bone marrow mesenchymal stem cells through WNT4/NF-κB pathway. J Orthop Surg Res 2023; 18:876. [PMID: 37980502 PMCID: PMC10656925 DOI: 10.1186/s13018-023-04335-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Accepted: 11/01/2023] [Indexed: 11/20/2023] Open
Abstract
BACKGROUND Human bone marrow mesenchymal stem cells (hBMSCs) are a major source of osteoblast precursor cells and are directly involved in osteoporosis (OP) progression. Bromodomain-containing protein 4 (BRD4) is an important regulator for osteogenic differentiation. Therefore, its role and mechanism in osteogenic differentiation process deserve further investigation. METHODS hBMSCs osteogenic differentiation was evaluated by flow cytometry, alkaline phosphatase assay and alizarin red staining. Western blot was used to test osteogenic differentiation-related proteins, BRD4 protein, WNT family members-4 (WNT4)/NF-κB-related proteins, and glycolysis-related proteins. Metabolomics techniques were used to detect metabolite changes and metabolic pathways. BRD4 and WNT4 mRNA levels were determined using quantitative real-time PCR. Dual-luciferase reporter assay and chromatin immunoprecipitation assay were performed to detect BRD4 and WNT4 interaction. Glycolysis ability was assessed by testing glucose uptake, lactic acid production, and ATP levels. RESULTS After successful induction of osteogenic differentiation, the expression of BRD4 was increased significantly. BRD4 knockdown inhibited hBMSCs osteogenic differentiation. Metabolomics analysis showed that BRD4 expression was related to glucose metabolism in osteogenic differentiation. Moreover, BRD4 could directly bind to the promoter of the WNT4 gene. Further experiments confirmed that recombinant WNT4 reversed the inhibition effect of BRD4 knockdown on glycolysis, and NF-κB inhibitors (Bardoxolone Methyl) overturned the suppressive effect of BRD4 knockdown on hBMSCs osteogenic differentiation. CONCLUSION BRD4 promoted hBMSCs osteogenic differentiation by inhibiting NF-κB pathway via enhancing WNT4 expression.
Collapse
Affiliation(s)
- Tao Ning
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Jinan University, Guangzhou City, 510630, Guangdong Province, People's Republic of China
- Department of Orthopedics, Fuyang People's Hospital, No.501 Sanqing Road, Fuyang City, 236000, Anhui Province, People's Republic of China
| | - Huihui Guo
- Department of Orthopedics, Fuyang People's Hospital, No.501 Sanqing Road, Fuyang City, 236000, Anhui Province, People's Republic of China
| | - Mingming Ma
- Department of Orthopedics, Fuyang People's Hospital, No.501 Sanqing Road, Fuyang City, 236000, Anhui Province, People's Republic of China
| | - Zhengang Zha
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Jinan University, Guangzhou City, 510630, Guangdong Province, People's Republic of China.
| |
Collapse
|
11
|
Liang G, Zhao J, Zhao D, Dou Y, Huang H, Yang W, Zhou G, Gu Z, Pan J, Liu J. Longbie capsules reduce bone loss in the subchondral bone of rats with comorbid osteoporosis and osteoarthritis by regulating metabolite alterations. Front Med (Lausanne) 2023; 10:1256238. [PMID: 37915330 PMCID: PMC10616798 DOI: 10.3389/fmed.2023.1256238] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Accepted: 09/25/2023] [Indexed: 11/03/2023] Open
Abstract
Background and objective With the development of global population aging, comorbidity (≥2 diseases) is a common health problem among elderly people. Osteoarthritis (OA) and osteoporosis (OP) are common in elderly individuals. There is a lack of drug therapy for OA and OP comorbidities. The purpose of this study was to explore the efficacy and mechanism of Longbie capsule (LBJN), which contains various plant herbs, in treating OA and OP comorbidities (OA + OP) in rats using metabolomics techniques. Methods We created an OA + OP rat model through bilateral oophorectomy combined with meniscus instability surgery. Thirty SD rats were randomly divided into five groups (six in each group), namely, the sham group, OA group, OA + OP group, LBJN low-dose group (0.625 g/kg, OA + OP+LB-L group) and LBJN high-dose group (1.25 g/kg, OA + OP+LB-H group). After 8 weeks of intervention, we used micro-CT to detect bone microstructure status, ELISA to measure bone metabolism indicators, and UPLC-MS technology for metabolomics analysis. Finally, the screened differentially expressed metabolites were subjected to Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway and functional enrichment analysis. Results The micro-CT results showed that LBJN significantly improved the bone mineral density (BMD) and bone quality of subchondral bone in OA + OP rats, and LBJN regulated the expression of bone alkaline phosphatase (BALP), osteoprotegerin (OPG), and tartrate-resistant acid phosphatase (TRACP) in serum to maintain bone metabolism balance. Metabolomics analysis showed that the metabolic trajectory of OA + OP rats after intervention in the OA + OP+LB-H group showed significant changes, and 107 potential biomarkers could be identified. Among them, 50 metabolites were upregulated (such as zeranol) and 57 were downregulated (such as vanillactic acid). The KEGG functional enrichment results indicated that the differentially expressed metabolites are mainly involved in amino acid metabolism, lipid metabolism, and carbohydrate metabolism. The KEGG pathway enrichment results indicated that LBJN may exert therapeutic effects on OA + OP rats by regulating the cAMP signaling pathway, and the FoxO signaling pathway. Conclusion LBJN can maintain bone metabolism balance by regulating serum lipid metabolism, amino acid metabolism, carbohydrate metabolism, and estrogen, thereby reducing bone loss in subchondral bone, which may be a potential mechanism through which LBJN treats OA + OP.
Collapse
Affiliation(s)
- Guihong Liang
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Research Team on Bone and Joint Degeneration and Injury of Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Jinlong Zhao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Research Team on Bone and Joint Degeneration and Injury of Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Di Zhao
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yaoxing Dou
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Research Team on Bone and Joint Degeneration and Injury of Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Hetao Huang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Weiyi Yang
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
| | - Guanghui Zhou
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Zhuoxu Gu
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Jianke Pan
- The Second Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Provincial Hospital of Chinese Medicine, Guangzhou, China
- The Research Team on Bone and Joint Degeneration and Injury of Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
| | - Jun Liu
- The Research Team on Bone and Joint Degeneration and Injury of Guangdong Provincial Academy of Chinese Medical Sciences, Guangzhou, China
- The Fifth Clinical College of Guangzhou University of Chinese Medicine, Guangzhou, China
- Guangdong Second Chinese Medicine Hospital (Guangdong Province Enginering Technology Research Institute of Traditional Chinese Medicine), Guangzhou, China
| |
Collapse
|
12
|
Badillo-Sanchez D, Serrano Ruber M, Davies-Barrett A, Jones DJ, Hansen M, Inskip S. Metabolomics in archaeological science: A review of their advances and present requirements. SCIENCE ADVANCES 2023; 9:eadh0485. [PMID: 37566664 PMCID: PMC10421062 DOI: 10.1126/sciadv.adh0485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Accepted: 07/11/2023] [Indexed: 08/13/2023]
Abstract
Metabolomics, the study of metabolites (small molecules of <1500 daltons), has been posited as a potential tool to explore the past in a comparable manner to other omics, e.g., genomics or proteomics. Archaeologists have used metabolomic approaches for a decade or so, mainly applied to organic residues adhering to archaeological materials. Because of advances in sensitivity, resolution, and the increased availability of different analytical platforms, combined with the low mass/volume required for analysis, metabolomics is now becoming a more feasible choice in the archaeological sector. Additional approaches, as presented by our group, show the versatility of metabolomics as a source of knowledge about the human past when using human osteoarchaeological remains. There is tremendous potential for metabolomics within archaeology, but further efforts are required to position it as a routine technique.
Collapse
Affiliation(s)
| | - Maria Serrano Ruber
- School of Archaeology and Ancient History, University of Leicester, Leicester, UK
| | - Anna Davies-Barrett
- School of Archaeology and Ancient History, University of Leicester, Leicester, UK
| | - Donald J. L. Jones
- Leicester Cancer Research Centre, RKCSB, University of Leicester, Leicester, UK
- The Leicester van Geest MultiOmics Facility, University of Leicester, Leicester, UK
| | - Martin Hansen
- Environmental Metabolomics Lab, Department of Environmental Science, Aarhus University, Roskilde, Denmark
| | - Sarah Inskip
- School of Archaeology and Ancient History, University of Leicester, Leicester, UK
| |
Collapse
|
13
|
Murali R, Wanjari UR, Mukherjee AG, Gopalakrishnan AV, Kannampuzha S, Namachivayam A, Madhyastha H, Renu K, Ganesan R. Crosstalk between COVID-19 Infection and Kidney Diseases: A Review on the Metabolomic Approaches. Vaccines (Basel) 2023; 11:vaccines11020489. [PMID: 36851366 PMCID: PMC9959335 DOI: 10.3390/vaccines11020489] [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: 01/15/2023] [Revised: 02/16/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
The novel severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes COVID-19, a respiratory disorder. Various organ injuries have been reported in response to this virus, including kidney injury and, in particular, kidney tubular injury. It has been discovered that infection with the virus does not only cause new kidney disease but also increases treatment difficulty and mortality rates in people with kidney diseases. In individuals hospitalized with COVID-19, urinary metabolites from several metabolic pathways are used to distinguish between patients with acute kidney injury (AKI) and those without. This review summarizes the pathogenesis, pathophysiology, treatment strategies, and role of metabolomics in relation to AKI in COVID-19 patients. Metabolomics is likely to play a greater role in predicting outcomes for patients with kidney disease and COVID-19 with varying levels of severity in the near future as data on metabolic profiles expand rapidly. Here, we also discuss the correlation between COVID-19 and kidney diseases and the available metabolomics approaches.
Collapse
Affiliation(s)
- Reshma Murali
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Uddesh Ramesh Wanjari
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Anirban Goutam Mukherjee
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Abilash Valsala Gopalakrishnan
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
- Correspondence: (A.V.G.); (R.G.)
| | - Sandra Kannampuzha
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Arunraj Namachivayam
- Department of Biomedical Sciences, School of Biosciences and Technology, Vellore Institute of Technology (VIT), Vellore 632014, Tamil Nadu, India
| | - Harishkumar Madhyastha
- Department of Cardiovascular Physiology, Faculty of Medicine, University of Miyazaki, Miyazaki 889-1692, Japan
| | - Kaviyarasi Renu
- Center of Molecular Medicine and Diagnostics (COMMAND), Department of Biochemistry, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600077, Tamil Nadu, India
| | - Raja Ganesan
- Institute for Liver and Digestive Diseases, College of Medicine, Hallym University, Chuncheon 24252, Republic of Korea
- Correspondence: (A.V.G.); (R.G.)
| |
Collapse
|
14
|
Tian J, Bao X, Yang F, Tang X, Jiang Q, Li Y, Yao K, Yin Y. Elevation of Intracellular Alpha-Ketoglutarate Levels Inhibits Osteoclastogenesis by Suppressing the NF-κB Signaling Pathway in a PHD1-Dependent Manner. Nutrients 2023; 15:nu15030701. [PMID: 36771407 PMCID: PMC9921543 DOI: 10.3390/nu15030701] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/20/2023] [Accepted: 01/24/2023] [Indexed: 02/03/2023] Open
Abstract
Age-related osteoporosis, a high-prevalence disease in the aged population, is generally attributed to the excessive activity of osteoclasts. Most approved drugs treat osteoporosis by inhibition of osteoclasts. Although in vivo studies have shown that alpha-ketoglutarate (AKG), an intermediate in the TCA cycle, can ameliorate age-related osteoporosis, the effects of AKG on osteoclastogenesis and the underlying mechanism of its action have not been studied yet. Here, we showed that the elevation of intracellular AKG levels by supplementing dimethyl AKG (DM-AKG, a cell-permeable derivative of AKG) inhibits the receptor activator of NF-κB ligand (RANKL)-induced osteoclasts differentiation from primary bone marrow-derived macrophages (BMMs) and RAW264.7 cells in vitro. We further found that DM-AKG treatment suppresses NF-κB signaling and oxidative phosphorylation (OXPHOS) during RANKL-induced osteoclastogenesis in RAW264.7 cells. Interestingly, dimethyl oxalylglycine (DMOG), an AKG competitive inhibitor of AKG-dependent prolyl hydroxylases (PHDs), antagonizes the suppression of the RANKL-activated NF-κB signaling pathway caused by DM-AKG treatment. Furthermore, blocked PHD1 expression (also known as EglN2), instead of PHD2 or PHD3, was confirmed to reverse the DM-AKG treatment-induced suppression of the RANKL-activated NF-κB signaling pathway. Accordingly, blocked PHD1 expression antagonized the inhibitory effects of DM-AKG on osteoclastogenesis. Together, our finding suggests that the elevation of intracellular AKG levels inhibits osteoclastogenesis by suppressing RANKL-activated NF-κB signaling in a PHD1-dependent manner, which may provide a novel nutritional strategy for osteoporosis treatment.
Collapse
Affiliation(s)
- Junquan Tian
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Xuetai Bao
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Fan Yang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
| | - Xiongzhuo Tang
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410000, China
| | - Qian Jiang
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410000, China
- Correspondence: (Q.J.); (K.Y.)
| | - Yuying Li
- Institute of Bast Fiber Crops, Chinese Academy of Agricultural Sciences, Changsha 410205, China
| | - Kang Yao
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
- Correspondence: (Q.J.); (K.Y.)
| | - Yulong Yin
- Laboratory of Animal Nutritional Physiology and Metabolic Process, Key Laboratory of Agro-Ecological Processes in Subtropical Region, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- University of Chinese Academy of Sciences, Beijing 100008, China
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410000, China
| |
Collapse
|
15
|
Bow AJ, Rifkin RE, Priester C, Christopher CJ, Grzeskowiak RM, Hecht S, Adair SH, Mulon PY, Castro HF, Campagna SR, Anderson DE. Temporal metabolic profiling of bone healing in a caprine tibia segmental defect model. Front Vet Sci 2023; 9:1023650. [PMID: 36733424 PMCID: PMC9886884 DOI: 10.3389/fvets.2022.1023650] [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: 08/20/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023] Open
Abstract
Bone tissue engineering is an emerging field of regenerative medicine, with a wide array of biomaterial technologies and therapeutics employed. However, it is difficult to objectively compare these various treatments during various stages of tissue response. Metabolomics is rapidly emerging as a powerful analytical tool to establish broad-spectrum metabolic signatures for a target biological system. Developing an effective biomarker panel for bone repair from small molecule data would provide an objective metric to readily assess the efficacy of novel therapeutics in relation to natural healing mechanisms. In this study we utilized a large segmental bone defect in goats to reflect trauma resulting in substantial volumetric bone loss. Characterization of the native repair capacity was then conducted over a period of 12 months through the combination of standard (radiography, computed tomography, histology, biomechanics) data and ultra-high-performance liquid chromatography-high resolution mass spectrometry (UHPLC-HRMS) metabolic profiling. Standard metrics demonstrated that samples formed soft callus structures that later mineralized. Small molecule profiles showed distinct temporal patterns associated with the bone tissue repair process. Specifically, increased lactate and amino acid levels at early time points indicated an environment conducive to osteoblast differentiation and extracellular matrix formation. Citrate and pyruvate abundances increased at later time points indicating increasing mineral content within the defect region. Taurine, shikimate, and pantothenate distribution profiles appeared to represent a shift toward a more homeostatic remodeling environment with the differentiation and activity of osteoclasts offsetting the earlier deposition phases of bone repair. The generation of a comprehensive metabolic reference portfolio offers a potent mechanism for examining novel biomaterials and can serve as guide for the development of new targeted therapeutics to improve the rate, magnitude, and quality of bone regeneration.
Collapse
Affiliation(s)
- Austin J. Bow
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States,*Correspondence: Austin J. Bow ✉
| | - Rebecca E. Rifkin
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Caitlin Priester
- Department of Animal Science, University of Tennessee, Knoxville, Knoxville, TN, United States
| | | | - Remigiusz M. Grzeskowiak
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Silke Hecht
- Department of Small Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Steve H. Adair
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Pierre-Yves Mulon
- Department of Large Animal Clinical Sciences, University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States
| | - Hector F. Castro
- Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States,Biological and Small Molecule Mass Spectrometry Core and the Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - Shawn R. Campagna
- Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States,Biological and Small Molecule Mass Spectrometry Core and the Department of Chemistry, University of Tennessee, Knoxville, Knoxville, TN, United States
| | - David E. Anderson
- University of Tennessee College of Veterinary Medicine, Knoxville, TN, United States,David E. Anderson ✉
| |
Collapse
|
16
|
Examination of human osteoarchaeological remains as a feasible source of polar and apolar metabolites to study past conditions. Sci Rep 2023; 13:696. [PMID: 36639564 PMCID: PMC9839756 DOI: 10.1038/s41598-023-27401-0] [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: 06/20/2022] [Accepted: 01/02/2023] [Indexed: 01/15/2023] Open
Abstract
Metabolomics is a modern tool that aids in our understanding of the molecular changes in organisms. Archaeological science is a branch of archaeology that explores different archaeological materials using modern analytical tools. Human osteoarchaeological material are a frequent finding in archaeological contexts and have the potential to offer information about previous human populations, which can be illuminating about our current condition. Using a set of samples comprising different skeletal elements and bone structures, here we explore for the first time the possibility of extracting metabolites from osteoarchaeological material. Here, a protocol for extraction and measurement of extracted polar and less-polar/apolar metabolites by ultra-high performance liquid chromatography hyphenated to high resolution mass spectrometry is presented to measure the molecules separated after a reversed phase and hydrophilic interaction liquid chromatography column. Molecular information was obtained, showing that osteoarchaeological material is a viable source of molecular information for metabolomic studies.
Collapse
|
17
|
Galvez-Fernandez M, Rodriguez-Hernandez Z, Grau-Perez M, Chaves FJ, Garcia-Garcia AB, Amigo N, Monleon D, Garcia-Barrera T, Gomez-Ariza JL, Briongos-Figuero LS, Perez-Castrillon JL, Redon J, Tellez-Plaza M, Martin-Escudero JC. Metabolomic patterns, redox-related genes and metals, and bone fragility endpoints in the Hortega Study. Free Radic Biol Med 2023; 194:52-61. [PMID: 36370960 DOI: 10.1016/j.freeradbiomed.2022.11.007] [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: 09/13/2022] [Revised: 11/06/2022] [Accepted: 11/06/2022] [Indexed: 11/10/2022]
Abstract
BACKGROUND The potential joint influence of metabolites on bone fragility has been rarely evaluated. We assessed the association of plasma metabolic patterns with bone fragility endpoints (primarily, incident osteoporosis-related bone fractures, and, secondarily, bone mineral density BMD) in the Hortega Study participants. Redox balance plays a key role in bone metabolism. We also assessed differential associations in participant subgroups by redox-related metal exposure levels and candidate genetic variants. MATERIAL AND METHODS In 467 participants older than 50 years from the Hortega Study, a representative sample from a region in Spain, we estimated metabolic principal components (mPC) for 54 plasma metabolites from NMR-spectrometry. Metals biomarkers were measured in plasma by AAS and in urine by HPLC-ICPMS. Redox-related SNPs (N = 341) were measured by oligo-ligation assay. RESULTS The prospective association with incident bone fractures was inverse for mPC1 (non-essential and essential amino acids, including branched-chain, and bacterial co-metabolites, including isobutyrate, trimethylamines and phenylpropionate, versus fatty acids and VLDL) and mPC4 (HDL), but positive for mPC2 (essential amino acids, including aromatic, and bacterial co-metabolites, including isopropanol and methanol). Findings from BMD models were consistent. Participants with decreased selenium and increased antimony, arsenic and, suggestively, cadmium exposures showed higher mPC2-associated bone fractures risk. Genetic variants annotated to 19 genes, with the strongest evidence for NCF4, NOX4 and XDH, showed differential metabolic-related bone fractures risk. CONCLUSIONS Metabolic patterns reflecting amino acids, microbiota co-metabolism and lipid metabolism were associated with bone fragility endpoints. Carriers of redox-related variants may benefit from metabolic interventions to prevent the consequences of bone fragility depending on their antimony, arsenic, selenium, and, possibly, cadmium, exposure levels.
Collapse
Affiliation(s)
- Marta Galvez-Fernandez
- Department of Preventive Medicine and Microbiology, School of Medicine, Universidad Autónoma de Madrid, Arzobispo Morcillo, 4, 28029, Madrid, Spain; Department of Preventive Medicine, Hospital Universitario Severo Ochoa, Avenida de Orellana, s/n, 28911, Madrid, Spain; Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Instituto de Salud Carlos III, Monforte de Lemos, 5, 28029, Madrid, Spain
| | - Zulema Rodriguez-Hernandez
- Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Instituto de Salud Carlos III, Monforte de Lemos, 5, 28029, Madrid, Spain; Department of Biotechnology, Universitat Politècnica de València, Camí de Vera, s/n, 46022, Valencia, Spain
| | - Maria Grau-Perez
- Department of Preventive Medicine and Microbiology, School of Medicine, Universidad Autónoma de Madrid, Arzobispo Morcillo, 4, 28029, Madrid, Spain; INCLIVA Biomedical Research Institute, Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - F Javier Chaves
- INCLIVA Biomedical Research Institute, Menéndez y Pelayo, 4, 46010, Valencia, Spain; CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Ana Barbara Garcia-Garcia
- INCLIVA Biomedical Research Institute, Menéndez y Pelayo, 4, 46010, Valencia, Spain; CIBER of Diabetes and Associated Metabolic Diseases (CIBERDEM), Madrid, Spain
| | - Nuria Amigo
- Biosfer Teslab, Plaça de Prim, 10, 43201, Tarragona, Spain; Department of Basic Medical Sciences, Universidad de Rovira I virgili, Carrer de Sant Llorenç, 21, 43201, Tarragona, Spain
| | - Daniel Monleon
- INCLIVA Biomedical Research Institute, Menéndez y Pelayo, 4, 46010, Valencia, Spain; Department of Pathology, School of Medicine, Universidad de Valencia, Avenida de Blasco Ibáñez, 15, 46010, Valencia, Spain; Center for Biomedical Research Network on Frailty and Health Aging (CIBERFES), Madrid, Spain
| | - Tamara Garcia-Barrera
- Department of Chemistry, Universidad de Huelva, Avenida de las Fuerzas Armadas, 21007, Huelva, Spain
| | - Jose L Gomez-Ariza
- Department of Chemistry, Universidad de Huelva, Avenida de las Fuerzas Armadas, 21007, Huelva, Spain
| | - Laisa S Briongos-Figuero
- Department of Internal Medicine, Hospital Universitario Rio Hortega, Calle Dulzaina, 2, 47012, Valladolid, Spain
| | - Jose L Perez-Castrillon
- Department of Internal Medicine, Hospital Universitario Rio Hortega, Calle Dulzaina, 2, 47012, Valladolid, Spain
| | - Josep Redon
- INCLIVA Biomedical Research Institute, Menéndez y Pelayo, 4, 46010, Valencia, Spain
| | - Maria Tellez-Plaza
- Department of Preventive Medicine and Microbiology, School of Medicine, Universidad Autónoma de Madrid, Arzobispo Morcillo, 4, 28029, Madrid, Spain; Department of Chronic Diseases Epidemiology, National Center for Epidemiology, Instituto de Salud Carlos III, Monforte de Lemos, 5, 28029, Madrid, Spain; INCLIVA Biomedical Research Institute, Menéndez y Pelayo, 4, 46010, Valencia, Spain.
| | - Juan C Martin-Escudero
- Department of Internal Medicine, Hospital Universitario Rio Hortega, Calle Dulzaina, 2, 47012, Valladolid, Spain
| |
Collapse
|
18
|
Podgórska B, Wielogórska-Partyka M, Godzień J, Siemińska J, Ciborowski M, Szelachowska M, Krętowski A, Siewko K. Applications of Metabolomics in Calcium Metabolism Disorders in Humans. Int J Mol Sci 2022; 23:ijms231810407. [PMID: 36142318 PMCID: PMC9499180 DOI: 10.3390/ijms231810407] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 08/26/2022] [Accepted: 09/05/2022] [Indexed: 12/13/2022] Open
Abstract
The pathogenesis of the disorders of calcium metabolism is not fully understood. This review discusses the studies in which metabolomics was applied in this area. Indeed, metabolomics could play an essential role in discovering biomarkers and elucidating pathological mechanisms. Despite the limited bibliography, the present review highlights the potential of metabolomics in identifying the biomarkers of some of the most common endocrine disorders, such as primary hyperparathyroidism (PHPT), secondary hyperparathyroidism (SHPT), calcium deficiency, osteoporosis and vitamin D supplementation. Metabolites related to above-mentioned diseorders were grouped into specific classes and mapped into metabolic pathways. Furthermore, disturbed metabolic pathways can open up new directions for the in-depth exploration of the basic mechanisms of these diseases at the molecular level.
Collapse
Affiliation(s)
- Beata Podgórska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
- Correspondence: ; Tel.: +48-85-831-83-12
| | - Marta Wielogórska-Partyka
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Joanna Godzień
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Julia Siemińska
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Michał Ciborowski
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Małgorzata Szelachowska
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Adam Krętowski
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
- Clinical Research Centre, Medical University of Bialystok, 15-276 Bialystok, Poland
| | - Katarzyna Siewko
- Department of Endocrinology, Diabetology and Internal Medicine, Medical University of Bialystok, 15-276 Bialystok, Poland
| |
Collapse
|
19
|
Fan J, Abedi-Dorcheh K, Sadat Vaziri A, Kazemi-Aghdam F, Rafieyan S, Sohrabinejad M, Ghorbani M, Rastegar Adib F, Ghasemi Z, Klavins K, Jahed V. A Review of Recent Advances in Natural Polymer-Based Scaffolds for Musculoskeletal Tissue Engineering. Polymers (Basel) 2022; 14:polym14102097. [PMID: 35631979 PMCID: PMC9145843 DOI: 10.3390/polym14102097] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 05/09/2022] [Accepted: 05/17/2022] [Indexed: 02/01/2023] Open
Abstract
The musculoskeletal (MS) system consists of bone, cartilage, tendon, ligament, and skeletal muscle, which forms the basic framework of the human body. This system plays a vital role in appropriate body functions, including movement, the protection of internal organs, support, hematopoiesis, and postural stability. Therefore, it is understandable that the damage or loss of MS tissues significantly reduces the quality of life and limits mobility. Tissue engineering and its applications in the healthcare industry have been rapidly growing over the past few decades. Tissue engineering has made significant contributions toward developing new therapeutic strategies for the treatment of MS defects and relevant disease. Among various biomaterials used for tissue engineering, natural polymers offer superior properties that promote optimal cell interaction and desired biological function. Natural polymers have similarity with the native ECM, including enzymatic degradation, bio-resorb and non-toxic degradation products, ability to conjugate with various agents, and high chemical versatility, biocompatibility, and bioactivity that promote optimal cell interaction and desired biological functions. This review summarizes recent advances in applying natural-based scaffolds for musculoskeletal tissue engineering.
Collapse
Affiliation(s)
- Jingzhi Fan
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
| | - Keyvan Abedi-Dorcheh
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Asma Sadat Vaziri
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Fereshteh Kazemi-Aghdam
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Saeed Rafieyan
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Masoume Sohrabinejad
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Mina Ghorbani
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Fatemeh Rastegar Adib
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Zahra Ghasemi
- Biomedical Engineering Division, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran 14117-13116, Iran; (K.A.-D.); (A.S.V.); (F.K.-A.); (S.R.); (M.S.); (M.G.); (F.R.A.); (Z.G.)
| | - Kristaps Klavins
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
- Correspondence: (K.K.); (V.J.)
| | - Vahid Jahed
- Rudolfs Cimdins Riga Biomaterials Innovations and Development Centre of RTU, Institute of General Chemical Engineering, Faculty of Materials Science and Applied Chemistry, Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia;
- Baltic Biomaterials Centre of Excellence, Headquarters at Riga Technical University, Pulka St 3, LV-1007 Riga, Latvia
- Correspondence: (K.K.); (V.J.)
| |
Collapse
|
20
|
Dias DB, Fritsche-Guenther R, Gutmann F, Duda GN, Kirwan J, Poh PSP. A Comparison of Solvent-Based Extraction Methods to Assess the Central Carbon Metabolites in Mouse Bone and Muscle. Metabolites 2022; 12:453. [PMID: 35629956 PMCID: PMC9144563 DOI: 10.3390/metabo12050453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 05/05/2022] [Accepted: 05/06/2022] [Indexed: 02/01/2023] Open
Abstract
The identification of endogenous metabolites has great potential for understanding the underlying tissue processes occurring in either a homeostatic or a diseased state. The application of gas chromatography-mass spectrometry (GC-MS)-based metabolomics on musculoskeletal tissue samples has gained traction. However, limited comparison studies exist evaluating the sensitivity, reproducibility, and robustness of the various existing extraction protocols for musculoskeletal tissues. Here, we evaluated polar metabolite extraction from bone and muscle of mouse origin. The extraction methods compared were (1) modified Bligh-Dyer (mBD), (2) low chloroform (CHCl3)-modified Bligh-Dyer (mBD-low), and (3) modified Matyash (mMat). In particular, the central carbon metabolites (CCM) appear to be relevant for musculoskeletal regeneration, given their role in energy metabolism. However, the sensitivity, reproducibility, and robustness of these methods for detecting targeted polar CCM remains unknown. Overall, the extraction of metabolites using the mBD, mBD-low, and mMat methods appears sufficiently robust and reproducible for bone, with the mBD method slightly bettering the mBD-low and mMat methods. Furthermore, mBD, mBD-low, and mMat were sufficiently sensitive in detecting polar metabolites extracted from mouse muscle; however, they lacked repeatability. This study highlights the need for a re-thinking, towards a tissue-specific optimization of methods for metabolite extractions, ensuring sufficient sensitivity, repeatability, and robustness.
Collapse
Affiliation(s)
- Daniela B. Dias
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (D.B.D.); (G.N.D.)
| | - Raphaela Fritsche-Guenther
- Berlin Institute of Health at Charité—BIH Metabolomics Platform, 10178 Berlin, Germany; (R.F.-G.); (F.G.); (J.K.)
| | - Friederike Gutmann
- Berlin Institute of Health at Charité—BIH Metabolomics Platform, 10178 Berlin, Germany; (R.F.-G.); (F.G.); (J.K.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str 10, 13125 Berlin, Germany
- Charité—Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, ECRC Experimental and Clinical Research Center, Lindenberger Weg 80, 13125 Berlin, Germany
| | - Georg N. Duda
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (D.B.D.); (G.N.D.)
- Berlin Institute of Health Center for Regenerative Therapies, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Jennifer Kirwan
- Berlin Institute of Health at Charité—BIH Metabolomics Platform, 10178 Berlin, Germany; (R.F.-G.); (F.G.); (J.K.)
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Robert-Rössle-Str 10, 13125 Berlin, Germany
| | - Patrina S. P. Poh
- Julius Wolff Institute, Berlin Institute of Health at Charité—Universitätsmedizin Berlin, 13353 Berlin, Germany; (D.B.D.); (G.N.D.)
| |
Collapse
|
21
|
Wang W, Wang Y, Hu J, Duan H, Wang Z, Yin L, He F. Untargeted Metabolomics Reveal the Protective Effect of Bone Marrow Mesenchymal Stem Cell Transplantation Against Ovariectomy-Induced Osteoporosis in Mice. Cell Transplant 2022; 31:9636897221079745. [PMID: 35225020 PMCID: PMC8891838 DOI: 10.1177/09636897221079745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Bone marrow mesenchymal stem cell transplantation (BMSCT) is a potential treatment for osteoporosis, capable of contributing to bone tissue repair. BMSCT has demonstrated osteoinductive effects and the ability to regulate microenvironmental metabolism; however, its role and mechanisms in bone loss due to reduced estrogen levels remain unclear. In this study, the effect of BMSCT on ovariectomy (OVX)-induced osteoporosis in mice was assessed, and liquid chromatography–mass spectrometry (LC-MS) metabolomic studies of bone tissue were conducted to identify potential metabolic molecular markers. The results revealed that BMSCT reduces OVX-induced bone loss in mice while improving the mechanical properties of mouse femurs and increasing the expression of osteogenic markers in peripheral blood. In a metabolomic study, 18 metabolites were screened as potential biomarkers of the anti-osteoporotic effect of BMSCT. These metabolites are mainly involved in arachidonic acid metabolism, taurine and hypotaurine metabolism, and pentose and glucuronate interconversions. Collectively, these results illustrate the correlation between metabolites and the underlying mechanisms of osteoporosis development and are important for understanding the role and mechanisms of exogenous bone marrow mesenchymal stem cells (BMSCs) in osteoporosis management. This study lays the foundation for research on BMSCs as a treatment strategy for osteoporosis.
Collapse
Affiliation(s)
- Weizhou Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Yanghao Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Jun Hu
- Kunming First People’s Hospital, Kunming, China
| | - Hao Duan
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Zhihua Wang
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Liang Yin
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| | - Fei He
- Department of Orthopedics, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Province Stem Cell Technology Application Research Center, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Provincial Clinical Medical Center for Bone and Joint Diseases, The First Affiliated Hospital of Kunming Medical University, Kunming, China
| |
Collapse
|
22
|
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: 1.3] [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
|