1
|
Wang C, Wang J, Chen S, Li K, Wan S, Yang L. COL10A1 as a Prognostic Biomarker in Association with Immune Infiltration in Prostate Cancer. Curr Cancer Drug Targets 2024; 24:340-353. [PMID: 37592784 DOI: 10.2174/1568009623666230817101809] [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: 11/16/2022] [Revised: 03/19/2023] [Accepted: 06/06/2023] [Indexed: 08/19/2023]
Abstract
BACKGROUND The collagen type X alpha 1 (COL10A1) has recently been found to play an important role in the development and progression of cancer. However, the link between COL10A1 and the tumor immune microenvironment remains understood scantily. METHODS In the current study, the pan-cancer data of The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were used to investigate the expression mode, the clinical prognostic and diagnostic value of COL10A1 in different tumors. We used TCGA data to assess the correlations between COL10A1 and clinical symptoms of prostate cancer. The R packages "edgR" and "clusterProfiler" were used for differential expression gene and enrichment analysis of COL10A1. Immunohistochemistry was further employed to corroborate the expression of COL10A1 gene in prostate cancer. After that, we used TIMER to evaluate the pertinence of COL10A1 expression to immune infiltration level in prostate cancer. RESULTS On the whole, COL10A1 was expressed at significantly higher levels in a variety of tumor tissues than in the corresponding normal tissues. Besides, significant correlations with tumor prognosis and relative exactitude in predicting tumors show that COL10A1 may be a probable prognostic and diagnostic biomarker of prostate cancer. In addition, the evidence indicates a significant correlation between COL10A1 and clinical symptoms of prostate cancer. Furthermore, the main molecular functions of COL10A1 included humoral immune response, complement activation, immunoglobulin, regulation of complement activation, and regulation of humoral immune response. Finally, we found that COL10A1 expression is positively correlated with enhanced macrophage and M2 macrophage infiltration in prostate cancer. CONCLUSION The study indicates that COL10A1 might participate in M2 macrophage polarization in prostate cancer. COL10A1 might be an innovative biomarker to evaluate tumor microenvironment immune cell infiltration and prognosis in prostate cancer.
Collapse
Affiliation(s)
- Chenyang Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou, China
| | - Jirong Wang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou, China
| | - Siyu Chen
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou, China
| | - Kunpeng Li
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou, China
| | - Shun Wan
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou, China
| | - Li Yang
- Department of Urology, The Second Hospital of Lanzhou University, Lanzhou, China
- Gansu Province Clinical Research Center for Urology, Lanzhou, China
| |
Collapse
|
2
|
Yang J, Pei T, Su G, Duan P, Liu X. AnnexinA6: a potential therapeutic target gene for extracellular matrix mineralization. Front Cell Dev Biol 2023; 11:1201200. [PMID: 37727505 PMCID: PMC10506415 DOI: 10.3389/fcell.2023.1201200] [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: 04/06/2023] [Accepted: 08/10/2023] [Indexed: 09/21/2023] Open
Abstract
The mineralization of the extracellular matrix (ECM) is an essential and crucial process for physiological bone formation and pathological calcification. The abnormal function of ECM mineralization contributes to the worldwide risk of developing mineralization-related diseases; for instance, vascular calcification is attributed to the hyperfunction of ECM mineralization, while osteoporosis is due to hypofunction. AnnexinA6 (AnxA6), a Ca2+-dependent phospholipid-binding protein, has been extensively reported as an essential target in mineralization-related diseases such as osteoporosis, osteoarthritis, atherosclerosis, osteosarcoma, and calcific aortic valve disease. To date, AnxA6, as the largest member of the Annexin family, has attracted much attention due to its significant contribution to matrix vesicles (MVs) production and release, MVs-ECM interaction, cytoplasmic Ca2+ influx, and maturation of hydroxyapatite, making it an essential target in ECM mineralization. In this review, we outlined the recent advancements in the role of AnxA6 in mineralization-related diseases and the potential mechanisms of AnxA6 under normal and mineralization-related pathological conditions. AnxA6 could promote ECM mineralization for bone regeneration in the manner described previously. Therefore, AnxA6 may be a potential osteogenic target for ECM mineralization.
Collapse
Affiliation(s)
| | | | | | | | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
3
|
Lv Y, Zhang J, Li C, Wang L, Lei L, Huang X. Network pharmacological analysis to reveal the mechanism governing the effect of Qin Xi Tong on osteoarthritis and rheumatoid arthritis. Clin Rheumatol 2023:10.1007/s10067-023-06625-5. [PMID: 37162694 DOI: 10.1007/s10067-023-06625-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2022] [Revised: 04/03/2023] [Accepted: 05/03/2023] [Indexed: 05/11/2023]
Abstract
INTRODUCTION Qin Xi Tong (QXT), produced by water extracts of Caulis Sinomenii, is clinically effective in the therapy of rheumatoid arthritis (RA). It is also a complementary agent for osteoarthritis (OA). This study aimed to screen the candidate targets and identify the potential mechanisms of QXT against RA and OA. METHOD The active ingredients contained in QXT were queried from the TCMSP database. Their predicted targets were obtained through web-based databases, including TCMSP, BATMAN-TCM, CTD, and PharmMapper. The OA and RA targets were collected from the Genecards database and the GSE55235 dataset. Based on the DAVID database, GO and KEGG enrichment analyses of disease-drug common targets predicted potential signaling pathways for QXT. In addition, core targets were identified by mapping component-target-disease interaction networks with Cytoscape 3.9.1 and STRING. The Swissdock and Pymol tools further validate the predicted results. RESULTS A total of 161 genes were put forward as potential targets for treating RA and OA. These genes might be involved in joint inflammation, including the IL-17 signaling pathway, MAPK signaling pathway, and TNF signaling pathway. They also regulated the progression of joint injuries, such as apoptosis, Th17 cell differentiation, and osteoclast differentiation. In addition, we identified 12 core targets of QXT. Molecular docking results showed that QXT has a high affinity with these core targets. CONCLUSIONS This study reveals the mechanism governing the effect of QXT on RA and OA, predicts the direct target, and provides new ideas for clinical treatment. Key Points • Our study reveals the underlying mechanism of QXT in the treatment of RA and OA. • Further research into the effects of compounds in QXT alone would be of interest.
Collapse
Affiliation(s)
- Yanyan Lv
- Department of Rheumatology and Immunology, Xi'an No. 5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China
| | - Jie Zhang
- Department of Rheumatology and Immunology, Xi'an No. 5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China
| | - Chao Li
- Department of Rheumatology and Immunology, Xi'an No. 5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China
| | - Li Wang
- Department of Rheumatology and Immunology, Xi'an No. 5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China
| | - Lei Lei
- School of Life Sciences and Medicine, Northwest University, No. 229 Taibai North Road, Xi'an, China
| | - Xiaoqiang Huang
- Department of Orthopedics, Xi'an No.5 Hospital, No. 112 Xi Guan Zheng Jie, Xi'an, China.
| |
Collapse
|
4
|
Xiang C, Liao Y, Chen Z, Xiao B, Zhao Z, Li A, Xia Y, Wang P, Li H, Xiao T. Network Pharmacology and Molecular Docking to Elucidate the Potential Mechanism of Ligusticum Chuanxiong Against Osteoarthritis. Front Pharmacol 2022; 13:854215. [PMID: 35496280 PMCID: PMC9050356 DOI: 10.3389/fphar.2022.854215] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 03/24/2022] [Indexed: 12/04/2022] Open
Abstract
Background: Osteoarthritis (OA) is a degenerative disease which serious affects patients. Ligusticum chuanxiong (CX) has been shown to have a certain curative effect on osteoarthritis in traditional Chinese medicine therapy. This study is based on network pharmacology and molecular docking technology to explore the potential mechanism of CX. Methods: Components of CX to treat osteoarthritis were screened in the TCMSP database and targets were predicted by the PharmMapper database, the osteoarthritis targets were collected from the GeneCards database, and intersection genes were found to be the possible targets of CX anti-OA. The STRING database and Cytoscape software were utilized for protein-protein interaction analysis and further screening of core targets. The Metascape database was used for KEGG and GO enrichment analyses. Then, the top 10 pathways were selected to construct “drug-compound-target-pathway-disease” network analysis. Finally, molecular docking was used to analyze the binding affinity of seven compounds with core targets and TNF-α. Results: Seven compounds with 253 non-repetitive targets of CX were screened from the TCMSP database and 60 potential intersection targets of CX anti-OA were found. PPI network analysis showed that the core targets were ALB, AKT1, IGF1, CASP3, MAPK1, ANXA5, and MAPK14, while GO and KEGG pathway enrichment analyses showed that the relevant biological processes involved in the treatment of osteoarthritis by CX might include the MAPK cascade and reactive oxygen species metabolic process. The KEGG pathway analysis result was mainly associated with the MAPK signaling pathway and PI3K-AKT signaling pathway. We further docked seven ingredients with MAPK1 and MAPK14 enriched in the MAPK pathway, and TNF-α as the typical inflammatory cytokine. The results also showed good binding affinity, especially FA, which may be the most important component of CX anti-OA. Conclusion: Our research revealed the potential mechanism of CX in the treatment of OA, and our findings can also pave the way for subsequent basic experimental verification and a new research direction.
Collapse
Affiliation(s)
- Cheng Xiang
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yilin Liao
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Zhuoyuan Chen
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Bo Xiao
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Ziyue Zhao
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Aoyu Li
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu Xia
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Pingxiao Wang
- Second Xiangya Hospital, Central South University, Changsha, China
| | - Hui Li
- Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Hui Li, ; Tao Xiao,
| | - Tao Xiao
- Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Hui Li, ; Tao Xiao,
| |
Collapse
|
5
|
Wang Y, Weremiejczyk L, Strzelecka‐Kiliszek A, Maniti O, Amabile Veschi E, Bolean M, Ramos AP, Ben Trad L, Magne D, Bandorowicz‐Pikula J, Pikula S, Millán JL, Bottini M, Goekjian P, Ciancaglini P, Buchet R, Dou WT, Tian H, Mebarek S, He XP, Granjon T. Fluorescence evidence of annexin A6 translocation across membrane in model matrix vesicles during apatite formation. JOURNAL OF EXTRACELLULAR BIOLOGY 2022; 1:e38. [PMID: 38939118 PMCID: PMC11080897 DOI: 10.1002/jex2.38] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/20/2022] [Accepted: 03/21/2022] [Indexed: 06/29/2024]
Abstract
Matrix vesicles (MVs) are 100-300 nm spherical structures released by mineralization competent cells to initiate formation of apatite, the mineral component in bones. Among proteins present in MVs, annexin A6 (AnxA6) is thought to be ubiquitously distributed in the MVs' lumen, on the surface of the internal and external leaflets of the membrane and also inserted in the lipid bilayer. To determine the molecular mechanism(s) that lead to the different locations of AnxA6, we hypothesized the occurrence of a pH drop during the mineralization. Such a change would induce the AnxA6 protonation, which in turn, and because of its isoelectric point of 5.41, would change the protein hydrophobicity facilitating its insertion into the MVs' bilayer. The various distributions of AnxA6 are likely to disturb membrane phospholipid organization. To examine this possibility, we used fluorescein as pH reporter, and established that pH decreased inside MVs during apatite formation. Then, 4-(14-phenyldibenzo[a,c]phenazin-9(14H)-yl)-phenol, a vibration-induced emission fluorescent probe, was used as a reporter of changes in membrane organization occurring with the varying mode of AnxA6 binding. Proteoliposomes containing AnxA6 and 1,2-Dimyristoyl-sn-glycero-3phosphocholine (DMPC) or 1,2-Dimyristoyl-sn-glycero-3phosphocholine: 1,2-Dipalmitoyl-sn-glycero-3-phosphoserine (DMPC:DPPS 9:1), to mimic the external and internal MV membrane leaflet, respectively, served as biomimetic models to investigate the nature of AnxA6 binding. Addition of Anx6 to DMPC at pH 7.4 and 5.4, or DMPC:DPPS (9:1) at pH 7.4 induced a decrease in membrane fluidity, consistent with AnxA6 interactions with the bilayer surface. In contrast, AnxA6 addition to DMPC:DPPS (9:1) at pH 5.4 increased the fluidity of the membrane. This latest result was interpreted as reflecting the insertion of AnxA6 into the bilayer. Taken together, these findings point to a possible mechanism of AnxA6 translocation in MVs from the surface of the internal leaflet into the phospholipid bilayer stimulated upon acidification of the MVs' lumen during formation of apatite.
Collapse
Affiliation(s)
- Yubo Wang
- Univ LyonUCBLCNRSICBMS UMR 5246IMBLLyonFrance
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular EngineeringFeringa Nobel Prize Scientist Joint Research CentreEast China University of Science and TechnologyShanghaiChina
| | - Liliana Weremiejczyk
- Laboratory of Biochemistry of LipidsNencki Institute of Experimental BiologyWarsawPoland
| | | | | | - Ekeveliny Amabile Veschi
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Mayte Bolean
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - Ana Paula Ramos
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | | | - David Magne
- Univ LyonUCBLCNRSICBMS UMR 5246IMBLLyonFrance
| | | | - Slawomir Pikula
- Laboratory of Biochemistry of LipidsNencki Institute of Experimental BiologyWarsawPoland
| | - Jose Luis Millán
- Sanford Burnham Prebys Medical Discovery InstituteLa JollaCaliforniaUSA
| | - Massimo Bottini
- Department of Experimental MedicineUniversity of Rome Tor VergataRomeItaly
| | | | - Pietro Ciancaglini
- Departamento de QuímicaFaculdade de FilosofiaCiências e Letras de Ribeirão Preto da Universidade de São Paulo (FFCLRP‐USP)Ribeirão PretoSão PauloBrazil
| | - René Buchet
- Univ LyonUCBLCNRSICBMS UMR 5246IMBLLyonFrance
| | - Wei Tao Dou
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular EngineeringFeringa Nobel Prize Scientist Joint Research CentreEast China University of Science and TechnologyShanghaiChina
| | - He Tian
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular EngineeringFeringa Nobel Prize Scientist Joint Research CentreEast China University of Science and TechnologyShanghaiChina
| | | | - Xiao P. He
- Key Laboratory for Advanced Materials and Joint International Research Laboratory of Precision Chemistry and Molecular EngineeringFeringa Nobel Prize Scientist Joint Research CentreEast China University of Science and TechnologyShanghaiChina
| | | |
Collapse
|
6
|
Transcriptomics of Wet Skin Biopsies Predict Early Radiation-Induced Hematological Damage in a Mouse Model. Genes (Basel) 2022; 13:genes13030538. [PMID: 35328091 PMCID: PMC8952434 DOI: 10.3390/genes13030538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/28/2022] [Accepted: 03/16/2022] [Indexed: 12/04/2022] Open
Abstract
The lack of an easy and fast radiation-exposure testing method with a dosimetric ability complicates triage and treatment in response to a nuclear detonation, radioactive material release, or clandestine exposure. The potential of transcriptomics in radiation diagnosis and prognosis were assessed here using wet skin (blood/skin) biopsies obtained at hour 2 and days 4, 7, 21, and 28 from a mouse radiation model. Analysis of significantly differentially transcribed genes (SDTG; p ≤ 0.05 and FC ≥ 2) during the first post-exposure week identified the glycoprotein 6 (GP-VI) signaling, the dendritic cell maturation, and the intrinsic prothrombin activation pathways as the top modulated pathways with stable inactivation after lethal exposures (20 Gy) and intermittent activation after sublethal (1, 3, 6 Gy) exposure time points (TPs). Interestingly, these pathways were inactivated in the late TPs after sublethal exposure in concordance with a delayed deleterious effect. Modulated transcription of a variety of collagen types, laminin, and peptidase genes underlay the modulated functions of these hematologically important pathways. Several other SDTGs related to platelet and leukocyte development and functions were identified. These results outlined genetic determinants that were crucial to clinically documented radiation-induced hematological and skin damage with potential countermeasure applications.
Collapse
|
7
|
The extracellular matrix of hematopoietic stem cell niches. Adv Drug Deliv Rev 2022; 181:114069. [PMID: 34838648 PMCID: PMC8860232 DOI: 10.1016/j.addr.2021.114069] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 11/18/2021] [Accepted: 11/21/2021] [Indexed: 12/21/2022]
Abstract
Comprehensive overview of different classes of ECM molecules in the HSC niche. Overview of current knowledge on role of biophysics of the HSC niche. Description of approaches to create artificial stem cell niches for several application. Importance of considering ECM in drug development and testing.
Hematopoietic stem cells (HSCs) are the life-long source of all types of blood cells. Their function is controlled by their direct microenvironment, the HSC niche in the bone marrow. Although the importance of the extracellular matrix (ECM) in the niche by orchestrating niche architecture and cellular function is widely acknowledged, it is still underexplored. In this review, we provide a comprehensive overview of the ECM in HSC niches. For this purpose, we first briefly outline HSC niche biology and then review the role of the different classes of ECM molecules in the niche one by one and how they are perceived by cells. Matrix remodeling and the emerging importance of biophysics in HSC niche function are discussed. Finally, the application of the current knowledge of ECM in the niche in form of artificial HSC niches for HSC expansion or targeted differentiation as well as drug testing is reviewed.
Collapse
|
8
|
Bian H, Zhu T, Liang Y, Hei R, Zhang X, Li X, Chen J, Lu Y, Gu J, Qiao L, Zheng Q. Expression Profiling and Functional Analysis of Candidate Col10a1 Regulators Identified by the TRAP Program. Front Genet 2021; 12:683939. [PMID: 34276786 PMCID: PMC8283764 DOI: 10.3389/fgene.2021.683939] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Accepted: 05/24/2021] [Indexed: 11/13/2022] Open
Abstract
Hypertrophic chondrocytes and their specific marker, the type X collagen gene (Col10a1), are critical components of endochondral bone formation during skeletal development. We previously found that Runx2 is an indispensable mouse Col10a1 gene regulator and identified many other transcription factors (TFs) that potentially interact with the 150-bp Col10a1 cis-enhancer. However, the roles of these candidate TFs in Col10a1 expression and chondrocyte hypertrophy have not been elucidated. Here, we focus on 32 candidate TFs recently identified by analyzing the 150-bp Col10a1 enhancer using the transcription factor affinity prediction (TRAP) program. We found that 12 TFs (Hoxa3, Lsx, Evx2, Dlx5, S8, Pax2, Egr2, Mef2a, Barhl2, GKlf, Sox17, and Crx) were significantly upregulated and four TFs (Lhx4, Tbx5, Mef2c, and Hb9) were significantly downregulated in hypertrophic MCT cells, which show upregulation of Col10a1 expression. Most of the differential expression pattern of these TFs conformed with the results obtained from ATDC5 cell model and primary mouse chondrocytes. Notably, Tbx5 was downregulated upon Col10a1 upregulation, overexpression of Tbx5 decreased Col10a1 expression, and knock-down of Tbx5 increased Col10a1 expression in hypertrophic chondrocytes, suggesting that Tbx5 is a negative regulator of Col10a1. We further generated a stable Tbx5-overexpressing ATDC5 cell line and ColX-Tbx5 transgenic mice driven by Col10a1-specific enhancers and promoters. Tbx5 overexpression decreased Col10a1 expression in ATDC5 cells cultured as early as day 7 and in limb tissue on post-natal day 1. Slightly weaker alkaline phosphatase staining was also observed in cell culture on day 7 and in limb digits on embryonic day 17.5, indicating mildly delayed ossification. Further characterization of these candidate Col10a1 transcriptional regulators could help identify novel therapeutic targets for skeletal diseases associated with abnormal chondrocyte hypertrophy.
Collapse
Affiliation(s)
- Huiqin Bian
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Ting Zhu
- Laboratory of Clinical Medicine, Huai'an Women & Children Hospital, Affiliated to Yangzhou University, Huai'an, China
| | - Yuting Liang
- Center of Clinical Laboratory, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Ruoxuan Hei
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaojing Zhang
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Xiaochen Li
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Jinnan Chen
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Yaojuan Lu
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.,Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd., Shenzhen, China
| | - Junxia Gu
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China
| | - Longwei Qiao
- Suzhou Affiliated to State Key Laboratory of Reproductive Medicine, School of Gusu, The Affiliated Suzhou Hospital of Nanjing Medical University, Nanjing Medical University, Suzhou, China
| | - Qiping Zheng
- Department of Hematology and Hematological Laboratory Science, Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, China.,Shenzhen Academy of Peptide Targeting Technology at Pingshan and Shenzhen Tyercan Bio-Pharm Co., Ltd., Shenzhen, China
| |
Collapse
|
9
|
Grewal T, Rentero C, Enrich C, Wahba M, Raabe CA, Rescher U. Annexin Animal Models-From Fundamental Principles to Translational Research. Int J Mol Sci 2021; 22:ijms22073439. [PMID: 33810523 PMCID: PMC8037771 DOI: 10.3390/ijms22073439] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 03/18/2021] [Accepted: 03/24/2021] [Indexed: 02/07/2023] Open
Abstract
Routine manipulation of the mouse genome has become a landmark in biomedical research. Traits that are only associated with advanced developmental stages can now be investigated within a living organism, and the in vivo analysis of corresponding phenotypes and functions advances the translation into the clinical setting. The annexins, a family of closely related calcium (Ca2+)- and lipid-binding proteins, are found at various intra- and extracellular locations, and interact with a broad range of membrane lipids and proteins. Their impacts on cellular functions has been extensively assessed in vitro, yet annexin-deficient mouse models generally develop normally and do not display obvious phenotypes. Only in recent years, studies examining genetically modified annexin mouse models which were exposed to stress conditions mimicking human disease often revealed striking phenotypes. This review is the first comprehensive overview of annexin-related research using animal models and their exciting future use for relevant issues in biology and experimental medicine.
Collapse
Affiliation(s)
- Thomas Grewal
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| | - Carles Rentero
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Carlos Enrich
- Departament de Biomedicina, Unitat de Biologia Cel·lular, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, 08036 Barcelona, Spain; (C.R.); (C.E.)
- Centre de Recerca Biomèdica CELLEX, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain
| | - Mohamed Wahba
- School of Pharmacy, Faculty of Medicine and Health, University of Sydney, Sydney, NSW 2006, Australia;
| | - Carsten A. Raabe
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
| | - Ursula Rescher
- Research Group Regulatory Mechanisms of Inflammation, Center for Molecular Biology of Inflammation (ZMBE) and Cells in Motion Interfaculty Center (CiM), Institute of Medical Biochemistry, University of Muenster, 48149 Muenster, Germany;
- Correspondence: (T.G.); (U.R.); Tel.: +61-(0)2-9351-8496 (T.G.); +49-(0)251-83-52121 (U.R.)
| |
Collapse
|
10
|
Gene Expression Profiling of the Extracellular Matrix Signature in Macrophages of Different Activation Status: Relevance for Skin Wound Healing. Int J Mol Sci 2019; 20:ijms20205086. [PMID: 31615030 PMCID: PMC6829210 DOI: 10.3390/ijms20205086] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Revised: 10/03/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
The extracellular matrix (ECM) provides structural support for tissue architecture and is a major effector of cell behavior during skin repair and inflammation. Macrophages are involved in all stages of skin repair but only limited knowledge exists about macrophage-specific expression and regulation of ECM components. In this study, we used transcriptome profiling and bioinformatic analysis to define the unique expression of ECM-associated genes in cultured macrophages. Characterization of the matrisome revealed that most genes were constitutively expressed and that several genes were uniquely regulated upon interferon gamma (IFNγ) and dexamethasone stimulation. Among those core matrisome and matrisome-associated components transforming growth factor beta (TGFβ)-induced, matrix metalloproteinase 9 (MMP9), elastin microfibril interfacer (EMILIN)-1, netrin-1 and gliomedin were also present within the wound bed at time points that are characterized by profound macrophage infiltration. Hence, macrophages are a source of ECM components in vitro as well as during skin wound healing, and identification of these matrisome components is a first step to understand the role and therapeutic value of ECM components in macrophages and during wound healing.
Collapse
|
11
|
New Insights into the Roles of Monocytes/Macrophages in Cardiovascular Calcification Associated with Chronic Kidney Disease. Toxins (Basel) 2019; 11:toxins11090529. [PMID: 31547340 PMCID: PMC6784181 DOI: 10.3390/toxins11090529] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Revised: 09/06/2019] [Accepted: 09/09/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular disease (CVD) is an important cause of death in patients with chronic kidney disease (CKD), and cardiovascular calcification (CVC) is one of the strongest predictors of CVD in this population. Cardiovascular calcification results from complex cellular interactions involving the endothelium, vascular/valvular cells (i.e., vascular smooth muscle cells, valvular interstitial cells and resident fibroblasts), and monocyte-derived macrophages. Indeed, the production of pro-inflammatory cytokines and oxidative stress by monocyte-derived macrophages is responsible for the osteogenic transformation and mineralization of vascular/valvular cells. However, monocytes/macrophages show the ability to modify their phenotype, and consequently their functions, when facing environmental modifications. This plasticity complicates efforts to understand the pathogenesis of CVC-particularly in a CKD setting, where both uraemic toxins and CKD treatment may affect monocyte/macrophage functions and thereby influence CVC. Here, we review (i) the mechanisms by which each monocyte/macrophage subset either promotes or prevents CVC, and (ii) how both uraemic toxins and CKD therapies might affect these monocyte/macrophage functions.
Collapse
|
12
|
Holzer T, Probst K, Etich J, Auler M, Georgieva VS, Bluhm B, Frie C, Heilig J, Niehoff A, Nüchel J, Plomann M, Seeger JM, Kashkar H, Baris OR, Wiesner RJ, Brachvogel B. Respiratory chain inactivation links cartilage-mediated growth retardation to mitochondrial diseases. J Cell Biol 2019; 218:1853-1870. [PMID: 31085560 PMCID: PMC6548139 DOI: 10.1083/jcb.201809056] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Revised: 01/12/2019] [Accepted: 04/12/2019] [Indexed: 12/17/2022] Open
Abstract
Children with mitochondrial diseases often present with slow growth and short stature, but the underlying mechanism remains unclear. In this study, Holzer et al. provide in vivo evidence that mitochondrial respiratory chain dysfunction induces cartilage degeneration coincident with altered metabolism, impaired extracellular matrix formation, and cell death at the cartilage–bone junction. In childhood, skeletal growth is driven by transient expansion of cartilage in the growth plate. The common belief is that energy production in this hypoxic tissue mainly relies on anaerobic glycolysis and not on mitochondrial respiratory chain (RC) activity. However, children with mitochondrial diseases causing RC dysfunction often present with short stature, which indicates that RC activity may be essential for cartilage-mediated skeletal growth. To elucidate the role of the mitochondrial RC in cartilage growth and pathology, we generated mice with impaired RC function in cartilage. These mice develop normally until birth, but their later growth is retarded. A detailed molecular analysis revealed that metabolic signaling and extracellular matrix formation is disturbed and induces cell death at the cartilage–bone junction to cause a chondrodysplasia-like phenotype. Hence, the results demonstrate the overall importance of the metabolic switch from fetal glycolysis to postnatal RC activation in growth plate cartilage and explain why RC dysfunction can cause short stature in children with mitochondrial diseases.
Collapse
Affiliation(s)
- Tatjana Holzer
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Cologne, Germany.,Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Kristina Probst
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Cologne, Germany.,Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Julia Etich
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Cologne, Germany.,Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Markus Auler
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Cologne, Germany.,Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Veronika S Georgieva
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Cologne, Germany.,Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Björn Bluhm
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Cologne, Germany.,Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Christian Frie
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Cologne, Germany.,Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Juliane Heilig
- Institute of Biomechanics and Orthopedics, German Sport University Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics, University of Cologne, Cologne, Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopedics, German Sport University Cologne, Cologne, Germany.,Cologne Center for Musculoskeletal Biomechanics, University of Cologne, Cologne, Germany
| | - Julian Nüchel
- Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Markus Plomann
- Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Jens M Seeger
- Institute for Medical Microbiology, Immunology, and Hygiene, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Hamid Kashkar
- Institute for Medical Microbiology, Immunology, and Hygiene, Faculty of Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.,Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Olivier R Baris
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany
| | - Rudolf J Wiesner
- Center for Physiology and Pathophysiology, Institute of Vegetative Physiology, Faculty of Medicine, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases, University of Cologne, Cologne, Germany.,Center of Molecular Medicine Cologne, University of Cologne, Cologne, Germany
| | - Bent Brachvogel
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Faculty of Medicine, University of Cologne, Cologne, Germany .,Center for Biochemistry, Faculty of Medicine, University of Cologne, Cologne, Germany
| |
Collapse
|
13
|
Kunitomi Y, Hara ES, Okada M, Nagaoka N, Kuboki T, Nakano T, Kamioka H, Matsumoto T. Biomimetic mineralization using matrix vesicle nanofragments. J Biomed Mater Res A 2019; 107:1021-1030. [PMID: 30675987 PMCID: PMC6594056 DOI: 10.1002/jbm.a.36618] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 11/02/2018] [Accepted: 11/26/2018] [Indexed: 01/10/2023]
Abstract
In vitro synthesis of bone tissue has been paid attention in recent years; however, current methods to fabricate bone tissue are still ineffective due to some remaining gaps in the understanding of real in vivo bone formation process, and application of the knowledge in bone synthesis. Therefore, the objectives of this study were first, to perform a systematic and ultrastructural investigation of the initial mineral formation during intramembranous ossification of mouse calvaria from a material scientists' viewpoint, and to develop novel mineralization methods based on the in vivo findings. First, the very initial mineral deposition was found to occur at embryonic day E14.0 in mouse calvaria. Analysis of the initial bone formation process showed that it involved the following distinct steps: collagen secretion, matrix vesicle (MV) release, MV mineralization, MV rupture, and collagen fiber mineralization. Next, we performed in vitro mineralization experiments using MVs and hydrogel scaffolds. Intact MVs embedded in collagen gel did not mineralize, whereas, interestingly, MV nanofragments obtained by ultrasonication could promote rapid mineralization. These results indicate that mechanically ruptured MV membrane can be a promising material for in vitro bone tissue synthesis. © 2019 The Authors. journal Of Biomedical Materials Research Part A Published By Wiley Periodicals, Inc. J Biomed Mater Res Part A: 107A: 1021-1030, 2019.
Collapse
Affiliation(s)
- Yosuke Kunitomi
- Department of BiomaterialsOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
- Department of OrthodonticsOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Emilio Satoshi Hara
- Department of BiomaterialsOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Masahiro Okada
- Department of BiomaterialsOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Noriyuki Nagaoka
- Advanced Research Center for Oral and Craniofacial SciencesOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Takuo Kuboki
- Department of Oral Rehabilitation and Regenerative MedicineOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Takayoshi Nakano
- Division of Materials and Manufacturing ScienceGraduate School of Engineering, Osaka UniversityOsakaJapan
| | - Hiroshi Kamioka
- Department of OrthodonticsOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| | - Takuya Matsumoto
- Department of BiomaterialsOkayama University Graduate School of Medicine, Dentistry and Pharmaceutical SciencesOkayamaJapan
| |
Collapse
|
14
|
Boot-Handford RP. Gene cloning to clinical trials-the trials and tribulations of a life with collagen. Int J Exp Pathol 2019; 100:4-11. [PMID: 30912609 DOI: 10.1111/iep.12311] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Revised: 02/19/2019] [Accepted: 02/24/2019] [Indexed: 12/17/2022] Open
Abstract
This review, based on the BSMB Fell-Muir Lecture I presented in July 2018 at the Matrix Biology Europe Conference in Manchester, gives a personal perspective of my own laboratory's contributions to research into type X collagen, metaphyseal chondrodysplasia type Schmid and potential treatments for this disorder that are currently entering clinical trial. I have tried to set the advances made in the context of the scientific technologies available at the time and how these have changed over the more than three decades of this research.
Collapse
Affiliation(s)
- Raymond P Boot-Handford
- Wellcome Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| |
Collapse
|
15
|
Recent Advances on Relationship Between Inorganic Phosphate and Pathologic Calcification: Is Calcification After Breast Augmentation with Fat Grafting Correlated with Locally Increased Concentration of Inorganic Phosphate? Aesthetic Plast Surg 2019; 43:243-252. [PMID: 30552471 DOI: 10.1007/s00266-018-1285-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2018] [Accepted: 11/24/2018] [Indexed: 12/29/2022]
Abstract
BACKGROUND Pathologic calcification has frequently occurred after breast augmentation with fat grafting as well as other conditions such as breast cancer, trauma, myocardial infarction, arteriosclerosis and even after reduction mammoplasty. Inorganic phosphate, correlated with fat metabolism, is an important factor that induces pathologic calcification such as vascular calcification. METHODS A literature search was conducted using PubMed with the keywords: calcification, inorganic phosphate, fat. Studies related to the process of pathologic calcification, correlation between inorganic phosphate and pathologic calcification, between inorganic phosphate and fat metabolism in pathologic calcification were collected. RESULTS Various mechanisms were referred to in pathologic calcification among which inorganic phosphate played an important role. Inorganic phosphate could be liberated, under the effect of various enzymes, in the process of fat metabolism. The authors hypothesized that a large-scale necrotizing zone, which could occur in fat grafting with large amounts per cannula, might provide a high-phosphate environment which might contribute to differentiation of surrounding cells such as stem cells or regenerated vessel cells into osteoblast-like cells that induce pathologic calcification. CONCLUSION Inorganic phosphate, which was correlated with fat metabolism, played a significant role in pathologic calcification. We firstly hypothesize that calcification after fat grafting may be related to locally increasing concentrations of phosphate in a necrotizing zone. Further research should be conducted to verify this hypothesis. LEVEL OF EVIDENCE V This journal requires that authors assign a level of evidence to each article. For a full description of these Evidence-Based Medicine ratings, please refer to the Table of Contents or the online Instructions to Authors www.springer.com/00266 .
Collapse
|
16
|
Zhang J, Xu S, Zhang Y, Zou S, Li X. Effects of equibiaxial mechanical stretch on extracellular matrix-related gene expression in human calvarial osteoblasts. Eur J Oral Sci 2018; 127:10-18. [PMID: 30474904 DOI: 10.1111/eos.12595] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Mechanical stretch commonly promotes craniofacial suture remodeling during interceptive orthodontics. The mechanical responses of osteoblasts in craniofacial sutures play a role in suture remodeling. Moreover, the extracellular matrix (ECM) produced by osteoblasts is crucial for the transduction of mechanical signals that promote cell differentiation. Therefore, we aimed to investigate the effect of mechanical stretch on cell viability and ECM-related gene-expression changes in human osteoblasts. Human calvarial osteoblasts (HCObs) were subjected to 2% deformation. Caspase activity, MTT, and cell viability assays were used to estimate osteoblast apoptosis, proliferation, and viability, respectively. Real-time RT-PCR (RT2 -PCR) arrays were used to assess expression of cytoskeletal-, apoptosis-, osteogenesis-, and ECM-related genes. We found that mechanical stretch significantly increased osteoblast viability and cell proliferation, and decreased the activities of caspases 3 and 7. Moreover, the expression of 18 genes related to osteoblast differentiation, apoptosis, and ECM remodeling changed by more than two-fold in a time-dependent manner. Therefore, mechanical stretch promotes HCOb viability and alters expression of genes that are closely related to suture remodeling under mechanical stretch.
Collapse
Affiliation(s)
- Jiawei Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shuhao Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yanggen Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Xiaobing Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Department of Pediatric Dentistry, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
17
|
Michigami T, Kawai M, Yamazaki M, Ozono K. Phosphate as a Signaling Molecule and Its Sensing Mechanism. Physiol Rev 2018; 98:2317-2348. [DOI: 10.1152/physrev.00022.2017] [Citation(s) in RCA: 78] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In mammals, phosphate balance is maintained by influx and efflux via the intestines, kidneys, bone, and soft tissue, which involves multiple sodium/phosphate (Na+/Pi) cotransporters, as well as regulation by several hormones. Alterations in the levels of extracellular phosphate exert effects on both skeletal and extra-skeletal tissues, and accumulating evidence has suggested that phosphate itself evokes signal transduction to regulate gene expression and cell behavior. Several in vitro studies have demonstrated that an elevation in extracellular Piactivates fibroblast growth factor receptor, Raf/MEK (mitogen-activated protein kinase/ERK kinase)/ERK (extracellular signal-regulated kinase) pathway and Akt pathway, which might involve the type III Na+/Picotransporter PiT-1. Excessive phosphate loading can lead to various harmful effects by accelerating ectopic calcification, enhancing oxidative stress, and dysregulating signal transduction. The responsiveness of mammalian cells to altered extracellular phosphate levels suggests that they may sense and adapt to phosphate availability, although the precise mechanism for phosphate sensing in mammals remains unclear. Unicellular organisms, such as bacteria and yeast, use some types of Pitransporters and other molecules, such as kinases, to sense the environmental Piavailability. Multicellular animals may need to integrate signals from various organs to sense the phosphate levels as a whole organism, similarly to higher plants. Clarification of the phosphate-sensing mechanism in humans may lead to the development of new therapeutic strategies to prevent and treat diseases caused by phosphate imbalance.
Collapse
Affiliation(s)
- Toshimi Michigami
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Masanobu Kawai
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Miwa Yamazaki
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| | - Keiichi Ozono
- Department of Bone and Mineral Research, Research Institute, Osaka Women’s and Children’s Hospital, Osaka Prefectural Hospital Organization, Izumi, Osaka, Japan; and Department of Pediatrics, Osaka University Graduate School of Medicine, Suita, Osaka, Japan
| |
Collapse
|
18
|
Probst K, Stermann J, von Bomhard I, Etich J, Pitzler L, Niehoff A, Bluhm B, Xu HC, Lang PA, Chmielewski M, Abken H, Blissenbach B, Machova A, Papadopoulou N, Brachvogel B. Depletion of Collagen IX Alpha1 Impairs Myeloid Cell Function. Stem Cells 2018; 36:1752-1763. [DOI: 10.1002/stem.2892] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 06/06/2018] [Accepted: 07/02/2018] [Indexed: 12/22/2022]
Affiliation(s)
- Kristina Probst
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Jacek Stermann
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Inga von Bomhard
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Julia Etich
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Lena Pitzler
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Anja Niehoff
- Institute of Biomechanics and Orthopaedics; German Sport University Cologne; Cologne Germany
- Cologne Center for Musculoskeletal Biomechanics (CCMB); University of Cologne; Cologne Germany
| | - Björn Bluhm
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| | - Haifeng C. Xu
- Department of Molecular Medicine II, Medical Faculty; Heinrich Heine University; Düsseldorf Germany
| | - Philipp A. Lang
- Department of Molecular Medicine II, Medical Faculty; Heinrich Heine University; Düsseldorf Germany
| | - Markus Chmielewski
- Center for Molecular Medicine Cologne (CMMC); University of Cologne; Cologne Germany
- Department I Internal Medicine, Medical Faculty; Cologne Germany
- RCI, Chair Gene-Immunotherapy; University Hospital Regensburg; Regensburg Germany
| | - Hinrich Abken
- Center for Molecular Medicine Cologne (CMMC); University of Cologne; Cologne Germany
- Department I Internal Medicine, Medical Faculty; Cologne Germany
- RCI, Chair Gene-Immunotherapy; University Hospital Regensburg; Regensburg Germany
| | - Birgit Blissenbach
- Institute for Medical Microbiology, Immunology and Hygiene, Medical Faculty; University of Cologne; Cologne Germany
| | - Alzbeta Machova
- Institute for Medical Microbiology, Immunology and Hygiene, Medical Faculty; University of Cologne; Cologne Germany
| | - Nikoletta Papadopoulou
- Institute for Medical Microbiology, Immunology and Hygiene, Medical Faculty; University of Cologne; Cologne Germany
| | - Bent Brachvogel
- Department of Pediatrics and Adolescent Medicine, Experimental Neonatology, Medical Faculty; University of Cologne; Cologne Germany
- Center for Biochemistry, Medical Faculty; University of Cologne; Cologne Germany
| |
Collapse
|
19
|
Bottini M, Mebarek S, Anderson KL, Strzelecka-Kiliszek A, Bozycki L, Simão AMS, Bolean M, Ciancaglini P, Pikula JB, Pikula S, Magne D, Volkmann N, Hanein D, Millán JL, Buchet R. Matrix vesicles from chondrocytes and osteoblasts: Their biogenesis, properties, functions and biomimetic models. Biochim Biophys Acta Gen Subj 2018; 1862:532-546. [PMID: 29108957 PMCID: PMC5801150 DOI: 10.1016/j.bbagen.2017.11.005] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2017] [Revised: 10/28/2017] [Accepted: 11/01/2017] [Indexed: 01/01/2023]
Abstract
BACKGROUND Matrix vesicles (MVs) are released from hypertrophic chondrocytes and from mature osteoblasts, the cells responsible for endochondral and membranous ossification. Under pathological conditions, they can also be released from cells of non-skeletal tissues such as vascular smooth muscle cells. MVs are extracellular vesicles of approximately 100-300nm diameter harboring the biochemical machinery needed to induce mineralization. SCOPE OF THE REVIEW The review comprehensively delineates our current knowledge of MV biology and highlights open questions aiming to stimulate further research. The review is constructed as a series of questions addressing issues of MVs ranging from their biogenesis and functions, to biomimetic models. It critically evaluates experimental data including their isolation and characterization methods, like lipidomics, proteomics, transmission electron microscopy, atomic force microscopy and proteoliposome models mimicking MVs. MAJOR CONCLUSIONS MVs have a relatively well-defined function as initiators of mineralization. They bind to collagen and their composition reflects the composition of lipid rafts. We call attention to the as yet unclear mechanisms leading to the biogenesis of MVs, and how minerals form and when they are formed. We discuss the prospects of employing upcoming experimental models to deepen our understanding of MV-mediated mineralization and mineralization disorders such as the use of reconstituted lipid vesicles, proteoliposomes and, native sample preparations and high-resolution technologies. GENERAL SIGNIFICANCE MVs have been extensively investigated owing to their roles in skeletal and ectopic mineralization. MVs serve as a model system for lipid raft structures, and for the mechanisms of genesis and release of extracellular vesicles.
Collapse
Affiliation(s)
- Massimo Bottini
- University of Rome Tor Vergata, Department of Experimental Medicine and Surgery, 00133 Roma, Italy; Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Saida Mebarek
- Universite Lyon 1, UFR Chimie Biochimie, 69 622 Villeurbanne Cedex, France; ICBMS UMR 5246 CNRS, 69 622 Villeurbanne Cedex, France; INSA, Lyon, 69 622 Villeurbanne Cedex, France; CPE, Lyon, 69 622 Villeurbanne Cedex, France; Universite de Lyon, 69 622 Villeurbanne Cedex, France
| | - Karen L Anderson
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Agnieszka Strzelecka-Kiliszek
- Nencki Institute of Experimental Biology, Department of Biochemistry, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Lukasz Bozycki
- Nencki Institute of Experimental Biology, Department of Biochemistry, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Ana Maria Sper Simão
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, USP, Departamento de Química, 14040-901 Ribeirão Preto, SP, Brazil
| | - Maytê Bolean
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, USP, Departamento de Química, 14040-901 Ribeirão Preto, SP, Brazil
| | - Pietro Ciancaglini
- Faculdade de Filosofia, Ciências e Letras de Ribeirão Preto, USP, Departamento de Química, 14040-901 Ribeirão Preto, SP, Brazil
| | - Joanna Bandorowicz Pikula
- Nencki Institute of Experimental Biology, Department of Biochemistry, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - Slawomir Pikula
- Nencki Institute of Experimental Biology, Department of Biochemistry, Polish Academy of Sciences, 02-093 Warsaw, Poland
| | - David Magne
- Universite Lyon 1, UFR Chimie Biochimie, 69 622 Villeurbanne Cedex, France; ICBMS UMR 5246 CNRS, 69 622 Villeurbanne Cedex, France; INSA, Lyon, 69 622 Villeurbanne Cedex, France; CPE, Lyon, 69 622 Villeurbanne Cedex, France; Universite de Lyon, 69 622 Villeurbanne Cedex, France
| | - Niels Volkmann
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Dorit Hanein
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - José Luis Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, USA
| | - Rene Buchet
- Universite Lyon 1, UFR Chimie Biochimie, 69 622 Villeurbanne Cedex, France; ICBMS UMR 5246 CNRS, 69 622 Villeurbanne Cedex, France; INSA, Lyon, 69 622 Villeurbanne Cedex, France; CPE, Lyon, 69 622 Villeurbanne Cedex, France; Universite de Lyon, 69 622 Villeurbanne Cedex, France.
| |
Collapse
|
20
|
Grewal T, Wason SJ, Enrich C, Rentero C. Annexins - insights from knockout mice. Biol Chem 2017; 397:1031-53. [PMID: 27318360 DOI: 10.1515/hsz-2016-0168] [Citation(s) in RCA: 64] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2016] [Accepted: 06/14/2016] [Indexed: 12/23/2022]
Abstract
Annexins are a highly conserved protein family that bind to phospholipids in a calcium (Ca2+) - dependent manner. Studies with purified annexins, as well as overexpression and knockdown approaches identified multiple functions predominantly linked to their dynamic and reversible membrane binding behavior. However, most annexins are found at multiple locations and interact with numerous proteins. Furthermore, similar membrane binding characteristics, overlapping localizations and shared interaction partners have complicated identification of their precise functions. To gain insight into annexin function in vivo, mouse models deficient of annexin A1 (AnxA1), A2, A4, A5, A6 and A7 have been generated. Interestingly, with the exception of one study, all mice strains lacking one or even two annexins are viable and develop normally. This suggested redundancy within annexins, but examining these knockout (KO) strains under stress conditions revealed striking phenotypes, identifying underlying mechanisms specific for individual annexins, often supporting Ca2+ homeostasis and membrane transport as central for annexin biology. Conversely, mice lacking AnxA1 or A2 show extracellular functions relevant in health and disease that appear independent of membrane trafficking or Ca2+ signaling. This review will summarize the mechanistic insights gained from studies utilizing mouse models lacking members of the annexin family.
Collapse
|
21
|
Nasi S, Ea HK, Lioté F, So A, Busso N. Sodium Thiosulfate Prevents Chondrocyte Mineralization and Reduces the Severity of Murine Osteoarthritis. PLoS One 2016; 11:e0158196. [PMID: 27391970 PMCID: PMC4938519 DOI: 10.1371/journal.pone.0158196] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/13/2016] [Indexed: 12/02/2022] Open
Abstract
Objectives Calcium-containing crystals participate in the pathogenesis of OA. Sodium thiosulfate (STS) has been shown to be an effective treatment in calcification disorders such as calciphylaxis and vascular calcification. This study investigated the effects and mechanisms of action of STS in a murine model of OA and in chondrocyte calcification. Methods Hydroxyapatite (HA) crystals-stimulated murine chondrocytes and macrophages were treated with STS. Mineralization and cellular production of IL-6, MCP-1 and reactive oxygen species (ROS) were assayed. STS's effects on genes involved in calcification, inflammation and cartilage matrix degradation were studied by RT-PCR. STS was administered in the menisectomy model of murine OA, and the effect on periarticular calcific deposits and cartilage degeneration was investigated by micro-CT-scan and histology. Results In vitro, STS prevented in a dose-dependent manner calcium crystal deposition in chondrocytes and inhibited Annexin V gene expression. In addition, there was a reduction in crystal-induced IL-6 and MCP-1 production. STS also had an antioxidant effect, diminished HA-induced ROS generation and abrogated HA-induced catabolic responses in chondrocytes. In vivo, administration of STS reduced the histological severity of OA, by limiting the size of new periarticular calcific deposits and reducing the severity of cartilage damage. Conclusions STS reduces the severity of periarticular calcification and cartilage damage in an animal model of OA via its effects on chondrocyte mineralization and its attenuation of crystal-induced inflammation as well as catabolic enzymes and ROS generation. Our study suggests that STS may be a disease-modifying drug in crystal-associated OA.
Collapse
Affiliation(s)
- Sonia Nasi
- Service of Rheumatology, Department of Musculoskeletal Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
- * E-mail:
| | - Hang-Korng Ea
- Hospital Lariboisière, Service of Rheumatology, University School of Medicine, Paris VII, Paris, France
| | - Frédéric Lioté
- Hospital Lariboisière, Service of Rheumatology, University School of Medicine, Paris VII, Paris, France
| | - Alexander So
- Service of Rheumatology, Department of Musculoskeletal Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| | - Nathalie Busso
- Service of Rheumatology, Department of Musculoskeletal Medicine, Centre Hospitalier Universitaire Vaudois and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
22
|
[Role of cartilage extracellular matrix for the development and function of the immune system]. Z Rheumatol 2015; 74:711-3. [PMID: 26450435 DOI: 10.1007/s00393-015-1650-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
23
|
Etich J, Holzer T, Pitzler L, Bluhm B, Brachvogel B. MiR-26a modulates extracellular matrix homeostasis in cartilage. Matrix Biol 2015; 43:27-34. [PMID: 25766405 DOI: 10.1016/j.matbio.2015.02.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2015] [Revised: 02/27/2015] [Accepted: 02/28/2015] [Indexed: 01/18/2023]
Abstract
MicroRNAs (miRNAs) may represent new therapeutic targets for bone and joint diseases. We hypothesized that several cartilage-specific proteins are targeted by a single miRNA and used bioinformatics to identify a miRNA that can modulate extracellular matrix (ECM) homeostasis in cartilage. Bioinformatic analysis of miRNA binding sequences in the 3'-untranslated region (3'-UTR) of target genes was performed to identify a miRNA that could bind to the 3'-UTR of cartilage matrix-related genes. MiRNA expression was studied by quantitative PCR of microdissected growth plate cartilage and binding to the 3'-UTR sequences was analyzed by luciferase interaction studies. Levels of proteins encoded by target genes in cultures of miR-26a mimic- or inhibitor-transfected chondrocytes were determined by FACS or immunoblot analysis. The complementary binding sequence of miR-26a and miR-26b was found in the 3'-UTR of the prehypertrophic/hypertrophic-specific genes Cd200, Col10a1 as well as Col9a1 and Ctgf. Both miRNAs were expressed in cartilage and only miR-26a was downregulated in hypertrophic growth plate cartilage. MiR-26a could interact with the 3'-UTR of Cd200 and Col10a1 in luciferase binding studies, but not with Col9a1 and Ctgf. However, protein expression of target genes and the ECM adaptor genes matrilin-3 and COMP was significantly altered in miR-26a mimic- or inhibitor-transfected chondrocytes, whereas the abundance of the cell surface receptor for insulin was not changed. In conclusion, miR-26a suppresses hypertrophic and ECM adaptor protein production. Dysregulation of miR-26a expression could contribute to ECM changes in cartilage diseases and this miRNA may therefore act as a therapeutic target.
Collapse
Affiliation(s)
- Julia Etich
- Experimental Neonatology, Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, Cologne, Germany; Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Tatjana Holzer
- Experimental Neonatology, Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, Cologne, Germany; Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Lena Pitzler
- Experimental Neonatology, Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, Cologne, Germany; Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Björn Bluhm
- Experimental Neonatology, Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, Cologne, Germany; Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany
| | - Bent Brachvogel
- Experimental Neonatology, Department of Pediatrics and Adolescent Medicine, Medical Faculty, University of Cologne, Cologne, Germany; Center for Biochemistry, Medical Faculty, University of Cologne, Cologne, Germany.
| |
Collapse
|
24
|
Gu J, Lu Y, Li F, Qiao L, Wang Q, Li N, Borgia JA, Deng Y, Lei G, Zheng Q. Identification and characterization of the novel Col10a1 regulatory mechanism during chondrocyte hypertrophic differentiation. Cell Death Dis 2014; 5:e1469. [PMID: 25321476 PMCID: PMC4649528 DOI: 10.1038/cddis.2014.444] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2014] [Revised: 09/02/2014] [Accepted: 09/03/2014] [Indexed: 02/03/2023]
Abstract
The majority of human skeleton develops through the endochondral pathway, in which cartilage-forming chondrocytes proliferate and enlarge into hypertrophic chondrocytes that eventually undergo apoptosis and are replaced by bone. Although at a terminal differentiation stage, hypertrophic chondrocytes have been implicated as the principal engine of bone growth. Abnormal chondrocyte hypertrophy has been seen in many skeletal dysplasia and osteoarthritis. Meanwhile, as a specific marker of hypertrophic chondrocytes, the type X collagen gene (COL10A1) is also critical for endochondral bone formation, as mutation and altered COL10A1 expression are often accompanied by abnormal chondrocyte hypertrophy in many skeletal diseases. However, how the type X collagen gene is regulated during chondrocyte hypertrophy has not been fully elucidated. We have recently demonstrated that Runx2 interaction with a 150-bp mouse Col10a1 cis-enhancer is required but not sufficient for its hypertrophic chondrocyte-specific reporter expression in transgenic mice, suggesting requirement of additional Col10a1 regulators. In this study, we report in silico sequence analysis of this 150-bp enhancer and identification of its multiple binding factors, including AP1, MEF2, NFAT, Runx1 and TBX5. Using this enhancer as bait, we performed yeast one-hybrid assay and identified multiple candidate Col10a1-interacting genes, including cyclooxygenase 1 (Cox-1) and Cox-2. We have also performed mass spectrometry analysis and detected EF1-alpha, Fus, GdF7 and Runx3 as components of the specific complex formed by the cis-enhancer and nuclear extracts from hypertrophic MCT (mouse chondrocytes immortalized with large T antigen) cells that express Col10a1 abundantly. Notably, some of the candidate genes are differentially expressed in hypertrophic MCT cells and have been associated with chondrocyte hypertrophy and Runx2, an indispensible Col10a1 regulator. Intriguingly, we detected high-level Cox-2 expression in hypertrophic MCT cells. Electrophoretic mobility shift assay and chromatin immunoprecipitation assays confirmed the interaction between Cox-2 and Col10a1 cis-enhancer, supporting its role as a candidate Col10a1 regulator. Together, our data support a Cox-2-containing, Runx2-centered Col10a1 regulatory mechanism, during chondrocyte hypertrophic differentiation.
Collapse
Affiliation(s)
- J Gu
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, China
| | - Y Lu
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, China
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| | - F Li
- Department of Pathophysiology, Anhui Medical University, Hefei, China
| | - L Qiao
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, China
| | - Q Wang
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, China
| | - N Li
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, China
| | - J A Borgia
- Department of Pathology and Department of Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - Y Deng
- Department of Internal Medicine and Biochemistry, Rush University Medical Center, Chicago, IL, USA
| | - G Lei
- Department of Orthopaedic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Q Zheng
- Department of Hematology and Hematological Laboratory Science, School of Medical Science and Laboratory Medicine, Jiangsu University, Zhenjiang, China
- Department of Anatomy and Cell Biology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
25
|
Lu Y, Qiao L, Lei G, Mira RR, Gu J, Zheng Q. Col10a1 gene expression and chondrocyte hypertrophy during skeletal development and disease. ACTA ACUST UNITED AC 2014. [DOI: 10.1007/s11515-014-1310-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
26
|
Abstract
PURPOSE OF REVIEW Osteoarthritis is the most common form of rheumatologic disease, with numerous factors increasing the risk of developing the condition; calcification of cartilage is common place in osteoarthritis. Regardless of these risk factors, certain disorders predispose individuals to developing arthritis. Pathologic mechanisms in cartilage calcification and advances in their understanding are reviewed alongside metabolic and endocrine arthritis. RECENT FINDINGS There is growing evidence suggesting that changes in chondrocytes and the extracellular environment both contribute to the calcification. Further evidence suggests that signaling cascades that are involved in physiological mineralisation are involved in the pathological process(es); data in mouse models continue to add weight to these hypotheses and correlate with human osteoarthritis data. Recent study of rare forms of arthritis is adding useful information that may help understand joint diseases in the general population and how therapies may be targeted. SUMMARY There is little doubt that calcium-containing crystals are involved in the osteoarthritis process contributing both biomechanically and biochemically. Understanding the processes involved provides important therapeutic opportunities. Furthermore, important information is often discovered in studying rare conditions in which these pathologies are inevitable.
Collapse
|
27
|
New SE, Goettsch C, Aikawa M, Marchini JF, Shibasaki M, Yabusaki K, Libby P, Shanahan CM, Croce K, Aikawa E. Macrophage-derived matrix vesicles: an alternative novel mechanism for microcalcification in atherosclerotic plaques. Circ Res 2013; 113:72-7. [PMID: 23616621 PMCID: PMC3703850 DOI: 10.1161/circresaha.113.301036] [Citation(s) in RCA: 343] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
RATIONALE We previously showed that early calcification of atherosclerotic plaques associates with macrophage accumulation. Chronic renal disease and mineral imbalance accelerate calcification and the subsequent release of matrix vesicles (MVs), precursors of microcalcification. OBJECTIVE We tested the hypothesis that macrophage-derived MVs contribute directly to microcalcification. METHODS AND RESULTS Macrophages associated with regions of calcified vesicular structures in human carotid plaques (n=136 patients). In vitro, macrophages released MVs with high calcification and aggregation potential. MVs expressed exosomal markers (CD9 and TSG101) and contained S100A9 and annexin V. Silencing S100A9 in vitro and genetic deficiency in S100A9-/- mice reduced MV calcification, whereas stimulation with S100A9 increased calcification potential. Externalization of phosphatidylserine after Ca/P stimulation and interaction of S100A9 and annexin V indicated that a phosphatidylserine-annexin V-S100A9 membrane complex facilitates hydroxyapatite nucleation within the macrophage-derived MV membrane. CONCLUSIONS Our results support the novel concept that macrophages release calcifying MVs enriched in S100A9 and annexin V, which contribute to accelerated microcalcification in chronic renal disease.
Collapse
Affiliation(s)
- Sophie E. New
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Claudia Goettsch
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Masanori Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division and Center for Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Julio F. Marchini
- Cardiovascular Division and Center for Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Manabu Shibasaki
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Katsumi Yabusaki
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Peter Libby
- Cardiovascular Division and Center for Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | | | - Kevin Croce
- Cardiovascular Division and Center for Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
- Cardiovascular Division and Center for Excellence in Vascular Biology, Cardiovascular Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA
| |
Collapse
|
28
|
New SEP, Aikawa E. Role of extracellular vesicles in de novo mineralization: an additional novel mechanism of cardiovascular calcification. Arterioscler Thromb Vasc Biol 2013; 33:1753-8. [PMID: 23766262 DOI: 10.1161/atvbaha.112.300128] [Citation(s) in RCA: 103] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Extracellular vesicles are membrane micro/nanovesicles secreted by many cell types into the circulation and the extracellular milieu in physiological and pathological conditions. Evidence suggests that extracellular vesicles, known as matrix vesicles, play a role in the mineralization of skeletal tissue, but emerging ultrastructural and in vitro studies have demonstrated their contribution to cardiovascular calcification as well. Cells involved in the progression of cardiovascular calcification release active vesicles capable of nucleating hydroxyapatite on their membranes. This review discusses the role of extracellular vesicles in cardiovascular calcification and elaborates on this additional mechanism of calcification as an alternative pathway to the currently accepted mechanism of biomineralization via osteogenic differentiation.
Collapse
Affiliation(s)
- Sophie E P New
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | | |
Collapse
|
29
|
Jiang L, Cui Y, Luan J, Zhou X, Zhou X, Han J. A comparative proteomics study on matrix vesicles of osteoblast-like Saos-2 and U2-OS cells. Intractable Rare Dis Res 2013; 2:59-62. [PMID: 25343104 PMCID: PMC4204581 DOI: 10.5582/irdr.2013.v2.2.59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2013] [Revised: 05/16/2013] [Accepted: 05/17/2013] [Indexed: 01/15/2023] Open
Abstract
Matrix vesicles (MVs) play an important role in the initial stage of the process of bone mineralization, and are involved in multiple rare skeletal diseases with pathological mineralization or calcification. The aim of the study was to compare the proteomic profiling of osteoblast-like cells with and without mineralization ability (Saos-2 and U2-OS), and to identify novel mineralization-associated MV proteins. MVs were extracted using ExoQuick solution from mineralization-induced Saos-2 and U2-OS cells, and then were validated by transmission electron microscopy. A label-free quantitative proteomic method was used to compare the protein profiling of MVs from Saos-2 and U2-OS cells. Western-blots were used to confirm the expression of MVs proteins identified in proteomic studies. In our proteomic studies, we identified that 89 mineralization-related proteins were significantly up-regulated in Saos-2 MVs compared with U2-OS MVs. We further validated that two MVs proteins, protein kinase C α and ras-related protein Ral-A, were up-regulated in MVs of Saos-2 cells compared to those of U2-OS cells under mineralization-induction. Our findings suggest that protein kinase C α and ras-related protein Ral-A might be involved in bone mineralization as MVs components.
Collapse
Affiliation(s)
- Liang Jiang
- Shandong Academy of Medical Sciences, Shandong Medical Biotechnology Center, Key Laboratory for Biotech-Drugs of the Ministry of Health, Ji'nan, Shandong, China
- School of Medicine and Life Sciences, University of Ji'nan-Shandong Academy of Medical Science, Ji'nan, Shandong, China
| | - Yazhou Cui
- Shandong Academy of Medical Sciences, Shandong Medical Biotechnology Center, Key Laboratory for Biotech-Drugs of the Ministry of Health, Ji'nan, Shandong, China
| | - Jing Luan
- Shandong Academy of Medical Sciences, Shandong Medical Biotechnology Center, Key Laboratory for Biotech-Drugs of the Ministry of Health, Ji'nan, Shandong, China
| | - Xiaoyan Zhou
- Shandong Academy of Medical Sciences, Shandong Medical Biotechnology Center, Key Laboratory for Biotech-Drugs of the Ministry of Health, Ji'nan, Shandong, China
| | - Xiaoying Zhou
- Shandong Academy of Medical Sciences, Shandong Medical Biotechnology Center, Key Laboratory for Biotech-Drugs of the Ministry of Health, Ji'nan, Shandong, China
| | - Jinxiang Han
- Shandong Academy of Medical Sciences, Shandong Medical Biotechnology Center, Key Laboratory for Biotech-Drugs of the Ministry of Health, Ji'nan, Shandong, China
- Address correspondence to: Dr. Jinxiang Han, Shandong Academy of Medical Sciences, No. 18877 Jing-shi Road, Ji'nan, 250062, Shandong, China. E-mail:
| |
Collapse
|
30
|
Cox RF, Morgan MP. Microcalcifications in breast cancer: Lessons from physiological mineralization. Bone 2013; 53:437-50. [PMID: 23334083 DOI: 10.1016/j.bone.2013.01.013] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2012] [Revised: 01/07/2013] [Accepted: 01/08/2013] [Indexed: 02/02/2023]
Abstract
Mammographic mammary microcalcifications are routinely used for the early detection of breast cancer, however the mechanisms by which they form remain unclear. Two species of mammary microcalcifications have been identified; calcium oxalate and hydroxyapatite. Calcium oxalate is mostly associated with benign lesions of the breast, whereas hydroxyapatite is associated with both benign and malignant tumors. The way in which hydroxyapatite forms within mammary tissue remains largely unexplored, however lessons can be learned from the process of physiological mineralization. Normal physiological mineralization by osteoblasts results in hydroxyapatite deposition in bone. This review brings together existing knowledge from the field of physiological mineralization and juxtaposes it with our current understanding of the genesis of mammary microcalcifications. As an increasing number of breast cancers are being detected in their non-palpable stage through mammographic microcalcifications, it is important that future studies investigate the underlying mechanisms of their formation in order to fully understand the significance of this unique early marker of breast cancer.
Collapse
Affiliation(s)
- Rachel F Cox
- Molecular and Cellular Therapeutics, Royal College of Surgeons in Ireland, 123 St. Stephen's Green, Dublin 2, Ireland
| | | |
Collapse
|