1
|
Hayward SJ, Chesnaye NC, Hole B, Aylward R, Meuleman Y, Torino C, Porto G, Szymczak M, Drechsler C, Dekker FW, Evans M, Jager KJ, Wanner C, Caskey FJ. Protein Biomarkers and Major Cardiovascular Events in Older People With Advanced CKD: The European Quality (EQUAL) Study. Kidney Med 2024; 6:100745. [PMID: 38162538 PMCID: PMC10757029 DOI: 10.1016/j.xkme.2023.100745] [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] [Indexed: 01/03/2024] Open
Abstract
Rationale & Objective Cardiovascular disease is the leading cause of morbidity and mortality in chronic kidney disease (CKD). We investigated 184 inflammatory and cardiovascular proteins to determine their potential as biomarkers for major cardiovascular events (MACEs). Study Design The European Quality (EQUAL) is an observational cohort study that enrolled people aged ≥65 years with an estimated glomerular filtration rate ≤20 mL/min/1.73 m2. Setting & Participants Recruited participants were split into the discovery (n = 611) and replication cohorts (n = 292). Exposure Levels of 184 blood proteins were measured at the baseline visit, and each protein was analyzed individually. Outcome MACE. Analytical Approach Cox proportional hazard models adjusted for age, sex, estimated glomerular filtration rate, previous MACE, and country were used to determine the risk of MACE. Proteins with false discovery rate adjusted P values of <0.05 in the discovery cohort were tested in the replication cohort. Sensitivity analyses were performed by adjusting for traditional risk factors, CKD-specific risk factors, and level of proteinuria and segregating atherosclerotic and nonatherosclerotic MACE. Results During a median follow-up of 2.9 years, 349 people (39%) experienced a MACE. Forty-eight proteins were associated with MACE in the discovery cohort; 9 of these were reproduced in the replication cohort. Three of these proteins maintained a strong association with MACE after adjustment for traditional and CKD-specific risk factors and proteinuria. Tenascin (TNC), fibroblast growth factor-23 (FGF-23), and V-set and immunoglobulin domain-containing protein 2 (VSIG2) were associated with both atherosclerotic and nonatherosclerotic MACE. All replicated proteins except carbonic anhydrase 1 and carbonic anhydrase 3 were associated with nonatherosclerotic MACE. Limitations Single protein concentration measurements and limited follow-up time. Conclusions Our findings corroborate previously reported relationships between FGF-23, vascular cell adhesion protein-1, TNC, and placental growth factor with cardiovascular outcomes in CKD. We identify 5 proteins not previously linked with MACE in CKD that may be targets for future therapies. Plain-Language Summary Kidney disease increases the risk of heart disease, stroke, and other vascular conditions. Blood tests that predict the likelihood of these problems may help to guide treatment, but studies are needed in people with kidney disease. We analyzed blood tests from older people with kidney disease, looking for proteins associated with higher risk of these conditions. Nine proteins were identified, of which 3 showed a strong effect after all other information was considered. This work supports previous research regarding 4 of these proteins and identifies 5 additional proteins that may be associated with higher risk. Further work is needed to confirm our findings and to determine whether these proteins can be used to guide treatment.
Collapse
Affiliation(s)
- Samantha J.L. Hayward
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
| | - Nicholas C. Chesnaye
- Amsterdam UMC, University of Amsterdam, ERA Registry, Medical Informatics, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Quality of Care, Amsterdam, The Netherlands
| | - Barnaby Hole
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - Ryan Aylward
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
| | - Yvette Meuleman
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Claudia Torino
- Institute of Clinical Physiology, National Research Council, Reggio Calabria, Italy
| | - Gaetana Porto
- GOM Bianchi Melacrino Morelli, Reggio Calabria, Italy
| | - Maciej Szymczak
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Wroclaw, Poland
| | | | - Friedo W. Dekker
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marie Evans
- Department of Clinical Sciences Intervention and Technology, Karolinska Institutet and Karolinska, Stockholm, Sweden
| | - Kitty J. Jager
- Amsterdam UMC, University of Amsterdam, ERA Registry, Medical Informatics, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Quality of Care, Amsterdam, The Netherlands
| | - Christoph Wanner
- Division of Nephrology, University Hospital of Wurzburg, Wurzburg, Germany
| | - Fergus J. Caskey
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
| | - EQUAL investigators
- Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Southmead Hospital, North Bristol NHS Trust, Bristol, United Kingdom
- Amsterdam UMC, University of Amsterdam, ERA Registry, Medical Informatics, Amsterdam, The Netherlands
- Amsterdam Public Health Research Institute, Quality of Care, Amsterdam, The Netherlands
- Population Health Sciences, Bristol Medical School, University of Bristol, Bristol, United Kingdom
- Department of Medicine, Faculty of Health Sciences, University of Cape Town, South Africa
- Department of Clinical Epidemiology, Leiden University Medical Center, Leiden, The Netherlands
- Institute of Clinical Physiology, National Research Council, Reggio Calabria, Italy
- GOM Bianchi Melacrino Morelli, Reggio Calabria, Italy
- Department of Nephrology and Transplantation Medicine, Wroclaw Medical University, Wroclaw, Poland
- Division of Nephrology, University Hospital of Wurzburg, Wurzburg, Germany
- Department of Clinical Sciences Intervention and Technology, Karolinska Institutet and Karolinska, Stockholm, Sweden
| |
Collapse
|
2
|
Tin A, Sullivan KJ, Walker KA, Bressler J, Talluri R, Yu B, Simino J, Gudmundsdottir V, Emilsson V, Jennings LL, Launer L, Mei H, Boerwinkle E, Windham BG, Gottesman R, Gudnason V, Coresh J, Fornage M, Mosley TH. Proteomic Analysis Identifies Circulating Proteins Associated With Plasma Amyloid-β and Incident Dementia. BIOLOGICAL PSYCHIATRY GLOBAL OPEN SCIENCE 2023; 3:490-499. [PMID: 37519456 PMCID: PMC10382706 DOI: 10.1016/j.bpsgos.2022.04.005] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 04/13/2022] [Accepted: 04/27/2022] [Indexed: 11/30/2022] Open
Abstract
Background Plasma amyloid-β (Aβ) (Aβ42, Aβ40, and Aβ42/Aβ40), biomarkers of the Alzheimer's form of dementia, are under consideration for clinical use. The associations of these peptides with circulating proteins may identify novel plasma biomarkers of dementia and inform peripheral factors influencing the levels of these peptides. Methods We analyzed the association of these 3 plasma Aβ measures with 4638 circulating proteins among a subset of the participants of the Atherosclerosis Risk in Communities (ARIC) study (midlife: n = 1955; late life: n = 2082), related the Aβ-associated proteins with incident dementia in the overall ARIC cohort (midlife: n = 11,069, late life: n = 4110) with external replication in the Age, Gene/Environment Susceptibility (AGES)-Reykjavik Study (n = 4973), estimated the proportion of Aβ variance explained, and conducted enrichment analyses to characterize the proteins associated with the plasma Aβ peptides. Results At midlife, of the 296 Aβ-associated proteins, 8 were associated with incident dementia from midlife and late life in the ARIC study, and NPPB, IBSP, and THBS2 were replicated in the AGES-Reykjavik Study. At late life, of the 34 Aβ-associated proteins, none were associated with incident dementia at midlife, and kidney function explained 10%, 12%, and 0.2% of the variance of Aβ42, Aβ40, and Aβ42/Aβ40, respectively. Aβ42-associated proteins at midlife were found to be enriched in the liver, and those at late life were found to be enriched in the spleen. Conclusions This study identifies circulating proteins associated with plasma Aβ levels and incident dementia and informs peripheral factors associated with plasma Aβ levels.
Collapse
Affiliation(s)
- Adrienne Tin
- Memory Impairment and Neurodegenerative Dementia (MIND) Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Kevin J. Sullivan
- Memory Impairment and Neurodegenerative Dementia (MIND) Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Keenan A. Walker
- Laboratory of Behavioral Neuroscience, Intramural Research Program, National Institute on Aging, Baltimore, Maryland
| | - Jan Bressler
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
| | - Rajesh Talluri
- Department of Data Science, University of Mississippi Medical Center, Jackson, Mississippi
| | - Bing Yu
- Department of Epidemiology, Human Genetics and Environmental Sciences, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
| | - Jeanette Simino
- Department of Data Science, University of Mississippi Medical Center, Jackson, Mississippi
| | - Valborg Gudmundsdottir
- Faculty of Medicine, University of Iceland, Reykjavik
- Heart Association, Kopavogur, Iceland
| | | | - Lori L. Jennings
- Novartis Institutes for Biomedical Research, Cambridge, Massachusetts
| | - Lenore Launer
- Laboratory of Epidemiology and Population Sciences, Intramural Research Program, National Institute on Aging, Bethesda, Maryland
| | - Hao Mei
- Department of Data Science, University of Mississippi Medical Center, Jackson, Mississippi
| | - Eric Boerwinkle
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
- Human Genome Sequencing Center, Baylor College of Medicine, Houston, Texas
| | - B. Gwen Windham
- Memory Impairment and Neurodegenerative Dementia (MIND) Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| | - Rebecca Gottesman
- Stroke Branch, National Institute of Neurological Disorders and Stroke Intramural Program, National Institutes of Health, Bethesda, Maryland
| | - Vilmundur Gudnason
- Faculty of Medicine, University of Iceland, Reykjavik
- Heart Association, Kopavogur, Iceland
| | - Josef Coresh
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland
| | - Myriam Fornage
- Human Genetics Center, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas
- Institute of Molecular Medicine, McGovern Medical School, University of Texas Health Science at Houston, Houston, Texas
| | - Thomas H. Mosley
- Memory Impairment and Neurodegenerative Dementia (MIND) Center and Department of Medicine, University of Mississippi Medical Center, Jackson, Mississippi
| |
Collapse
|
3
|
Zhou W, Bai Y, Chen J, Li H, Zhang B, Liu H. Revealing the Critical Regulators of Modulated Smooth Muscle Cells in Atherosclerosis in Mice. Front Genet 2022; 13:900358. [PMID: 35677564 PMCID: PMC9168464 DOI: 10.3389/fgene.2022.900358] [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] [Received: 03/20/2022] [Accepted: 04/15/2022] [Indexed: 01/23/2023] Open
Abstract
Background: There are still residual risks for atherosclerosis (AS)-associated cardiovascular diseases to be resolved. Considering the vital role of phenotypic switching of smooth muscle cells (SMCs) in AS, especially in calcification, targeting SMC phenotypic modulation holds great promise for clinical implications. Methods: To perform an unbiased and systematic analysis of the molecular regulatory mechanism of phenotypic switching of SMCs during AS in mice, we searched and included several publicly available single-cell datasets from the GEO database, resulting in an inclusion of more than 80,000 cells. Algorithms implemented in the Seurat package were used for cell clustering and cell atlas depiction. The pySCENIC and SCENIC packages were used to identify master regulators of interested cell groups. Monocle2 was used to perform pseudotime analysis. clusterProfiler was used for Gene Ontology enrichment analysis. Results: After dimensionality reduction and clustering, reliable annotation was performed. Comparative analysis between cells from normal artery and AS lesions revealed that three clusters emerged as AS progression, designated as mSMC1, mSMC2, and mSMC3. Transcriptional and functional enrichment analysis established a continuous transitional mode of SMCs’ transdifferentiation to mSMCs, which is further supported by pseudotime analysis. A total of 237 regulons were identified with varying activity scores across cell types. A potential core regulatory network was constructed for SMC and mSMC subtypes. In addition, module analysis revealed a coordinate regulatory mode of regulons for a specific cell type. Intriguingly, consistent with gain of ossification-related transcriptional and functional characteristics, a corresponding small set of regulators contributing to osteochondral reprogramming was identified in mSMC3, including Dlx5, Sox9, and Runx2. Conclusion: Gene regulatory network inference indicates a hierarchical organization of regulatory modules that work together in fine-tuning cellular states. The analysis here provides a valuable resource that can provide guidance for subsequent biological experiments.
Collapse
Affiliation(s)
- Wenli Zhou
- Medical School of Chinese PLA, Beijing, China
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Yongyi Bai
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Jianqiao Chen
- Medical School of Chinese PLA, Beijing, China
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Huiying Li
- Medical School of Chinese PLA, Beijing, China
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Baohua Zhang
- Medical School of Chinese PLA, Beijing, China
- Department of Health Care, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Hongbin Liu
- Department of Cardiology, The Second Medical Center, Chinese PLA General Hospital, Beijing, China
- *Correspondence: Hongbin Liu,
| |
Collapse
|
4
|
Chen C, Wu Y, Lu HL, Liu K, Qin X. Identification of potential biomarkers of vascular calcification using bioinformatics analysis and validation in vivo. PeerJ 2022; 10:e13138. [PMID: 35313524 PMCID: PMC8934046 DOI: 10.7717/peerj.13138] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 02/28/2022] [Indexed: 01/12/2023] Open
Abstract
Background Vascular calcification (VC) is the most widespread pathological change in diseases of the vascular system. However, we know poorly about the molecular mechanisms and effective therapeutic approaches of VC. Methods The VC dataset, GSE146638, was downloaded from the Gene Expression Omnibus (GEO) database. Using the edgeR package to screen Differentially expressed genes (DEGs). Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) were used to find pathways affecting VC. Gene ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) were performed on the DEGs. Meanwhile, using the String database and Cytoscape software to construct protein-protein interaction (PPI) networks and identify hub genes with the highest module scores. Correlation analysis was performed for hub genes. Receiver operating characteristic (ROC) curves, expression level analysis, GSEA, and subcellular localization were performed for each hub gene. Expression of hub genes in normal and calcified vascular tissues was verified by quantitative reverse transcription PCR (RT-qPCR) and immunohistochemistry (IHC) experiments. The hub gene-related miRNA-mRNA and TF-mRNA networks were constructed and functionally enriched for analysis. Finally, the DGIdb database was utilized to search for alternative drugs targeting VC hub genes. Results By comparing the genes with normal vessels, there were 64 DEGs in mildly calcified vessels and 650 DEGs in severely calcified vessels. Spp1, Sost, Col1a1, Fn1, and Ibsp were central in the progression of the entire VC by the MCODE plug-in. These hub genes are primarily enriched in ossification, extracellular matrix, and ECM-receptor interactions. Expression level results showed that Spp1, Sost, Ibsp, and Fn1 were significantly highly expressed in VC, and Col1a1 was incredibly low. RT-qPCR and IHC validation results were consistent with bioinformatic analysis. We found multiple pathways of hub genes acting in VC and identified 16 targeting drugs. Conclusions This study perfected the molecular regulatory mechanism of VC. Our results indicated that Spp1, Sost, Col1a1, Fn1, and Ibsp could be potential novel biomarkers for VC and promising therapeutic targets.
Collapse
Affiliation(s)
- Chuanzhen Chen
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Yinteng Wu
- Department of Orthopedic and Trauma Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Hai-lin Lu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Kai Liu
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| | - Xiao Qin
- Department of Vascular Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Province, China
| |
Collapse
|
5
|
Jeong K, Murphy JM, Kim JH, Campbell PM, Park H, Rodriguez Y, Choi C, Kim JS, Park S, Kim HJ, Scammell JG, Weber DS, Honkanen RE, Schlaepfer DD, Ahn EYE, Lim STS. FAK Activation Promotes SMC Dedifferentiation via Increased DNA Methylation in Contractile Genes. Circ Res 2021; 129:e215-e233. [PMID: 34702049 DOI: 10.1161/circresaha.121.319066] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Rationale: Vascular smooth muscle cells (SMCs) exhibit remarkable plasticity and can undergo dedifferentiation upon pathological stimuli associated with disease and interventions. Objective: Although epigenetic changes are critical in SMC phenotype switching, a fundamental regulator that governs the epigenetic machineries regulating the fate of SMC phenotype has not been elucidated. Methods and Results: Using SMCs, mouse models, and human atherosclerosis specimens, we found that focal adhesion kinase (FAK) activation elicits SMC dedifferentiation by stabilizing DNA methyltransferase 3A (DNMT3A). FAK in SMCs is activated in the cytoplasm upon serum stimulation in vitro or vessel injury and active FAK prevents DNMT3A from nuclear FAK-mediated degradation. However, pharmacological or genetic FAK catalytic inhibition forced FAK nuclear localization, which reduced DNMT3A protein via enhanced ubiquitination and proteasomal degradation. Reduced DNMT3A protein led to DNA hypomethylation in contractile gene promoters, which increased SMC contractile protein expression. RNA sequencing identified SMC contractile genes as a foremost upregulated group by FAK inhibition from injured femoral artery samples compared to vehicle group. DNMT3A knockdown in injured arteries reduced DNA methylation and enhanced contractile gene expression supports the notion that nuclear FAK-mediated DNMT3A degradation via E3 ligase TRAF6 drives differentiation of SMCs. Furthermore, we observed that SMCs of human atherosclerotic lesions exhibited decreased nuclear FAK, which was associated with increased DNMT3A levels and decreased contractile gene expression. Conclusions: This study reveals that nuclear FAK induced by FAK catalytic inhibition specifically suppresses DNMT3A expression in injured vessels resulting in maintaining SMC differentiation by promoting the contractile gene expression. Thus, FAK inhibitors may provide a new treatment option to block SMC phenotypic switching during vascular remodeling and atherosclerosis.
Collapse
Affiliation(s)
- Kyuho Jeong
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| | - James M Murphy
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| | - Jung-Hyun Kim
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| | | | - Hyeonsoo Park
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, KOREA, REPUBLIC OF
| | - Yelitza Rodriguez
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| | - Chungsik Choi
- Physiology, University of South Alabama College of Medicine, UNITED STATES
| | - Jun-Sub Kim
- Biotechnology, Korea National University of Transportation, KOREA, REPUBLIC OF
| | - Sangwon Park
- Pharmacology, Gyeongsang National University, KOREA, REPUBLIC OF
| | - Hyun Joon Kim
- Anatomy and Convergence Medical Sciences, Gyeongsang National University
| | - Jonathan G Scammell
- Comparative Medicine, University of South Alabama College of Medicine, UNITED STATES
| | - David S Weber
- Physiology and Cell Biology, University of South Alabama College of Medicine, UNITED STATES
| | - Richard E Honkanen
- Biochemistry and Molecualr Biology, University of South Alabama College of Medicine, UNITED STATES
| | - David D Schlaepfer
- Obstetrics, Gynecology, and Reproductive Medicine, University of California, San Diego Moores Cancer Center, UNITED STATES
| | | | - Ssang-Taek Steve Lim
- Biochemistry and Molecular Biology, University of South Alabama College of Medicine, UNITED STATES
| |
Collapse
|
6
|
Hu L, Liu J, Xue H, Panayi AC, Xie X, Lin Z, Wang T, Xiong Y, Hu Y, Yan C, Chen L, Abududilibaier A, Zhou W, Mi B, Liu G. miRNA-92a-3p regulates osteoblast differentiation in patients with concomitant limb fractures and TBI via IBSP/PI3K-AKT inhibition. MOLECULAR THERAPY. NUCLEIC ACIDS 2021; 23:1345-1359. [PMID: 33717654 PMCID: PMC7920808 DOI: 10.1016/j.omtn.2021.02.008] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 02/08/2021] [Indexed: 01/20/2023]
Abstract
Patients who sustain concomitant fractures and traumatic brain injury (TBI) are known to have significantly quicker fracture-healing rates than patients with isolated fractures. The mechanisms underlying this phenomenon have yet to be identified. In the present study, we found that the upregulation of microRNA-92a-3p (miRNA-92a-3p) induced by TBI correlated with a decrease in integrin binding sialoprotein (IBSP) expression in callus formation. In vitro, overexpressing miRNA-92a-3p inhibited IBSP expression and accelerated osteoblast differentiation, whereas silencing of miRNA-92a-3p inhibited osteoblast activity. A decrease in IBSP facilitated osteoblast differentiation via the Phosphatidylinositol 3-kinase/threonine kinase 1 (PI3K/AKT) signaling pathway. Through luciferase assays, we found evidence that IBSP is a miRNA-92a-3p target gene that negatively regulates osteoblast differentiation. Moreover, the present study confirmed that pre-injection of agomiR-92a-3p leads to increased bone formation. Collectively, these results indicate that miRNA-92a-3p overexpression may be a key factor underlying the improved fracture healing observed in TBI patients. Upregulation of miRNA-92a-3p may therefore be a promising therapeutic strategy for promoting fracture healing and preventing nonunion.
Collapse
Affiliation(s)
- Liangcong Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Jing Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Hang Xue
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Adriana C Panayi
- Division of Plastic Surgery, Brigham and Women's Hospital, Harvard Medical School, Boston 02215, USA
| | - Xudong Xie
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Ze Lin
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Tiantian Wang
- Department of Emergency, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yuan Xiong
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Yiqiang Hu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Chengcheng Yan
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Lang Chen
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Abudula Abududilibaier
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Wu Zhou
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Bobin Mi
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| | - Guohui Liu
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, China
| |
Collapse
|
7
|
Qian Y, Li L, Sun Z, Liu J, Yuan W, Wang Z. A multi-omics view of the complex mechanism of vascular calcification. Biomed Pharmacother 2021; 135:111192. [PMID: 33401220 DOI: 10.1016/j.biopha.2020.111192] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 12/19/2020] [Accepted: 12/26/2020] [Indexed: 02/07/2023] Open
Abstract
Vascular calcification is a high incidence and high risk disease with increasing morbidity and high mortality, which is considered the consequence of smooth muscle cell transdifferentiation initiating the mechanism of accumulation of hydroxyl calcium phosphate. Vascular calcification is also thought to be strongly associated with poor outcomes in diabetes and chronic kidney disease. Numerous studies have been accomplished; however, the specific mechanism of the disease remains unclear. Development of the genome project enhanced the understanding of life science and has entered the post-genomic era resulting in a variety of omics techniques used in studies and a large amount of available data; thus, a new perspective on data analysis has been revealed. Omics has a broader perspective and is thus advantageous over a single pathway analysis in the study of complex vascular calcification mechanisms. This paper reviews in detail various omics studies including genomics, proteomics, transcriptomics, metabolomics and multiple group studies on vascular calcification. Advances and deficiencies in the use of omics to study vascular calcification are presented in a comprehensive view. We also review the methodology of the omics studies and omics data analysis and processing. In addition, the methodology and data processing presented here can be applied to other areas. An omics landscape perspective across the boundaries between genomics, transcriptomics, proteomics and metabolomics is used to examine the mechanisms of vascular calcification. The perspective combined with various technologies also provides a direction for the subsequent exploration of clinical significance.
Collapse
Affiliation(s)
- Yongjiang Qian
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Lihua Li
- Department of Pathology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Zhen Sun
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Jia Liu
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Wei Yuan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, 212000, Zhenjiang, China.
| |
Collapse
|
8
|
Argan O, Çıkrıkçı K, Baltacı A, Gencer N. The effects of cardiac drugs on human erythrocyte carbonic anhydrase I and II isozymes. J Enzyme Inhib Med Chem 2020; 35:1359-1362. [PMID: 32567385 PMCID: PMC7717712 DOI: 10.1080/14756366.2020.1781844] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2020] [Revised: 06/05/2020] [Accepted: 06/05/2020] [Indexed: 11/14/2022] Open
Abstract
Cardiovascular diseases are the leading cause of mortality worldwide. In recent years, the relationship between carbonic anhydrase inhibitors and atherosclerosis has attracted attention. In this study, we aimed to determine the in vitro effects of 35 frequently used cardiac drugs on human carbonic anhydrase I (hCA I) and II (hCA II). The inhibitory effects of the drugs on hCA I and hCA II were determined with both the hydratase and esterase methods. The most potent inhibitors observed were propafenone (hCA I: 2.8 µM and hCA II: 3.02 µM) and captopril (hCA I: 1.58 µM and hCA II: 6.25 µM). Isosorbide mononitrate, propranolol, furosemide, and atorvastatin were also potent inhibitors. The inhibitor constant, Ki, value from the Lineweaver-Burk plot for propafenone was 2.38 µM for hCA I and 2.97 µM for hCA II. The tested cardiac drugs showed potent in vitro inhibition of the hCA I and II isozymes. Especially, in patients with atherosclerotic heart disease, these drugs may be preferred primarily due to the beneficial effects of carbonic anhydrase inhibition on atherosclerosis.
Collapse
Affiliation(s)
- Onur Argan
- Department of Cardiology, Faculty of Medicine, Balikesir University, Balikesir, Turkey
| | - Kübra Çıkrıkçı
- Department of Chemistry, Science and Art Faculty, Balikesir University, Balikesir, Turkey
| | - Aybike Baltacı
- Department of Chemistry, Science and Art Faculty, Balikesir University, Balikesir, Turkey
| | - Nahit Gencer
- Department of Chemistry, Science and Art Faculty, Balikesir University, Balikesir, Turkey
| |
Collapse
|
9
|
Duan M, Zhao WL, Zhou L, Novák P, Zhu X, Yin K. Omics research in vascular calcification. Clin Chim Acta 2020; 511:319-328. [PMID: 33096035 DOI: 10.1016/j.cca.2020.10.022] [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: 08/31/2020] [Revised: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023]
Abstract
Vascular calcification (VC), the pathological process of hydroxyapatite mineral deposition in the vascular system, is closely associated with aging, atherosclerotic plaque formation, cardiovascular disease (CVD) and diabetes mellitus (DM). Studies have shown that VC is related to cellular phenotypic changes, extracellular vesicles, disordered calcium and phosphate homeostasis, and an imbalance between inducers and inhibitors of VC. Unfortunately, there is currently no effective preventive or targeted treatment for pathologic condition. The rapid evolution of omics technology (genomics, epigenomics, transcriptomics, proteomics and metabolomics) has provided a novel approach for elucidation of pathophysiologic mechanisms in general and those associated with VC specifically. Here, we review articles published over the last twenty years and focus on the current state, challenges, limitations and future of omics in VC research and clinical practice. Highlighting potential targets based on omics technology will improve our understanding of this pathologic condition and assist in the development of potential treatment options for VC related disease.
Collapse
Affiliation(s)
- Meng Duan
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Wen-Li Zhao
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| |
Collapse
|
10
|
Duan M, Zhao WL, Zhou L, Novák P, Zhu X, Yin K. Omics research in vascular calcification. Clin Chim Acta 2020; 511:198-207. [PMID: 33096032 DOI: 10.1016/j.cca.2020.10.021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 10/15/2020] [Indexed: 02/07/2023]
Abstract
Vascular calcification (VC), the pathological process of hydroxyapatite mineral deposition in the vascular system, is closely associated with aging, atherosclerotic plaque formation, cardiovascular disease (CVD) and diabetes mellitus (DM). Studies have shown that VC is related to cellular phenotypic changes, extracellular vesicles, disordered calcium phosphate homeostasis and an imbalance between inducers and inhibitors of VC. Unfortunately, there is currently no effective preventive or targeted treatment for this disorder. Recently, the evolution of omics technology (genomics, epigenomics, transcriptomics, proteomics and metabolomics) has paved the way for elucidation of complex biochemical processes and, as such, may provide new insight on VC. Accordingly, we conducted a review of articles published over the last twenty years and herein focus on current and future potential of omics technology in clarifying mechanisms of this disease process. Identification of new biomarkers will provide additional tools in characterizing this pathology and will further assist in the development of potential therapeutic targets.
Collapse
Affiliation(s)
- Meng Duan
- Research Lab of Translational Medicine, Hengyang Medical School, University of South China, Hengyang 421001, China; Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Wen-Li Zhao
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Le Zhou
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Petr Novák
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China
| | - Xiao Zhu
- Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| | - Kai Yin
- The Second Affiliated Hospital of Guilin Medical University, Guangxi Key Laboratory of Diabetic Systems Medicine, Guilin Medical University, Guilin 541100, China.
| |
Collapse
|
11
|
Varennes O, Mentaverri R, Duflot T, Kauffenstein G, Objois T, Lenglet G, Avondo C, Morisseau C, Brazier M, Kamel S, Six I, Bellien J. The Metabolism of Epoxyeicosatrienoic Acids by Soluble Epoxide Hydrolase Is Protective against the Development of Vascular Calcification. Int J Mol Sci 2020; 21:ijms21124313. [PMID: 32560362 PMCID: PMC7352784 DOI: 10.3390/ijms21124313] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 06/15/2020] [Indexed: 12/13/2022] Open
Abstract
This study addressed the hypothesis that soluble epoxide hydrolase (sEH), which metabolizes endothelium-derived epoxyeicosatrienoic acids, plays a role in vascular calcification. The sEH inhibitor trans-4-(4-(3-adamantan-1-yl-ureido)-cyclohexyloxy)-benzoic acid (t-AUCB) potentiated the increase in calcium deposition of rat aortic rings cultured in high-phosphate conditions. This was associated with increased tissue-nonspecific alkaline phosphatase activity and mRNA expression level of the osteochondrogenic marker Runx2. The procalcifying effect of t-AUCB was prevented by mechanical aortic deendothelialization or inhibition of the production and action of epoxyeicosatrienoic acids using the cytochrome P450 inhibitor fluconazole and the antagonist 14,15-epoxyeicosa-5(Z)-enoic acid (14,15-EEZE), respectively. Similarly, exogenous epoxyeicosatrienoic acids potentiated the calcification of rat aortic rings through a protein kinase A (PKA)-dependent mechanism and of human aortic vascular smooth muscle cells when sEH was inhibited by t-AUCB. Finally, a global gene expression profiling analysis revealed that the mRNA expression level of sEH was decreased in human carotid calcified plaques compared to adjacent lesion-free sites and was inversely correlated with Runx2 expression. These results show that sEH hydrolase plays a protective role against vascular calcification by reducing the bioavailability of epoxyeicosatrienoic acids.
Collapse
Affiliation(s)
- Olivier Varennes
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Romuald Mentaverri
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
- Department of Biochemistry, Amiens-Picardie University Hospital, 80054 Amiens, France
| | - Thomas Duflot
- Department of Pharmacology, Rouen University Hospital, CEDEX 1, 76031 Rouen, France;
- INSERM U1096, Normandy University, UNIROUEN, F-76000 Rouen, France
| | | | - Thibaut Objois
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Gaëlle Lenglet
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Carine Avondo
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Christophe Morisseau
- Department of Entomology and Nematology and UCD Comprehensive Cancer Center, University of California, Davis, CA 95616, USA;
| | - Michel Brazier
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
| | - Saïd Kamel
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
- Department of Biochemistry, Amiens-Picardie University Hospital, 80054 Amiens, France
| | - Isabelle Six
- MP3CV, EA7517, CURS (Centre de Recherche Universitaire en Santé), University of Picardie Jules Verne, 80025 Amiens, France; (O.V.); (R.M.); (T.O.); (G.L.); (C.A.); (M.B.); (S.K.)
- Correspondence: (I.S.); (J.B.); Tel.: +33-2-32-88-90-30 (J.B.); Fax: +33-2-32-88-91-16 (J.B)
| | - Jeremy Bellien
- Department of Pharmacology, Rouen University Hospital, CEDEX 1, 76031 Rouen, France;
- INSERM U1096, Normandy University, UNIROUEN, F-76000 Rouen, France
- Correspondence: (I.S.); (J.B.); Tel.: +33-2-32-88-90-30 (J.B.); Fax: +33-2-32-88-91-16 (J.B)
| |
Collapse
|
12
|
Abstract
Over the last decades, the association between vascular calcification (VC) and all-cause/cardiovascular mortality, especially in patients with high atherogenic status, such as those with diabetes and/or chronic kidney disease, has been repeatedly highlighted. For over a century, VC has been noted as a passive, degenerative, aging process without any treatment options. However, during the past decades, studies confirmed that mineralization of the arteries is an active, complex process, similar to bone genesis and formation. The main purpose of this review is to provide an update of the existing biomarkers of VC in serum and develop the various pathogenetic mechanisms underlying the calcification process, including the pivotal roles of matrix Gla protein, osteoprotegerin, bone morphogenetic proteins, fetuin-a, fibroblast growth-factor-23, osteocalcin, osteopontin, osteonectin, sclerostin, pyrophosphate, Smads, fibrillin-1 and carbonic anhydrase II.
Collapse
|
13
|
Novikova OA, Nazarkina ZK, Cherepanova AV, Laktionov PP, Chelobanov BP, Murashov IS, Deev RV, Pokushalov EA, Karpenko AA, Laktionov PP. Isolation, culturing and gene expression profiling of inner mass cells from stable and vulnerable carotid atherosclerotic plaques. PLoS One 2019; 14:e0218892. [PMID: 31242269 PMCID: PMC6594632 DOI: 10.1371/journal.pone.0218892] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 06/11/2019] [Indexed: 12/14/2022] Open
Abstract
The connective tissue components that form the atherosclerotic plaque body are produced by the plaque inner mass cells (PIMC), located inside the plaque. We report an approach to isolate and culture cells from the connective tissue of stable and vulnerable human atherosclerotic plaques based on elimination of non-connective tissue cells such as blood and non-plaque intima cells with a lysis buffer. The resulting plaque cells were characterized by growth capacity, morphology, transcriptome profiling and specific protein expression. Plaque cells slowly proliferated for up to three passages unaffected by the use of proliferation stimulants or changes of culture media composition. Stable plaques yielded more cells than vulnerable ones. Plaque cell cultures also contained several morphological cellular types. RNA-seq profiles of plaque cells were different from any of the cell types known to be involved in atherogenesis. The expression of the following proteins was observed in cultured plaque cells: smooth muscle cells marker α-SMA, macrophage marker CD14, extracellular matrix proteins aggrecan, fibronectin, neovascularisation markers VEGF-A, CD105, cellular adhesion receptor CD31 and progenitor/dedifferentiation receptor CD34. Differential expression of several notable transcripts in cells from stable and vulnerable plaques suggests the value of plaque cell culture studies for the search of plaque vulnerability markers.
Collapse
Affiliation(s)
- Olga A. Novikova
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Zhanna K. Nazarkina
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | - Anna V. Cherepanova
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
- * E-mail:
| | - Petr P. Laktionov
- Laboratory of Genomics, SB RAS Institute of Molecular and Cellular Biology, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Boris P. Chelobanov
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
- Novosibirsk State University, Novosibirsk, Russia
| | - Ivan S. Murashov
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | | | - Evgeny A. Pokushalov
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| | - Andrey A. Karpenko
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Pavel P. Laktionov
- “E. Meshalkin National Medical Research Center”, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Laboratory of Molecular Medicine, SB RAS Institute of Chemical Biology and Fundamental Medicine, Novosibirsk, Russia
| |
Collapse
|
14
|
Varennes O, Mary A, Bricca G, Kamel S, Bellien J. Dipeptidyl Peptidase-4 Inhibition Prevents Vascular Calcification by Potentiating the Insulin-Like Growth Factor-1 Signaling Pathway. JACC Basic Transl Sci 2019; 4:113-115. [PMID: 30847425 PMCID: PMC6390676 DOI: 10.1016/j.jacbts.2018.11.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
|
15
|
Xiang L, Xin N, Yuan Y, Hou X, Chen J, Wei N, Gong P. Effect of follicular dendritic cell secreted protein on gene expression of human periodontal ligament cells. Arch Oral Biol 2017; 81:151-159. [PMID: 28544936 DOI: 10.1016/j.archoralbio.2017.05.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 04/27/2017] [Accepted: 05/14/2017] [Indexed: 02/05/2023]
Abstract
OBJECTIVE The objective of this study was to investigate the specific roles of follicular dendritic cell secreted protein (FDC-SP), a protein exists in saliva, in the inhibition of calcium precipitation during periodontal regeneration, as well as affect phenotype expression of human periodontal ligament cells (hPDLCs) during the differentiation process. DESIGN To investigate this, we applied microarray technology to identify gene expression changes in hPDLCs transfected with FDC-SP and then clustered them according to their biological functions. RESULTS One hundred seventy-one genes were found differentially expressed by at least two-fold between FDC-SP -transfected and empty vector-transfected cells. Besides, genes encoding cell-cycle proteins, blood-related and cell differentiation-related proteins tended to be up-regulated after FDC-SP transfection, whereas cytokine/growth factors, signal transduction and metabolism-related genes tended to be down-regulated in hPDLCs overexpression FDC-SP. CONCLUSIONS The present study investigated FDC-SP's roles in hPDLCs' phenotype expression, via comparing the gene expression profiles between FDC-SP -transfected hPDLCs and empty vector-transfected cells upon microarray analysis. hPDLCs overexpression FDC-SP appear to display different gene expression patterns. In all, these observations showed a potential of FDC-SP in the maintenance of PDL homeostasis and its ultimate contribution to periodontal would-healing processes.
Collapse
Affiliation(s)
- Lin Xiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Na Xin
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Ying Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Xiaogang Hou
- College of Hydraulic and Hydroelectric Engineering, Sichuan University, Chengdu 610041, China.
| | - Junwei Chen
- Department of Energy and Resources Engineering, College of Engineering, Peking University, Beijing 100000, China.
| | - Na Wei
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| | - Ping Gong
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China; Department of Oral Implantology, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
16
|
Nehme A, Cerutti C, Zibara K. Transcriptomic Analysis Reveals Novel Transcription Factors Associated With Renin-Angiotensin-Aldosterone System in Human Atheroma. Hypertension 2016; 68:1375-1384. [PMID: 27754866 DOI: 10.1161/hypertensionaha.116.08070] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2016] [Revised: 07/16/2016] [Accepted: 09/20/2016] [Indexed: 11/16/2022]
Abstract
Despite the well-known role of the renin-angiotensin-aldosterone system (RAAS) in atheroma, its global local organization is poorly understood. In this study, we used transcriptomic meta-analysis to reveal the local transcriptional organization and regulation of 37 extended RAAS (extRAAS) genes in atheroma. Expression analysis and hierarchical clustering were done on extRAAS genes in 32 paired early and advanced atherosclerotic lesions. Contrary to receptor-coding transcripts, multiple angiotensin-metabolizing enzymes showed higher expression in advance, in comparison to early lesions. Interestingly, similar results were obtained from GEO data sets containing human (n=839) and mouse (n=18) atherosclerotic samples, but different from normal human (n=11) arterial tissues. The expression and coordination patterns were then used to construct transcriptional maps of extRAAS, displaying favored pathways in atheroma. Three coexpression modules (M1, M2, and M3) with >80% reproducibility across human atheroma data sets were identified. M1 and M3 contained angiotensin-metabolizing enzymes transcripts, whereas M2 contained proatherogenic receptor-coding transcripts. Interestingly, M1 and M2 were negatively correlated. A total of 21 transcription factors with enriched binding sites in the promoters of coordinated genes were extracted, among which IRF5, MAX, and ETV5 showed significant positive correlations with M1, but negative correlations with M2. However, ETS1 and SMAD1 transcripts were positively correlated to receptor-coding genes in M2. Despite sharing some similarities in extRAAS organization with kidney and adipose, atheroma showed specific correlations between extRAAS and transcription factors. In conclusion, our transcriptional map helps in designing more efficient treatments for atherosclerosis. In addition, the identified transcription factors provide a basis for the discovery of atheroma-specific modulators of extRAAS.
Collapse
Affiliation(s)
- Ali Nehme
- From the EA4173, Functional Genomics of Arterial Hypertension, UCBL-1, Lyon, France (A.N., C.C.); and ER045, Laboratory of Stem Cells, DSST (A.N., K.Z.) and Department of Biology, Faculty of Sciences-I (K.Z.), Lebanese University, Beirut, Lebanon
| | - Catherine Cerutti
- From the EA4173, Functional Genomics of Arterial Hypertension, UCBL-1, Lyon, France (A.N., C.C.); and ER045, Laboratory of Stem Cells, DSST (A.N., K.Z.) and Department of Biology, Faculty of Sciences-I (K.Z.), Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- From the EA4173, Functional Genomics of Arterial Hypertension, UCBL-1, Lyon, France (A.N., C.C.); and ER045, Laboratory of Stem Cells, DSST (A.N., K.Z.) and Department of Biology, Faculty of Sciences-I (K.Z.), Lebanese University, Beirut, Lebanon.
| |
Collapse
|
17
|
Adeva-Andany MM, Fernández-Fernández C, Sánchez-Bello R, Donapetry-García C, Martínez-Rodríguez J. The role of carbonic anhydrase in the pathogenesis of vascular calcification in humans. Atherosclerosis 2015; 241:183-91. [PMID: 26005791 DOI: 10.1016/j.atherosclerosis.2015.05.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/04/2015] [Accepted: 05/07/2015] [Indexed: 11/19/2022]
Abstract
Carbonic anhydrases are a group of isoenzymes that catalyze the reversible conversion of carbon dioxide into bicarbonate. They participate in a constellation of physiological processes in humans, including respiration, bone metabolism, and the formation of body fluids, including urine, bile, pancreatic juice, gastric secretion, saliva, aqueous humor, cerebrospinal fluid, and sweat. In addition, carbonic anhydrase may provide carbon dioxide/bicarbonate to carboxylation reactions that incorporate carbon dioxide to substrates. Several isoforms of carbonic anhydrase have been identified in humans, but their precise physiological role and the consequences of their dysfunction are mostly unknown. Carbonic anhydrase isoenzymes are involved in calcification processes in a number of biological systems, including the formation of calcareous spicules from sponges, the formation of shell in some animals, and the precipitation of calcium salts induced by several microorganisms, particularly urease-producing bacteria. In human tissues, carbonic anhydrase is implicated in calcification processes either directly by facilitating calcium carbonate deposition which in turn serves to facilitate calcium phosphate mineralization, or indirectly via its action upon γ-glutamyl-carboxylase, a carboxylase that enables the biological activation of proteins involved in calcification, such as matrix Gla protein, bone Gla protein, and Gla-rich protein. Carbonic anhydrase is implicated in calcification of human tissues, including bone and soft-tissue calcification in rheumatological disorders such as ankylosing spondylitis and dermatomyositis. Carbonic anhydrase may be also involved in bile and kidney stone formation and carcinoma-associated microcalcifications. The aim of this review is to evaluate the possible association between carbonic anhydrase isoenzymes and vascular calcification in humans.
Collapse
Affiliation(s)
- María M Adeva-Andany
- Nephrology Division, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406 Ferrol, Spain.
| | | | - Rocío Sánchez-Bello
- Nephrology Division, Hospital General Juan Cardona, c/ Pardo Bazán s/n, 15406 Ferrol, Spain
| | | | | |
Collapse
|
18
|
Oxidized low-density lipoprotein increases bone sialoprotein expression in vascular smooth muscle cells via runt-related transcription factor 2. Am J Med Sci 2015; 349:240-3. [PMID: 25504218 DOI: 10.1097/maj.0000000000000381] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND Vascular calcification is a pivotal stage in atherosclerosis. During vascular calcification, vascular smooth muscle cells (VSMCs) synthesize many osteogenic factors such as bone sialoprotein (BSP). Oxidative stress plays a critical role in progression of atherosclerosis and also increases extracellular matrix proteins expression. BSP overexpression has been observed during vascular calcification by oxidative stress. However, the regulatory mechanism of oxidized low-density lipoprotein (oxLDL)-mediated vascular calcification has not yet been fully defined. In this study, we aimed to investigate whether runt-related transcription factor 2 (Runx2) affects the oxLDL-induced BSP expression or not. METHODS In this experimental study, we cultured VSMCs in F12K media and then treated them with oxLDL. The expression of Runx2 and BSP genes was determined by real-time polymerase chain reaction method. Protein level of each gene was investigated by Western blotting technique. To determine whether Runx2 regulates BSP gene expression at VSMCs induced by oxLDL, we suppressed Runx2 mRNA using siRNA. Transfected cells then were treated with oxLDL and expression of Runx2 and BSP genes was determined again. RESULTS oxLDL increased Runx2 and BSP expression (4.8 ± 0.47-fold and 4.91 ± 0.56-fold, respectively) after 48 hours. Western blotting method confirmed the increased levels of Runx2 and BSP proteins after 48 hours. Runx2 overexpression alone induced BSP expression, whereas knockdown of Runx2 with small interfering siRNA blocked oxLDL-induced BSP expression. CONCLUSIONS Our results showed that oxLDL-induced BSP expression was dependent on Runx2 expression, suggesting that Runx2 is required for oxLDL-induced BSP expression.
Collapse
|
19
|
Farrokhi E, Samani KG, Chaleshtori MH. Oxidized Low-Density Lipoprotein and Upregulated Expression of Osteonectin and Bone Sialoprotein in Vascular Smooth Muscle Cells. Lab Med 2014; 45:297-301. [DOI: 10.1309/lmujwvqfw6cjmsoq] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
|
20
|
Wolak T. Osteopontin - a multi-modal marker and mediator in atherosclerotic vascular disease. Atherosclerosis 2014; 236:327-37. [PMID: 25128758 DOI: 10.1016/j.atherosclerosis.2014.07.004] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 06/26/2014] [Accepted: 07/07/2014] [Indexed: 02/07/2023]
Abstract
Atherosclerosis is a chronic inflammatory process of the vessel wall with systemic correlates. It is now well established that patients' outcome is tightly linked to atherosclerotic plaque stability, potentially more so than to the mere plaque size. Osteopontin (OPN) is an integrin-binding ligand, N-linked glycoprotein, which was recognized as a significant participant in the atherosclerotic inflammatory milieu. Evidence from several genetic mouse models suggests that OPN is an enhancer of atherosclerosis. This may be mediated by its capacity to enhance inflammation in the atherosclerotic plaque. Interestingly, OPN may also possess potentially protective vascular effects, such as attenuation of vascular calcification. In humans circulating levels of OPN were found to be independently associated with the severity of coronary atherosclerosis. Moreover, several studies report that high plasma OPN levels were associated with increased risk for major adverse cardiac events. This review aims to critically assess current understanding of the role of OPN in the atherosclerotic process, from animal models to clinical practice. Specific focus is given to evaluating whether OPN could serve as a marker for monitoring coronary atherosclerosis severity, and in parallel, assess the evidence for its role as a mediator in the pathogenic pathways leading to atherosclerotic vascular disease.
Collapse
Affiliation(s)
- Talya Wolak
- Hypertension Unit Faculty of Health Sciences, Soroka University Medical Center, Ben-Gurion University of the Negev, Be'er-Sheva, Israel.
| |
Collapse
|
21
|
Wang W, Shen H, Xie J, Zhou Q, Chen Y, Lu H. Bioinformatics analysis of time-series genes profiling to explore key genes affected by age in fracture healing. Mol Biol Rep 2014; 41:3881-9. [PMID: 24627361 DOI: 10.1007/s11033-014-3255-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2013] [Accepted: 02/08/2014] [Indexed: 01/23/2023]
Abstract
The present study was aimed to explore possible key genes and bioprocess affected by age during fracture healing. GSE589, GSE592 and GSE1371 were downloaded from gene expression omnibus database. The time-series genes of three age levels rats were firstly identified with hclust function in R. Then functional and pathway enrichment analysis for selected time-series genes were performed. Finally, the VennDiagram package of R language was used to screen overlapping n time-series genes. The expression changes of time-series genes in the rats of three age levels were classified into two types: one was higher expressed at 0 day, decreased at 3 day to 2 week, and increased from 4 to 6 week; the other was the opposite. Functional and pathways enrichment analysis showed that 12 time-series genes of adult and old rats were significantly involved in ECM-receptor interaction pathway. The expression changes of 11 genes were consistent with time axis, 10 genes were up-regulated at 3 days after fracture, and increased slowly in 6 week, while Itga2b was down-regulated. The functions of 106 overlapping genes were all associated with growth and development of bone after fracture. The key genes in ECM-receptor interaction pathway including Spp1, Ibsp, Tnn and Col3a1 have been reported to be related to fracture in literatures. The difference during fracture healing in three age levels rats is mainly related to age. The Spp1, Ibsp, Tnn and Col3a1 are possible potential age-related genes and ECM-receptor interaction pathway is the potential age-related process during fracture healing.
Collapse
Affiliation(s)
- Wei Wang
- Department of Orthopaedics, Xinhua Hospital Affiliated to Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, China
| | | | | | | | | | | |
Collapse
|