1
|
Yoodee S, Malaitad T, Plumworasawat S, Thongboonkerd V. E53, E96, D162, E247 and D322 in Ca 2+-binding domains of annexin A2 are essential for regulating intracellular [Ca 2+] and crystal adhesion to renal cells via ERK1/2 and JNK signaling pathways. Arch Biochem Biophys 2025; 769:110410. [PMID: 40189002 DOI: 10.1016/j.abb.2025.110410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/31/2025] [Accepted: 04/04/2025] [Indexed: 04/10/2025]
Abstract
Annexin A2 (ANXA2) is expressed inside the cytoplasm and on the surface of renal tubular epithelial cells (RTECs) and is documented as a calcium oxalate monohydrate (COM) crystal-binding protein. Nevertheless, its molecular mechanism involved in kidney stone disease (KSD) remains underinvestigated. Herein, we performed various molecular assays to unravel the roles of ANXA2 and core residues (E53, E96, D162, E247 and D322) in its Ca2+-binding domains in the stone formation mechanism, particularly at crystal-cell adhesion step and downstream signaling cascade. ANXA2 was up-regulated in apical membranes, not cytosol, of RTECs after COM crystal exposure. Neutralizing the surface expression of ANXA2 by a specific monoclonal antibody and silencing its expression by small interfering RNA (siRNA) significantly decreased COM crystal-cell adhesion. siRNA also suppressed the COM-induced up-regulation of phospho-ERK1/2 and phospho-JNK, but not that of phospho-p38. Overexpression of ANXA2 wild-type (WT), but not that of E53A, E96A, D162A, E247A and D322A mutants of its Ca2+-binding domains, significantly increased intracellular [Ca2+], COM-cell adhesion, and phospho-ERK1/2 level. Therefore, E53, E96, D162, E247 and D322 in the Ca2+-binding domains of annexin A2 are essential for regulating intracellular [Ca2+] and COM crystal-cell adhesion via ERK1/2 and JNK signaling pathways.
Collapse
Affiliation(s)
- Sunisa Yoodee
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Thanyalak Malaitad
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Sirikanya Plumworasawat
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
2
|
Allgayer R, Kabir RF, Bergeron A, Demers P, Mantovani D, Cerruti M. A collagen-based laboratory model to mimic sex-specific features of calcific aortic valve disease. Acta Biomater 2025; 194:204-218. [PMID: 39864641 DOI: 10.1016/j.actbio.2025.01.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 01/08/2025] [Accepted: 01/21/2025] [Indexed: 01/28/2025]
Abstract
Calcific aortic valve disease (CAVD) shows in the deposition of calcium phosphates in the collagen-rich layer of the valve leaflets. This stiffens the leaflets and eventually leads to heart failure. Recent research suggests that CAVD follows sex-specific pathways: at the same severity of the disease, women tend to have fewer and less crystalline calcifications, and the phases of their calcifications are decidedly different than those of men; namely, dicalcium phosphate dihydrate (DCPD) - one of the mineral phases in CAVD - occurs almost exclusively in females. Furthermore, the morphologies of heart valve calcifications might be sex-specific, but the sex dependence of the morphologies has not been systematically investigated. Herein, we first show that male CAVD patients have more compact and less fibrous calcifications than females, establishing sex-dependent morphological features of heart valve calcification. We then build a model that recapitulates the sex differences of the calcifications in CAVD, which is based on a collagen gel that we calcify in simulated body fluid with varying fetuin A concentrations. With increasing fetuin A concentration, the calcifications become less crystalline and more fibrous, and more DCPD deposits in the collagen matrix, resembling the physicochemical characteristics of the calcifications in female valves. Lower fetuin A concentrations give rise to a model that replicates male-specific mineral characteristics. The models could be used to develop sex-specific detection and treatment methods for CAVD. STATEMENT OF SIGNIFICANCE: Although calcific aortic valve disease (CAVD) affects ∼10 million people globally, researchers have only discovered recently that the disease follows sex-specific pathways, and many of its sex-specific features remain unknown. To further our understanding of sex differences in CAVD and to develop better detection and treatment methods, there is an urgent need to establish models for CAVD that account for its sex-specific manifestations. In this study, we first show that CAVD calcifications in men and women take on different morphologies. Second, we present a model that can replicate physicochemical calcification characteristics of male or female valves, including morphology, and that can help to develop sex-specific detection and treatment methods for CAVD.
Collapse
Affiliation(s)
- Raphaela Allgayer
- Department of Mining and Materials Engineering, McGill University, 3610 Rue University, Montreal, QC H3A 0C5, Canada
| | - Reefah Fahmida Kabir
- Department of Bioengineering, McGill University, 3480 Rue University, Montreal, QC H2A 0E9, Canada
| | - Alexandre Bergeron
- Department of Surgery, Montreal Heart Institute, 5000 Rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Philippe Demers
- Department of Surgery, Montreal Heart Institute, 5000 Rue Bélanger, Montreal, QC H1T 1C8, Canada
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, CRC-I, Department of Min-Met-Materials Engineering & Research Center of CHU de Quebec, Division of Regenerative Medicine, Laval University, Quebec City, QC G1V 0A6, Canada
| | - Marta Cerruti
- Department of Mining and Materials Engineering, McGill University, 3610 Rue University, Montreal, QC H3A 0C5, Canada.
| |
Collapse
|
3
|
Shi M, Su X, Xiang H, Song Q, Yang S. Advances in the mechanism of urinary proteins in calcium oxalate kidney stone formation. Urolithiasis 2025; 53:27. [PMID: 39932538 DOI: 10.1007/s00240-025-01703-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 01/28/2025] [Indexed: 04/30/2025]
Abstract
Kidney stones are a common urological disease worldwide, imposing a significant burden on healthcare systems. Calcium oxalate stones are the predominant form of urinary calculi, with two main theoretical models explaining their pathogenesis: the fixed particle and free particle models. Regardless of the model, the formation of calcium oxalate kidney stones is inseparably linked to crystal nucleation, growth, aggregation, and adhesion in urine. Growing evidence highlights the significant role of urinary proteins, particularly matrix proteins, in the development of calcium oxalate stones. The review classifies urinary proteins impacting calcium oxalate stone formation into three groups: inhibitors, promoters, and dual-regulators, outlining their contributions to the formation process.
Collapse
Affiliation(s)
- Mingcheng Shi
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Xiaozhe Su
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Heng Xiang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China
| | - Qianlin Song
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China.
| | - Sixing Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan, Hubei Province, 430060, People's Republic of China.
| |
Collapse
|
4
|
Mougkogiannis P, Adamatzky A. Memfractance of Proteinoids. ACS OMEGA 2024; 9:15085-15100. [PMID: 38585073 PMCID: PMC10993267 DOI: 10.1021/acsomega.3c09330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2023] [Revised: 02/22/2024] [Accepted: 03/05/2024] [Indexed: 04/09/2024]
Abstract
Proteinoids, or thermal proteins, are amino acid polymers formed at high temperatures by nonbiological processes. The objective of this study is to examine the memfractance characteristics of proteinoids within a supersaturated hydroxyapatite solution. The ionic solution utilized for the current-voltage (I-V) measurements possessed an ionic strength of 0.15 mol/L, a temperature of 37 °C, and a pH value of 7.4. The I-V curves exhibited distinct spikes, which are hypothesized to arise from the capacitive charging and discharging of the proteinoid-hydroxyapatite media. The experimental results demonstrated a positive correlation between the concentration of proteinoids and the observed number of spikes in the I-V curves. This observation provides evidence in favor of the hypothesis that the spikes originate from the proteinoids' capacitive characteristics. The memfractance behavior exemplifies the capacity of proteinoids to retain electrical charge within the hydrated hydroxyapatite media. Additional investigation is required in order to comprehensively identify the memcapacitive phenomena and delve into their implications for models of protocellular membranes. In a nutshell, this study provides empirical support for the existence of capacitive membrane-memfractance mechanisms in ensembles of proteinoids.
Collapse
Affiliation(s)
| | - Andrew Adamatzky
- Unconventional Computing
Laboratory, UWE, Bristol BS16 1QY, U.K.
| |
Collapse
|
5
|
Sassanarakkit S, Peerapen P, Thongboonkerd V. StoneMod 2.0: Database and prediction of kidney stone modulatory proteins. Int J Biol Macromol 2024; 261:129912. [PMID: 38309384 DOI: 10.1016/j.ijbiomac.2024.129912] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Revised: 01/29/2024] [Accepted: 01/31/2024] [Indexed: 02/05/2024]
Abstract
Stone modulators are various kinds of molecules that play crucial roles in promoting/inhibiting kidney stone formation. Several recent studies have extensively characterized the stone modulatory proteins with the ultimate goal of preventing kidney stone formation. Herein, we introduce the StoneMod 2.0 database (https://www.stonemod.org), which has been dramatically improved from the previous version by expanding the number of the modulatory proteins in the list (from 32 in the initial version to 17,130 in this updated version). The stone modulatory proteins were recruited from solid experimental evidence (via PubMed) and/or predicted evidence (via UniProtKB, QuickGO, ProRule, STITCH and OxaBIND to retrieve calcium-binding and oxalate-binding proteins). Additionally, StoneMod 2.0 has implemented a scoring system that can be used to determine the likelihood and to classify the potential stone modulatory proteins as either "solid" (modulator score ≥ 50) or "weak" (modulator score < 50) modulators. Furthermore, the updated version has been designed with more user-friendly interfaces and advanced visualization tools. In addition to the monthly scheduled update, the users can directly submit their experimental evidence online anytime. Therefore, StoneMod 2.0 is a powerful database with prediction scores that will be very useful for many future studies on the stone modulatory proteins.
Collapse
Affiliation(s)
- Supatcha Sassanarakkit
- Medical Proteomics Unit, Research Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Paleerath Peerapen
- Medical Proteomics Unit, Research Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand
| | - Visith Thongboonkerd
- Medical Proteomics Unit, Research Development, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok 10700, Thailand.
| |
Collapse
|
6
|
Chmiel JA, Stuivenberg GA, Al KF, Akouris PP, Razvi H, Burton JP, Bjazevic J. Vitamins as regulators of calcium-containing kidney stones - new perspectives on the role of the gut microbiome. Nat Rev Urol 2023; 20:615-637. [PMID: 37161031 PMCID: PMC10169205 DOI: 10.1038/s41585-023-00768-5] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2023] [Indexed: 05/11/2023]
Abstract
Calcium-based kidney stone disease is a highly prevalent and morbid condition, with an often complicated and multifactorial aetiology. An abundance of research on the role of specific vitamins (B6, C and D) in stone formation exists, but no consensus has been reached on how these vitamins influence stone disease. As a consequence of emerging research on the role of the gut microbiota in urolithiasis, previous notions on the contribution of these vitamins to urolithiasis are being reconsidered in the field, and investigation into previously overlooked vitamins (A, E and K) was expanded. Understanding how the microbiota influences host vitamin regulation could help to determine the role of vitamins in stone disease.
Collapse
Affiliation(s)
- John A Chmiel
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
| | - Gerrit A Stuivenberg
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
| | - Kait F Al
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
| | - Polycronis P Akouris
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
| | - Hassan Razvi
- Division of Urology, Department of Surgery, Western University, London, Ontario, Canada
| | - Jeremy P Burton
- Department of Microbiology & Immunology, Western University, London, Ontario, Canada
- Canadian Centre for Human Microbiome and Probiotic Research, London, Ontario, Canada
- Division of Urology, Department of Surgery, Western University, London, Ontario, Canada
| | - Jennifer Bjazevic
- Division of Urology, Department of Surgery, Western University, London, Ontario, Canada.
| |
Collapse
|
7
|
Abstract
Patients with chronic kidney disease (CKD) exhibit tremendously elevated risk for cardiovascular disease, particularly ischemic heart disease, due to premature vascular and cardiac aging and accelerated ectopic calcification. The presence of cardiovascular calcification associates with increased risk in patients with CKD. Disturbed mineral homeostasis and diverse comorbidities in these patients drive increased systemic cardiovascular calcification in different manifestations with diverse clinical consequences, like plaque instability, vessel stiffening, and aortic stenosis. This review outlines the heterogeneity in calcification patterning, including mineral type and location and potential implications on clinical outcomes. The advent of therapeutics currently in clinical trials may reduce CKD-associated morbidity. Development of therapeutics for cardiovascular calcification begins with the premise that less mineral is better. While restoring diseased tissues to a noncalcified homeostasis remains the ultimate goal, in some cases, calcific mineral may play a protective role, such as in atherosclerotic plaques. Therefore, developing treatments for ectopic calcification may require a nuanced approach that considers individual patient risk factors. Here, we discuss the most common cardiac and vascular calcification pathologies observed in CKD, how mineral in these tissues affects function, and the potential outcomes and considerations for therapeutic strategies that seek to disrupt the nucleation and growth of mineral. Finally, we discuss future patient-specific considerations for treating cardiac and vascular calcification in patients with CKD-a population in need of anticalcification therapies.
Collapse
Affiliation(s)
- Joshua D. Hutcheson
- Department of Biomedical Engineering, Florida International University, Miami, FL (J.D.H.)
| | - Claudia Goettsch
- Department of Internal Medicine I, Division of Cardiology, Medical Faculty, RWTH Aachen University, Germany (C.G.)
| |
Collapse
|
8
|
Yan Q, Zhang T, O'Connor C, Barlow JW, Walsh J, Scalabrino G, Xu F, Sheridan H. The biological responses of vitamin K2: A comprehensive review. Food Sci Nutr 2023; 11:1634-1656. [PMID: 37051359 PMCID: PMC10084986 DOI: 10.1002/fsn3.3213] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/14/2022] [Accepted: 12/19/2022] [Indexed: 01/09/2023] Open
Abstract
Vitamin K1 (VitK1) and Vitamin K2 (VitK2), two important naturally occurring micronutrients in the VitK family, found, respectively, in green leafy plants and algae (VitK1) and animal and fermented foods (VitK2). The present review explores the multiple biological functions of VitK2 from recently published in vitro and in vivo studies, including promotion of osteogenesis, prevention of calcification, relief of menopausal symptoms, enhancement of mitochondrial energy release, hepato- and neuro-protective effects, and possible use in treatment of coronavirus disease. The mechanisms of action associated with these biological effects are also explored. Overall, the findings presented here suggest that VitK, especially VitK2, is an important nutrient family for the normal functioning of human health. It acts on almost all major body systems and directly or indirectly participates in and regulates hundreds of physiological or pathological processes. However, as biological and clinical data are still inconsistent and conflicting, more in-depth investigations are warranted to elucidate its potential as a therapeutic strategy to prevent and treat a range of disease conditions.
Collapse
Affiliation(s)
- Quanxiang Yan
- Institute of Science and TechnologyShenyang Open UniversityShenyangChina
| | - Tao Zhang
- School of Food Science & Environmental HealthTechnological University DublinDublin 7Ireland
- NatPro Centre, School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublin 2Ireland
| | - Christine O'Connor
- School of Food Science & Environmental HealthTechnological University DublinDublin 7Ireland
| | - James W. Barlow
- Department of ChemistryRCSI University of Medicine and Health SciencesDublin 2Ireland
| | - John Walsh
- NatPro Centre, School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublin 2Ireland
- School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublin 2Ireland
| | - Gaia Scalabrino
- NatPro Centre, School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublin 2Ireland
| | - Feng Xu
- The Centre of Vitamin K2 ResearchShenyang Pharmaceutical UniversityShenyangChina
| | - Helen Sheridan
- NatPro Centre, School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublin 2Ireland
- School of Pharmacy and Pharmaceutical SciencesTrinity College DublinDublin 2Ireland
| |
Collapse
|
9
|
Parashar A, Gourgas O, Lau K, Li J, Muiznieks L, Sharpe S, Davis E, Cerruti M, Murshed M. Elastin calcification in in vitro models and its prevention by MGP's N-terminal peptide. J Struct Biol 2021; 213:107637. [PMID: 33059036 DOI: 10.1016/j.jsb.2020.107637] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Revised: 10/01/2020] [Accepted: 10/03/2020] [Indexed: 01/17/2023]
Abstract
Medial calcification has been associated with diabetes, chronic kidney disease, and genetic disorders like pseudoxanthoma elasticum. Recently, we showed that genetic reduction of arterial elastin content reduces the severity of medial calcification in matrix Gla protein (MGP)-deficient and Eln haploinsufficient Mgp-/-;Eln+/- mice. This study suggests that there might be a direct effect of elastin amount on medial calcification. We studied this using novel in vitro systems, which are based on elastin or elastin-like polypeptides. We first examined the mineral deposition properties of a transfected pigmented epithelial cell line that expresses elastin and other elastic lamina proteins. When grown in inorganic phosphate-supplemented medium, these cells deposited calcium phosphate minerals, which could be prevented by an N'-terminal peptide of MGP (m3pS) carrying phosphorylated serine residues. We next confirmed these findings using a cell-free elastin-like polypeptide (ELP3) scaffold, where the peptide prevented mineral maturation. Overall, this work describes a novel cell culture model for elastocalcinosis and examines the inhibition of mineral deposition by the m3pS peptide in this and a cell-free elastin-based scaffold. Our study provides strong evidence suggesting the critical functional roles of MGP's phosphorylated serine residues in the prevention of elastin calcification and proposes a possible mechanism of their action.
Collapse
Affiliation(s)
- Abhinav Parashar
- Faculty of Dentistry, McGill University, Montreal, Québec, Canada
| | - Ophélie Gourgas
- Department of Medicine, McGill University, Montreal, Québec, Canada
| | - Kirk Lau
- Materials Engineering, McGill University, Montreal, Québec, Canada
| | - Jingjing Li
- Department of Medicine, McGill University, Montreal, Québec, Canada
| | - Lisa Muiznieks
- Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Simon Sharpe
- Molecular Medicine, Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Elaine Davis
- Department of Anatomy and Cell Biology, McGill University, Montreal, Québec, Canada
| | - Marta Cerruti
- Materials Engineering, McGill University, Montreal, Québec, Canada
| | - Monzur Murshed
- Faculty of Dentistry, McGill University, Montreal, Québec, Canada; Department of Medicine, McGill University, Montreal, Québec, Canada; Shriners Hospital for Children, Montreal, Quebec, Canada.
| |
Collapse
|
10
|
Sarosiak A, Oziębło D, Udziela M, Vermeer C, Malejczyk J, Szaflik JP, Ołdak M. High expression of Matrix Gla Protein in Schnyder corneal dystrophy patients points to an active role of vitamin K in corneal health. Acta Ophthalmol 2021; 99:e171-e177. [PMID: 32602245 DOI: 10.1111/aos.14533] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2020] [Revised: 05/28/2020] [Accepted: 06/02/2020] [Indexed: 12/19/2022]
Abstract
PURPOSE Schnyder corneal dystrophy (SCD) is a rare autosomal dominant disorder characterized by corneal lipid accumulation and caused by UBIAD1 pathogenic variants. UBIAD1 encodes a vitamin K (VK) biosynthetic enzyme. To assess the corneal and vascular VK status in SCD patients, we focused on matrix Gla protein (MGP), a VK-dependent protein. METHODS Conformation-specific immunostainings of different MGP maturation forms were performed on corneal sections and primary keratocytes from corneal buttons of two SCD patients with UBIAD1 p.Asp112Asn and p.Asn102Ser pathogenic variants and unrelated donors. Native or UBIAD1-transfected keratocytes were used for gene expression analysis. Plasma samples from SCD patients (n = 12) and control individuals (n = 117) were subjected for inactive desphospho-uncarboxylated MGP level measurements with an ELISA assay. RESULTS Substantial amounts of MGP were identified in human cornea and most of it in its fully matured and active form. The level of mature MGP did not differ between SCD and control corneas. In primary keratocytes from SCD patients, a highly increased MGP expression and presence of immature MGP forms were detected. Significantly elevated plasma concentration of inactive MGP was found in SCD patients. CONCLUSION High amount of MGP and the predominance of mature MGP forms in human cornea indicate that VK metabolism is active in the visual system. Availability of MGP seems of vital importance for a healthy cornea and may be related to protection against corneal calcification. Systemic MGP findings reveal a poor vascular VK status in SCD patients and indicate that SCD may lead to cardiovascular consequences.
Collapse
Affiliation(s)
- Anna Sarosiak
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Dominika Oziębło
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
- Postgraduate School of Molecular Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Monika Udziela
- Department of Ophthalmology, Medical University of Warsaw, Warsaw, Poland
| | - Cees Vermeer
- R&D Group VitaK and Cardiovascular Research Institute CARIM, Maastricht University, Maastricht, The Netherlands
| | - Jacek Malejczyk
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| | - Jacek P Szaflik
- Department of Ophthalmology, Medical University of Warsaw, Warsaw, Poland
| | - Monika Ołdak
- Department of Histology and Embryology, Center of Biostructure Research, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
11
|
Dayekh K, Mequanint K. The effects of progenitor and differentiated cells on ectopic calcification of engineered vascular tissues. Acta Biomater 2020; 115:288-298. [PMID: 32853805 DOI: 10.1016/j.actbio.2020.08.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 08/15/2020] [Accepted: 08/18/2020] [Indexed: 12/17/2022]
Abstract
Ectopic vascular calcification associated with aging, diabetes mellitus, atherosclerosis, and chronic kidney disease is a considerable risk factor for cardiovascular events and death. Although vascular smooth muscle cells are primarily implicated in calcification, the role of progenitor cells is less known. In this study, we engineered tubular vascular tissues from embryonic multipotent mesenchymal progenitor cells either without differentiating or after differentiating them into smooth muscle cells and studied ectopic calcification through targeted gene analysis. Tissues derived from both differentiated and undifferentiated cells calcified in response to hyperphosphatemic inorganic phosphate (Pi) treatment suggesting that a single cell-type (progenitor cells or differentiated cells) may not be the sole cause of the process. We also demonstrated that Vitamin K, which is the matrix gla protein activator, had a protective role against calcification in engineered vascular tissues. Addition of partially-soluble elastin upregulated osteogenic marker genes suggesting a calcification process. Furthermore, partially-soluble elastin downregulated smooth muscle myosin heavy chain (Myh11) gene which is a late-stage differentiation marker. This latter point, in turn, suggests that SMC may be switching into a synthetic phenotype which is one feature of vascular calcification. Taken together, our approach presents a valuable tool to study ectopic calcification and associated gene expressions relevant to clinical therapeutic targets.
Collapse
|
12
|
Abstract
Kidney stone disease (nephrolithiasis) is a common problem that can be associated with alterations in urinary solute composition including hypercalciuria. Studies suggest that the prevalence of monogenic kidney stone disorders, including renal tubular acidosis with deafness, Bartter syndrome, primary hyperoxaluria and cystinuria, in patients attending kidney stone clinics is ∼15%. However, for the majority of individuals, nephrolithiasis has a multifactorial aetiology involving genetic and environmental factors. Nonetheless, the genetic influence on stone formation in these idiopathic stone formers remains considerable and twin studies estimate a heritability of >45% for nephrolithiasis and >50% for hypercalciuria. The contribution of polygenic influences from multiple loci have been investigated by genome-wide association and candidate gene studies, which indicate that a number of genes and molecular pathways contribute to the risk of stone formation. Genetic approaches, studying both monogenic and polygenic factors in nephrolithiasis, have revealed that the following have important roles in the aetiology of kidney stones: transporters and channels; ions, protons and amino acids; the calcium-sensing receptor (a G protein-coupled receptor) signalling pathway; and the metabolic pathways for vitamin D, oxalate, cysteine, purines and uric acid. These advances, which have increased our understanding of the pathogenesis of nephrolithiasis, will hopefully facilitate the future development of targeted therapies for precision medicine approaches in patients with nephrolithiasis.
Collapse
Affiliation(s)
- Sarah A Howles
- Nuffield Department of Surgical Sciences, University of Oxford, Oxford, UK.,Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Rajesh V Thakker
- Academic Endocrine Unit, Radcliffe Department of Medicine, University of Oxford, Oxford, UK.
| |
Collapse
|
13
|
Castiglione V, Pottel H, Lieske JC, Lukas P, Cavalier E, Delanaye P, Rule AD. Evaluation of inactive Matrix-Gla-Protein (MGP) as a biomarker for incident and recurrent kidney stones. J Nephrol 2020; 33:101-107. [PMID: 31222647 DOI: 10.1007/s40620-019-00623-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 06/17/2019] [Indexed: 01/07/2023]
Abstract
BACKGROUND Matrix-Gla-protein (MGP) is an inhibitor of vascular calcification. Its dephosphorylated and uncarboxylated inactive form, dpucMGP, is a marker of vitamin K status and of cardio-vascular outcomes in chronic kidney disease. We hypothesized that higher serum dpucMGP would be a biomarker of kidney stone disease. METHODS We measured serum dpucMGP in incident symptomatic kidney stone-formers and non-stone formers at a baseline visit. Symptomatic stone recurrence was assessed in the stones formers over a 5-year period. The association of dpucMGP with incident or recurrent kidney stones was assessed with and without adjustment for clinical, blood, and urine characteristics. RESULTS There was no significant difference in serum dpucMGP level between 498 stone formers and 395 non-stone former (510 vs 501 pmol/L; p = 0.66). In a multivariable model adjusting for clinical, blood and urine chemistries, higher MGP was associated with lower risk of stone formation (OR = 0.674, 95% CI 0.522-0.870), contrary to previous reports. Among 375 stone formers with 5 years of follow-up, 79 (21%) had symptomatic recurrence. No difference in serum dpucMGP was evident in recurrent versus non-recurrent stone-formers (482 vs 502 pmol/L; p = 0.26). Serum dpucMGP was correlated with cystatin C levels in non stone-formers, incident stone-formers and recurrent stone-formers (r > 0.3, p < 0.0001). CONCLUSION Elevated serum dpucMGP was not associated with incident or recurrent symptomatic kidney stone events. However, higher level of dpucMGP was associated with lower risk of kidney stone in a multivariable logistic regression model.
Collapse
Affiliation(s)
- Vincent Castiglione
- Department of Clinical Chemistry, CHU of Liège, University of Liège (ULg CHU), Liège, Belgium.
| | - Hans Pottel
- Department of Public Health and Primary Care, KU Leuven Campus Kulak Kortrijk, Kortrijk, Belgium
| | | | - Pierre Lukas
- Department of Clinical Chemistry, CHU of Liège, University of Liège (ULg CHU), Liège, Belgium
| | - Etienne Cavalier
- Department of Clinical Chemistry, CHU of Liège, University of Liège (ULg CHU), Liège, Belgium
| | - Pierre Delanaye
- Department of Nephrology, Dialysis and Transplantation, CHU of Liège, University of Liège (ULg CHU), Liège, Belgium
| | - Andrew David Rule
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
14
|
Incorporation of osteopontin peptide into kidney stone-related calcium oxalate monohydrate crystals: a quantitative study. Urolithiasis 2018; 47:425-440. [DOI: 10.1007/s00240-018-01105-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2018] [Accepted: 12/11/2018] [Indexed: 10/27/2022]
|
15
|
Urothelium proliferation is a trigger for renal crystal deposits in a murine lithogenesis model. Sci Rep 2018; 8:16319. [PMID: 30397242 PMCID: PMC6218513 DOI: 10.1038/s41598-018-34734-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2018] [Accepted: 10/15/2018] [Indexed: 11/20/2022] Open
Abstract
Most mouse kidney stone models induce nephrocalcinosis rather than urolithiasis. The aim of our study was to find an accelerated experimental model in order to study the early events of stone formation, that is, at the time of crystal binding to intrarenal urothelium. C57B6 mice exposed to vitamin D supplements and water containing hydroxyl-L-proline, ammonium chloride and calcium chloride were studied for 42 days. A group receiving urothelial cell mitogen Fibroblast Growth Factor 7 (FGF7) was compared to control group receiving saline. Calcium oxalate monohydrate (COM) crystals were detected in urines by day 2 and within urinary spaces in specialized fornix areas in both groups as soon as day 14 with enhanced deposits in FGF7 group compared to controls at day 21. Urothelial cells proliferation, uroplakin III downregulation and de novo expression of osteopontin receptor CD44 detected in FGF7 group, were delayed in the control group (day 42). Crystal aggregates within specialized fornix areas by day 42 were located in urinary spaces but also within and under a multilayered metaplastic urothelium, simultaneous to macrophages influx. Point of note, administration of a normal diet by day 21 was responsible for a spontaneous crystal clearance. Our data show that under supersaturation conditions, urothelial cell proliferation and calcium oxalate crystal retention occur within specialized fornix areas. Enhanced crystal deposits following FGF7 administration suggest that urothelium proliferation would be a relevant trigger for renal stone formation.
Collapse
|
16
|
Lin B, Srikanth P, Castle AC, Nigwekar S, Malhotra R, Galloway JL, Sykes DB, Rajagopal J. Modulating Cell Fate as a Therapeutic Strategy. Cell Stem Cell 2018; 23:329-341. [PMID: 29910150 PMCID: PMC6128730 DOI: 10.1016/j.stem.2018.05.009] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
In injured tissues, regeneration is often associated with cell fate plasticity, in that cells deviate from their normal lineage paths. It is becoming increasingly clear that this plasticity often creates alternative strategies to restore damaged or lost cells. Alternatively, cell fate plasticity is also part and parcel of pathologic tissue transformations that accompany disease. In this Perspective, we summarize a few illustrative examples of physiologic and aberrant cellular plasticity. Then, we speculate on how one could enhance endogenous plasticity to promote regeneration and reverse pathologic plasticity, perhaps inspiring interest in a new class of therapies targeting cell fate modulation.
Collapse
Affiliation(s)
- Brian Lin
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Priya Srikanth
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Pathways Program, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Alison C Castle
- Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Pathways Program, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Sagar Nigwekar
- Pathways Program, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Nephrology, Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
| | - Rajeev Malhotra
- Pathways Program, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Cardiology, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Jenna L Galloway
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Orthopaedic Surgery, Massachusetts General Hospital, Boston, MA 02114, USA
| | - David B Sykes
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA
| | - Jayaraj Rajagopal
- Center for Regenerative Medicine, Massachusetts General Hospital, 185 Cambridge Street, Boston, MA 02114, USA; Harvard Stem Cell Institute, Cambridge, MA 02138, USA; Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Pathways Program, Department of Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Division of Pulmonary and Critical Care Medicine, Massachusetts General Hospital, Boston, MA 02114, USA; Pediatric Pulmonary Medicine, Massachusetts General Hospital for Children, Boston, MA 02114, USA.
| |
Collapse
|
17
|
Wang Q, Hu H, Dirie NI, Lu Y, Zhang J, Cui L, Qin B, Wang Y, Zhu J, Xun Y, Zhu Y, Wu Y, Wang S. High Concentration of Calcium Promotes Mineralization in NRK-52E Cells Via Inhibiting the Expression of Matrix Gla Protein. Urology 2018; 119:161.e1-161.e7. [PMID: 29935264 DOI: 10.1016/j.urology.2018.06.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 05/27/2018] [Accepted: 06/05/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To address whether matrix Gla protein (MGP) can inhibit mineralization in normal rat kidney tubular cells (NRK-52E) under high concentration of calcium. MATERIALS AND METHODS NRK-52E cells were treated with high concentration of calcium. The viability and apoptosis of cells were detected by cell counting kit-8 and flow cytology, respectively. Real-time-polymerase chain, Western blotting, and immunofluorescence analysis were conducted to detect the expression of MGP. Cells were transfected with plasmid-MGP or siRNA-MGP for up- or down-regulation of the expression of MGP, respectively. Rat recombinant MGP was also used as supplementation of exogenous MGP. Alizarin red staining was conducted to detect the adherent and deposition of calcium salt. RESULTS High concentration of calcium suppressed MGP expression in NRK-52E cells. There was significant mineralization when NRK-52E cells were treated with high concentration of calcium. Supplementation with exogenous rat recombinant MGP and overexpression of endogenous MGP both decreased the adherent and deposition of calcium salt to NRK-52E cells, while silence of MGP showed reverse results. CONCLUSION MGP plays an inhibitory role in the stone formation. However, high concentration of calcium significantly inhibits the expression of MGP and then promotes mineralization in NRK-52E cells.
Collapse
Affiliation(s)
- Qing Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Henglong Hu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Najib Isse Dirie
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yuchao Lu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jiaqiao Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Cui
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Baolong Qin
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yufeng Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Jianning Zhu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yang Xun
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yunpeng Zhu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yue Wu
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Shaogang Wang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China.
| |
Collapse
|
18
|
Wei FF, Trenson S, Thijs L, Huang QF, Zhang ZY, Yang WY, Moliterno P, Allegaert K, Boggia J, Janssens S, Verhamme P, Vermeer C, Staessen JA. Desphospho-uncarboxylated matrix Gla protein is a novel circulating biomarker predicting deterioration of renal function in the general population. Nephrol Dial Transplant 2018; 33:1122-1128. [PMID: 28992263 PMCID: PMC6030862 DOI: 10.1093/ndt/gfx258] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2017] [Accepted: 07/05/2017] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Recent studies showing an inverse association between estimated glomerular filtration rate (eGFR), a microvascular trait, and inactive desphospho-uncarboxylated matrix Gla protein (dp-ucMGP) support the hypothesis that after vitamin K-dependent activation, matrix Gla protein (MGP) is renoprotective, but these were limited by their cross-sectional design. METHODS In 1009 randomly recruited Flemish (50.6% women), we assessed the association between eGFR and plasma dp-ucMGP, using multivariable-adjusted analyses. RESULTS From baseline to follow-up 8.9 years later (median), dp-ucMGP increased by 23.0% whereas eGFR decreased by 4.05 mL/min/1.73 m2 (P < 0.001). In 938 participants with baseline eGFR ≥60 mL/min/1.73 m2, the incidence of eGFR <60 mL/min/1.73 m2 at follow-up was 8.0% versus 4.1% in the top versus the bottom halve of baseline dp-ucMGP. For a 5-fold higher plasma dp-ucMGP at baseline, eGFR at follow-up decreased by 3.15 mL/min/1.73 m2 [95% confidence interval (CI) 1.26-5.05; P = 0.001]. The hazard ratio expressing the risk of progression to eGFR <60 mL/min/1.73 m2 was 3.49 (95% CI 1.45-8.40; P = 0.005). The hazard ratio relating the presence of microalbuminuria at follow-up to baseline dp-ucMGP was 4.70 (95% CI 1.57-14.1; P = 0.006). CONCLUSIONS In conclusion, circulating inactive dp-ucMGP, a biomarker of poor vitamin K status, predicts renal dysfunction. Possible underlying mechanisms include protection by activated MGP against calcification and inhibition of the bone morphogenetic protein-signalling pathway.
Collapse
Affiliation(s)
- Fang-Fei Wei
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Sander Trenson
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Lutgarde Thijs
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Qi-Fang Huang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Zhen-Yu Zhang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Wen-Yi Yang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Paula Moliterno
- Escuela de Nutrición, Universidad de la República, Montevideo, Uruguay
| | - Karel Allegaert
- Research Unit Organ Systems, KU Leuven Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - José Boggia
- Centro de Nefrología and Departamento de Fisiopatología, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Stefan Janssens
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Peter Verhamme
- Centre for Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Cees Vermeer
- R&D Group VitaK, Maastricht University, Maastricht, The Netherlands
| | - Jan A Staessen
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
- R&D Group VitaK, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
19
|
Wei FF, Trenson S, Monney P, Yang WY, Pruijm M, Zhang ZY, Bouatou Y, Huang QF, Ponte B, Martin PY, Thijs L, Kuznetsova T, Allegaert K, Janssens S, Vermeer C, Verhamme P, Burnier M, Bochud M, Ehret G, Staessen JA. Epidemiological and histological findings implicate matrix Gla protein in diastolic left ventricular dysfunction. PLoS One 2018; 13:e0193967. [PMID: 29529056 PMCID: PMC5846787 DOI: 10.1371/journal.pone.0193967] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Accepted: 02/22/2018] [Indexed: 01/07/2023] Open
Abstract
OBJECTIVES A novel paradigm of diastolic left ventricular (LV) dysfunction proposes involvement of the cardiac microvasculature. Vitamin K dependent matrix Gla protein (MGP) plays a role in preserving microcirculatory integrity. We hypothesized that LV filling pressure-a measure of diastolic LV dysfunction-increases with higher plasma level of inactive desphospho-uncarboxylated MGP (dp-ucMGP). We also studied the distribution of active and inactive MGP in human myocardium. METHODS We measured echocardiographic diastolic LV function and plasma dp-ucMGP (ELISA) in 668 Flemish and for replication in 386 Swiss. RESULTS Among Flemish and Swiss, E/e' (6.78 vs. 6.73) and dp-ucMGP (3.94 μg/L vs. 4.20 μg/L) were similarly distributed. In multivariable-adjusted models, for each doubling of dp-ucMGP, E/e' increased by 0.26, 0.33 and 0.31 in Flemish, Swiss and both cohorts combined (P≤0.026); the odds ratios for having E/e' ≥ 8.5 were 1.99, 3.29 and 2.36, respectively (P≤0.017). Cardiac biopsies from patients with ischemic or dilated cardiomyopathy and healthy hearts (n = 4 for each) were stained with conformation-specific MGP antibodies. In diseased compared with normal hearts, uncarboxylated inactive MGP was more prevalent (P≤0.004) in the perivascular matrix and interstitium (204.4 vs. 8.6 μm2 per field) and phosphorylated active MGP in and around capillaries and interstitial cells (31.3 vs. 6.6 number of positive capillaries and cells per field). CONCLUSIONS Our study supports a role of activated MGP in maintaining myocardial integrity and diastolic LV performance and can potentially be translated into new strategies for managing diastolic LV dysfunction and preventing its progression to heart failure.
Collapse
Affiliation(s)
- Fang-Fei Wei
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Sander Trenson
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Pierre Monney
- Department of Cardiology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Lausanne, Switzerland
| | - Wen-Yi Yang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Menno Pruijm
- Department of Nephrology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Zhen-Yu Zhang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Yassine Bouatou
- Department of Pathology, Academisch Medisch Centrum, Universiteit van Amsterdam, Amsterdam, The Netherlands
| | - Qi-Fang Huang
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Belen Ponte
- Department of Nephrology, University Hospital of Geneva, Geneva, Switzerland
| | - Pierre-Yves Martin
- Department of Nephrology, University Hospital of Geneva, Geneva, Switzerland
| | - Lutgarde Thijs
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Tatiana Kuznetsova
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Karel Allegaert
- Research Unit Organ Systems, Department of Development and Regeneration, University of Leuven, Leuven, Belgium
- Department of Pediatric Surgery, Erasmus Medical Center, Sophia Children’s Hospital, Rotterdam, The Netherlands
| | - Stefan Janssens
- Division of Cardiology, University Hospitals Leuven, Leuven, Belgium
| | - Cees Vermeer
- R&D Group VitaK, Maastricht University, Maastricht, The Netherlands
| | - Peter Verhamme
- Research Unit Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Michel Burnier
- Department of Nephrology, University Hospital of Lausanne, Lausanne, Switzerland
| | - Murielle Bochud
- Division of Chronic Disease, Institute of Social and Preventive Medicine, University Hospital of Lausanne, Lausanne, Switzerland
| | - Georg Ehret
- Department of Cardiology, University Hospital of Geneva, Geneva, Switzerland
- Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, John Hopkins University, Baltimore, Maryland, United States of America
| | - Jan A. Staessen
- Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
- R&D Group VitaK, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
20
|
Wei FF, Thijs L, Zhang ZY, Jacobs L, Yang WY, Salvi E, Citterio L, Cauwenberghs N, Kuznetsova T, E A Drummen N, Hara A, Manunta P, Li Y, Verhamme P, Allegaert K, Cusi D, Vermeer C, Staessen JA. The risk of nephrolithiasis is causally related to inactive matrix Gla protein, a marker of vitamin K status: a Mendelian randomization study in a Flemish population. Nephrol Dial Transplant 2018; 33:514-522. [PMID: 28340119 DOI: 10.1093/ndt/gfx014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Accepted: 01/15/2017] [Indexed: 01/12/2023] Open
Abstract
BACKGROUND Vitamin K (VK)-dependent γ-glutamate carboxylation and serine phosphorylation activate matrix Gla protein (MGP) to a potent locally acting inhibitor of calcification. Nephrolithiasis represents a process of unwanted calcification associated with substantial mortality and high recurrence rates. We hypothesized that the risk of nephrolithiasis increases with VK shortage, as exemplified by higher plasma levels of desphospho-uncarboxylated MGP (dp-ucMGP). METHODS In 1748 randomly recruited Flemish individuals (51.1% women; mean age 46.8 years), we determined dp-ucMGP and the prevalence of nephrolithiasis at baseline (April 1996-February 2015) and its incidence during follow-up until March 2016. We estimated the multivariable-adjusted relative risk associated with the doubling of dp-ucMGP, using logistic or Cox regression. We did a Mendelian randomization analysis using four MGP genotypes as instrumental variables. RESULTS With adjustments applied for sex, age and 24-h urinary volume and calcium excretion, the odds of having prevalent nephrolithiasis [n = 144 (8.2%)] associated with dp-ucMGP was 1.31 [95% confidence interval (CI) 1.04-1.64; P = 0.022]. dp-ucMGP levels were associated (P ≤ 0.001) with MGP variants rs2098435, rs4236 and rs2430692. In the Mendelian analysis, the causal odds ratio was 3.82 (95% CI 1.15-12.7; P = 0.029). The incidence of nephrolithiasis over 12.0 years (median) was 37 cases (0.2%). With similar adjustments as before, the hazard ratio in relation to dp-ucMGP was 2.48 (95% CI 1.71-3.61; P < 0.001). Additional adjustment for a nephrolithiasis propensity score produced consistent results. CONCLUSION Higher levels of inactive dp-ucMGP may be causally associated with the risk of nephrolithiasis. Whether or not VK deficiency plays a role in these observations remains to be firmly established.
Collapse
Affiliation(s)
- Fang-Fei Wei
- Studies Coordinating Centre, Research Unit of Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Lutgarde Thijs
- Studies Coordinating Centre, Research Unit of Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Zhen-Yu Zhang
- Studies Coordinating Centre, Research Unit of Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Lotte Jacobs
- Studies Coordinating Centre, Research Unit of Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Wen-Yi Yang
- Studies Coordinating Centre, Research Unit of Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Erika Salvi
- Genomics and Bioinformatics Platform at Filarete Foundation, Department of Health Sciences and Graduate School of Nephrology, Division of Nephrology, San Paolo Hospital, University of Milan, Italy
| | - Lorena Citterio
- Division of Nephrology and Dialysis, IRCCS San Raffaele Scientific Institute and School of Nephrology, University Vita-Salute San Raffaele, Milan, Italy
| | - Nicholas Cauwenberghs
- Studies Coordinating Centre, Research Unit of Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Tatiana Kuznetsova
- Studies Coordinating Centre, Research Unit of Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | | | - Azusa Hara
- Department of Social Pharmacy and Public Health, Showa Pharmaceutical University, Tokyo, Japan
| | - Paolo Manunta
- Division of Nephrology and Dialysis, IRCCS San Raffaele Scientific Institute and School of Nephrology, University Vita-Salute San Raffaele, Milan, Italy
| | - Yan Li
- Center for Epidemiological Studies and Clinical Trials and Center for Vascular Evaluations, Shanghai Institute of Hypertension, Shanghai Key Laboratory of Hypertension, Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Peter Verhamme
- Research Unit of Molecular and Vascular Biology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Karel Allegaert
- Research Unit of Organ Systems, KU Leuven Department of Development and Regeneration, University of Leuven, Leuven, Belgium
| | - Daniele Cusi
- Genomics and Bioinformatics Platform at Filarete Foundation, Department of Health Sciences and Graduate School of Nephrology, Division of Nephrology, San Paolo Hospital, University of Milan, Italy
| | - Cees Vermeer
- R&D Group VitaK, Maastricht University, Maastricht, The Netherlands
| | - Jan A Staessen
- Studies Coordinating Centre, Research Unit of Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
- R&D Group VitaK, Maastricht University, Maastricht, The Netherlands
| |
Collapse
|
21
|
Effect of Blumea balsamifera extract on the phase and morphology of calcium oxalate crystals. Asian J Urol 2018; 4:201-207. [PMID: 29387552 PMCID: PMC5772838 DOI: 10.1016/j.ajur.2016.08.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Revised: 03/23/2016] [Accepted: 06/01/2016] [Indexed: 11/23/2022] Open
Abstract
Objective Calcium oxalate crystals are found in majority of kidney stones with calcium oxalate monohydrate (COM) as one of the primary types of kidney stones. Various methods of treatment exist, including herbal treatment in the Philippines that uses the medicinal herb Blumea balsamifera (B. balsamifera). Methods The effect of B. balsamifera extract on the morphology of calcium oxalate crystals was studied by light microscopy, image analysis, powder X-ray diffraction and scanning electron microscopy. Results The extract decreased the crystal size by 5.22%–82.62% depending on the degree of supersaturation. Through analysis of the projected area of the crystals, the extract was found to shift the phase of the crystals from COM to calcium oxalate dihydrate (COD). This shift in phase is significant with a COM to COD shift of 88.26% at 0.5 mg/mL of extract and 91.53% at 1.0 mg/mL of extract. Scanning election microscopic (SEM) images revealed aggregation of crystals at 0 mg/mL of extract. At 1.0 mg/mL of extract, the scanning electron micrographs showed discernible crystal unit boundaries. Conclusion B. balsamifera extract was observed to have decreased crystal size, shifted crystal phase from COM to COD and prevented the aggregation of calcium oxalate crystals.
Collapse
|
22
|
Grohe B. RETRACTED: Synthetic peptides derived from salivary proteins and the control of surface charge densities of dental surfaces improve the inhibition of dental calculus formation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 77:58-68. [DOI: 10.1016/j.msec.2017.03.229] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 01/09/2017] [Accepted: 03/24/2017] [Indexed: 11/30/2022]
|
23
|
|
24
|
Robertson WG. Do "inhibitors of crystallisation" play any role in the prevention of kidney stones? A critique. Urolithiasis 2016; 45:43-56. [PMID: 27900407 DOI: 10.1007/s00240-016-0953-y] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Accepted: 11/24/2016] [Indexed: 01/09/2023]
Abstract
A critical examination of data in the literature and in as yet unpublished laboratory records on the possible role of so-called inhibitors of crystallisation in preventing the formation of calcium-containing kidney stones leads to the following conclusions. So-called inhibitors of spontaneous "self-nucleation" are unlikely to play any role in the initiation of the crystallisation of CaOx or CaP in urine because excessive urinary supersaturation of urine with respect to these salts dominates the onset of "self-nucleation" within the normal time frame of the transit of tubular fluid through the nephron (3-4 min). Inhibitors of the crystal growth of CaOx crystals may or may not play a significant role in the prevention of CaOx stone-formation since once again excessive supersaturation of urine can overwhelm any potential effect of the inhibitors on the growth process. However, they may play a role as inhibitors of crystal growth at lower levels of metastable supersaturation when the balance between supersaturation and inhibitors is more equal. Inhibitors of CaOx crystal aggregation may play a significant role in the prevention of stones, since they do not appear to be strongly affected by excessive supersaturation, either in vitro or in vivo. Inhibitors of CaOx crystal binding to renal tubular epithelium may exist but further studies are necessary to elucidate their importance in reducing the risk of initiating stones in the renal tubules. Inhibitors of CaOx crystal binding to Randall's Plaques and Randall's Plugs may exist but further studies are necessary to elucidate their importance in reducing the risk of initiating stones on renal papillae. There may be an alternative explanation other than a deficiency in the excretion of inhibitors for the observations that there is a difference between CaOx crystal size and degree of aggregation in the fresh, warm urines of normal subjects compared those in urine from patients with recurrent CaOx stones. This difference may depend more on the site of "self-nucleation" of CaOx crystals in the renal tubule rather than on a deficiency in the excretion of so-called inhibitors of crystallisation by patients with CaOx stones. The claim that administration of potassium citrate, potassium magnesium citrate or magnesium hydroxide reduces the rate of stone recurrence may be due to the effect of these forms of medication on the supersaturation of urine with respect to CaOx and CaP rather than to any increase in "inhibitory activity" attributed to these forms of treatment. In summary, there is a competition between supersaturation and so-called inhibitors of crystallisation which ultimately determines the pattern of crystalluria in stone-formers and normals. If the supersaturation of urine with respect to CaOx reaches or exceeds the 3-4 min formation product of that salt, then it dominates the crystallisation process both in terms of "self-nucleation" and crystal growth but appears to have little or no effect on the degree of aggregation of the crystals produced. At supersaturation levels of urine with respect to CaOx well below the 3-4 min formation product of that salt, the influence of inhibitors increases and some may affect not only the degree of aggregation but also the crystal growth of any pre-formed crystals of CaOx at these lower levels of metastability.
Collapse
Affiliation(s)
- William G Robertson
- Visiting Professor at the University of Oxford, Nuffield Department of Surgical Sciences, John Radcliffe Hospital, Headley Way, Headington, Oxford, OX3 9DU, UK.
| |
Collapse
|
25
|
Langdon A, Grohe B. The osteopontin-controlled switching of calcium oxalate monohydrate morphologies in artificial urine provides insights into the formation of papillary kidney stones. Colloids Surf B Biointerfaces 2016; 146:296-306. [PMID: 27362921 DOI: 10.1016/j.colsurfb.2016.06.030] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Revised: 06/15/2016] [Accepted: 06/16/2016] [Indexed: 11/30/2022]
Abstract
The protein osteopontin (OPN) plays an important role in preventing the formation of calcium oxalate monohydrate (COM) kidney stones. To gain insight into these mechanisms, crystallization was induced by addition of human kidney OPN to artificial urine (ionic strength comparable to urine; without citrate), and the OPN-COM interaction studied using a combination of scanning electron (SEM) and confocal microscopy. By SEM, we found that increasing OPN concentrations formed large monoclinic penetration twins (no protein added) and, at higher concentrations (1-, 2μg/ml OPN), super and hyper twins with crystal habits not found in previous studies. For instance, the hyper twins indicate well-facetted gearwheel-like habits with "teeth" developed in all crystallographic <h0l> directions. At OPN concentrations ≥2μg/ml, a switching to small dumbbell-shaped COM habits with fine-textured surfaces occurred. Confocal microscopy of these dumbbells indicates protein incorporation in almost the entire crystal structure (in contrast to facetted COM), proposing a threshold concentration of ∼2μg/ml OPN for the facetted to the non-facetted habit transformation. Both the gearwheel-like and the dumbbell-shaped habit are again found side-by-side (presumably triggered by OPN concentration gradients within the sample) in in-vitro formed conglomerates, which resemble cross-sections of papillary kidney stones. The abrupt transformation from facetted to non-facetted habits and the unique compliance of the two in-vitro formed habits with the two main morphologies found in papillary kidney stones propose that OPN is a main effector in direct stone-forming processes. Moreover, stone structures which exhibit these two morphologies side-by-side might serve as a novel indicator for OPN concentrations surrounding those structures.
Collapse
Affiliation(s)
- Aaron Langdon
- Schulich School of Medicine & Dentistry, School of Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada, Canada.
| | - Bernd Grohe
- Schulich School of Medicine & Dentistry, School of Dentistry, University of Western Ontario, London, ON N6A 5C1, Canada, Canada; Department of Chemical & Biochemical Engineering, University of Western Ontario, London, ON N6A 5B9, Canada, Canada.
| |
Collapse
|
26
|
Pivin E, Ponte B, Pruijm M, Ackermann D, Guessous I, Ehret G, Liu YP, Drummen NEA, Knapen MHJ, Pechere-Bertschi A, Paccaud F, Mohaupt M, Vermeer C, Staessen JA, Vogt B, Martin PY, Burnier M, Bochud M. Inactive Matrix Gla-Protein Is Associated With Arterial Stiffness in an Adult Population-Based Study. Hypertension 2015; 66:85-92. [PMID: 25987667 DOI: 10.1161/hypertensionaha.115.05177] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2015] [Accepted: 04/27/2015] [Indexed: 01/07/2023]
Abstract
Increased pulse wave velocity (PWV) is a marker of aortic stiffness and an independent predictor of mortality. Matrix Gla-protein (MGP) is a vascular calcification inhibitor that needs vitamin K to be activated. Inactive MGP, known as desphospho-uncarboxylated MGP (dp-ucMGP), can be measured in plasma and has been associated with various cardiovascular markers, cardiovascular outcomes, and mortality. In this study, we hypothesized that high levels of dp-ucMGP are associated with increased PWV. We recruited participants via a multicenter family-based cross-sectional study in Switzerland. Dp-ucMGP was quantified in plasma by sandwich ELISA. Aortic PWV was determined by applanation tonometry using carotid and femoral pulse waveforms. Multiple regression analysis was performed to estimate associations between PWV and dp-ucMGP adjusting for age, renal function, and other cardiovascular risk factors. We included 1001 participants in our analyses (475 men and 526 women). Mean values were 7.87±2.10 m/s for PWV and 0.43±0.20 nmol/L for dp-ucMGP. PWV was positively associated with dp-ucMGP both before and after adjustment for sex, age, body mass index, height, systolic and diastolic blood pressure (BP), heart rate, renal function, low- and high-density lipoprotein, glucose, smoking status, diabetes mellitus, BP and cholesterol lowering drugs, and history of cardiovascular disease (P≤0.01). In conclusion, high levels of dp-ucMGP are independently and positively associated with arterial stiffness after adjustment for common cardiovascular risk factors, renal function, and age. Experimental studies are needed to determine whether vitamin K supplementation slows arterial stiffening by increasing MGP carboxylation.
Collapse
Affiliation(s)
- Edward Pivin
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Belen Ponte
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Menno Pruijm
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Daniel Ackermann
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Idris Guessous
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Georg Ehret
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Yan-Ping Liu
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Nadja E A Drummen
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Marjo H J Knapen
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Antoinette Pechere-Bertschi
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Fred Paccaud
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Markus Mohaupt
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Cees Vermeer
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Jan A Staessen
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Bruno Vogt
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Pierre-Yves Martin
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Michel Burnier
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.)
| | - Murielle Bochud
- From the Division of Chronic Disease, University Institute of Social and Preventive Medicine (IUMSP) (E.P., F.P., M. Bochud, I.G.), and Department of Medicine, Service of Nephrology and Hypertension, (M.P., M. Burnier), University Hospital of Lausanne (CHUV), Lausanne, Switzerland; Service of Nephrology, Department of Specialties (B.P., P.-Y.M.), and Unit of Population Epidemiology, Department of Community Medicine and Primary Care and Emergency Medicine (I.G.), University Hospital of Geneva, Geneva, Switzerland; University Clinic for Nephrology, Hypertension and Clinical Pharmacology, Inselspital, Bern University Hospital and University of Bern, Bern, Switzerland (D.A., M.M., B.V.); Cardiology, Department of Specialties of Internal Medicine, Geneva University Hospitals, Geneva, Switzerland and Center for Complex Disease Genomics, McKusick-Nathans Institute of Genetic Medicine, Johns Hopkins University, Baltimore, MD (G.E.); Studies Coordinating Centre, Research Unit Hypertension and Cardiovascular Epidemiology, KU Leuven Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium (Y.-P.L., J.A.S.); R&D Group VitaK, Maastricht University, Maastricht, The Netherlands (N.E.A.D., M.H.J.K., C.V.); and Hypertension Unit, Department of Specialties, University Hospitals of Geneva, Geneva, Switzerland (A.P.-B.).
| |
Collapse
|
27
|
Wang J, Yang G, Wang Y, Du Y, Liu H, Zhu Y, Mao C, Zhang S. Chimeric Protein Template-Induced Shape Control of Bone Mineral Nanoparticles and Its Impact on Mesenchymal Stem Cell Fate. Biomacromolecules 2015; 16:1987-1996. [PMID: 26079683 DOI: 10.1021/acs.biomac.5b00419] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Protein-mediated molecular self-assembly has become a powerful strategy to fabricate biomimetic biomaterials with controlled shapes. Here we designed a novel chimeric molecular template made of two proteins, silk fibroin (SF) and albumin (ALB), which serve as a promoter and an inhibitor for hydroxyapatite (HA) formation, respectively, to synthesize HA nanoparticles with controlled shapes. HA nanospheres were produced by the chimeric ALB-SF template, whereas HA nanorods were generated by the SF template alone. The success in controlling the shape of HA nanoparticles allowed us to further study the effect of the shape of HA nanoparticles on the fate of rat mesenchymal stem cells (MSCs). We found that the nanoparticle shape had a crucial impact on the cellular uptake and HA nanospheres were internalized in MSCs at a faster rate. Both HA nanospheres and nanorods showed no significant influence on cell proliferation and migration. However, HA nanospheres significantly promoted the osteoblastic differentiation of MSCs in comparison to HA nanorods. Our work suggests that a chimeric combination of promoter and inhibitor proteins is a promising approach to tuning the shape of nanoparticles. It also sheds new light into the role of the shape of the HA nanoparticles in directing stem cell fate.
Collapse
Affiliation(s)
- Jianglin Wang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China.,Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Gaojie Yang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Yifan Wang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Yinying Du
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Haoming Liu
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| | - Ye Zhu
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Chuanbin Mao
- Department of Chemistry and Biochemistry, Stephenson Life Sciences Research Center, University of Oklahoma, Norman, Oklahoma 73019, United States
| | - Shengmin Zhang
- Advanced Biomaterials and Tissue Engineering Center, Huazhong University of Science and Technology, Wuhan, Hubei, 430074, China
| |
Collapse
|
28
|
Malhotra R, Burke MF, Martyn T, Shakartzi HR, Thayer TE, O’Rourke C, Li P, Derwall M, Spagnolli E, Kolodziej SA, Hoeft K, Mayeur C, Jiramongkolchai P, Kumar R, Buys ES, Yu PB, Bloch KD, Bloch DB. Inhibition of bone morphogenetic protein signal transduction prevents the medial vascular calcification associated with matrix Gla protein deficiency. PLoS One 2015; 10:e0117098. [PMID: 25603410 PMCID: PMC4300181 DOI: 10.1371/journal.pone.0117098] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2014] [Accepted: 12/18/2014] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Matrix Gla protein (MGP) is reported to inhibit bone morphogenetic protein (BMP) signal transduction. MGP deficiency is associated with medial calcification of the arterial wall, in a process that involves both osteogenic transdifferentiation of vascular smooth muscle cells (VSMCs) and mesenchymal transition of endothelial cells (EndMT). In this study, we investigated the contribution of BMP signal transduction to the medial calcification that develops in MGP-deficient mice. APPROACH AND RESULTS MGP-deficient mice (MGP(-/-)) were treated with one of two BMP signaling inhibitors, LDN-193189 or ALK3-Fc, beginning one day after birth. Aortic calcification was assessed in 28-day-old mice by measuring the uptake of a fluorescent bisphosphonate probe and by staining tissue sections with Alizarin red. Aortic calcification was 80% less in MGP(-/-) mice treated with LDN-193189 or ALK3-Fc compared with vehicle-treated control animals (P<0.001 for both). LDN-193189-treated MGP(-/-) mice survived longer than vehicle-treated MGP(-/-) mice. Levels of phosphorylated Smad1/5 and Id1 mRNA (markers of BMP signaling) did not differ in the aortas from MGP(-/-) and wild-type mice. Markers of EndMT and osteogenesis were increased in MGP(-/-) aortas, an effect that was prevented by LDN-193189. Calcification of isolated VSMCs was also inhibited by LDN-193189. CONCLUSIONS Inhibition of BMP signaling leads to reduced vascular calcification and improved survival in MGP(-/-) mice. The EndMT and osteogenic transdifferentiation associated with MGP deficiency is dependent upon BMP signaling. These results suggest that BMP signal transduction has critical roles in the development of vascular calcification in MGP-deficient mice.
Collapse
Affiliation(s)
- Rajeev Malhotra
- Cardiovascular Research Center and Cardiology Division of the Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Megan F. Burke
- Cardiovascular Research Center and Cardiology Division of the Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Trejeeve Martyn
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Hannah R. Shakartzi
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Timothy E. Thayer
- Cardiovascular Research Center and Cardiology Division of the Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Caitlin O’Rourke
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Pingcheng Li
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Matthias Derwall
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
- Department of Anesthesiology, Uniklinik Aachen, RWTH Aachen University, Aachen, Germany
| | - Ester Spagnolli
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Starsha A. Kolodziej
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Konrad Hoeft
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Claire Mayeur
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Pawina Jiramongkolchai
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Ravindra Kumar
- Acceleron Pharma, Inc. Cambridge, MA, United States of America
| | - Emmanuel S. Buys
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Paul B. Yu
- Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Kenneth D. Bloch
- Cardiovascular Research Center and Cardiology Division of the Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| | - Donald B. Bloch
- Anesthesia Center for Critical Care Research of the Department of Anesthesia, Critical Care, and Pain Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
- Center for Immunology and Inflammatory Diseases and the Division of Rheumatology, Allergy, and Immunology of the Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, MA, United States of America
| |
Collapse
|
29
|
Church AT, Hughes ZE, Walsh TR. Improving the description of interactions between Ca2+ and protein carboxylate groups, including γ-carboxyglutamic acid: revised CHARMM22* parameters. RSC Adv 2015. [DOI: 10.1039/c5ra11268k] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
We show that the CHARMM22* force-field over-binds the interaction between aqueous carboxylates and Ca2+, and introduce a modification that can recover experimentally-determined binding free energies for these systems.
Collapse
Affiliation(s)
- Andrew T. Church
- Institute for Frontier Materials
- Deakin University
- Geelong
- Australia
| | - Zak E. Hughes
- Institute for Frontier Materials
- Deakin University
- Geelong
- Australia
| | - Tiffany R. Walsh
- Institute for Frontier Materials
- Deakin University
- Geelong
- Australia
| |
Collapse
|
30
|
Arcidiacono T, Mingione A, Macrina L, Pivari F, Soldati L, Vezzoli G. Idiopathic calcium nephrolithiasis: a review of pathogenic mechanisms in the light of genetic studies. Am J Nephrol 2014; 40:499-506. [PMID: 25504362 DOI: 10.1159/000369833] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND Calcium nephrolithiasis is a multifactorial disease with a polygenic milieu. Association studies identified genetic polymorphisms potentially implicated in the pathogenesis of calcium nephrolithiasis. The present article reviews the mechanisms of calcium stone formation and the potential contribution of gene polymorphisms to lithogenic mechanisms. SUMMARY Endoscopy observations suggested that precipitation of calcium-oxalate on the Randall's plaque at the papilla surface may cause idiopathic calcium-oxalate stones. The Randall's plaque is a hydroxyapatite deposit in the interstitium of the kidney medulla, which resembles a soft tissue calcification. Conversely, calcium-phosphate stones may develop from crystalline deposits located at the tip of the Bellini duct. Polymorphisms of eleven genes have been associated with stones in genome-wide association studies and replicated candidate-gene association studies: VDR, SLC34A1, SLC34A4, CLDN14, and CaSR genes coding for proteins regulating tubular phosphate and calcium reabsorption; CaSR, MGP, OPN, PLAU, and UMOD genes coding for proteins preventing calcium salt precipitation; AQP1 gene coding for a water channel in the proximal tubule. The renal activity of the last gene, DGKH, is unknown. Polymorphisms in these genes may predispose to calcium-oxalate and -phosphate stones by increasing the risk of calcium-phosphate precipitation in the tubular fluid. Key Messages: Genetic findings suggest that tubular fluid supersaturation with respect to calcium and phosphate predisposes to calcium-oxalate stones by triggering cellular mechanisms that lead to the Randall's plaque formation.
Collapse
Affiliation(s)
- Teresa Arcidiacono
- Nephrology and Dialysis Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | | | | | | | | |
Collapse
|
31
|
Cancela ML, Laizé V, Conceição N. Matrix Gla protein and osteocalcin: from gene duplication to neofunctionalization. Arch Biochem Biophys 2014; 561:56-63. [PMID: 25068814 DOI: 10.1016/j.abb.2014.07.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Revised: 07/04/2014] [Accepted: 07/15/2014] [Indexed: 12/31/2022]
Abstract
Osteocalcin (OC or bone Gla protein, BGP) and matrix Gla protein (MGP) are two members of the growing family of vitamin K-dependent (VKD) proteins. They were the first VKD proteins found not to be involved in coagulation and synthesized outside the liver. Both proteins were isolated from bone although it is now known that only OC is synthesized by bone cells under normal physiological conditions, but since both proteins can bind calcium and hydroxyapatite, they can also accumulate in bone. Both OC and MGP share similar structural features, both in terms of protein domains and gene organization. OC gene is likely to have appeared from MGP through a tandem gene duplication that occurred concomitantly with the appearance of the bony vertebrates. Despite their relatively close relationship and the fact that both can bind calcium and affect mineralization, their functions are not redundant and they also have other unrelated functions. Interestingly, these two proteins appear to have followed quite different evolutionary strategies in order to acquire novel functionalities, with OC following a gene duplication strategy while MGP variability was obtained mostly by the use of multiple promoters and alternative splicing, leading to proteins with additional functional characteristics and alternative gene regulatory pathways.
Collapse
Affiliation(s)
- M Leonor Cancela
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal; Department of Biomedical Sciences and Medicine, University of Algarve, 8005-139 Faro, Portugal.
| | - Vincent Laizé
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
| | - Natércia Conceição
- Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
| |
Collapse
|