1
|
Li L, Cheng M, Jin J, Zhao Y, Bai W, Zhang D, Zhang S, Bai Y, Xu J. The m6A reader YTHDF2 protects vascular smooth muscle cells against the osteogenic differentiation through targeting Runx2. Ren Fail 2025; 47:2488876. [PMID: 40230077 PMCID: PMC12001846 DOI: 10.1080/0886022x.2025.2488876] [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: 08/10/2024] [Revised: 03/12/2025] [Accepted: 03/31/2025] [Indexed: 04/16/2025] Open
Abstract
Vascular calcification (VC) is an important pathological development progress in chronic kidney disease (CKD) and may increase mortality but lacks effective treatments. N6-methyladenosine (m6A) has been verified to be the most prevalent internal chemical RNA modification in mammalian mRNAs. The M6A-modified mRNA degradation process is mediated by the reader YTHDF2 in an m6A-dependent manner. Nevertheless, the exact role and molecular mechanism of YTHDF2 in VC remain unclear. This study aimed to investigate the potential role of YTHDF2 in the osteogenic differentiation of vascular smooth muscle cells (VSMCs). It was found that YTHDF2 was markedly downregulated in both in vivo and in vitro calcified models. Functionally, YTHDF2 plays a protective role in VC. The overexpression of YTHDF2 inhibited the transdifferentiation of VSMCs from a contractile to an osteogenic phenotype, thus decreasing the expression of mineralization regulatory proteins and calcium deposition. Conversely, YTHDF2 deficiency aggravated this process. At the mechanistic level, YTHDF2 suppressed osteogenic transdifferentiation of VSMCs by regulating the Runt-related transcription factor 2 (Runx2). RNA immunoprecipitation-qPCR (RIP-qPCR) confirmed the binding of YTHDF2 to Runx2, and luciferase reporter assays confirmed the presence of the m6A site in Runx2. In addition, an actinomycin D assay showed that the half-life of Runx2 mRNA was dramatically shortened in VSMCs overexpressing YTHDF2. These results suggest that YTHDF2 directly binds to the m6A modification site of Runx2 to mediate the mRNA degradation that prevents VC by inhibiting the osteogenic development of VSMCs. Therefore, YTHDF2 can be considered a potential therapeutic target for managing VC.
Collapse
Affiliation(s)
- Lanmei Li
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Meijuan Cheng
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Jingjing Jin
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Yunfeng Zhao
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Weiwei Bai
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Dongxue Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Shenglei Zhang
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Yaling Bai
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| | - Jinsheng Xu
- Department of Nephrology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, China
- Hebei Clinical Research Center for Chronic Kidney Disease, Hebei Key Laboratory of Vascular Calcification in Kidney Disease, Shijiazhuang, China
| |
Collapse
|
2
|
Minisola S, Cipriani C, Colangelo L, Labbadia G, Pepe J, Magnusson P. Diagnostic Approach to Abnormal Alkaline Phosphatase Value. Mayo Clin Proc 2025; 100:712-728. [PMID: 40019430 DOI: 10.1016/j.mayocp.2024.11.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/04/2024] [Accepted: 11/20/2024] [Indexed: 03/01/2025]
Abstract
Alkaline phosphatase (ALP) is abundantly represented in nature, being fundamental for a number of processes. In addition to its fundamental function in skeletal mineralization, its roles in the pathogenesis of other diseases are being explored. The measurement of total ALP activity in serum or in plasma is a useful biomarker in clinical practice. Indeed, routine measurement of serum total ALP is a long-standing established part of initial biochemical evaluation of patients both in the hospital setting and on an ambulatory basis. Raised or reduced values of this enzyme activity are indicative of a number of diseases, most commonly affecting the skeleton and the biliary tract. Electrophoretic assays are preferable for visualizing and investigating the cause of increased serum total ALP activities, and bone ALP immunoassays are preferable for investigating and monitoring individuals with bone and mineral metabolic abnormalities. Here, we give a holistic vision of this fundamental enzyme, suggesting a clinical approach to the identification of diseases causing abnormal values. Finally, a therapeutic role has emerged as substitutive therapy in patients with hypophosphatasia, even though ongoing and future studies are exploring its role in other therapeutic areas. This narrative review was based on articles found by searching PubMed from its inception until July 2024 for the terms alkaline phosphatases, isozymes, isoforms, bone alkaline phosphatase, liver alkaline phosphatase, intestinal alkaline phosphatase, placental alkaline phosphatase, liver function tests, γ-glutamyltransferase, skeletal diseases, and liver diseases. We limited our research to papers published in the English language, with emphasis placed on those describing differential diagnosis whenever available.
Collapse
Affiliation(s)
- Salvatore Minisola
- Department of Clinical, Internal, Anesthesiologic, and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy.
| | - Cristiana Cipriani
- Department of Clinical, Internal, Anesthesiologic, and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Luciano Colangelo
- Department of Clinical, Internal, Anesthesiologic, and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Giancarlo Labbadia
- Department of Clinical, Internal, Anesthesiologic, and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Jessica Pepe
- Department of Clinical, Internal, Anesthesiologic, and Cardiovascular Sciences, "Sapienza" University of Rome, Rome, Italy
| | - Per Magnusson
- Department of Clinical Chemistry and Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| |
Collapse
|
3
|
Tan B, Tang W, Zeng Y, Liu J, Du X, Su H, Pang X, Liao L, Hu Q. Development of animal models with chronic kidney disease-mineral and bone disorder based on clinical characteristics and pathogenesis. Front Endocrinol (Lausanne) 2025; 16:1549562. [PMID: 40201764 PMCID: PMC11975589 DOI: 10.3389/fendo.2025.1549562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Chronic kidney disease-mineral and bone disorder (CKD-MBD) is a systemic complication of chronic kidney disease (CKD), resulting in high morbidity and mortality. However, effective treatment strategies are lacking. The pathogenesis of CKD-MBD is unclear but involves feedback mechanisms between calcium, phosphorus, parathyroid hormone, vitamin D and other factors, in addition to FGF23, Klotho, Wnt inhibitors, and activin A. Construction of a perfect animal model of CKD-MBD with clinical characteristics is important for in-depth study of disease development, pathological changes, targeted drug screening, and management of patients. Currently, the modeling methods of CKD-MBD include surgery, feeding and radiation. Additionally, the method of CKD-MBD modeling by surgical combined feeding is worth promoting because of short time, simplicity, and low mortality. Therefore, this review based on the pathogenesis and clinical features of CKD-MBD, combined with the current status of animal models, outlines the advantages and disadvantages of modeling methods, and provides a reference for further CKD-MBD research.
Collapse
Affiliation(s)
- Biyu Tan
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Weili Tang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Sichuan, China
| | - Yan Zeng
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Jian Liu
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Xiaomei Du
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Hongwei Su
- Department of Urology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Xianlun Pang
- Department of Orthopedics, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Sichuan, China
| | - Lishang Liao
- Department of Neurosurgery, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| | - Qiongdan Hu
- Department of Nephrology, The Affiliated Traditional Chinese Medicine Hospital, Southwest Medical University, Sichuan, China
| |
Collapse
|
4
|
Furgoł T, Karska K, Miciak M, Jureczko J, Gigoń K, Jezierzański M, Jureczko P. Coronary artery disease with heavily calcified lesions - literature review of novel therapeutic methods. Folia Med (Plovdiv) 2025; 67. [PMID: 40270140 DOI: 10.3897/folmed.67.e141763] [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: 11/13/2024] [Accepted: 02/11/2025] [Indexed: 04/25/2025] Open
Abstract
Coronary artery disease and atherosclerosis are a very significant and widespread problem in modern medicine. The development of current diagnostics and treatment of atherosclerotic lesions is moving towards minimally invasive methods. The purpose of this article is to present selected novel methods of treating coronary atherosclerosis, comparing their effectiveness, indications, contraindications, and possible complications. A literature review of selected treatment methods for calcified atherosclerotic lesions was conducted in the online databases of PubMed, PubMed Central, Google Scholar, and NCBI. It includes original and review papers. The main language of the articles was English. The search used keywords such as "coronary artery disease," "calcified atherosclerotic lesions," "rotational atherectomy," "intravascular lithotripsy," and "RotaTripsy," as well as related phrases. After analyzing the abstracts, the papers that most closely matched the stated topic were selected. Atherosclerosis is the leading cause of coronary heart disease incidence. Several risk factors, both non-modifiable and modifiable, predispose to its occurrence. Heavily calcified atherosclerotic plaques are associated with a higher risk of coronary artery disease consequences. Currently, methods such as CT coronary angiography and optical coherence tomography are used for diagnosis. Endovascular therapies are now recommended for the treatment of atherosclerosis with heavily calcified plaques. Rotational atherectomy, intravascular lithotripsy and RotaTripsy are promising methods for treating high-grade atherosclerosis with calcified deposits. However, especially in the case of RotaTripsy, further clinical studies are required to better evaluate the efficacy of this novel method.
Collapse
|
5
|
Guan C, Chen SX, Huang CL, Du YP, Wang KH, Li PX, Liu SR, Liu ZY, Huang Z. Utilizing Integrated Bioinformatics Analysis to Explore Potential Alterations in Mitochondrial Function Within Immune Cells Associated with Thoracic Aortic Aneurysms. Bioengineering (Basel) 2025; 12:197. [PMID: 40001716 PMCID: PMC11852063 DOI: 10.3390/bioengineering12020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
Thoracic aortic aneurysm (TAA) is a life-threatening peripheral vascular disease with a complex pathogenesis. Altered mitochondrial function in vascular smooth muscle cells has been implicated in TAA development. However, the link between mitochondrial functional status and immune cell behavior in TAA patients remains largely unexplored. In this study, we analyzed several bulk RNA-seq and snRNA-seq datasets of TAA from the NCBI-GEO and Genome Sequence Archive database, identifying differentially expressed mitochondrial-related genes (DE-MRGs). To assess mitochondrial function, we calculated a mitoscore to represent the overall expression level of MRGs. Our analysis revealed mitochondrial-mediated apoptosis occurring in M1 macrophages, while CD4 + T cells demonstrated the activation of quality control mechanisms, such as mitochondrial fission. Through LASSO regression and SVM-RFE, we identified key MRGs, including MUCB, ARRB2, FRG, and ALPL, which we further validated using TAA mouse models. Additionally, we found that DE-MRGs were closely linked to methionine metabolism. In conclusion, this study highlights mitochondrial dysfunction in immune cells associated with TAA, shedding light on potential mitochondrial roles in TAA pathogenesis.
Collapse
Affiliation(s)
- Chang Guan
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Si-Xu Chen
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (S.-X.C.); (C.-L.H.)
| | - Chun-Ling Huang
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (S.-X.C.); (C.-L.H.)
| | - Yi-Peng Du
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Kai-Hao Wang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Pei-Xin Li
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Shen-Rong Liu
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| | - Zhao-Yu Liu
- Medical Research Center, Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China; (S.-X.C.); (C.-L.H.)
| | - Zheng Huang
- Department of Cardiology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou 510120, China; (C.G.); (Y.-P.D.); (K.-H.W.); (P.-X.L.); (S.-R.L.)
| |
Collapse
|
6
|
She C, Liu H. Construction and validation of a predictive model for the risk of osteoporosis in patients with chronic kidney disease based on NHANES data. PLoS One 2025; 20:e0316494. [PMID: 39913394 PMCID: PMC11801546 DOI: 10.1371/journal.pone.0316494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 12/10/2024] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Chronic kidney disease (CKD) patients tend to exhibit a heightened susceptibility to osteoporosis owing to abnormalities in mineral and bone metabolism. The objective of this study was to develop and validate a nomogram for the prediction of osteoporosis risk in patients with CKD. METHODS 1498 patients diagnosed with CKD were enrolled from the National Health and Nutrition Examination Survey (NHANES) data spanning 2005-2010, 2013-2014, and 2017-2018. The dataset was randomly divided into a training set and a validation set in a ratio of 7:3. Utilizing the least absolute shrinkage and selection operator (LASSO) regression technique for predictor identification, followed by employing multivariate logistic regression based on the selected predictors to construct a nomogram. The performance of the prediction model was assessed using various metrics, including the area under the receiver operating characteristic curve (AUC), calibration curve, the Hosmer-Lemeshow test, and decision curve analysis (DCA). RESULTS The construction of the nomogram was based on five predictors, namely age, height, weight, alkaline phosphatase (ALP), and history of fracture. The AUC of 0.8511 in the training set and 0.8184 in the validation set demonstrates robust discriminability. Furthermore, the excellent calibration and clinical applicability of the model have been thoroughly validated. CONCLUSIONS Our study suggests a nomogram, providing nephrologists with a convenient and effective tool for identifying individuals at high risk of osteoporosis and avoiding adverse outcomes related to CKD.
Collapse
Affiliation(s)
- Chunjie She
- Department of Orthopaedics, Chaohu Hospital affiliated to Anhui Medical University, Chaohu, Anhui, China
| | - Hefeng Liu
- Department of Orthopaedics, Chaohu Hospital affiliated to Anhui Medical University, Chaohu, Anhui, China
| |
Collapse
|
7
|
Razazian M, Bahiraii S, Jannat I, Tiffner A, Beilhack G, Levkau B, Voelkl J, Alesutan I. Sphingosine kinase 1 inhibition aggravates vascular smooth muscle cell calcification. Pflugers Arch 2025:10.1007/s00424-025-03068-6. [PMID: 39899071 DOI: 10.1007/s00424-025-03068-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 01/19/2025] [Accepted: 01/20/2025] [Indexed: 02/04/2025]
Abstract
Medial vascular calcification is common in chronic kidney disease patients and linked to hyperphosphatemia. Upon phosphate exposure, intricate signaling events orchestrate pro-calcific effects in the vasculature mediated by vascular smooth muscle cells (VSMCs). Sphingosine kinase 1 (SPHK1) produces sphingosine-1-phosphate (S1P) and is associated with complex effects in the vascular system. The present study investigated a possible involvement of SPHK1 in VSMC calcification. Experiments were performed in primary human aortic VSMCs under pro-calcific conditions, with pharmacological inhibition or knockdown of SPHK1 or SPNS2 (a lysolipid transporter involved in cellular S1P export), as well as in Sphk1-deficient and wild-type mice treated with cholecalciferol. In VSMCs, SPHK1 expression was up-regulated by pro-calcific conditions. Calcification medium up-regulated osteogenic marker mRNA expression and activity as well as calcification of VSMCs, effects significantly augmented by co-treatment with the SPHK1 inhibitor SK1-IN-1. SK1-IN-1 alone was sufficient to up-regulate osteogenic signaling in VSMCs during control conditions. Similarly, the SPHK1 inhibitor PF-543 and SPHK1 knockdown up-regulated osteogenic signaling in VSMCs and aggravated VSMC calcification. In contrast, co-treatment with the SPNS2 inhibitor SLF1081851 suppressed osteogenic signaling and calcification of VSMCs, effects abolished by silencing of SPHK1. In addition, Sphk1 deficiency aggravated vascular calcification and aortic osteogenic marker expression in mice after cholecalciferol overload. In conclusion, SPHK1 inhibition, knockdown, or deficiency aggravates vascular pro-calcific signaling and calcification. The reduced calcification after inhibition of S1P export suggests a possible involvement of intracellular S1P, but further studies are required to elucidate the complex roles of SPHKs and S1P signaling in calcifying VSMCs.
Collapse
Affiliation(s)
- Mehdi Razazian
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
| | - Sheyda Bahiraii
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
| | - Isratul Jannat
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
| | - Adéla Tiffner
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
- Institute of Biophysics, Johannes Kepler University Linz, Linz, Austria
| | - Georg Beilhack
- Division of Nephrology and Dialysis, Department of Medicine III, Medical University of Vienna, Vienna, Austria
| | - Bodo Levkau
- Institute of Molecular Medicine III, University Hospital and Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Jakob Voelkl
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria.
- Department of Nephrology and Medical Intensive Care, Charité-Universitätsmedizin Berlin, Berlin, Germany.
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany.
| | - Ioana Alesutan
- Institute for Physiology and Pathophysiology, Johannes Kepler University Linz, Krankenhausstrasse 5, 4020, Linz, Austria
| |
Collapse
|
8
|
Wang S, Yang L, Bai G, Gu Y, Fan Q, Guan X, Yuan J, Liu J. A preliminary study on calcifying nanoparticles in dental plaque: Isolation, characterization, and potential mineralization mechanism. Clin Exp Dent Res 2024; 10:e885. [PMID: 38798048 PMCID: PMC11128756 DOI: 10.1002/cre2.885] [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: 11/01/2023] [Revised: 01/31/2024] [Accepted: 04/02/2024] [Indexed: 05/29/2024] Open
Abstract
OBJECTIVES Calcifying nanoparticles (CNPs), referred to as nanobacteria (NB), are recognized to be associated with ectopic calcification. This study aims to isolate and culture CNPs from the dental plaque of patients with periodontal disease and investigate their possible role in unravelling the aetiology of periodontal disease. MATERIAL AND METHODS Supragingival and subgingival plaques were sampled from 30 periodontitis patients for CNPs isolation and culture. Alkaline phosphatase (ALP) content changes were tracked over time. Positive samples underwent thorough morphological identification via hematoxylin and eosin (HE) staining, Alizarin red S (ARS), and transmission electron microscopy (TEM). The chemical composition of CNPs analysis involved calcium (Ca) and phosphorus (P) content determination, Fourier transform infrared spectroscopy (FTIR), and X-ray diffraction (XRD). RESULTS The subgingival plaque dental group exhibited a higher CNPs isolation rate at 36.67% (11/30) compared to the supragingival dental plaque group at 66.67% (20/30). ALP activity varied among the positive, negative and control groups. Morphological observation characterized the CNPs as round, oval, and ellipsoid particles with Ca deposits. Chemical analysis revealed the Ca/P ratio was 0.6753. Hydroxyl, methyl, carbonate, phosphate, hydrogen phosphate, and dihydrogen phosphate were detected by FTIR; the main chemical components detected by XRD were hydroxyapatite and tricalcium phosphate. CONCLUSION CNPs were found in periodontitis-related dental plaque and exhibited the potential to develop calcified structures resembling dental calculus. However, the potential involvement of ALP in CNPs formation requires deeper exploration, as does the precise nature of its role and the interrelation with periodontitis demand a further comprehensive investigation.
Collapse
Affiliation(s)
- Siwei Wang
- Department of Dental Implantology, The Affiliated Stomatological HospitalZunyi Medical UniversityZunyiChina
| | - Lan Yang
- Department of Prosthodontics, The Affiliated Stomatological HospitalZunyi Medical UniversityZunyiChina
| | - Guohui Bai
- Key Laboratory of Oral Disease of Higher Schools in Guizhou ProvinceZunyi Medical UniversityZunyiChina
| | - Yu Gu
- Department of StomatologyZunyi Medical UniversityZhuhaiChina
| | - Qin Fan
- Department of Dental Implantology, The Affiliated Stomatological HospitalZunyi Medical UniversityZunyiChina
| | - Xiaoyan Guan
- Department of Orthodontics, The Affiliated Stomatological HospitalZunyi Medical UniversityZunyiChina
| | - Jie Yuan
- Department of Pain MedicineThe Affiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Jianguo Liu
- Key Laboratory of Oral Disease of Higher Schools in Guizhou ProvinceZunyi Medical UniversityZunyiChina
| |
Collapse
|
9
|
Ma W, Jia K, Cheng H, Xu H, Li Z, Zhang H, Xie H, Sun H, Yi L, Chen Z, Duan S, Sano M, Fukuda K, Lu L, Gao F, Zhang R, Yan X. Orphan Nuclear Receptor NR4A3 Promotes Vascular Calcification via Histone Lactylation. Circ Res 2024; 134:1427-1447. [PMID: 38629274 DOI: 10.1161/circresaha.123.323699] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Accepted: 04/02/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND Medial arterial calcification is a chronic systemic vascular disorder distinct from atherosclerosis and is commonly observed in patients with chronic kidney disease, diabetes, and aging individuals. We previously showed that NR4A3 (nuclear receptor subfamily 4 group A member 3), an orphan nuclear receptor, is a key regulator in apo (apolipoprotein) A-IV-induced atherosclerosis progression; however, its role in vascular calcification is poorly understood. METHODS We generated NR4A3-/- mice and 2 different types of medial arterial calcification models to investigate the biological roles of NR4A3 in vascular calcification. RNA-seq was performed to determine the transcriptional profile of NR4A3-/- vascular smooth muscle cells under β-glycerophosphate treatment. We integrated Cleavage Under Targets and Tagmentation analysis and RNA-seq data to further investigate the gene regulatory mechanisms of NR4A3 in arterial calcification and target genes regulated by histone lactylation. RESULTS NR4A3 expression was upregulated in calcified aortic tissues from chronic kidney disease mice, 1,25(OH)2VitD3 overload-induced mice, and human calcified aorta. NR4A3 deficiency preserved the vascular smooth muscle cell contractile phenotype, inhibited osteoblast differentiation-related gene expression, and reduced calcium deposition in the vasculature. Further, NR4A3 deficiency lowered the glycolytic rate and lactate production during the calcification process and decreased histone lactylation. Mechanistic studies further showed that NR4A3 enhanced glycolysis activity by directly binding to the promoter regions of the 2 glycolysis genes ALDOA and PFKL and driving their transcriptional initiation. Furthermore, histone lactylation promoted medial calcification both in vivo and in vitro. NR4A3 deficiency inhibited the transcription activation and expression of Phospho1 (phosphatase orphan 1). Consistently, pharmacological inhibition of Phospho1 attenuated calcium deposition in NR4A3-overexpressed vascular smooth muscle cells, whereas overexpression of Phospho1 reversed the anticalcific effect of NR4A3 deficiency in vascular smooth muscle cells. CONCLUSIONS Taken together, our findings reveal that NR4A3-mediated histone lactylation is a novel metabolome-epigenome signaling cascade mechanism that participates in the pathogenesis of medial arterial calcification.
Collapse
MESH Headings
- Animals
- Vascular Calcification/metabolism
- Vascular Calcification/genetics
- Vascular Calcification/pathology
- Mice
- Mice, Knockout
- Humans
- Histones/metabolism
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Mice, Inbred C57BL
- Nuclear Receptor Subfamily 4, Group A, Member 3/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 3/genetics
- Male
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Cells, Cultured
- DNA-Binding Proteins
- Nerve Tissue Proteins
- Receptors, Steroid
- Receptors, Thyroid Hormone
Collapse
Affiliation(s)
- Wenqi Ma
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
- Institute of Cardiovascular Diseases (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Kangni Jia
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
- Institute of Cardiovascular Diseases (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Haomai Cheng
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
- Institute of Cardiovascular Diseases (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Hong Xu
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Zhigang Li
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
- Institute of Cardiovascular Diseases (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Hang Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
- Institute of Cardiovascular Diseases (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Hongyang Xie
- Institute of Cardiovascular Diseases (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Hang Sun
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Lei Yi
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Zhiyong Chen
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Shengzhong Duan
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
- Laboratory of Oral Microbiota and Systemic Diseases, Shanghai Ninth People's Hospital, College of Stomatology (S.D.), Shanghai Jiao Tong University School of Medicine, China
- Department of Cardiovascular Medicine, State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Ruijin Hospital (S.D.), Shanghai Jiao Tong University School of Medicine, China
| | - Motoaki Sano
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (M.S., K.F.)
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Tokyo, Japan (M.S., K.F.)
| | - Lin Lu
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Fei Gao
- Beijing Anzhen Hospital, Capital Medical University, China (F.G.)
| | - Ruiyan Zhang
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| | - Xiaoxiang Yan
- Department of Cardiovascular Medicine, Ruijin Hospital (W.M., K.J., H.C., Z.L., H.Z., H.X., L.Z., Z.W., Y.C., H.S., L.Y., Z.C., L.L., R.Z., X.Y.), Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
10
|
Turner ME, Beck L, Hill Gallant KM, Chen Y, Moe OW, Kuro-o M, Moe S, Aikawa E. Phosphate in Cardiovascular Disease: From New Insights Into Molecular Mechanisms to Clinical Implications. Arterioscler Thromb Vasc Biol 2024; 44:584-602. [PMID: 38205639 PMCID: PMC10922848 DOI: 10.1161/atvbaha.123.319198] [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] [Indexed: 01/12/2024]
Abstract
Hyperphosphatemia is a common feature in patients with impaired kidney function and is associated with increased risk of cardiovascular disease. This phenomenon extends to the general population, whereby elevations of serum phosphate within the normal range increase risk; however, the mechanism by which this occurs is multifaceted, and many aspects are poorly understood. Less than 1% of total body phosphate is found in the circulation and extracellular space, and its regulation involves multiple organ cross talk and hormones to coordinate absorption from the small intestine and excretion by the kidneys. For phosphate to be regulated, it must be sensed. While mostly enigmatic, various phosphate sensors have been elucidated in recent years. Phosphate in the circulation can be buffered, either through regulated exchange between extracellular and cellular spaces or through chelation by circulating proteins (ie, fetuin-A) to form calciprotein particles, which in themselves serve a function for bulk mineral transport and signaling. Either through direct signaling or through mediators like hormones, calciprotein particles, or calcifying extracellular vesicles, phosphate can induce various cardiovascular disease pathologies: most notably, ectopic cardiovascular calcification but also left ventricular hypertrophy, as well as bone and kidney diseases, which then propagate phosphate dysregulation further. Therapies targeting phosphate have mostly focused on intestinal binding, of which appreciation and understanding of paracellular transport has greatly advanced the field. However, pharmacotherapies that target cardiovascular consequences of phosphate directly, such as vascular calcification, are still an area of great unmet medical need.
Collapse
Affiliation(s)
- Mandy E. Turner
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Laurent Beck
- Nantes Université, CNRS, Inserm, l’institut du thorax, F-44000 Nantes, France
| | - Kathleen M Hill Gallant
- Department of Food Science and Nutrition, University of Minnesota, St. Paul, Minnesota, USA
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Yabing Chen
- Department of Pathology, University of Alabama at Birmingham
- Research Department, Veterans Affairs Birmingham Medical Center, Birmingham, AL, USA
| | - Orson W Moe
- Charles and Jane Pak Center for Mineral Metabolism and Clinical Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Makoto Kuro-o
- Division of Anti-aging Medicine, Center for Molecular Medicine, Jichi Medical University 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Sharon Moe
- Division of Nephrology, Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
11
|
Villa-Bellosta R. Vascular Calcification: A Passive Process That Requires Active Inhibition. BIOLOGY 2024; 13:111. [PMID: 38392329 PMCID: PMC10886409 DOI: 10.3390/biology13020111] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/07/2024] [Accepted: 02/08/2024] [Indexed: 02/24/2024]
Abstract
The primary cause of worldwide mortality and morbidity stems from complications in the cardiovascular system resulting from accelerated atherosclerosis and arterial stiffening. Frequently, both pathologies are associated with the pathological calcification of cardiovascular structures, present in areas such as cardiac valves or blood vessels (vascular calcification). The accumulation of hydroxyapatite, the predominant form of calcium phosphate crystals, is a distinctive feature of vascular calcification. This phenomenon is commonly observed as a result of aging and is also linked to various diseases such as diabetes, chronic kidney disease, and several genetic disorders. A substantial body of evidence indicates that vascular calcification involves two primary processes: a passive process and an active process. The physicochemical process of hydroxyapatite formation and deposition (a passive process) is influenced significantly by hyperphosphatemia. However, the active synthesis of calcification inhibitors, including proteins and low-molecular-weight inhibitors such as pyrophosphate, is crucial. Excessive calcification occurs when there is a loss of function in enzymes and transporters responsible for extracellular pyrophosphate metabolism. Current in vivo treatments to prevent calcification involve addressing hyperphosphatemia with phosphate binders and implementing strategies to enhance the availability of pyrophosphate.
Collapse
Affiliation(s)
- Ricardo Villa-Bellosta
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), Campus Vida, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- Department of Biochemistry and Molecular Biology, University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
- The Health Research Institute of Santiago de Compostela (IDIS), Travesia da Choupana S/N, 15706 Santiago de Compostela, Spain
| |
Collapse
|
12
|
Kofotolios I, Bonios MJ, Adamopoulos M, Mourouzis I, Filippatos G, Boletis JN, Marinaki S, Mavroidis M. The Han:SPRD Rat: A Preclinical Model of Polycystic Kidney Disease. Biomedicines 2024; 12:362. [PMID: 38397964 PMCID: PMC10887417 DOI: 10.3390/biomedicines12020362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2023] [Revised: 01/27/2024] [Accepted: 01/30/2024] [Indexed: 02/25/2024] Open
Abstract
Autosomal Dominant Polycystic Kidney Disease (ADPKD) stands as the most prevalent hereditary renal disorder in humans, ultimately culminating in end-stage kidney disease. Animal models carrying mutations associated with polycystic kidney disease have played an important role in the advancement of ADPKD research. The Han:SPRD rat model, carrying an R823W mutation in the Anks6 gene, is characterized by cyst formation and kidney enlargement. The mutated protein, named Samcystin, is localized in cilia of tubular epithelial cells and seems to be involved in cystogenesis. The homozygous Anks6 mutation leads to end-stage renal disease and death, making it a critical factor in kidney development and function. This review explores the utility of the Han:SPRD rat model, highlighting its phenotypic similarity to human ADPKD. Specifically, we discuss its role in preclinical trials and its importance for investigating the pathogenesis of the disease and developing new therapeutic approaches.
Collapse
Affiliation(s)
- Ioannis Kofotolios
- Clinic of Nephrology and Renal Tranplantation, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece (M.M.)
| | - Michael J. Bonios
- Heart Failure and Transplant Unit, Onassis Cardiac Surgery Center, 17674 Athens, Greece;
| | - Markos Adamopoulos
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece (M.M.)
| | - Iordanis Mourouzis
- Department of Pharmacology, National and Kapodistrian University of Athens, 11527 Athens, Greece;
| | - Gerasimos Filippatos
- Department of Cardiology, Attikon University Hospital, Medical School, National and Kapodistrian University of Athens, 12462 Athens, Greece
| | - John N. Boletis
- Clinic of Nephrology and Renal Tranplantation, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Smaragdi Marinaki
- Clinic of Nephrology and Renal Tranplantation, Laiko Hospital, Medical School, National and Kapodistrian University of Athens, 11527 Athens, Greece
| | - Manolis Mavroidis
- Center of Basic Research, Biomedical Research Foundation, Academy of Athens, 11527 Athens, Greece (M.M.)
| |
Collapse
|
13
|
Yang X, Liu Y, Zhu X, Chen P, Xie X, Xu T, Zhang X, Zhao Y. Vascular, valvular and kidney calcification manifested in mouse models of adenine-induced chronic kidney disease. Ren Fail 2023; 45:2228920. [PMID: 37369635 DOI: 10.1080/0886022x.2023.2228920] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/19/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
BACKGROUND Ectopic calcification (EC) involves multiple organ systems in chronic kidney disease (CKD). Previous CKD-animal models primarily focused on a certain histological abnormality but did not show the correlation with calcified development among various tissues. This study compared calcified deposition in various tissues during CKD progression in mice. METHODS Male 8-week-old C57BL/6J mice were randomly allocated to the seven groups: a basic, adenine, high-phosphorus, or adenine and high-phosphorus diet for 12-16 weeks (Ctl16, A12, P16, or AP16, respectively); an adenine diet for 4-6 weeks; and a high-phosphorus or adenine and high-phosphorus diet for 10-12 weeks (A6 + P10, A4 + P12, or A4 + AP12, respectively). RESULTS Compared to the Ctl16 mice, the P16 mice only displayed a slight abnormality in serum calcium and phosphorus; the A12 mice had the most serious kidney impairment; the A4 + P12 and A6 + P10 mice had similar conditions of CKD, mineral abnormalities, and mild calcification in the kidney and aortic valves; the A4 + AP12 and AP16 groups had severe kidney impairment, mineral abnormalities and calcification in the kidneys, aortic valves and aortas. Furthermore, calcium-phosphate particles were deposited not only in the tubulointerstitial compartment but in the glomerular and tubular basement membrane. The elemental composition of EC in various tissues matched the calcification of human cardiovascular tissue as determined by energy dispersive spectroscopy. CONCLUSIONS The severity of CKD was unparalleled with the progression of mineral metabolism disorder and EC. Calcification was closely related in different tissues and observed in the glomerular and tubular basement membranes.
Collapse
Affiliation(s)
- Xin Yang
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yuqiu Liu
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaodong Zhu
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Pingsheng Chen
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaotong Xie
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Tian Xu
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Xiaoliang Zhang
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| | - Yu Zhao
- Department of Nephrology, Zhong Da Hospital, Southeast University School of Medicine, Nanjing, China
| |
Collapse
|
14
|
Li X, Chan YT, Jiang Y. Development of an image processing software for quantification of histological calcification staining images. PLoS One 2023; 18:e0286626. [PMID: 37797053 PMCID: PMC10553316 DOI: 10.1371/journal.pone.0286626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 05/22/2023] [Indexed: 10/07/2023] Open
Abstract
Quantification of the histological staining images gives important insights in biomedical research. In wet lab, it is common to have some stains off the target to become unwanted noisy stains during the generation of histological staining images. The current tools designed for quantification of histological staining images do not consider such situations; instead, the stained region is identified based on assumptions that the background is pure and clean. The goal of this study is to develop a light software named Staining Quantification (SQ) tool which could handle the image quantification job with features for removing a large amount of unwanted stains blended or overlaid with Region of Interest (ROI) in complex scenarios. The core algorithm was based on the method of higher order statistics transformation, and local density filtering. Compared with two state-of-art thresholding methods (i.e. Otsu's method and Triclass thresholding method), the SQ tool outperformed in situations such as (1) images with weak positive signals and experimental caused dirty stains; (2) images with experimental counterstaining by multiple colors; (3) complicated histological structure of target tissues. The algorithm was developed in R4.0.2 with over a thousand in-house histological images containing Alizarin Red (AR) and Von Kossa (VK) staining, and was validated using external images. For the measurements of area and intensity in total and stained region, the average mean of difference in percentage between SQ and ImageJ were all less than 0.05. Using this as a criterion of successful image recognition, the success rate for all measurements in AR, VK and external validation batch were above 0.8. The test of Pearson's coefficient, difference between SQ and ImageJ, and difference of proportions between SQ and ImageJ were all significant at level of 0.05. Our results indicated that the SQ tool is well established for automatic histological staining image quantification.
Collapse
Affiliation(s)
- Xinrui Li
- School of Medicine, Northwest University, Xi’an, Shaanxi, China
| | - Yau Tsz Chan
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Faculty of Medicine, Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
| | - Yangzi Jiang
- Faculty of Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Faculty of Medicine, Institute for Tissue Engineering and Regenerative Medicine, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
- Key Laboratory for Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, New Territories, Hong Kong, China
- Faculty of Medicine, Department of Orthopaedics & Traumatology, The Chinese University of Hong Kong, and Prince of Wales Hospital, Shatin, Hong Kong SAR, China
| |
Collapse
|
15
|
Cozzolino M, Maffei Faccioli F, Cara A, Boni Brivio G, Rivela F, Ciceri P, Magagnoli L, Galassi A, Barbuto S, Speciale S, Minicucci C, Cianciolo G. Future treatment of vascular calcification in chronic kidney disease. Expert Opin Pharmacother 2023; 24:2041-2057. [PMID: 37776230 DOI: 10.1080/14656566.2023.2266381] [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: 07/07/2023] [Accepted: 09/29/2023] [Indexed: 10/02/2023]
Abstract
INTRODUCTION Cardiovascular disease (CVD) is one of the global leading causes of morbidity and mortality in chronic kidney disease (CKD) patients. Vascular calcification (VC) is a major cause of CVD in this population and is the consequence of complex interactions between inhibitor and promoter factors leading to pathological deposition of calcium and phosphate in soft tissues. Different pathological landscapes are associated with the development of VC, such as endothelial dysfunction, oxidative stress, chronic inflammation, loss of mineralization inhibitors, release of calcifying extracellular vesicles (cEVs) and circulating calcifying cells. AREAS COVERED In this review, we examined the literature and summarized the pathophysiology, biomarkers and focused on the treatments of VC. EXPERT OPINION Even though there is no consensus regarding specific treatment options, we provide the currently available treatment strategies that focus on phosphate balance, correction of vitamin D and vitamin K deficiencies, avoidance of both extremes of bone turnover, normalizing calcium levels and reduction of inflammatory response and the potential and promising therapeutic approaches liketargeting cellular mechanisms of calcification (e.g. SNF472, TNAP inhibitors).Creating novel scores to detect in advance VC and implementing targeted therapies is crucial to treat them and improve the future management of these patients.
Collapse
Affiliation(s)
- Mario Cozzolino
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Federico Maffei Faccioli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Anila Cara
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Giulia Boni Brivio
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Francesca Rivela
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Paola Ciceri
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Lorenza Magagnoli
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Andrea Galassi
- Renal Division, Department of Health Sciences, ASST Santi Paolo e Carlo, University of Milan, Milan, Italy
| | - Simona Barbuto
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Serena Speciale
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Carlo Minicucci
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| | - Giuseppe Cianciolo
- Nephrology, Dialysis and Renal Transplant Unit, IRCCS-Azienda Ospedaliero-Universitaria di Bologna, Alma Mater Studiorum University of Bologna, Bologna, Italy
| |
Collapse
|
16
|
Mohamed FF, de Oliveira FA, Kinoshita Y, Yalamanchili RR, Eltilib LA, Andras NL, Narisawa S, Tani T, Chu EY, Millán JL, Foster BL. Dentoalveolar Alterations in an Adenine-Induced Chronic Kidney Disease Mouse Model. J Bone Miner Res 2023; 38:1192-1207. [PMID: 37191192 PMCID: PMC10524958 DOI: 10.1002/jbmr.4829] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 05/02/2023] [Accepted: 05/08/2023] [Indexed: 05/17/2023]
Abstract
Chronic kidney disease (CKD) is characterized by kidney damage and loss of renal function. CKD mineral and bone disorder (CKD-MBD) describes the dysregulation of mineral homeostasis, including hyperphosphatemia and elevated parathyroid hormone (PTH) secretion, skeletal abnormalities, and vascular calcification. CKD-MBD impacts the oral cavity, with effects including salivary gland dysfunction, enamel hypoplasia and damage, increased dentin formation, decreased pulp volume, pulp calcifications, and altered jaw bones, contributing to clinical manifestations of periodontal disease and tooth loss. Underlying mechanisms are not fully understood, and CKD mouse models commonly require invasive procedures with high rates of infection and mortality. We aimed to characterize the dentoalveolar effects of an adenine diet (AD)-induced CKD (AD-CKD) mouse model. Eight-week-old C57BL/6J mice were provided either a normal phosphorus diet control (CTR) or adenine and high-phosphorus diet CKD to induce kidney failure. Mice were euthanized at 15 weeks old, and mandibles were collected for micro-computed tomography and histology. CKD mice exhibited kidney failure, hyperphosphatemia, and hyperparathyroidism in association with porous cortical bone in femurs. CKD mice showed a 30% decrease in molar enamel volume compared to CTR mice. Enamel wear was associated with reduced ductal components, ectopic calcifications, and altered osteopontin (OPN) deposition in submandibular salivary glands of CKD mice. Molar cusps in CKD mice were flattened, exposing dentin. Molar dentin/cementum volume increased 7% in CKD mice and pulp volume decreased. Histology revealed excessive reactionary dentin and altered pulp-dentin extracellular matrix proteins, including increased OPN. Mandibular bone volume fraction decreased 12% and bone mineral density decreased 9% in CKD versus CTR mice. Alveolar bone in CKD mice exhibited increased tissue-nonspecific alkaline phosphatase localization, OPN deposition, and greater osteoclast numbers. AD-CKD recapitulated key aspects reported in CKD patients and revealed new insights into CKD-associated oral defects. This model has potential for studying mechanisms of dentoalveolar defects or therapeutic interventions. © 2023 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Fatma F. Mohamed
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Flavia Amadeu de Oliveira
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Yuka Kinoshita
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Riti R. Yalamanchili
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Leena A. Eltilib
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Natalie L. Andras
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Sonoko Narisawa
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Takashi Tani
- Department of Endocrinology, Metabolism and Nephrology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Emily Y. Chu
- Department of General Dentistry, Operative Division, University of Maryland School of Dentistry, Baltimore, Maryland, USA
| | - José Luis Millán
- Sanford Children’s Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Brian L. Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
17
|
Ma J, Li Y, Yang X, Liu K, Zhang X, Zuo X, Ye R, Wang Z, Shi R, Meng Q, Chen X. Signaling pathways in vascular function and hypertension: molecular mechanisms and therapeutic interventions. Signal Transduct Target Ther 2023; 8:168. [PMID: 37080965 PMCID: PMC10119183 DOI: 10.1038/s41392-023-01430-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 37.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 03/03/2023] [Accepted: 03/31/2023] [Indexed: 04/22/2023] Open
Abstract
Hypertension is a global public health issue and the leading cause of premature death in humans. Despite more than a century of research, hypertension remains difficult to cure due to its complex mechanisms involving multiple interactive factors and our limited understanding of it. Hypertension is a condition that is named after its clinical features. Vascular function is a factor that affects blood pressure directly, and it is a main strategy for clinically controlling BP to regulate constriction/relaxation function of blood vessels. Vascular elasticity, caliber, and reactivity are all characteristic indicators reflecting vascular function. Blood vessels are composed of three distinct layers, out of which the endothelial cells in intima and the smooth muscle cells in media are the main performers of vascular function. The alterations in signaling pathways in these cells are the key molecular mechanisms underlying vascular dysfunction and hypertension development. In this manuscript, we will comprehensively review the signaling pathways involved in vascular function regulation and hypertension progression, including calcium pathway, NO-NOsGC-cGMP pathway, various vascular remodeling pathways and some important upstream pathways such as renin-angiotensin-aldosterone system, oxidative stress-related signaling pathway, immunity/inflammation pathway, etc. Meanwhile, we will also summarize the treatment methods of hypertension that targets vascular function regulation and discuss the possibility of these signaling pathways being applied to clinical work.
Collapse
Affiliation(s)
- Jun Ma
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Yanan Li
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xiangyu Yang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Kai Liu
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xin Zhang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Xianghao Zuo
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Runyu Ye
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Ziqiong Wang
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Rufeng Shi
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China
| | - Qingtao Meng
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| | - Xiaoping Chen
- Department of Cardiology, West China Hospital, Sichuan University, No. 37, Guo Xue District, Chengdu, Sichuan, 610041, People's Republic of China.
| |
Collapse
|
18
|
Mouse Models of Mineral Bone Disorders Associated with Chronic Kidney Disease. Int J Mol Sci 2023; 24:ijms24065325. [PMID: 36982400 PMCID: PMC10048881 DOI: 10.3390/ijms24065325] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 02/27/2023] [Accepted: 03/03/2023] [Indexed: 03/14/2023] Open
Abstract
Patients with chronic kidney disease (CKD) inevitably develop mineral and bone disorders (CKD–MBD), which negatively impact their survival and quality of life. For a better understanding of underlying pathophysiology and identification of novel therapeutic approaches, mouse models are essential. CKD can be induced by surgical reduction of a functional kidney mass, by nephrotoxic compounds and by genetic engineering specifically interfering with kidney development. These models develop a large range of bone diseases, recapitulating different types of human CKD–MBD and associated sequelae, including vascular calcifications. Bones are usually studied by quantitative histomorphometry, immunohistochemistry and micro-CT, but alternative strategies have emerged, such as longitudinal in vivo osteoblast activity quantification by tracer scintigraphy. The results gained from the CKD–MBD mouse models are consistent with clinical observations and have provided significant knowledge on specific pathomechanisms, bone properties and potential novel therapeutic strategies. This review discusses available mouse models to study bone disease in CKD.
Collapse
|
19
|
Ralph D, Levine M, Millán JL, Uitto J, Li Q. Weighing the Evidence for the Roles of Plasma Versus Local Pyrophosphate in Ectopic Calcification Disorders. J Bone Miner Res 2023; 38:457-463. [PMID: 36807615 PMCID: PMC10365072 DOI: 10.1002/jbmr.4791] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 02/02/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023]
Abstract
Ectopic calcification is characterized by inappropriate deposition of calcium mineral in nonskeletal connective tissues and can cause significant morbidity and mortality, particularly when it affects the cardiovascular system. Identification of the metabolic and genetic determinants of ectopic calcification could help distinguish individuals at the greatest risk of developing these pathological calcifications and could guide development of medical interventions. Inorganic pyrophosphate (PPi ) has long been recognized as the most potent endogenous inhibitor of biomineralization. It has been intensively studied as both a marker and a potential therapeutic for ectopic calcification. Decreased extracellular concentrations of PPi have been proposed to be a unifying pathophysiological mechanism for disorders of ectopic calcification, both genetic and acquired. However, are reduced plasma concentrations of PPi a reliable predictor of ectopic calcification? This perspective article evaluates the literature in favor and against a pathophysiological role of plasma versus tissue PPi dysregulation as a determinant of, and as a biomarker for, ectopic calcification. © 2023 American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Douglas Ralph
- Genetics, Genomics and Cancer Biology Ph.D. Program, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA, USA.,Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA.,PXE International Center of Excellence in Research and Clinical Care, Thomas Jefferson University, Philadelphia, PA, USA
| | - Michael Levine
- Division of Endocrinology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jouni Uitto
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA.,PXE International Center of Excellence in Research and Clinical Care, Thomas Jefferson University, Philadelphia, PA, USA
| | - Qiaoli Li
- Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA, USA.,PXE International Center of Excellence in Research and Clinical Care, Thomas Jefferson University, Philadelphia, PA, USA
| |
Collapse
|
20
|
Padalkar MV, Tsivitis AH, Gelfman Y, Kasiyanyk M, Kaungumpillil N, Ma D, Gao M, Borges KA, Dhaliwal P, Nasruddin S, Saji S, Gilani H, Schram EJ, Singh M, Plummer MM, Savinova OV. Paradoxical reduction of plasma lipids and atherosclerosis in mice with adenine-induced chronic kidney disease and hypercholesterolemia. Front Cardiovasc Med 2023; 10:1088015. [PMID: 36844738 PMCID: PMC9947538 DOI: 10.3389/fcvm.2023.1088015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Accepted: 01/23/2023] [Indexed: 02/11/2023] Open
Abstract
Background Atherosclerotic cardiovascular disease is prevalent among patients with chronic kidney disease (CKD). In this study, we initially aimed to test whether vascular calcification associated with CKD can worsen atherosclerosis. However, a paradoxical finding emerged from attempting to test this hypothesis in a mouse model of adenine-induced CKD. Methods We combined adenine-induced CKD and diet-induced atherosclerosis in mice with a mutation in the low-density lipoprotein receptor gene. In the first study, mice were co-treated with 0.2% adenine in a western diet for 8 weeks to induce CKD and atherosclerosis simultaneously. In the second study, mice were pre-treated with adenine in a regular diet for 8 weeks, followed by a western diet for another 8 weeks. Results Co-treatment with adenine and a western diet resulted in a reduction of plasma triglycerides and cholesterol, liver lipid contents, and atherosclerosis in co-treated mice when compared with the western-only group, despite a fully penetrant CKD phenotype developed in response to adenine. In the two-step model, renal tubulointerstitial damage and polyuria persisted after the discontinuation of adenine in the adenine-pre-treated mice. The mice, however, had similar plasma triglycerides, cholesterol, liver lipid contents, and aortic root atherosclerosis after being fed a western diet, irrespective of adenine pre-treatment. Unexpectedly, adenine pre-treated mice consumed twice the calories from the diet as those not pre-treated without showing an increase in body weight. Conclusion The adenine-induced CKD model does not recapitulate accelerated atherosclerosis, limiting its use in pre-clinical studies. The results indicate that excessive adenine intake impacts lipid metabolism.
Collapse
Affiliation(s)
- Mugdha V. Padalkar
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Alexandra H. Tsivitis
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Ylona Gelfman
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Mariya Kasiyanyk
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Neil Kaungumpillil
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Danyang Ma
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Michael Gao
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Kelly A. Borges
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Puneet Dhaliwal
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Saud Nasruddin
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Sruthi Saji
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Hina Gilani
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Eric J. Schram
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Mohnish Singh
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Maria M. Plummer
- Department of Clinical Specialties, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| | - Olga V. Savinova
- Department of Biomedical Sciences, New York Institute of Technology College of Osteopathic Medicine, Old Westbury, NY, United States
| |
Collapse
|
21
|
Zhang X, Li T, Wang L, Li Y, Ruan T, Guo X, Wang Q, Meng X. Relative comparison of chronic kidney disease-mineral and bone disorder rat models. Front Physiol 2023; 14:1083725. [PMID: 36818435 PMCID: PMC9936098 DOI: 10.3389/fphys.2023.1083725] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Accepted: 01/18/2023] [Indexed: 02/05/2023] Open
Abstract
Objective: The aim of this study is to establish a suitable animal model of chronic kidney disease-mineral and bone disorder (CKD-MBD) by comparing CKD-MBD rat models induced by 5/6 Nx, AN, and UUO, accompanied by a low-calcium and high-phosphorus diet. Methods: Sprague‒Dawley rats were randomly divided into four groups: control group, 5/6 nephrectomy (5/6 Nx) group, Adriamycin nephropathy (AN) group, and unilateral ureteral obstruction (UUO) group. Serum biochemical indices were measured to evaluate renal function, mineral and bone metabolism, the severity of CKD-MBD, and the status of bone transformation. Hematoxylin-eosin staining (HE) and Masson's trichrome (Masson) staining were used for histopathological analysis of the kidney. Goldner's trichrome (Goldner) and tartrate-resistant acid phosphatase (TRAP) staining were utilized to observe bone mineralization and osteoclasts in the femur, respectively. Micro-CT images were applied to study the structure of the femur. The expression levels of osterix and cathepsin K in the femur were measured by immunohistochemistry (IHC) to confirm the status of bone transformation. Results: The levels of serum creatinine (Scr) and blood urea nitrogen (BUN) in the 5/6 Nx and AN group rats were significantly higher than those in the control rats, and this change was accompanied by marked changes in the levels of calcium (Ca), phosphate (Pi), intact parathyroid hormone (i-PTH), fibroblast growth factor 23 (FGF23), osteocalcin (OC), and cross-linked C-telopeptide of type 1 collagen (CTX-1); UUO group rats exhibited slight and inconsistent variations in the levels of Scr, BUN, Ca, Pi, i-PTH, FGF23, OC, and CTX-1 in serum. Histopathological analysis of the kidney showed that the UUO group rats suffered serious fibrosis and 5/6 Nx group rats exhibited severe focal calcification. Histopathological analysis of the femur showed that the AN group rats had minimal bone mineralization and that the 5/6 Nx group rats had overactive osteoclasts. Micro-CT revealed that the AN model had the most severe bone destruction and that the 5/6 Nx model had the least severe bone loss among the three models. The expression of cathepsin K in the femur was significantly increased in all models, while the expression of osterix in the femur was only significantly increased in the 5/6 Nx model. Conclusion: 5/6 Nx, AN, and UUO accompanied by a low-calcium and high-phosphorus diet successfully induced CKD-MBD in rats. The 5/6 NX model presented the progression of high-turnover bone disease, with consistency between biochemical indices in serum and histomorphometric analysis of the femur, and the AN and UUO models developed a severe deterioration in bone quantity and severe bone resorption; however, the changes in biochemical indices were subtle in the UUO model, and liver injury was obvious in the AN model.
Collapse
Affiliation(s)
- Xiaoqiong Zhang
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China,Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional Chinese Medicine, Chongqing, China
| | - Ting Li
- School of Pharmacy, Chongqing University of Medical Sciences, Chongqing, China
| | - Lijuan Wang
- Department of Pathology, Chongqing Hospital of Traditional Chinese Medicine, The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional, Chongqing, China
| | - Yanhui Li
- Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing Hospital of Traditional Chinese Medicine, The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional Chinese Medicine, Chongqing, China
| | - Taoren Ruan
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional Chinese Medicine, Chongqing, China
| | - Xiaohong Guo
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional Chinese Medicine, Chongqing, China
| | - Qin Wang
- Department of Pharmacy, Chongqing Hospital of Traditional Chinese Medicine, The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional Chinese Medicine, Chongqing, China,Chongqing Key Laboratory of Traditional Chinese Medicine to Prevent and Treat Autoimmune Diseases, Chongqing Hospital of Traditional Chinese Medicine, The Fourth Affiliated Clinical Medical College of Chengdu University of Traditional Chinese Medicine, Chongqing, China,*Correspondence: Qin Wang, ; Xianli Meng,
| | - Xianli Meng
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China,Innovative Institute of Chinese Medicine and Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China,*Correspondence: Qin Wang, ; Xianli Meng,
| |
Collapse
|
22
|
Amadeu de Oliveira F, Mohamed FF, Kinoshita Y, Narisawa S, Farquharson C, Miyake K, Foster BL, Millan JL. Gene Therapy Using Recombinant AAV Type 8 Vector Encoding TNAP-D 10 Improves the Skeletal Phenotypes in Murine Models of Osteomalacia. JBMR Plus 2023; 7:e10709. [PMID: 36699639 PMCID: PMC9850441 DOI: 10.1002/jbm4.10709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/25/2022] [Accepted: 11/08/2022] [Indexed: 12/05/2022] Open
Abstract
Hypophosphatasia (HPP), caused by loss-of-function mutations in the ALPL gene encoding tissue-nonspecific alkaline phosphatase (TNAP), is characterized by skeletal and dental hypomineralization that can vary in severity from life-threatening to milder manifestations only in adulthood. PHOSPHO1 deficiency leads to early-onset scoliosis, osteomalacia, and fractures that mimic pseudo-HPP. Asfotase alfa, a life-saving enzyme replacement therapy approved for pediatric-onset HPP, requires subcutaneous injections 3 to 6 times per week. We recently showed that a single injection of an adeno-associated virus vector serotype 8 harboring TNAP-D10 (AAV8-TNAP-D10) effectively prevented skeletal disease and prolonged life in Alpl -/- mice phenocopying infantile HPP. Here, we aimed to determine the efficacy of AAV8-TNAP-D10 in improving the skeletal and dental phenotype in the Alpl Prx1/Prx1 and Phospho1 -/- mouse models of late-onset (adult) HPP and pseudo-HPP, respectively. A single dose of 3 × 1011 vector genomes per body (vg/b) was injected intramuscularly into 8-week-old Alpl Prx1/Prx1 and wild-type (WT) littermates, or into 3-day-old Phospho1 -/- and WT mice, and treatment efficacy was evaluated after 60 days for late-onset HPP mice and after 90 days for Phospho1 -/- mice. Biochemical analysis showed sustained serum alkaline phosphatase activity and reduced plasma PPi levels, and radiographic images, micro-computed tomography (micro-CT) analysis, and hematoxylin and eosin (H&E) staining showed improvements in the long bones in the late-onset HPP mice and corrected scoliosis in the Phospho1 -/- mice. Micro-CT analysis of the dentoalveolar complex did not reveal significant changes in the phenotype of late-onset HPP and pseudo-HPP models. Moreover, alizarin red staining analysis showed that AAV8-TNAP-D10 treatment did not promote ectopic calcification of soft organs in adult HPP mice after 60 days of treatment, even after inducing chronic kidney disease. Overall, the AAV8-TNAP-D10 treatment improved the skeletal phenotype in both the adult HPP and pseudo-HPP mouse models. This preclinical study will contribute to the advancement of gene therapy for the improvement of skeletal disease in patients with heritable forms of osteomalacia. © 2022 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
| | - Fatma F. Mohamed
- Division of Biosciences, College of DentistryThe Ohio State UniversityColumbusOHUSA
| | - Yuka Kinoshita
- Human Genetics ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCAUSA
| | - Sonoko Narisawa
- Human Genetics ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCAUSA
| | - Colin Farquharson
- The Royal (Dick) School of Veterinary Studies (RDSVS), The Roslin InstituteUniversity of EdinburghEdinburghUK
| | - Koichi Miyake
- Department of Gene TherapyNippon Medical SchoolTokyoJapan
| | - Brian L Foster
- Division of Biosciences, College of DentistryThe Ohio State UniversityColumbusOHUSA
| | - Jose Luis Millan
- Human Genetics ProgramSanford Burnham Prebys Medical Discovery InstituteLa JollaCAUSA
| |
Collapse
|
23
|
Bessueille L, Kawtharany L, Quillard T, Goettsch C, Briolay A, Taraconat N, Balayssac S, Gilard V, Mebarek S, Peyruchaud O, Duboeuf F, Bouillot C, Pinkerton A, Mechtouff L, Buchet R, Hamade E, Zibara K, Fonta C, Canet-Soulas E, Millan JL, Magne D. Inhibition of alkaline phosphatase impairs dyslipidemia and protects mice from atherosclerosis. Transl Res 2023; 251:2-13. [PMID: 35724933 DOI: 10.1016/j.trsl.2022.06.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 06/10/2022] [Accepted: 06/10/2022] [Indexed: 11/16/2022]
Abstract
Calcium accumulation in atherosclerotic plaques predicts cardiovascular mortality, but the mechanisms responsible for plaque calcification and how calcification impacts plaque stability remain debated. Tissue-nonspecific alkaline phosphatase (TNAP) recently emerged as a promising therapeutic target to block cardiovascular calcification. In this study, we sought to investigate the effect of the recently developed TNAP inhibitor SBI-425 on atherosclerosis plaque calcification and progression. TNAP levels were investigated in ApoE-deficient mice fed a high-fat diet from 10 weeks of age and in plaques from the human ECLAGEN biocollection (101 calcified and 14 non-calcified carotid plaques). TNAP was inhibited in mice using SBI-425 administered from 10 to 25 weeks of age, and in human vascular smooth muscle cells (VSMCs) with MLS-0038949. Plaque calcification was imaged in vivo with 18F-NaF-PET/CT, ex vivo with osteosense, and in vitro with alizarin red. Bone architecture was determined with µCT. TNAP activation preceded and predicted calcification in human and mouse plaques, and TNAP inhibition prevented calcification in human VSMCs and in ApoE-deficient mice. More unexpectedly, TNAP inhibition reduced the blood levels of cholesterol and triglycerides, and protected mice from atherosclerosis, without impacting the skeletal architecture. Metabolomics analysis of liver extracts identified phosphocholine as a substrate of liver TNAP, who's decreased dephosphorylation upon TNAP inhibition likely reduced the release of cholesterol and triglycerides into the blood. Systemic inhibition of TNAP protects from atherosclerosis, by ameliorating dyslipidemia, and preventing plaque calcification.
Collapse
Affiliation(s)
- Laurence Bessueille
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Lynn Kawtharany
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Thibaut Quillard
- CNRS, INSERM, l'institut du thorax, Nantes Université, Nantes, France
| | - Claudia Goettsch
- Department of Internal Medicine I, Cardiology, Medical Faculty, RWTH Aachen University, Aachen Germany
| | - Anne Briolay
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Nirina Taraconat
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III, Paul Sabatier, France
| | - Stéphane Balayssac
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III, Paul Sabatier, France
| | - Véronique Gilard
- Laboratoire des IMRCP, Université de Toulouse, CNRS UMR 5623, Université Toulouse III, Paul Sabatier, France
| | - Saida Mebarek
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | | | | | | | | | - Laura Mechtouff
- Stroke Department, Hospices Civils de Lyon, France; CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | - René Buchet
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France
| | - Eva Hamade
- Laboratory of Cancer Biology and Molecular Immunology, Faculty of Sciences-I, Lebanese University, Beirut, Lebanon
| | - Kazem Zibara
- PRASE and Biology Department, Faculty of Sciences - I, Lebanese University, Beirut, Lebanon
| | - Caroline Fonta
- Brain and Cognition Research Center CerCo, CNRS UMR5549, Université de Toulouse, France
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, INSERM, INRA, INSA Lyon, Université Claude Bernard Lyon 1, Univ Lyon, Lyon, France
| | | | - David Magne
- Université Claude Bernard Lyon 1, UMR CNRS 5246, ICBMS, Univ Lyon, LYON, France.
| |
Collapse
|
24
|
Bernabei I, So A, Busso N, Nasi S. Cartilage calcification in osteoarthritis: mechanisms and clinical relevance. Nat Rev Rheumatol 2023; 19:10-27. [PMID: 36509917 DOI: 10.1038/s41584-022-00875-4] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2022] [Indexed: 12/14/2022]
Abstract
Pathological calcification of cartilage is a hallmark of osteoarthritis (OA). Calcification can be observed both at the cartilage surface and in its deeper layers. The formation of calcium-containing crystals, typically basic calcium phosphate (BCP) and calcium pyrophosphate dihydrate (CPP) crystals, is an active, highly regulated and complex biological process that is initiated by chondrocytes and modified by genetic factors, dysregulated mitophagy or apoptosis, inflammation and the activation of specific cellular-signalling pathways. The links between OA and BCP deposition are stronger than those observed between OA and CPP deposition. Here, we review the molecular processes involved in cartilage calcification in OA and summarize the effects of calcium crystals on chondrocytes, synovial fibroblasts, macrophages and bone cells. Finally, we highlight therapeutic pathways leading to decreased joint calcification and potential new drugs that could treat not only OA but also other diseases associated with pathological calcification.
Collapse
Affiliation(s)
- Ilaria Bernabei
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Alexander So
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.
| | - Nathalie Busso
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| | - Sonia Nasi
- Service of Rheumatology, Department of Musculoskeletal Medicine, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
25
|
Savic I, Farver C, Milovanovic P. Pathogenesis of Pulmonary Calcification and Homologies with Biomineralization in Other Tissues. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1496-1505. [PMID: 36030837 DOI: 10.1016/j.ajpath.2022.07.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/18/2022] [Accepted: 07/29/2022] [Indexed: 06/15/2023]
Abstract
Lungs often present tissue calcifications and even ossifications, both in the context of high or normal serum calcium levels. Precise mechanisms governing lung calcifications have not been explored. Herein, we emphasize recent advances about calcification processes in other tissues (especially vascular and bone calcifications) and discuss potential sources of calcium precipitates in the lungs, involvement of mineralization promoters and crystallization inhibitors, as well as specific cytokine milieu and cellular phenotypes characteristic for lung diseases, which may be involved in pulmonary calcifications. Further studies are necessary to demonstrate the exact mechanisms underlying calcifications in the lungs, document homologies in biomineralization processes between various tissues in physiological and pathologic conditions, and unravel any locally specific characteristics of mineralization processes that may be targeted to reduce or prevent functionally relevant lung calcifications without negatively affecting the skeleton.
Collapse
Affiliation(s)
- Ivana Savic
- Institute of Pathology, University of Belgrade Faculty of Medicine, Belgrade, Serbia
| | - Carol Farver
- Department of Pathology, Cleveland Clinic, Cleveland, Ohio
| | - Petar Milovanovic
- Laboratory of Bone Biology and Bioanthropology, Institute of Anatomy, University of Belgrade Faculty of Medicine, Belgrade, Serbia; Center of Bone Biology, University of Belgrade Faculty of Medicine, Belgrade, Serbia.
| |
Collapse
|
26
|
Soma K, Izumi M, Yamamoto Y, Miyazaki S, Watanabe K. In Vitro and In Vivo Pharmacological Profiles of DS-1211, a Novel Potent, Selective, and Orally Bioavailable Tissue-Nonspecific Alkaline Phosphatase Inhibitor. J Bone Miner Res 2022; 37:2033-2043. [PMID: 36054139 PMCID: PMC9826446 DOI: 10.1002/jbmr.4680] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 08/05/2022] [Accepted: 08/13/2022] [Indexed: 01/11/2023]
Abstract
Inhibition of tissue-nonspecific alkaline phosphatase (TNAP) may prevent ectopic soft tissue calcification by increasing endogenous pyrophosphate (PPi). DS-1211 is a potent and selective novel small molecule TNAP inhibitor with well-characterized pharmacokinetics (PKs) in rodent and monkey. Herein, we report a comprehensive summary of studies establishing the pharmaceutical profile of DS-1211. In vitro studies characterized the mode of inhibition and inhibitory effects of DS-1211 on three human alkaline phosphatase (ALP) isozymes-TNAP, human intestinal ALP, human placental ALP-and on ALP activity across species in mouse, monkey, and human plasma. In vivo PK and pharmacodynamic (PD) effects of a single oral dose of DS-1211 in mice and monkeys were evaluated, including biomarker changes in PPi and pyridoxal 5'-phosphate (PLP). Oral bioavailability (BA) was determined through administration of DS-1211 at a 0.3-mg/kg dose in monkeys. In vitro experiments demonstrated DS-1211 inhibited ALP activity through an uncompetitive mode of action. DS-1211 exhibited TNAP selectivity and potent inhibition of TNAP across species. In vivo studies in mice and monkeys after single oral administration of DS-1211 showed linear PKs, with dose-dependent inhibition of ALP activity and increases in plasma PPi and PLP. Inhibitory effects of DS-1211 were consistent in both mouse and monkey. Mean absolute oral BA was 73.9%. Overall, in vitro and in vivo studies showed DS-1211 is a potent and selective TNAP inhibitor across species. Further in vivo pharmacology studies in ectopic calcification animal models and clinical investigations of DS-1211 in patient populations are warranted. © 2022 Daiichi Sankyo, Inc. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
|
27
|
Uremic mouse model to study vascular calcification and "inflamm-aging". J Mol Med (Berl) 2022; 100:1321-1330. [PMID: 35916902 PMCID: PMC9402761 DOI: 10.1007/s00109-022-02234-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Revised: 05/19/2022] [Accepted: 07/05/2022] [Indexed: 11/05/2022]
Abstract
Calcification and chronic inflammation of the vascular wall is a high-risk factor for cardiovascular mortality, especially in patients with chronic uremia. For the reduction or prevention of rapid disease progression, no specific treatment options are currently available. This study aimed to evaluate an adenine-based uremic mouse model for studying medial vessel calcification and senescence-associated secretory phenotype (SASP) changes of aortic tissue to unravel molecular pathogenesis and provide a model for therapy testing. The dietary adenine administration induced a stable and similar degree of chronic uremia in DBA2/N mice with an increase of uremia blood markers such as blood urea nitrogen, calcium, creatinine, alkaline phosphatase, and parathyroid hormone. Also, renal fibrosis and crystal deposits were detected upon adenine feeding. The uremic condition is related to a moderate to severe medial vessel calcification and subsequent elastin disorganization. In addition, expression of osteogenic markers as Bmp-2 and its transcription factor Sox-9 as well as p21 as senescence marker were increased in uremic mice compared to controls. Pro-inflammatory uremic proteins such as serum amyloid A, interleukin (Il)-1β, and Il-6 increased. This novel model of chronic uremia provides a simple method for investigation of signaling pathways in vascular inflammation and calcification and therefore offers an experimental basis for the development of potential therapeutic intervention studies.
Collapse
|
28
|
Alkaline Phosphatase: An Old Friend as Treatment Target for Cardiovascular and Mineral Bone Disorders in Chronic Kidney Disease. Nutrients 2022; 14:nu14102124. [PMID: 35631265 PMCID: PMC9144546 DOI: 10.3390/nu14102124] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 11/17/2022] Open
Abstract
Alkaline phosphatase (ALP) is an evolutionary conserved enzyme and widely used biomarker in clinical practice. Tissue-nonspecific alkaline phosphatase (TNALP) is one of four human isozymes that are expressed as distinct TNALP isoforms after posttranslational modifications, mainly in bone, liver, and kidney tissues. Beyond the well-known effects on bone mineralization, the bone ALP (BALP) isoforms (B/I, B1, B1x, and B2) are also involved in the pathogenesis of ectopic calcification. This narrative review summarizes the recent clinical investigations and mechanisms that link ALP and BALP to inflammation, metabolic syndrome, vascular calcification, endothelial dysfunction, fibrosis, cardiovascular disease, and mortality. The association between ALP, vitamin K, bone metabolism, and fracture risk in patients with chronic kidney disease (CKD) is also discussed. Recent advances in different pharmacological strategies are highlighted, with the potential to modulate the expression of ALP directly and indirectly in CKD–mineral and bone disorder (CKD-MBD), e.g., epigenetic modulation, phosphate binders, calcimimetics, vitamin D, and other anti-fracture treatments. We conclude that the significant evidence for ALP as a pathogenic factor and risk marker in CKD-MBD supports the inclusion of concrete treatment targets for ALP in clinical guidelines. While a target value below 120 U/L is associated with improved survival, further experimental and clinical research should explore interventional strategies with optimal risk–benefit profiles. The future holds great promise for novel drug therapies modulating ALP.
Collapse
|
29
|
Cardiac Calcifications: Phenotypes, Mechanisms, Clinical and Prognostic Implications. BIOLOGY 2022; 11:biology11030414. [PMID: 35336788 PMCID: PMC8945469 DOI: 10.3390/biology11030414] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/21/2022] [Accepted: 03/07/2022] [Indexed: 12/20/2022]
Abstract
There is a growing interest in arterial and heart valve calcifications, as these contribute to cardiovascular outcome, and are leading predictors of cardiovascular and kidney diseases. Cardiovascular calcifications are often considered as one disease, but, in effect, they represent multifaced disorders, occurring in different milieus and biological phenotypes, following different pathways. Herein, we explore each different molecular process, its relative link with the specific clinical condition, and the current therapeutic approaches to counteract calcifications. Thus, first, we explore the peculiarities between vascular and valvular calcium deposition, as this occurs in different tissues, responds differently to shear stress, has specific etiology and time courses to calcification. Then, we differentiate the mechanisms and pathways leading to hyperphosphatemic calcification, typical of the media layer of the vessel and mainly related to chronic kidney diseases, to those of inflammation, typical of the intima vascular calcification, which predominantly occur in atherosclerotic vascular diseases. Finally, we examine calcifications secondary to rheumatic valve disease or other bacterial lesions and those occurring in autoimmune diseases. The underlying clinical conditions of each of the biological calcification phenotypes and the specific opportunities of therapeutic intervention are also considered and discussed.
Collapse
|
30
|
Huang J, Ralph D, Boraldi F, Quaglino D, Uitto J, Li Q. Inhibition of the DNA Damage Response Attenuates Ectopic Calcification in Pseudoxanthoma Elasticum. J Invest Dermatol 2022; 142:2140-2148.e1. [PMID: 35143822 PMCID: PMC9329183 DOI: 10.1016/j.jid.2022.01.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 12/27/2022]
Abstract
Pseudoxanthoma elasticum (PXE) is a heritable ectopic calcification disorder with multi-organ clinical manifestations. The gene at default, ABCC6, encodes an efflux transporter, ABCC6, which is a new player regulating the homeostasis of inorganic pyrophosphate (PPi), a potent endogenous anti-calcification factor. Previous studies suggested that systemic PPi deficiency is the major, but not the exclusive, cause of ectopic calcification in PXE. In this study, we demonstrate that the DNA damage response (DDR) and poly(ADP-ribose) (PAR) pathways are involved locally in PXE at sites of ectopic calcification. Genetic inhibition of PARP1, the predominant PAR-producing enzyme, showed a 54% reduction of calcification in the muzzle skin in Abcc6-/-Parp1-/- mice, as compared to age-matched Abcc6-/-Parp1+/+ littermates. Subsequently, oral administration of minocycline, an inhibitor of DDR/PAR signaling, resulted in an 86% reduction of calcification in the muzzle skin of Abcc6-/- mice. Minocycline treatment also attenuated the DDR/PAR signaling and reduced calcification of dermal fibroblasts derived from PXE patients. The anti-calcification effect of DDR/PAR inhibition was not accompanied by alterations in plasma PPi concentrations. These results suggest that local DDR/PAR signaling in calcification-prone tissues contributes to PXE pathogenesis, and its inhibition might provide a promising treatment strategy for ectopic calcification in PXE, a currently intractable disease.
Collapse
Affiliation(s)
- Jianhe Huang
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; PXE international Center of Excellence in Research and Clinical Care, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Douglas Ralph
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; PXE international Center of Excellence in Research and Clinical Care, Thomas Jefferson University, Philadelphia, PA 19107, USA; Genetics, Genomics and Cancer Biology Ph.D. Program, Jefferson College of Life Sciences, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Federica Boraldi
- Department of Life Science, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Daniela Quaglino
- Department of Life Science, University of Modena and Reggio Emilia, Modena 41125, Italy
| | - Jouni Uitto
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; PXE international Center of Excellence in Research and Clinical Care, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Qiaoli Li
- Jefferson Institute of Molecular Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA 19107, USA; PXE international Center of Excellence in Research and Clinical Care, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
31
|
Martin-Ventura JL, Roncal C, Orbe J, Blanco-Colio LM. Role of Extracellular Vesicles as Potential Diagnostic and/or Therapeutic Biomarkers in Chronic Cardiovascular Diseases. Front Cell Dev Biol 2022; 10:813885. [PMID: 35155428 PMCID: PMC8827403 DOI: 10.3389/fcell.2022.813885] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 01/10/2022] [Indexed: 12/12/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the first cause of death worldwide. In recent years, there has been great interest in the analysis of extracellular vesicles (EVs), including exosomes and microparticles, as potential mediators of biological communication between circulating cells/plasma and cells of the vasculature. Besides their activity as biological effectors, EVs have been also investigated as circulating/systemic biomarkers in different acute and chronic CVDs. In this review, the role of EVs as potential diagnostic and prognostic biomarkers in chronic cardiovascular diseases, including atherosclerosis (mainly, peripheral arterial disease, PAD), aortic stenosis (AS) and aortic aneurysms (AAs), will be described. Mechanistically, we will analyze the implication of EVs in pathological processes associated to cardiovascular remodeling, with special emphasis in their role in vascular and valvular calcification. Specifically, we will focus on the participation of EVs in calcium accumulation in the pathological vascular wall and aortic valves, involving the phenotypic change of vascular smooth muscle cells (SMCs) or valvular interstitial cells (IC) to osteoblast-like cells. The knowledge of the implication of EVs in the pathogenic mechanisms of cardiovascular remodeling is still to be completely deciphered but there are promising results supporting their potential translational application to the diagnosis and therapy of different CVDs.
Collapse
Affiliation(s)
- Jose Luis Martin-Ventura
- Vascular Research Laboratory, IIS-Fundación Jiménez-Díaz, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- *Correspondence: Jose Luis Martin-Ventura, ; Carmen Roncal,
| | - Carmen Roncal
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
- *Correspondence: Jose Luis Martin-Ventura, ; Carmen Roncal,
| | - Josune Orbe
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
- Laboratory of Atherothrombosis, Program of Cardiovascular Diseases, Cima Universidad de Navarra, Instituto de Investigación Sanitaria de Navarra, IdiSNA, Pamplona, Spain
| | - Luis Miguel Blanco-Colio
- Vascular Research Laboratory, IIS-Fundación Jiménez-Díaz, Madrid, Spain
- CIBER de Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| |
Collapse
|
32
|
Maruyama S, Visser H, Ito T, Limsakun T, Zahir H, Ford D, Tao B, Zamora CA, Stark JG, Chou HS. Phase I studies of the safety, tolerability, pharmacokinetics, and pharmacodynamics of DS-1211, a tissue-nonspecific alkaline phosphatase inhibitor. Clin Transl Sci 2022; 15:967-980. [PMID: 35021269 PMCID: PMC9010257 DOI: 10.1111/cts.13214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/18/2021] [Accepted: 12/02/2021] [Indexed: 01/15/2023] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) hydrolyzes and inactivates inorganic pyrophosphate (PPi), a potent inhibitor of calcification; therefore, TNAP inhibition is a potential target to treat ectopic calcification. These two first-in-human studies evaluated safety, tolerability, pharmacokinetics (PKs), and pharmacodynamics (PDs) of single (SAD) and multiple-ascending doses (MAD) of DS-1211, a TNAP inhibitor. Healthy adults were randomized 6:2 to DS-1211 or placebo, eight subjects per dose cohort. SAD study subjects received one dose of DS-1211 (range, 3-3000 mg) or placebo, whereas MAD study subjects received DS-1211 (range, 10-300 mg) once daily, 150 mg twice daily (b.i.d.), or placebo for 10 days. Primary end points were safety and tolerability. PK and PD assessments included plasma concentrations of DS-1211, alkaline phosphatase (ALP) activity, and TNAP substrates (PPi, pyridoxal 5'-phosphate [PLP], and phosphoethanolamine [PEA]). A total of 56 (DS-1211: n = 42; placebo: n = 14) and 40 (DS-1211: n = 30; placebo: n = 10) subjects enrolled in the SAD and MAD studies, respectively. In both studies, adverse events were mild or moderate and did not increase with dose. PKs of DS-1211 were linear up to 100 mg administered as a single dose and 150 mg b.i.d. administered as a multiple-dose regimen. In multiple dosing, there was minimal accumulation of DS-1211. Increased DS-1211 exposure correlated with dose-dependent ALP inhibition and concomitant increases in PPi, PLP, and PEA. In two phase I studies, DS-1211 appeared safe and well-tolerated. Post-treatment PD assessments were consistent with exposure-dependent TNAP inhibition. These data support further evaluation of DS-1211 for ectopic calcification diseases.
Collapse
Affiliation(s)
| | - Hester Visser
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| | | | | | - Hamim Zahir
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| | - Daniel Ford
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| | - Ben Tao
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| | | | | | - Hubert S Chou
- Daiichi Sankyo, Inc., Basking Ridge, New Jersey, USA
| |
Collapse
|
33
|
Goettsch C, Strzelecka-Kiliszek A, Bessueille L, Quillard T, Mechtouff L, Pikula S, Canet-Soulas E, Luis MJ, Fonta C, Magne D. TNAP as a therapeutic target for cardiovascular calcification: a discussion of its pleiotropic functions in the body. Cardiovasc Res 2022; 118:84-96. [PMID: 33070177 PMCID: PMC8752354 DOI: 10.1093/cvr/cvaa299] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Revised: 09/11/2020] [Accepted: 10/06/2020] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular calcification (CVC) is associated with increased morbidity and mortality. It develops in several diseases and locations, such as in the tunica intima in atherosclerosis plaques, in the tunica media in type 2 diabetes and chronic kidney disease, and in aortic valves. In spite of the wide occurrence of CVC and its detrimental effects on cardiovascular diseases (CVD), no treatment is yet available. Most of CVC involve mechanisms similar to those occurring during endochondral and/or intramembranous ossification. Logically, since tissue-nonspecific alkaline phosphatase (TNAP) is the key-enzyme responsible for skeletal/dental mineralization, it is a promising target to limit CVC. Tools have recently been developed to inhibit its activity and preclinical studies conducted in animal models of vascular calcification already provided promising results. Nevertheless, as its name indicates, TNAP is ubiquitous and recent data indicate that it dephosphorylates different substrates in vivo to participate in other important physiological functions besides mineralization. For instance, TNAP is involved in the metabolism of pyridoxal phosphate and the production of neurotransmitters. TNAP has also been described as an anti-inflammatory enzyme able to dephosphorylate adenosine nucleotides and lipopolysaccharide. A better understanding of the full spectrum of TNAP's functions is needed to better characterize the effects of TNAP inhibition in diseases associated with CVC. In this review, after a brief description of the different types of CVC, we describe the newly uncovered additional functions of TNAP and discuss the expected consequences of its systemic inhibition in vivo.
Collapse
Affiliation(s)
- Claudia Goettsch
- Department of Internal Medicine I, Cardiology, Medical Faculty, RWTH Aachen
University, Aachen, Germany
| | - Agnieszka Strzelecka-Kiliszek
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental
Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Laurence Bessueille
- Institute of Molecular and Supramolecular Chemistry and Biochemistry
(ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment
Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France
| | - Thibaut Quillard
- PHY-OS Laboratory, UMR 1238 INSERM, Université de Nantes, CHU
de Nantes, France
| | - Laura Mechtouff
- Stroke Department, Hospices Civils de Lyon, France
- CREATIS Laboratory, CNRS UMR 5220, Inserm U1044, Université Claude Bernard
Lyon 1, Lyon, France
| | - Slawomir Pikula
- Laboratory of Biochemistry of Lipids, Nencki Institute of Experimental
Biology, 3 Pasteur Street, 02-093 Warsaw, Poland
| | - Emmanuelle Canet-Soulas
- CarMeN Laboratory, Univ Lyon, INSERM, INRA, INSA Lyon, Université Claude
Bernard Lyon 1, Lyon, France
| | - Millan Jose Luis
- Human Genetics Program, Sanford Burnham Prebys Medical Discovery
Institute, La Jolla, CA 92037, USA
| | - Caroline Fonta
- Brain and Cognition Research Center CerCo, CNRS UMR5549, Université de
Toulouse, France
| | - David Magne
- Institute of Molecular and Supramolecular Chemistry and Biochemistry
(ICBMS), UMR CNRS 5246, Université Claude Bernard Lyon 1, Bâtiment
Raulin, 43 Bd du 11 novembre 1918, Lyon 69622 Villeurbanne Cedex, France
| |
Collapse
|
34
|
Sekaran S, Vimalraj S, Thangavelu L. The Physiological and Pathological Role of Tissue Nonspecific Alkaline Phosphatase beyond Mineralization. Biomolecules 2021; 11:1564. [PMID: 34827562 PMCID: PMC8615537 DOI: 10.3390/biom11111564] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/12/2021] [Accepted: 10/12/2021] [Indexed: 12/17/2022] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is a key enzyme responsible for skeletal tissue mineralization. It is involved in the dephosphorylation of various physiological substrates, and has vital physiological functions, including extra-skeletal functions, such as neuronal development, detoxification of lipopolysaccharide (LPS), an anti-inflammatory role, bile pH regulation, and the maintenance of the blood brain barrier (BBB). TNAP is also implicated in ectopic pathological calcification of soft tissues, especially the vasculature. Although it is the crucial enzyme in mineralization of skeletal and dental tissues, it is a logical clinical target to attenuate vascular calcification. Various tools and studies have been developed to inhibit its activity to arrest soft tissue mineralization. However, we should not neglect its other physiological functions prior to therapies targeting TNAP. Therefore, a better understanding into the mechanisms mediated by TNAP is needed for minimizing off targeted effects and aid in the betterment of various pathological scenarios. In this review, we have discussed the mechanism of mineralization and functions of TNAP beyond its primary role of hard tissue mineralization.
Collapse
Affiliation(s)
- Saravanan Sekaran
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai 600 077, Tamil Nadu, India;
| | - Selvaraj Vimalraj
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai 600 077, Tamil Nadu, India;
- Centre for Biotechnology, Anna University, Chennai 600 025, Tamil Nadu, India
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Institute of Medical and Technical Sciences, Saveetha Dental College and Hospitals, Saveetha University, Chennai 600 077, Tamil Nadu, India;
| |
Collapse
|
35
|
Tintut Y, Honda HM, Demer LL. Biomolecules Orchestrating Cardiovascular Calcification. Biomolecules 2021; 11:biom11101482. [PMID: 34680115 PMCID: PMC8533507 DOI: 10.3390/biom11101482] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 09/28/2021] [Accepted: 10/03/2021] [Indexed: 01/12/2023] Open
Abstract
Vascular calcification, once considered a degenerative, end-stage, and inevitable condition, is now recognized as a complex process regulated in a manner similar to skeletal bone at the molecular and cellular levels. Since the initial discovery of bone morphogenetic protein in calcified human atherosclerotic lesions, decades of research have now led to the recognition that the regulatory mechanisms and the biomolecules that control cardiovascular calcification overlap with those controlling skeletal mineralization. In this review, we focus on key biomolecules driving the ectopic calcification in the circulation and their regulation by metabolic, hormonal, and inflammatory stimuli. Although calcium deposits in the vessel wall introduce rupture stress at their edges facing applied tensile stress, they simultaneously reduce rupture stress at the orthogonal edges, leaving the net risk of plaque rupture and consequent cardiac events depending on local material strength. A clinically important consequence of the shared mechanisms between the vascular and bone tissues is that therapeutic agents designed to inhibit vascular calcification may adversely affect skeletal mineralization and vice versa. Thus, it is essential to consider both systems when developing therapeutic strategies.
Collapse
Affiliation(s)
- Yin Tintut
- Department of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA; (Y.T.); (H.M.H.)
- Department of Physiology, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Department of Orthopaedic Surgery, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
| | - Henry M. Honda
- Department of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA; (Y.T.); (H.M.H.)
| | - Linda L. Demer
- Department of Medicine, University of California-Los Angeles, Los Angeles, CA 90095, USA; (Y.T.); (H.M.H.)
- Department of Physiology, University of California-Los Angeles, Los Angeles, CA 90095, USA
- Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, CA 90073, USA
- Department of Bioengineering, University of California-Los Angeles, Los Angeles, CA 90095, USA
- The David Geffen School of Medicine, University of California-Los Angeles, 10833 Le Conte Ave, Los Angeles, CA 90095, USA
- Correspondence: ; Tel.: +1-(310)-206-2677
| |
Collapse
|
36
|
Kinoshita Y, Mohamed FF, Amadeu de Oliveira F, Narisawa S, Miyake K, Foster BL, Millán JL. Gene Therapy Using Adeno-Associated Virus Serotype 8 Encoding TNAP-D 10 Improves the Skeletal and Dentoalveolar Phenotypes in Alpl -/- Mice. J Bone Miner Res 2021; 36:1835-1849. [PMID: 34076297 PMCID: PMC8446309 DOI: 10.1002/jbmr.4382] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/25/2021] [Accepted: 05/28/2021] [Indexed: 12/26/2022]
Abstract
Hypophosphatasia (HPP) is caused by loss-of-function mutations in the ALPL gene that encodes tissue-nonspecific alkaline phosphatase (TNAP), whose deficiency results in the accumulation of extracellular inorganic pyrophosphate (PPi ), a potent mineralization inhibitor. Skeletal and dental hypomineralization characterizes HPP, with disease severity varying from life-threatening perinatal or infantile forms to milder forms that manifest in adulthood or only affect the dentition. Enzyme replacement therapy (ERT) using mineral-targeted recombinant TNAP (Strensiq/asfotase alfa) markedly improves the life span, skeletal phenotype, motor function, and quality of life of patients with HPP, though limitations of ERT include frequent injections due to a short elimination half-life of 2.28 days and injection site reactions. We tested the efficacy of a single intramuscular administration of adeno-associated virus 8 (AAV8) encoding TNAP-D10 to increase the life span and improve the skeletal and dentoalveolar phenotypes in TNAP knockout (Alpl-/- ) mice, a murine model for severe infantile HPP. Alpl-/- mice received 3 × 1011 vector genomes/body of AAV8-TNAP-D10 within 5 days postnatal (dpn). AAV8-TNAP-D10 elevated serum ALP activity and suppressed plasma PPi . Treatment extended life span of Alpl-/- mice, and no ectopic calcifications were observed in the kidneys, aorta, coronary arteries, or brain in the 70 dpn observational window. Treated Alpl-/- mice did not show signs of rickets, including bowing of long bones, enlargement of epiphyses, or fractures. Bone microstructure of treated Alpl-/- mice was similar to wild type, with a few persistent small cortical and trabecular defects. Histology showed no measurable osteoid accumulation but reduced bone volume fraction in treated Alpl-/- mice versus controls. Treated Alpl-/- mice featured normal molar and incisor dentoalveolar tissues, with the exceptions of slightly reduced molar enamel and alveolar bone density. Histology showed the presence of cementum and normal periodontal ligament attachment. These results support gene therapy as a promising alternative to ERT for the treatment of HPP. © 2021 The Authors. Journal of Bone and Mineral Research published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research (ASBMR).
Collapse
Affiliation(s)
- Yuka Kinoshita
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Fatma F Mohamed
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - Flavia Amadeu de Oliveira
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Sonoko Narisawa
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Koichi Miyake
- Department of Biochemistry and Molecular Biology, Nippon Medical School, Tokyo, Japan
| | - Brian L Foster
- Division of Biosciences, College of Dentistry, The Ohio State University, Columbus, OH, USA
| | - José Luis Millán
- Sanford Children's Health Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
37
|
Chronic Kidney Disease-Induced Arterial Media Calcification in Rats Prevented by Tissue Non-Specific Alkaline Phosphatase Substrate Supplementation Rather Than Inhibition of the Enzyme. Pharmaceutics 2021; 13:pharmaceutics13081138. [PMID: 34452102 PMCID: PMC8399849 DOI: 10.3390/pharmaceutics13081138] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2021] [Revised: 07/02/2021] [Accepted: 07/19/2021] [Indexed: 11/30/2022] Open
Abstract
Patients with chronic kidney disease (CKD) suffer from arterial media calcification and a disturbed bone metabolism. Tissue-nonspecific alkaline phosphatase (TNAP) hydrolyzes the calcification inhibitor pyrophosphate (PPi) into inorganic phosphate (Pi) and thereby stimulates arterial media calcification as well as physiological bone mineralization. This study investigates whether the TNAP inhibitor SBI-425, PPi or the combination of both inhibit arterial media calcification in an 0.75% adenine rat model of CKD. Treatments started with the induction of CKD, including (i) rats with normal renal function (control diet) treated with vehicle and CKD rats treated with either (ii) vehicle, (iii) 10 mg/kg/day SBI-425, (iv) 120 µmol/kg/day PPi and (v) 120 µmol/kg/day PPi and 10 mg/kg/day SBI-425. All CKD groups developed a stable chronic renal failure reflected by hyperphosphatemia, hypocalcemia and high serum creatinine levels. CKD induced arterial media calcification and bone metabolic defects. All treatments, except for SBI-425 alone, blocked CKD-related arterial media calcification. More important, SBI-425 alone and in combination with PPi increased osteoid area pointing to a less efficient bone mineralization. Clearly, potential side effects on bone mineralization will need to be assessed in any clinical trial aimed at modifying the Pi/PPi ratio in CKD patients who already suffer from a compromised bone status.
Collapse
|
38
|
Kim SH, Monticone RE, McGraw KR, Wang M. Age-associated proinflammatory elastic fiber remodeling in large arteries. Mech Ageing Dev 2021; 196:111490. [PMID: 33839189 PMCID: PMC8154723 DOI: 10.1016/j.mad.2021.111490] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 03/24/2021] [Accepted: 04/05/2021] [Indexed: 12/12/2022]
Abstract
Elastic fibers are the main components of the extracellular matrix of the large arterial wall. Elastic fiber remodeling is an intricate process of synthesis and degradation of the core elastin protein and microfibrils accompanied by the assembly and disassembly of accessory proteins. Age-related morphological, structural, and functional proinflammatory remodeling within the elastic fiber has a profound effect upon the integrity, elasticity, calcification, amyloidosis, and stiffness of the large arterial wall. An age-associated increase in arterial stiffness is a major risk factor for the pathogenesis of diseases of the large arteries such as hypertensive and atherosclerotic vasculopathy. This mini review is an update on the key molecular, cellular, functional, and structural mechanisms of elastic fiber proinflammatory remodeling in large arteries with aging. Targeting structural and functional integrity of the elastic fiber may be an effective approach to impede proinflammatory arterial remodeling with advancing age.
Collapse
Affiliation(s)
- Soo Hyuk Kim
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Robert E Monticone
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Kimberly R McGraw
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA
| | - Mingyi Wang
- Laboratory of Cardiovascular Science, Intramural Research Program, National Institution on Aging, National Institutes of Health, Biomedical Research Center (BRC), 251 Bayview Boulevard, Baltimore, MD, 21224, USA.
| |
Collapse
|
39
|
Liu X, Chen A, Liang Q, Yang X, Dong Q, Fu M, Wang S, Li Y, Ye Y, Lan Z, Chen Y, Ou J, Yang P, Lu L, Yan J. Spermidine inhibits vascular calcification in chronic kidney disease through modulation of SIRT1 signaling pathway. Aging Cell 2021; 20:e13377. [PMID: 33969611 PMCID: PMC8208796 DOI: 10.1111/acel.13377] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 03/31/2021] [Accepted: 04/18/2021] [Indexed: 12/20/2022] Open
Abstract
Vascular calcification is a common pathologic condition in patients with chronic kidney disease (CKD) and aging individuals. It has been established that vascular calcification is a gene‐regulated biological process resembling osteogenesis involving osteogenic differentiation. However, there is no efficient treatment available for vascular calcification so far. The natural polyamine spermidine has been demonstrated to increase life span and protect against cardiovascular disease. It is unclear whether spermidine supplementation inhibits vascular calcification in CKD. Alizarin red staining and quantification of calcium content showed that spermidine treatment markedly reduced mineral deposition in both rat and human vascular smooth muscle cells (VSMCs) under osteogenic conditions. Additionally, western blot analysis revealed that spermidine treatment inhibited osteogenic differentiation of rat and human VSMCs. Moreover, spermidine treatment remarkably attenuated calcification of rat and human arterial rings ex vivo and aortic calcification in rats with CKD. Furthermore, treatment with spermidine induced the upregulation of Sirtuin 1 (SIRT1) in VSMCs and resulted in the downregulation of endoplasmic reticulum (ER) stress signaling components, such as activating transcription factor 4 (ATF4) and CCAAT/enhancer‐binding protein homologous protein (CHOP). Both pharmacological inhibition of SIRT1 by SIRT1 inhibitor EX527 and knockdown of SIRT1 by siRNA markedly blocked the inhibitory effect of spermidine on VSMC calcification. Consistently, EX527 abrogated the inhibitory effect of spermidine on aortic calcification in CKD rats. We for the first time demonstrate that spermidine alleviates vascular calcification in CKD by upregulating SIRT1 and inhibiting ER stress, and this may develop a promising therapeutic treatment to ameliorate vascular calcification in CKD.
Collapse
Affiliation(s)
- Xiaoyu Liu
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - An Chen
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Qingchun Liang
- Department of Anesthesiology The Third Affiliated Hospital Southern Medical University Guangzhou China
| | - Xiulin Yang
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Qianqian Dong
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Mingwei Fu
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Siyi Wang
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Yining Li
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Yuanzhi Ye
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Zirong Lan
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Yanting Chen
- Department of Pathophysiolgy Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Jing‐Song Ou
- Division of Cardiac Surgery The First Affiliated Hospital Sun Yat‐Sen University Guangzhou China
| | - Pingzhen Yang
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| | - Lihe Lu
- Department of Pathophysiolgy Zhongshan School of Medicine Sun Yat‐Sen University Guangzhou China
| | - Jianyun Yan
- Department of Cardiology Laboratory of Heart Center Heart Center Zhujiang Hospital Southern Medical University Guangzhou China
- Guangdong Provincial Biomedical Engineering Technology Research Center for Cardiovascular Disease Guangzhou China
- Sino‐Japanese Cooperation Platform for Translational Research in Heart Failure Guangzhou China
| |
Collapse
|
40
|
Role of Uremic Toxins in Early Vascular Ageing and Calcification. Toxins (Basel) 2021; 13:toxins13010026. [PMID: 33401534 PMCID: PMC7824162 DOI: 10.3390/toxins13010026] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 12/14/2022] Open
Abstract
In patients with advanced chronic kidney disease (CKD), the accumulation of uremic toxins, caused by a combination of decreased excretion secondary to reduced kidney function and increased generation secondary to aberrant expression of metabolite genes, interferes with different biological functions of cells and organs, contributing to a state of chronic inflammation and other adverse biologic effects that may cause tissue damage. Several uremic toxins have been implicated in severe vascular smooth muscle cells (VSMCs) changes and other alterations leading to vascular calcification (VC) and early vascular ageing (EVA). The above mentioned are predominant clinical features of patients with CKD, contributing to their exceptionally high cardiovascular mortality. Herein, we present an update on pathophysiological processes and mediators underlying VC and EVA induced by uremic toxins. Moreover, we discuss their clinical impact, and possible therapeutic targets aiming at preventing or ameliorating the harmful effects of uremic toxins on the vasculature.
Collapse
|
41
|
Losurdo F, Ferraresi R, Ucci A, Zanetti A, Clerici G, Zambon A. Association of infrapopliteal medial arterial calcification with lower-limb amputations in high-risk patients: A systematic review and meta-analysis. Vasc Med 2020; 26:164-173. [PMID: 33375914 DOI: 10.1177/1358863x20979738] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
Medial arterial calcification (MAC) is a known risk factor for cardiovascular morbidity. The association between vascular calcifications and poor outcome in several vascular districts suggest that infrapopliteal MAC could be a risk factor for lower-limb amputation (LLA). This study's objective is to review the available literature focusing on the association between infrapopliteal MAC and LLA in high-risk patients. The PubMed and Embase databases were systematically searched. We selected original studies reporting the association between infrapopliteal MAC and LLAs in patients with diabetes and/or peripheral artery disease (PAD). Estimates were pooled using either a fixed-effects or a random-effects model meta-analysis. Heterogeneity was evaluated using the Q and I2 statistics. Publication bias was investigated with a funnel plot and Egger test. The trim-and-fill method was designed to estimate the possibly missing studies. Influence analysis was conducted to search studies influencing the final result. Test of moderators was used to compare estimates in good versus non-good-quality studies. Fifteen articles satisfied the selection criteria (n = 6489; median follow-up: 36 months). MAC was significantly associated with LLAs (pooled adjusted risk ratio (RR): 2.27; 95% CI: 1.89-2.74; I2 = 25.3%, Q-test: p = 0.17). This association was kept in the subgroup of patients with diabetes (RR: 2.37; 95% CI: 1.76-3.20) and patients with PAD (RR: 2.48; 95% CI: 1.72-3.58). The association was maintained if considering as outcome only major amputations (RR: 2.11; 95% CI: 1.46-3.06). Our results show that infrapopliteal MAC is associated with LLAs, thus suggesting MAC as a possible new marker of the at-risk limb.
Collapse
Affiliation(s)
- Fabrizio Losurdo
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy.,Diabetic Foot Outpatient Clinic, ASST Bergamo-Est, P.O.T. 'F.M. Passi', Calcinate, BG, Italy
| | - Roberto Ferraresi
- Centro per la cura del Piede Diabetico, Clinica San Carlo, Paderno Dugnano, MI, Italy
| | - Alessandro Ucci
- Vascular Surgery, University Hospital of Parma, Parma, Italy
| | - Anna Zanetti
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy
| | - Giacomo Clerici
- Centro per la cura del Piede Diabetico, Clinica San Carlo, Paderno Dugnano, MI, Italy
| | - Antonella Zambon
- Department of Statistics and Quantitative Methods, University of Milano-Bicocca, Milan, Italy.,Istituto Auxologico Italiano, IRCCS, Biostatistics Unit, Milan, Italy
| |
Collapse
|
42
|
Tissue-Nonspecific Alkaline Phosphatase-A Gatekeeper of Physiological Conditions in Health and a Modulator of Biological Environments in Disease. Biomolecules 2020; 10:biom10121648. [PMID: 33302551 PMCID: PMC7763311 DOI: 10.3390/biom10121648] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 11/30/2020] [Accepted: 12/05/2020] [Indexed: 12/15/2022] Open
Abstract
Tissue-nonspecific alkaline phosphatase (TNAP) is a ubiquitously expressed enzyme that is best known for its role during mineralization processes in bones and skeleton. The enzyme metabolizes phosphate compounds like inorganic pyrophosphate and pyridoxal-5′-phosphate to provide, among others, inorganic phosphate for the mineralization and transportable vitamin B6 molecules. Patients with inherited loss of function mutations in the ALPL gene and consequently altered TNAP activity are suffering from the rare metabolic disease hypophosphatasia (HPP). This systemic disease is mainly characterized by impaired bone and dental mineralization but may also be accompanied by neurological symptoms, like anxiety disorders, seizures, and depression. HPP characteristically affects all ages and shows a wide range of clinical symptoms and disease severity, which results in the classification into different clinical subtypes. This review describes the molecular function of TNAP during the mineralization of bones and teeth, further discusses the current knowledge on the enzyme’s role in the nervous system and in sensory perception. An additional focus is set on the molecular role of TNAP in health and on functional observations reported in common laboratory vertebrate disease models, like rodents and zebrafish.
Collapse
|
43
|
Laurain A, Rubera I, Duranton C, Rutsch F, Nitschke Y, Ray E, Vido S, Sicard A, Lefthériotis G, Favre G. Alkaline Phosphatases Account for Low Plasma Levels of Inorganic Pyrophosphate in Chronic Kidney Disease. Front Cell Dev Biol 2020; 8:586831. [PMID: 33425894 PMCID: PMC7793922 DOI: 10.3389/fcell.2020.586831] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Accepted: 11/16/2020] [Indexed: 01/19/2023] Open
Abstract
Introduction Patients on dialysis and kidney transplant recipients (KTR) present the syndrome of mineral and bone disorders (MBD), which share common traits with monogenic calcifying diseases related to disturbances of the purinergic system. Low plasma levels of inorganic pyrophosphate (PPi) and ectopic vascular calcifications belong to these two conditions. This suggests that the purinergic system may be altered in chronic kidney disease with MBD. Therefore, we perform a transversal pilot study in order to compare the determinants of PPi homeostasis and the plasma levels of PPi in patients on dialysis, in KTR and in healthy people. Patients and Methods We included 10 controls, 10 patients on maintenance dialysis, 10 early KTR 3 ± 1 months after transplantation and nine late KTR 24 ± 3 months after transplantation. We measured aortic calcifications, plasma and urine levels of PPi, the renal fractional excretion of PPi (FePPi), nucleoside triphosphate hydrolase (NPP) and ALP activities in plasma. Correlations and comparisons were assessed with non-parametric tests. Results Low PPi was found in patients on dialysis [1.11 (0.88–1.35), p = 0.004], in early KTR [0.91 (0.66–0.98), p = 0.0003] and in late KTR [1.16 (1.07–1.45), p = 0.02] compared to controls [1.66 (1.31–1.72) μmol/L]. Arterial calcifications were higher in patients on dialysis than in controls [9 (1–75) vs. 399 (25–526) calcium score/cm2, p < 0.05]. ALP activity was augmented in patients on dialysis [113 (74–160), p = 0.01] and in early KTR [120 (84–142), p = 0.002] compared to controls [64 (56–70) UI/L]. The activity of NPP and FePPi were not different between groups. ALP activity was negatively correlated with PPi (r = −0.49, p = 0.001). Discussion Patients on dialysis and KTR have low plasma levels of PPi, which are partly related to high ALP activity, but neither to low NPP activity, nor to increased renal excretion of PPi. Further work is necessary to explore comprehensively the purinergic system in chronic kidney disease.
Collapse
Affiliation(s)
- Audrey Laurain
- Faculty of Medicine, Côte d'Azur University, Nice, France.,UMR 7073, Laboratory of Physiology and Molecular Medicine (LP2M), Centre National de la Recherche Scientifique, Nice, France.,Nephrology Department, University Hospital, Nice, France
| | - Isabelle Rubera
- UMR 7073, Laboratory of Physiology and Molecular Medicine (LP2M), Centre National de la Recherche Scientifique, Nice, France
| | - Christophe Duranton
- UMR 7073, Laboratory of Physiology and Molecular Medicine (LP2M), Centre National de la Recherche Scientifique, Nice, France
| | - Frank Rutsch
- Department of General Pediatrics, Muenster University Children's Hospital, Muenster, Germany
| | - Yvonne Nitschke
- Department of General Pediatrics, Muenster University Children's Hospital, Muenster, Germany
| | - Elodie Ray
- Department of Vascular Medicine and Surgery, University Hospital, Nice, France
| | - Sandor Vido
- Nephrology Department, University Hospital, Nice, France
| | - Antoine Sicard
- Faculty of Medicine, Côte d'Azur University, Nice, France.,Nephrology Department, University Hospital, Nice, France
| | - Georges Lefthériotis
- Faculty of Medicine, Côte d'Azur University, Nice, France.,UMR 7073, Laboratory of Physiology and Molecular Medicine (LP2M), Centre National de la Recherche Scientifique, Nice, France.,Department of Vascular Medicine and Surgery, University Hospital, Nice, France
| | - Guillaume Favre
- Faculty of Medicine, Côte d'Azur University, Nice, France.,UMR 7073, Laboratory of Physiology and Molecular Medicine (LP2M), Centre National de la Recherche Scientifique, Nice, France.,Nephrology Department, University Hospital, Nice, France
| |
Collapse
|
44
|
Zimmermann H. History of ectonucleotidases and their role in purinergic signaling. Biochem Pharmacol 2020; 187:114322. [PMID: 33161020 DOI: 10.1016/j.bcp.2020.114322] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 11/03/2020] [Accepted: 11/03/2020] [Indexed: 12/15/2022]
Abstract
Ectonucleotidases are key for purinergic signaling. They control the duration of activity of purinergic receptor agonists. At the same time, they produce hydrolysis products as additional ligands of purinergic receptors. Due to the considerable diversity of enzymes, purinergic receptor ligands and purinergic receptors, deciphering the impact of extracellular purinergic receptor control has become a challenge. The first group of enzymes described were the alkaline phosphatases - at the time not as nucleotide-metabolizing but as nonspecific phosphatases. Enzymes now referred to as nucleoside triphosphate diphosphohydrolases and ecto-5'-nucleotidase were the first and only nucleotide-specific ectonucleotidases identified. And they were the first group of enzymes related to purinergic signaling. Additional research brought to light a surprising number of ectoenzymes with broad substrate specificity, which can also hydrolyze nucleotides. This short overview traces the development of the field and briefly highlights important results and benefits for therapies of human diseases achieved within nearly a century of investigations.
Collapse
Affiliation(s)
- Herbert Zimmermann
- Goethe University, Institute of Cell Biology and Neuroscience, Max-von-Laue-Str. 13, 60438 Frankfurt am Main, Germany.
| |
Collapse
|
45
|
Extracellular Nucleotides Regulate Arterial Calcification by Activating Both Independent and Dependent Purinergic Receptor Signaling Pathways. Int J Mol Sci 2020; 21:ijms21207636. [PMID: 33076470 PMCID: PMC7589647 DOI: 10.3390/ijms21207636] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/09/2020] [Accepted: 10/12/2020] [Indexed: 01/02/2023] Open
Abstract
Arterial calcification, the deposition of calcium-phosphate crystals in the extracellular matrix, resembles physiological bone mineralization. It is well-known that extracellular nucleotides regulate bone homeostasis raising an emerging interest in the role of these molecules on arterial calcification. The purinergic independent pathway involves the enzymes ecto-nucleotide pyrophosphatase/phosphodiesterases (NPPs), ecto-nucleoside triphosphate diphosphohydrolases (NTPDases), 5′-nucleotidase and alkaline phosphatase. These regulate the production and breakdown of the calcification inhibitor—pyrophosphate and the calcification stimulator—inorganic phosphate, from extracellular nucleotides. Maintaining ecto-nucleotidase activities in a well-defined range is indispensable as enzymatic hyper- and hypo-expression has been linked to arterial calcification. The purinergic signaling dependent pathway focusses on the activation of purinergic receptors (P1, P2X and P2Y) by extracellular nucleotides. These receptors influence arterial calcification by interfering with the key molecular mechanisms underlying this pathology, including the osteogenic switch and apoptosis of vascular cells and possibly, by favoring the phenotypic switch of vascular cells towards an adipogenic phenotype, a recent, novel hypothesis explaining the systemic prevention of arterial calcification. Selective compounds influencing the activity of ecto-nucleotidases and purinergic receptors, have recently been developed to treat arterial calcification. However, adverse side-effects on bone mineralization are possible as these compounds reasonably could interfere with physiological bone mineralization.
Collapse
|
46
|
Calcinosis in Systemic Sclerosis: Updates in Pathophysiology, Evaluation, and Treatment. Curr Rheumatol Rep 2020; 22:73. [DOI: 10.1007/s11926-020-00951-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
47
|
Opdebeeck B, Neven E, Millán JL, Pinkerton AB, D'Haese PC, Verhulst A. Pharmacological TNAP inhibition efficiently inhibits arterial media calcification in a warfarin rat model but deserves careful consideration of potential physiological bone formation/mineralization impairment. Bone 2020; 137:115392. [PMID: 32360899 PMCID: PMC8406684 DOI: 10.1016/j.bone.2020.115392] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/23/2020] [Accepted: 04/24/2020] [Indexed: 12/25/2022]
Abstract
Arterial media calcification is frequently seen in elderly and patients with chronic kidney disease (CKD), diabetes and osteoporosis. Pyrophosphate is a well-known calcification inhibitor that binds to nascent hydroxyapatite crystals and prevents further incorporation of inorganic phosphate into these crystals. However, the enzyme tissue-nonspecific alkaline phosphatase (TNAP), which is expressed in calcified arteries, degrades extracellular pyrophosphate into phosphate ions, by which pyrophosphate loses its ability to block vascular calcification. Here, we aimed to evaluate whether pharmacological TNAP inhibition is able to prevent the development of arterial calcification in a rat model of warfarin-induced vascular calcification. To investigate the effect of the pharmacological TNAP inhibitor SBI-425 on vascular calcification and bone metabolism, a 0.30% warfarin rat model was used. Warfarin exposure resulted in distinct calcification in the aorta and peripheral arteries. Daily administration of the TNAP inhibitor SBI-425 (10 mg/kg/day) for 7 weeks significantly reduced vascular calcification as indicated by a significant decrease in calcium content in the aorta (vehicle 3.84 ± 0.64 mg calcium/g wet tissue vs TNAP inhibitor 0.70 ± 0.23 mg calcium/g wet tissue) and peripheral arteries and a distinct reduction in area % calcification on Von Kossa stained aortic sections as compared to vehicle. Administration of SBI-425 resulted in decreased bone formation rate and mineral apposition rate, and increased osteoid maturation time and this without significant changes in osteoclast- and eroded perimeter. Administration of TNAP inhibitor SBI-425 significantly reduced the calcification in the aorta and peripheral arteries of a rat model of warfarin-induced vascular calcification. However, suppression of TNAP activity should be limited in order to maintain adequate physiological bone mineralization.
Collapse
Affiliation(s)
- Britt Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - Ellen Neven
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| | - José Luis Millán
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States of America
| | - Anthony B Pinkerton
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA 92037, United States of America
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium.
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Belgium
| |
Collapse
|
48
|
Opdebeeck B, D'Haese PC, Verhulst A. Inhibition of tissue non-specific alkaline phosphatase; a novel therapy against arterial media calcification? J Pathol 2020; 250:248-250. [PMID: 31859361 DOI: 10.1002/path.5377] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/16/2019] [Indexed: 11/10/2022]
Abstract
Arterial media calcification refers to ectopic mineralization in the arterial wall and favors arterial stiffness and cardiovascular events. Patients with chronic kidney disease (CKD), diabetes, or osteoporosis are highly vulnerable to the development of arterial media calcifications. Tissue non-specific alkaline phosphatase (TNAP) is upregulated in calcified arteries and plays a key role in the degradation of the calcification inhibitor pyrophosphate into inorganic phosphate ions. A recent study published in The Journal of Pathology showed that an oral dosage of 10 or 30 mg/kg/day SBI-425, a selective TNAP inhibitor, inhibited the development of arterial media calcification in mice with CKD, without affecting bone mineralization. Their results indicated that SBI-425 is an effective and safe treatment for arterial media calcification. However, additional studies regarding the effect of TNAP-inhibitor SBI-425 on the progression and even the reversion of pre-existing pathological arterial media calcifications without affecting physiological bone mineralization are deserved. Furthermore, investigating the extent to which SBI-425 inhibits arterial calcification in a non-CKD context would be of particular interest to treat this comorbidity in diabetes and osteoporosis patients. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Britt Opdebeeck
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Patrick C D'Haese
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| | - Anja Verhulst
- Laboratory of Pathophysiology, Department of Biomedical Sciences, University of Antwerp, Antwerp, Belgium
| |
Collapse
|