Next Article in Journal
Gut Molecules in Cardiometabolic Diseases: The Mechanisms behind the Story
Next Article in Special Issue
Lansoprazole Increases Inorganic Pyrophosphate in Patients with Pseudoxanthoma Elasticum: A Double-Blind, Randomized, Placebo-Controlled Crossover Trial
Previous Article in Journal
Hormonal Gut–Brain Signaling for the Treatment of Obesity
Previous Article in Special Issue
p53 Regulates Mitochondrial Dynamics in Vascular Smooth Muscle Cell Calcification
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Zebrafish Models to Study Ectopic Calcification and Calcium-Associated Pathologies

by
João M. A. Santos
1,*,
Vincent Laizé
1,
Paulo J. Gavaia
1,2,
Natércia Conceição
1,2,3 and
M. Leonor Cancela
1,2,3,*
1
Centre of Marine Sciences, University of Algarve, 8005-139 Faro, Portugal
2
Faculty of Medicine and Biomedical Sciences, University of Algarve, 8005-139 Faro, Portugal
3
Algarve Biomedical Center, University of Algarve, 8005-139 Faro, Portugal
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(4), 3366; https://doi.org/10.3390/ijms24043366
Submission received: 9 December 2022 / Revised: 20 January 2023 / Accepted: 1 February 2023 / Published: 8 February 2023

Abstract

:
Ectopic calcification refers to the pathological accumulation of calcium ions in soft tissues and is often the result of a dysregulated action or disrupted function of proteins involved in extracellular matrix mineralization. While the mouse has traditionally been the go-to model organism for the study of pathologies associated with abnormal calcium deposition, many mouse mutants often have exacerbated phenotypes and die prematurely, limiting the understanding of the disease and the development of effective therapies. Since the mechanisms underlying ectopic calcification share some analogy with those of bone formation, the zebrafish (Danio rerio)—a well-established model for studying osteogenesis and mineralogenesis—has recently gained momentum as a model to study ectopic calcification disorders. In this review, we outline the mechanisms of ectopic mineralization in zebrafish, provide insights into zebrafish mutants that share phenotypic similarities with human pathological mineralization disorders, list the compounds capable of rescuing mutant phenotypes, and describe current methods to induce and characterize ectopic calcification in zebrafish.

1. Introduction

Calcium is central to human physiology, being a major player in the homeostasis of mineralized tissues (e.g., bone and teeth), but also of soft tissues, where it is involved in blood clotting, muscle contraction, nerve function, and regulation of heart rhythm, among other processes [1,2]. Under pathological conditions, calcium may accumulate in soft tissues increasing their stiffness and affecting their function [3]. The pathological accumulation of calcium ions in soft tissues is defined as ectopic calcification and, while all soft tissues can be affected, vascular and cartilaginous tissues are among those most prone to calcium deposition leading to a pathological phenotype [4,5,6].
Ectopic calcification was long thought to be an aging-dependent disorder, but significant data has recently come forward indicating that it may also result from dysfunctional anti-mineralizing proteins [7,8]. In the latter case, circulating calcium in excess may precipitate, deposit, and accumulate in soft tissues. Deposits become progressively more crystalline and insoluble, hardening the tissue, and affecting its function [9]. Evidence of ectopic calcification at a young age is usually associated with a pathological condition, such as chronic kidney disease or autoimmune diseases [10,11].
Two distinct forms of ectopic calcifications have been described in humans. Metastatic calcification is characterized by an accumulation of calcium in soft tissues following an increase in serum levels, while dystrophic calcification results from an accumulation of calcium at pathologically altered sites [12]. Despite a better understanding of the pathologies behind disorders characterized by ectopic calcification, there is still a lack of efficient therapies capable of effectively preventing and treating ectopic mineral deposition [13].
Animal models of ectopic calcification are critical to better understand the disease pathophysiology and allow the screening of novel therapeutics [4]. While the mouse has often been the model of choice to investigate the mechanisms of ectopic calcification and has greatly contributed to a better understanding of calcium-associated pathologies [4,7], it has some issues that have prevented further advances in this field. When compared to many single-gene human diseases, loss-of-function mouse models often show exacerbated phenotypes and die prematurely, which limits our understanding of the pathological mechanisms [14,15]. For example, the matrix Gla protein (MGP) is a vitamin K-dependent protein present in the extracellular matrix and a well-documented inhibitor of ectopic mineralization, whose dysfunction is associated with the development of Keutel syndrome [16]. While only a few patients show vascular calcifications, the Mgp null mice die within the first two months of age due to extensive mineralization of arteries and subsequent rupture [17].
The zebrafish (Danio rerio) has emerged as a valuable model to gain insights into the mechanisms underlying the development of human diseases as it brings intrinsic advantages over mammalian models. Among those, it is worth mentioning that (i) a single breeding event can produce hundreds of embryos for testing, allowing a robust statistical analysis; (ii) embryos and larvae are optically clear and develop externally, allowing easy and direct visualization of the developmental process; (iii) amenability to genetic manipulation and availability of tools for genetic screening and editing; and (iv) a fast development throughout adulthood, allowing genetic experiments to be performed within a short period [18,19]. Zebrafish has gained momentum in the last decades for the study of bone disorders [20]—with tools and methodologies available to assess osteogenic and mineralogenic effects [21]—and, in the present review, we provide evidence that zebrafish is also a relevant model to study pathologies associated with tissue calcification. Based on the analysis of scientific articles referenced in the PubMed database “pubmed.ncbi.nlm.nih.gov (accessed on 7 December 2022)”, we also outline the current tools, techniques, and mutants available to gain insights into mechanisms of ectopic calcification and calcium-associated pathologies, and list compounds with the capacity to rescue mutant phenotypes and contribute to the next-generation therapeutics to prevent or treat ectopic calcification disorders.

2. Zebrafish In Vivo Models to Study Ectopic Mineralization Disorders

The successful modeling of ectopic mineralization disorders in zebrafish implies the conservation throughout vertebrate evolution of (i) the mechanisms underlying calcium metabolism, (ii) the sites of calcium-phosphate crystal (predominantly in the form of hydroxyapatite) deposition, and (iii) a phenotypic response that mimics human mineralization disorders.
Although calcium intake occurs in the kidney and intestine in mammals and the gills and yolk skin (during development) in fish, the major principles of calcium transport and hormonal control were found to be conserved from zebrafish to mammals [22,23]. For instance, a loss-of-function mutation in zebrafish Trpv6—a calcium channel expressed in the kidney and intestinal epithelia and central to calcium absorption in humans [24]—generated a 68% reduction in systemic calcium content and significantly inhibited calcium uptake in the yolk sac and gills in zebrafish embryos [25]. Similarly, adult zebrafish fed a diet rich in cholesterol develop lesions in the intimal layer region of the dorsal aorta and show macrophage infiltration mimicking the development of atherosclerosis and ectopic calcification observed in humans [12,13]. Similar pathological outcomes were observed in zebrafish mutants with impaired production of proteins important for lipid metabolism, such as the low-density lipoprotein (LDL) receptor [26], the lipoprotein lipase apolipoprotein C2 (APOC2) [27,28], or the cholesterol catabolism liver X receptor (LXR) [29].
Several zebrafish models of Mendelian genetic disorders share phenotypic similarities with the acquired forms of metastatic and dystrophic calcifications and may serve as genetically controlled systems to study human calcification disorders. Recent studies on small teleost fishes have allowed the identification of various genetic factors that contribute to ectopic calcification; they are further discussed below and summarized in Table 1.

2.1. Models of GACI and PXE

Generalized arterial calcification of infancy (GACI)—caused by mutations in the gene Ectonucleotide pyrophosphatase/phosphodiesterase 1 (ENPP1)—and pseudoxanthoma elasticum (PXE)—caused by mutations in the gene ATP binding cassette subfamily C member 6 (ABCC6)—are rare autosomal recessive genetic disorders associated with arterial and cartilage calcification, and ectopic calcifications of elastic fibers, respectively [41,42]. Zebrafish enpp1 mutants were shown to develop ectopic calcification of the skin, cartilage, heart, intracranial space, and notochord sheet, independently of osteoblast activity. They also displayed higher osteoclast activity at sites of ectopic calcification, and bisphosphonates could efficiently rescue associated phenotypes [30]. Zebrafish abcc6a mutants showed defects in vertebral calcification and displayed ectopic calcification in soft tissues [32]. Since Abcc6a is present in the eyes and heart of the zebrafish, extensive calcification of the ocular sclera and Bruch’s membrane and a fibrotic heart were also observed in the abcc6a mutants [33].
Vitamin K was proposed to be central to the pathophysiology of GACI and PXE as patients usually show a reduction in circulating vitamin K, which may be a countermeasure against pathological mineralization [43]. While the administration of vitamin K failed to rescue the ectopic calcification phenotype of Abcc6 knockout mice [44], it restored mineralization levels and lowered the sites of ectopic mineralization in zebrafish abcc6a mutants [33,43]. Interestingly, treatment with warfarin, a vitamin K antagonist, exacerbated the ectopic mineralization phenotype in abcc6a zebrafish mutants [43]. Aberrant mineralization in PXE pathogenesis is partly due to excessive DNA damage. In this regard, zebrafish abcc6a mutants treated with minocycline, a DNA damage response inhibitor, had reduced occurrences of pathological mineralization [45].

2.2. Models of Chronic Kidney Disease

Chronic kidney disease (CKD) patients are likely to suffer from ectopic calcifications due to a dysregulated mineral metabolism and pathological alterations in KLOTHO and Fibroblast Growth Factor (FGF) 23 [46]. Mice deficient for αKlotho or Fgf23 have age-related disorders, including abnormal mineral regulation and ectopic calcification [47,48]. As both mutants share similar phenotypes, a link between the two proteins was established, and αKLOTHO was found to function as a co-receptor of the FGF receptor and to be responsible for activating and controlling the production of FGF23 [49,50]. αKLOTHO and FGF23 are produced in the kidney and bone tissues, respectively, and both are central to mineral homeostasis. Changes in their expression are strongly associated with the development of chronic kidney disease (CKD) [51,52]. Brood stocks of αKlotho and Fgf23 null mice are difficult to maintain as they die by three months of age; this represents a major bottleneck in the use of these murine models [47,48].
In zebrafish, αklotho expression is detected in the adult kidney and fgf23 is continuously expressed in the corpuscles of Stannius, an endocrine gland close to the nephron that contributes to calcium homeostasis, where Fgf23 is responsible for adjusting and regulating calcium metabolism [53,54]. As for the mouse mutants, zebrafish mutants for αklotho and fgf23 have a short lifespan [34]. However, the disease phenotype only occurs at approximately five months of age, later than in the mouse model (i.e., as soon as one month of age). Thus, zebrafish mutants can reach adulthood and reproduce, allowing the maintenance of a mutant brood stock, a major drawback of the mouse model. Both zebrafish mutants display ectopic calcification of the vessels throughout the body, especially in the outflow tract of the heart and the bulbus arteriosus, a pathological calcification likely associated with premature aging, ectopic activation of osteoclast differentiation, and age-associated vascular calcification [34]. Indeed, in vivo studies using αklotho, fgf23, and ennp1 zebrafish mutants have consistently shown an increase in osteoclast activity around mineralized soft tissues, hinting at the existence of osteoclasts that develop as a response to ectopic calcifications [30,34,55,56].

2.3. Model of Primary Familial Brain Calcification

Primary familial brain calcification (PFBC) is a rare progressive neurodegenerative disorder characterized by bilateral brain calcifications and associated with symptoms of dementia, Parkinsonism, and dystonia [57,58]. In 2018, mutations in the gene Myogenesis regulating glycosidase (MYORG) have been linked to the development of autosomal recessive PFBC [59,60]. The central role of Myorg in the disease was confirmed by the observation of irregular bilateral brain calcifications in Myorg knockout mice at nine months of age [58]. Because the development of PFBC in the murine model requires a long period of time, alternative solutions were pursued, and zebrafish provided a timelier model. Zebrafish larvae where myorg expression was knocked-down using morpholinos (Table 1) exhibited multiple calcifications in the brain already at two days post-fertilization [60], demonstrating the suitability of zebrafish morphants over mouse mutants.

2.4. Other Models That Remain to Be Assessed

Mutations in the genes golgin b1 (golgb1) and activin A receptor, type 1 like (acvr1l) have been associated with the development of hyperphosphatemic familial tumoral calcinosis (HFTC) [37] and fibrodysplasia ossificans progressiva (FOP) [39]—rare disorders associated with the development of ectopic calcification [61,62]. While there is no data showing that zebrafish mutants for golgin b1 and acvr1l develop ectopic calcification, it is unknown whether these fish simply failed to do so, or it was not observed or reported as it was not the focus of these studies. So far, mutations in the zebrafish orthologs of many single-gene human diseases—associated with ectopic mineralization disorders—faithfully mimicked disease phenotype and show pathological calcium deposition. Therefore, it is likely that many zebrafish mutants for genes associated with calcium mineralization disorders—whose phenotype has yet to be published—may develop ectopic mineralization phenotypes. Zebrafish mutants that may develop ectopic calcifications, and are therefore promising targets for the development of new models, are summarized in Table 2.

3. Tools to Study Ectopic Calcification in Zebrafish

3.1. Genetic-Based Approaches to Develop Mutant Lines

Most zebrafish models initially used to study ectopic mineralization have been developed through forward genetic approaches. In a screening to identify novel regulators of osteogenesis and bone mineralization, a mutant named dragonfish (dgf) showed ectopic mineralization in the craniofacial and axial skeleton [63]. This mutant was later reported to have altered enpp1 expression and it remains today one of the most used and well-described models of ectopic calcification [30]. The Zebrafish Mutation Project, a large-scale N-ethyl-N-nitrosourea (ENU) mutagenesis project, has generated a mutant archive of over 40,000 alleles covering 60% of zebrafish protein-coding genes, with many mutants yet to be characterized [64]. In relation to this article, four zebrafish mutants for αklotho were identified and are available from zebrafish stock centers. Each mutant has an allele with one point mutant that induces a premature stop in four out of five exons. At this point, only αklothosa18644 studies—targeting exon 3—have been reported and the phenotype closely matches the loss-of-αKlotho function previously reported in zebrafish [35].
Reverse genetics has also been successfully applied to zebrafish. Morpholino antisense oligomers (MOs) are the preferred method of gene knockdown in zebrafish [65,66] and the initial approach taken at generating ectopic calcification in zebrafish models. Injection of abcc6a-specific MOs in zebrafish eggs induced cardiac malformations and developmental defects in 8 dpf morphant larvae similar to those observed in GACI patients [67]. Similarly, myorg-specific MOs induced brain calcification in 2 dpf morphant larvae [40]. CRISPR/Cas9 genome-editing technology has also been successfully applied to zebrafish [68,69] making it possible to phenotype F0 generation zebrafish (founders), also known as crispants, within weeks [70]. As many zebrafish mutants are already available, CRISPR/Cas9 is a promising tool to accelerate the study of ectopic mineralization in situations in which (i) a mutant is not available; (ii) to investigate a gain of function; (iii) and to target important functional domains.

3.2. Techniques for the Detection of Abnormal Calcium Deposition

The detection and quantification of calcium deposition in zebrafish can be achieved through a variety of techniques that are based on the optical detection of changes in luminescence, fluorescence, or absorbance of an organic indicator that specifically binds to calcium ions [71]. Several water-soluble dyes can be used to detect tissue calcification, calcein and alizarin red S—that emit green and red signals, respectively, when bound to calcium-based crystals—have been mostly used in zebrafish [72]. Both dyes can be used in vivo—e.g., live staining without the sacrifice of the animal—allowing the real-time detection of calcium deposition [73]. Repetitive alizarin red S staining performed at low concentrations can be applied to zebrafish without affecting bone growth and mineralization [74]. Furthermore, alizarin red S can be combined with alcian blue, a polyvalent basic dye that stains cartilage in blue, allowing the visualization of calcium deposition within cartilaginous tissues [74]. Calcein and alizarin red S have similar efficiency in staining calcium deposits, thus the choice of using either dye often depends on the usage of transgenic fish models to complement the fluorescent reporter protein. As most transgenic fish models use enhanced green fluorescent protein (eGFP) [75,76], alizarin red S staining is often preferred over calcein.
Von Kossa’s staining is also a very popular method to detect the presence of abnormal deposits of calcium-phosphate crystals in the body. This histological method is based on the transformation of calcium phosphate salts into silver phosphate salts, which undergo photochemical degradation when illuminated with a UV light, leading to the formation of dark silver deposits [77]. While its first usage dates back to more than a century, von Kossa’s staining remains a widely used method to detect the presence of vascular calcification in human tissue samples [78,79].

3.3. Radiographic Methods

Histological techniques used to detect ectopic calcification have limitations associated with the physical sectioning of hard tissues that often result in tissue loss or incomplete tissue sectioning. In addition, visualization of calcium deposition in elongated structures (e.g., blood vessels) and in a single section is extremely difficult. Micro-computed tomography (micro-CT) is commonly used for the 3D imaging and analysis of skeletal structures and other calcified tissues [80,81]; it utilizes X-rays to create cross-sections of a physical object to render a 3D image. Traditional micro-CT scans have been a reliable method for visualization of calcified bone tissues in zebrafish but it has lacked the resolution and tissue contrast to detect ectopic calcification [82]. However, recent advances in micro-CT and high-resolution imaging have reached a point in which direct visualization of nanoscale structures and the detection of ectopic calcification sites are possible [83,84]. Similarly, Raman spectroscopy, which is used to distinguish changes in biomolecules (e.g., minerals) present in tissues [85], has already been applied to the diagnosis of human calcification disorders [86,87] and represents a promising tool for the detection of ectopic calcification in zebrafish embryos, as it allows both the visualization and profiling of calcium deposits across mineralizing and soft tissues [88,89].

3.4. Ectopic Calcification-Inducing Drugs

The identification of compounds that induce phenotypical changes similar to those observed in pathological conditions is of the utmost interest as these compounds allow quick, easy, and cost-effective access to in vitro or in vivo disease models. Although the precise mechanisms that lead to ectopic calcification remain to be better understood, molecules that disrupt the balance between pro- and anti-mineralizing pathways have often been associated with pathological mineralization [90,91].
Vitamin K vitamers are central to bone mineralization through their role in the carboxylation of several vitamin K-dependent bone-related proteins, such as osteocalcin, matrix Gla protein, and Gla-rich protein [92,93,94]. Warfarin and other vitamin K antagonists inhibit the activity of vitamin K epoxide reductase and consenquently the recycling of vitamin K back to its active form. Sodium warfarin has been used in anticoagulation therapy in humans for seven decades [95,96], but long-term treatments have been associated with systemic arterial calcification [97,98], and warfarin is contraindicated during pregnancy as fetal exposure can lead to the development of warfarin embryopathy, a rare condition associated with abnormal bone and cartilage growth [99]. Mice treated with warfarin showed significant cardiovascular calcification that compromised cardiovascular function [100]. In zebrafish, embryonic exposure to warfarin has also been associated with ectopic calcification, and a decrease in γ-glutamyl carboxylase activity and embryonic lethality [101,102]. Long-term exposure of zebrafish to warfarin led to the development of warfarin embryopathy-associated phenotypes [101,103,104]. Similarly, adult zebrafish exposed to 25 mg/L of sodium warfarin for fifteen days developed vascular calcification, further supporting its suitability as an ectopic calcification-inducing drug (Figure 1).

3.5. Screening of Drugs to Rescue Pathological Calcification Disorders

Therapeutic strategies to efficiently counteract the accumulation of calcium ions in soft tissues responsible for ectopic mineralization disorders have yet to be developed. Antiresorptive drugs have been tried, but they were only able to delay and not prevent disease progression [105,106]. The inherent advantages and a similar response to anti-mineralizing compounds (Table 3)—when compared to mouse models—provide the proof-of-concept necessary to validate zebrafish as a fast high-throughput screening model for the identification of novel anti-mineralizing drugs.

4. Conclusions

Ectopic calcification disorders are medical conditions that affect the well-being and life expectancy of many people worldwide and for which therapeutics remain to be developed. To improve the current scenario, the complex mechanisms underlying these disorders must be better understood. Until recently, most of the information regarding pathological calcification was collected from human patients and mouse models, two biological systems with inherent limitations. Zebrafish models have recently emerged with the potential to provide new and fast insights into pathological mechanisms, as they can overcome some of these limitations (e.g., knockout mice being non-viable) and accelerate the collection of data with phenotypic alterations occurring at early stages of development. As for mammalian systems, zebrafish ectopic calcification models develop calcification in most soft tissues, and the detection of sites of ectopic calcification could be detected early during development (as soon as 2 dpf, the period at which zebrafish larvae hatch). Figure 2 summarizes the different zebrafish models available to study mechanisms of ectopic mineralization.
Thanks to the Zebrafish Mutation Project “zmp.buschlab.org (accessed on 7 December 2022)”, there are currently dozens of available mutants with genes associated with calcium mineralization disorders that remain to be analyzed and studied (Table 1) and eggs can be ordered from international repositories such as the Zebrafish International Resource Center (ZIRC), the European Zebrafish Resource Center (EZRC) or the China Zebrafish Resource Center (CZRC). Thanks to advances in gene-editing techniques, methods to induce and detect ectopic mineralization, and currently available mutant and transgenic lines, combined with some fish inherent advantages, the zebrafish has become the new go-to model organism to study the mechanisms of ectopic calcification and calcium-associated pathologies.

Author Contributions

Conceptualization, J.M.A.S., V.L., P.J.G., N.C. and M.L.C.; writing—original draft preparation, J.M.A.S.; writing—review and editing, J.M.A.S., V.L., P.J.G., N.C. and M.L.C.; funding acquisition V.L., P.J.G., N.C. and M.L.C. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the European Joint Program on Rare Diseases (EJP-RD) through the project PhysPath-KS EJPRD2019-290, COST through the actions EuroSoftCalcNet CA16115 and Ocean4Biotech CA18283, by the Portuguese Foundation for Science and Technology (FCT) through the projects UIDB/04326/2020, UIDP/04326/2020, and LA/P/0101/2020 and by the operational programmes CRESC Algarve 2020 and COMPETE 2020 through the project EMBRC.PT ALG-01-0145-FEDER-02212.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Beto, J.A. The role of calcium in human aging. Clin. Nutr. Res. 2015, 4, 1–8. [Google Scholar] [CrossRef] [PubMed]
  2. Berchtold, M.W.; Brinkmeier, H.; Muntener, M. Calcium ion in skeletal muscle: Its crucial role for muscle function, plasticity, and disease. Physiol. Rev. 2000, 80, 1215–1265. [Google Scholar] [CrossRef] [PubMed]
  3. Giachelli, C.M. Ectopic calcification: Gathering hard facts about soft tissue mineralization. Am. J. Pathol. 1999, 154, 671–675. [Google Scholar] [CrossRef] [PubMed]
  4. Li, Q.; Uitto, J. Mineralization/anti-mineralization networks in the skin and vascular connective tissues. Am. J. Pathol. 2013, 183, 10–18. [Google Scholar] [CrossRef] [PubMed]
  5. Kempf, H.; Komarova, S.; Murshed, M. Editorial: Ectopic Mineralization of Tissues: Mechanisms, Risk Factors, Diseases, and Prevention. Front. Cell Dev. Biol. 2021, 9, 759702. [Google Scholar] [CrossRef] [PubMed]
  6. Bala, Y.; Farlay, D.; Boivin, G. Bone mineralization: From tissue to crystal in normal and pathological contexts. Osteoporos. Int. 2013, 24, 2153–2166. [Google Scholar] [CrossRef] [PubMed]
  7. Li, Q.; Jiang, Q.; Uitto, J. Ectopic mineralization disorders of the extracellular matrix of connective tissue: Molecular genetics and pathomechanisms of aberrant calcification. Matrix Biol. 2014, 33, 23–28. [Google Scholar] [CrossRef] [PubMed]
  8. De Vilder, E.Y.; Vanakker, O.M. From variome to phenome: Pathogenesis, diagnosis and management of ectopic mineralization disorders. World J. Clin. Cases 2015, 3, 556–574. [Google Scholar] [CrossRef] [PubMed]
  9. Wuthier, R.E.; Lipscomb, G.F. Matrix vesicles: Structure, composition, formation and function in calcification. Front. Biosci. 2011, 16, 2812–2902. [Google Scholar] [CrossRef] [PubMed]
  10. Laycock, J.; Furmanik, M.; Sun, M.; Schurgers, L.J.; Shroff, R.; Shanahan, C.M. The role of chronic kidney disease in ectopic calcification. In Cardiovascular Calcification and Bone Mineralization; Springer: Cham, Switzerland, 2020; pp. 137–166. [Google Scholar]
  11. Stabley, J.N.; Towler, D.A. Arterial Calcification in Diabetes Mellitus: Preclinical Models and Translational Implications. Arterioscler. Thromb. Vasc. Biol. 2017, 37, 205–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Stoletov, K.; Fang, L.; Choi, S.H.; Hartvigsen, K.; Hansen, L.F.; Hall, C.; Pattison, J.; Juliano, J.; Miller, E.R.; Almazan, F.; et al. Vascular lipid accumulation, lipoprotein oxidation, and macrophage lipid uptake in hypercholesterolemic zebrafish. Circ. Res. 2009, 104, 952–960. [Google Scholar] [CrossRef]
  13. Tang, D.; Geng, F.; Yu, C.; Zhang, R. Recent Application of Zebrafish Models in Atherosclerosis Research. Front. Cell Dev. Biol. 2021, 9, 643697. [Google Scholar] [CrossRef] [PubMed]
  14. Goh, K.I.; Cusick, M.E.; Valle, D.; Childs, B.; Vidal, M.; Barabasi, A.L. The human disease network. Proc. Natl. Acad. Sci. USA 2007, 104, 8685–8690. [Google Scholar] [CrossRef]
  15. Liptak, N.; Gal, Z.; Biro, B.; Hiripi, L.; Hoffmann, O.I. Rescuing lethal phenotypes induced by disruption of genes in mice: A review of novel strategies. Physiol. Res. 2021, 70, 3–12. [Google Scholar] [CrossRef] [PubMed]
  16. Cancela, M.L.; Laizé, V.; Conceição, N.; Kempf, H.; Murshed, M. Keutel Syndrome, a Review of 50 Years of Literature. Front. Cell Dev. Biol. 2021, 9, 642136. [Google Scholar] [CrossRef]
  17. Luo, G.; Ducy, P.; McKee, M.D.; Pinero, G.J.; Loyer, E.; Behringer, R.R.; Karsenty, G. Spontaneous calcification of arteries and cartilage in mice lacking matrix GLA protein. Nature 1997, 386, 78–81. [Google Scholar] [CrossRef] [PubMed]
  18. Patton, E.E.; Zon, L.I.; Langenau, D.M. Zebrafish disease models in drug discovery: From preclinical modelling to clinical trials. Nat. Rev. Drug Discov. 2021, 20, 611–628. [Google Scholar] [CrossRef] [PubMed]
  19. Santoriello, C.; Zon, L.I. Hooked! Modeling human disease in zebrafish. J. Clin. Investig. 2012, 122, 2337–2343. [Google Scholar] [CrossRef] [PubMed]
  20. Carnovali, M.; Banfi, G.; Mariotti, M. Zebrafish Models of Human Skeletal Disorders: Embryo and Adult Swimming Together. Biomed. Res. Int. 2019, 2019, 1253710. [Google Scholar] [CrossRef] [PubMed]
  21. Rosa, J.T.; Tarasco, M.; Gavaia, P.J.; Cancela, M.L.; Laizé, V. Screening of Mineralogenic and Osteogenic Compounds in Zebrafish-Tools to Improve Assay Throughput and Data Accuracy. Pharmaceuticals 2022, 15, 983. [Google Scholar] [CrossRef]
  22. Lin, C.H.; Hwang, P.P. The Control of Calcium Metabolism in Zebrafish (Danio rerio). Int. J. Mol. Sci. 2016, 17, 1783. [Google Scholar] [CrossRef]
  23. Pozo-Morales, M.; Garteizgogeascoa, I.; Perazzolo, C.; So, J.; Shin, D.; Singh, S.P. In vivo imaging of calcium dynamics in zebrafish hepatocytes. Hepatology 2022. [Google Scholar] [CrossRef]
  24. Peng, J.B.; Suzuki, Y.; Gyimesi, G.; Hediger, M.A. TRPV5 and TRPV6 Calcium-Selective Channels. In Calcium Entry Channels in Non-Excitable Cells; Kozak, J.A., Putney, J.W., Jr., Eds.; Taylor & Francis Group: Boca Raton, FL, USA, 2018; pp. 241–274. [Google Scholar]
  25. Vanoevelen, J.; Janssens, A.; Huitema, L.F.; Hammond, C.L.; Metz, J.R.; Flik, G.; Voets, T.; Schulte-Merker, S. Trpv5/6 is vital for epithelial calcium uptake and bone formation. FASEB J. 2011, 25, 3197–3207. [Google Scholar] [CrossRef] [PubMed]
  26. Andersen, L.; Estrella, L.; Andersen, R. LDLR Variant Databases and Familial Hypercholesterolemia Population Studies. J. Am. Coll. Cardiol. 2017, 69, 754–755. [Google Scholar] [CrossRef]
  27. Wolska, A.; Dunbar, R.L.; Freeman, L.A.; Ueda, M.; Amar, M.J.; Sviridov, D.O.; Remaley, A.T. Apolipoprotein C-II: New findings related to genetics, biochemistry, and role in triglyceride metabolism. Atherosclerosis 2017, 267, 49–60. [Google Scholar] [CrossRef] [PubMed]
  28. Liu, C.; Gates, K.P.; Fang, L.; Amar, M.J.; Schneider, D.A.; Geng, H.; Huang, W.; Kim, J.; Pattison, J.; Zhang, J.; et al. Apoc2 loss-of-function zebrafish mutant as a genetic model of hyperlipidemia. Dis. Model Mech. 2015, 8, 989–998. [Google Scholar] [CrossRef]
  29. Ziegler, S.G.; Gahl, W.A.; Ferreira, C.R. Generalized arterial calcification of infancy. In GeneReviews; Adam, M.P., Ed.; University of Washington: Seattle, WA, USA, 1993. [Google Scholar]
  30. Apschner, A.; Huitema, L.F.; Ponsioen, B.; Peterson-Maduro, J.; Schulte-Merker, S. Zebrafish enpp1 mutants exhibit pathological mineralization, mimicking features of generalized arterial calcification of infancy (GACI) and pseudoxanthoma elasticum (PXE). Dis. Model Mech. 2014, 7, 811–822. [Google Scholar] [CrossRef] [PubMed]
  31. Van Gils, M.; Willaert, A.; De Vilder, E.Y.G.; Coucke, P.J.; Vanakker, O.M. Generation and Validation of a Complete Knockout Model of abcc6a in Zebrafish. J. Investig. Dermatol. 2018, 138, 2333–2342. [Google Scholar] [CrossRef]
  32. Czimer, D.; Porok, K.; Csete, D.; Gyure, Z.; Lavro, V.; Fulop, K.; Chen, Z.; Gyergyak, H.; Tusnady, G.E.; Burgess, S.M.; et al. A New Zebrafish Model for Pseudoxanthoma Elasticum. Front. Cell Dev. Biol. 2021, 9, 628699. [Google Scholar] [CrossRef]
  33. Sun, J.; She, P.; Liu, X.; Gao, B.; Jin, D.; Zhong, T.P. Disruption of Abcc6 Transporter in Zebrafish Causes Ocular Calcification and Cardiac Fibrosis. Int. J. Mol. Sci. 2020, 22, 278. [Google Scholar] [CrossRef]
  34. Singh, A.P.; Sosa, M.X.; Fang, J.; Shanmukhappa, S.K.; Hubaud, A.; Fawcett, C.H.; Molind, G.J.; Tsai, T.; Capodieci, P.; Wetzel, K.; et al. αKlotho Regulates Age-Associated Vascular Calcification and Lifespan in Zebrafish. Cell. Rep. 2019, 28, 2767–2776.e2765. [Google Scholar] [CrossRef]
  35. Ogura, Y.; Kaneko, R.; Ujibe, K.; Wakamatsu, Y.; Hirata, H. Loss of αklotho causes reduced motor ability and short lifespan in zebrafish. Sci. Rep. 2021, 11, 15090. [Google Scholar] [CrossRef] [PubMed]
  36. Stevenson, N.L.; Bergen, D.J.M.; Skinner, R.E.H.; Kague, E.; Martin-Silverstone, E.; Robson Brown, K.A.; Hammond, C.L.; Stephens, D.J. Giantin-knockout models reveal a feedback loop between Golgi function and glycosyltransferase expression. J. Cell. Sci. 2017, 130, 4132–4143. [Google Scholar] [CrossRef]
  37. Bergen, D.J.M.; Stevenson, N.L.; Skinner, R.E.H.; Stephens, D.J.; Hammond, C.L. The Golgi matrix protein giantin is required for normal cilia function in zebrafish. Biol. Open 2017, 6, 1180–1189. [Google Scholar] [CrossRef] [PubMed]
  38. Allen, R.S.; Tajer, B.; Shore, E.M.; Mullins, M.C. Fibrodysplasia ossificans progressiva mutant ACVR1 signals by multiple modalities in the developing zebrafish. eLife 2020, 9, e53761. [Google Scholar] [CrossRef]
  39. LaBonty, M.; Pray, N.; Yelick, P.C. A Zebrafish Model of Human Fibrodysplasia Ossificans Progressiva. Zebrafish 2017, 14, 293–304. [Google Scholar] [CrossRef]
  40. Zhao, M.; Lin, X.H.; Zeng, Y.H.; Su, H.Z.; Wang, C.; Yang, K.; Chen, Y.K.; Lin, B.W.; Yao, X.P.; Chen, W.J. Knockdown of myorg leads to brain calcification in zebrafish. Mol. Brain 2022, 15, 65. [Google Scholar] [CrossRef] [PubMed]
  41. Boyce, A.M.; Gafni, R.I.; Ferreira, C.R. Generalized Arterial Calcification of Infancy: New Insights, Controversies, and Approach to Management. Curr. Osteoporos. Rep. 2020, 18, 232–241. [Google Scholar] [CrossRef]
  42. Germain, D.P. Pseudoxanthoma elasticum. Orphanet. J. Rare Dis. 2017, 12, 85. [Google Scholar] [CrossRef]
  43. Mackay, E.W.; Apschner, A.; Schulte-Merker, S. Vitamin K reduces hypermineralisation in zebrafish models of PXE and GACI. Development 2015, 142, 1095–1101. [Google Scholar] [CrossRef] [Green Version]
  44. Jiang, Q.; Li, Q.; Grand-Pierre, A.E.; Schurgers, L.J.; Uitto, J. Administration of vitamin K does not counteract the ectopic mineralization of connective tissues in Abcc6 (-/-) mice, a model for pseudoxanthoma elasticum. Cell Cycle 2011, 10, 701–707. [Google Scholar] [CrossRef]
  45. Nollet, L.; Van Gils, M.; Willaert, A.; Coucke, P.J.; Vanakker, O.M. Minocycline Attenuates Excessive DNA Damage Response and Reduces Ectopic Calcification in Pseudoxanthoma Elasticum. J. Investig. Dermatol. 2022, 142, 1629–1638.e1626. [Google Scholar] [CrossRef]
  46. Lu, X.; Hu, M.C. Klotho/FGF23 Axis in Chronic Kidney Disease and Cardiovascular Disease. Kidney Dis. 2017, 3, 15–23. [Google Scholar] [CrossRef]
  47. Kuro-o, M.; Matsumura, Y.; Aizawa, H.; Kawaguchi, H.; Suga, T.; Utsugi, T.; Ohyama, Y.; Kurabayashi, M.; Kaname, T.; Kume, E.; et al. Mutation of the mouse klotho gene leads to a syndrome resembling ageing. Nature 1997, 390, 45–51. [Google Scholar] [CrossRef]
  48. Shimada, T.; Kakitani, M.; Yamazaki, Y.; Hasegawa, H.; Takeuchi, Y.; Fujita, T.; Fukumoto, S.; Tomizuka, K.; Yamashita, T. Targeted ablation of Fgf23 demonstrates an essential physiological role of FGF23 in phosphate and vitamin D metabolism. J. Clin. Investig. 2004, 113, 561–568. [Google Scholar] [CrossRef]
  49. Smith, R.C.; O’Bryan, L.M.; Farrow, E.G.; Summers, L.J.; Clinkenbeard, E.L.; Roberts, J.L.; Cass, T.A.; Saha, J.; Broderick, C.; Ma, Y.L.; et al. Circulating alphaKlotho influences phosphate handling by controlling FGF23 production. J. Clin. Investig. 2012, 122, 4710–4715. [Google Scholar] [CrossRef]
  50. Hu, M.C.; Shiizaki, K.; Kuro-o, M.; Moe, O.W. Fibroblast growth factor 23 and Klotho: Physiology and pathophysiology of an endocrine network of mineral metabolism. Annu. Rev. Physiol. 2013, 75, 503–533. [Google Scholar] [CrossRef]
  51. Kuro, O.M.; Moe, O.W. FGF23-αKlotho as a paradigm for a kidney-bone network. Bone 2017, 100, 4–18. [Google Scholar] [CrossRef]
  52. Smith, E.R.; Holt, S.G.; Hewitson, T.D. αKlotho-FGF23 interactions and their role in kidney disease: A molecular insight. Cell. Mol. Life Sci. 2019, 76, 4705–4724. [Google Scholar] [CrossRef]
  53. Mangos, S.; Amaral, A.P.; Faul, C.; Juppner, H.; Reiser, J.; Wolf, M. Expression of fgf23 and αklotho in developing embryonic tissues and adult kidney of the zebrafish, Danio rerio. Nephrol. Dial. Transplant. 2012, 27, 4314–4322. [Google Scholar] [CrossRef] [Green Version]
  54. Lin, C.H.; Hu, H.J.; Hwang, P.P. Molecular Physiology of the Hypocalcemic Action of Fibroblast Growth Factor 23 in Zebrafish (Danio rerio). Endocrinology 2017, 158, 1347–1358. [Google Scholar] [CrossRef]
  55. Min, H.; Morony, S.; Sarosi, I.; Dunstan, C.R.; Capparelli, C.; Scully, S.; Van, G.; Kaufman, S.; Kostenuik, P.J.; Lacey, D.L.; et al. Osteoprotegerin reverses osteoporosis by inhibiting endosteal osteoclasts and prevents vascular calcification by blocking a process resembling osteoclastogenesis. J. Exp. Med. 2000, 192, 463–474. [Google Scholar] [CrossRef] [PubMed]
  56. Bas, A.; Lopez, I.; Perez, J.; Rodriguez, M.; Aguilera-Tejero, E. Reversibility of calcitriol-induced medial artery calcification in rats with intact renal function. J. Bone Miner. Res. 2006, 21, 484–490. [Google Scholar] [CrossRef] [PubMed]
  57. Tadic, V.; Westenberger, A.; Domingo, A.; Alvarez-Fischer, D.; Klein, C.; Kasten, M. Primary familial brain calcification with known gene mutations: A systematic review and challenges of phenotypic characterization. JAMA Neurol. 2015, 72, 460–467. [Google Scholar] [CrossRef] [PubMed]
  58. Westenberger, A.; Balck, A.; Klein, C. Primary familial brain calcifications: Genetic and clinical update. Curr. Opin. Neurol. 2019, 32, 571–578. [Google Scholar] [CrossRef]
  59. Yao, X.P.; Cheng, X.; Wang, C.; Zhao, M.; Guo, X.X.; Su, H.Z.; Lai, L.L.; Zou, X.H.; Chen, X.J.; Zhao, Y.; et al. Biallelic Mutations in MYORG Cause Autosomal Recessive Primary Familial Brain Calcification. Neuron 2018, 98, 1116–1123.e1115. [Google Scholar] [CrossRef]
  60. Peng, Y.; Wang, P.; Chen, Z.; Jiang, H. A novel mutation in MYORG causes primary familial brain calcification with central neuropathic pain. Clin. Genet. 2019, 95, 433–435. [Google Scholar] [CrossRef]
  61. Sprecher, E. Familial tumoral calcinosis: From characterization of a rare phenotype to the pathogenesis of ectopic calcification. J. Investig. Dermatol. 2010, 130, 652–660. [Google Scholar] [CrossRef]
  62. Al Kaissi, A.; Kenis, V.; Ben Ghachem, M.; Hofstaetter, J.; Grill, F.; Ganger, R.; Kircher, S.G. The Diversity of the Clinical Phenotypes in Patients With Fibrodysplasia Ossificans Progressiva. J. Clin. Med. Res. 2016, 8, 246–253. [Google Scholar] [CrossRef]
  63. Huitema, L.F.; Apschner, A.; Logister, I.; Spoorendonk, K.M.; Bussmann, J.; Hammond, C.L.; Schulte-Merker, S. Entpd5 is essential for skeletal mineralization and regulates phosphate homeostasis in zebrafish. Proc. Natl. Acad. Sci. USA 2012, 109, 21372–21377. [Google Scholar] [CrossRef] [Green Version]
  64. Kettleborough, R.N.; Busch-Nentwich, E.M.; Harvey, S.A.; Dooley, C.M.; de Bruijn, E.; van Eeden, F.; Sealy, I.; White, R.J.; Herd, C.; Nijman, I.J.; et al. A systematic genome-wide analysis of zebrafish protein-coding gene function. Nature 2013, 496, 494–497. [Google Scholar] [CrossRef] [PubMed]
  65. Zakaria, Z.Z.; Eisa-Beygi, S.; Benslimane, F.M.; Ramchandran, R.; Yalcin, H.C. Design and Microinjection of Morpholino Antisense Oligonucleotides and mRNA into Zebrafish Embryos to Elucidate Specific Gene Function in Heart Development. J. Vis. Exp. 2022, 186. [Google Scholar] [CrossRef] [PubMed]
  66. Nasevicius, A.; Ekker, S.C. Effective targeted gene ‘knockdown’ in zebrafish. Nat. Genet. 2000, 26, 216–220. [Google Scholar] [CrossRef] [PubMed]
  67. Li, Q.; Sadowski, S.; Frank, M.; Chai, C.; Varadi, A.; Ho, S.Y.; Lou, H.; Dean, M.; Thisse, C.; Thisse, B.; et al. The abcc6a gene expression is required for normal zebrafish development. J. Investig. Dermatol. 2010, 130, 2561–2568. [Google Scholar] [CrossRef] [PubMed]
  68. Jinek, M.; Chylinski, K.; Fonfara, I.; Hauer, M.; Doudna, J.A.; Charpentier, E. A programmable dual-RNA-guided DNA endonuclease in adaptive bacterial immunity. Science 2012, 337, 816–821. [Google Scholar] [CrossRef]
  69. Ran, F.A.; Hsu, P.D.; Wright, J.; Agarwala, V.; Scott, D.A.; Zhang, F. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 2013, 8, 2281–2308. [Google Scholar] [CrossRef]
  70. Kroll, F.; Powell, G.T.; Ghosh, M.; Gestri, G.; Antinucci, P.; Hearn, T.J.; Tunbak, H.; Lim, S.; Dennis, H.W.; Fernandez, J.M.; et al. A simple and effective F0 knockout method for rapid screening of behaviour and other complex phenotypes. eLife 2021, 10, e59683. [Google Scholar] [CrossRef]
  71. Mertes, N.; Busch, M.; Huppertz, M.C.; Hacker, C.N.; Wilhelm, J.; Gurth, C.M.; Kuhn, S.; Hiblot, J.; Koch, B.; Johnsson, K. Fluorescent and Bioluminescent Calcium Indicators with Tuneable Colors and Affinities. J. Am. Chem. Soc. 2022, 144, 6928–6935. [Google Scholar] [CrossRef]
  72. Adkins, K.F. Alizarin Red S as an Intravital Fluorochrome in Mineralizing Tissues. Stain. Technol. 1965, 40, 69–70. [Google Scholar] [CrossRef]
  73. Walker, M.B.; Kimmel, C.B. A two-color acid-free cartilage and bone stain for zebrafish larvae. Biotech. Histochem. 2007, 82, 23–28. [Google Scholar] [CrossRef]
  74. Bensimon-Brito, A.; Cardeira, J.; Dionisio, G.; Huysseune, A.; Cancela, M.L.; Witten, P.E. Revisiting in vivo staining with alizarin red S—A valuable approach to analyse zebrafish skeletal mineralization during development and regeneration. BMC Dev. Biol. 2016, 16, 2. [Google Scholar] [CrossRef]
  75. DeLaurier, A.; Eames, B.F.; Blanco-Sanchez, B.; Peng, G.; He, X.; Swartz, M.E.; Ullmann, B.; Westerfield, M.; Kimmel, C.B. Zebrafish sp7:EGFP: A transgenic for studying otic vesicle formation, skeletogenesis, and bone regeneration. Genesis 2010, 48, 505–511. [Google Scholar] [CrossRef]
  76. Tonelli, F.; Bek, J.W.; Besio, R.; De Clercq, A.; Leoni, L.; Salmon, P.; Coucke, P.J.; Willaert, A.; Forlino, A. Zebrafish: A Resourceful Vertebrate Model to Investigate Skeletal Disorders. Front. Endocrinol. 2020, 11, 489. [Google Scholar] [CrossRef]
  77. Schneider, M.R. Von Kossa and his staining technique. Histochem. Cell. Biol. 2021, 156, 523–526. [Google Scholar] [CrossRef]
  78. Fadini, G.P.; Albiero, M.; Menegazzo, L.; Boscaro, E.; Vigili de Kreutzenberg, S.; Agostini, C.; Cabrelle, A.; Binotto, G.; Rattazzi, M.; Bertacco, E.; et al. Widespread increase in myeloid calcifying cells contributes to ectopic vascular calcification in type 2 diabetes. Circ. Res. 2011, 108, 1112–1121. [Google Scholar] [CrossRef]
  79. Wu, C.Y.; Martel, J.; Young, J.D. Ectopic calcification and formation of mineralo-organic particles in arteries of diabetic subjects. Sci. Rep. 2020, 10, 8545. [Google Scholar] [CrossRef]
  80. Rutges, J.P.; Duit, R.A.; Kummer, J.A.; Oner, F.C.; van Rijen, M.H.; Verbout, A.J.; Castelein, R.M.; Dhert, W.J.; Creemers, L.B. Hypertrophic differentiation and calcification during intervertebral disc degeneration. Osteoarthr. Cartil. 2010, 18, 1487–1495. [Google Scholar] [CrossRef]
  81. Silvent, J.; Akiva, A.; Brumfeld, V.; Reznikov, N.; Rechav, K.; Yaniv, K.; Addadi, L.; Weiner, S. Zebrafish skeleton development: High resolution micro-CT and FIB-SEM block surface serial imaging for phenotype identification. PLoS ONE 2017, 12, e0177731. [Google Scholar] [CrossRef]
  82. Cheng, K.C.; Xin, X.; Clark, D.P.; La Riviere, P. Whole-animal imaging, gene function, and the Zebrafish Phenome Project. Curr. Opin. Genet. Dev. 2011, 21, 620–629. [Google Scholar] [CrossRef]
  83. Ding, Y.; Vanselow, D.J.; Yakovlev, M.A.; Katz, S.R.; Lin, A.Y.; Clark, D.P.; Vargas, P.; Xin, X.; Copper, J.E.; Canfield, V.A.; et al. Computational 3D histological phenotyping of whole zebrafish by X-ray histotomography. eLife 2019, 8, 44898. [Google Scholar] [CrossRef]
  84. Katz, S.R.; Yakovlev, M.A.; Vanselow, D.J.; Ding, Y.; Lin, A.Y.; Parkinson, D.Y.; Wang, Y.; Canfield, V.A.; Ang, K.C.; Cheng, K.C. Whole-organism 3D quantitative characterization of zebrafish melanin by silver deposition micro-CT. eLife 2021, 10, e68920. [Google Scholar] [CrossRef] [PubMed]
  85. Nemecek, D.; Stepanek, J.; Thomas, G.J., Jr. Raman spectroscopy of proteins and nucleoproteins. Curr. Protoc. Protein Sci. 2013, 71, 17–18. [Google Scholar] [CrossRef] [PubMed]
  86. Ghita, A.; Matousek, P.; Stone, N. High sensitivity non-invasive detection of calcifications deep inside biological tissue using Transmission Raman Spectroscopy. J. Biophotonics 2018, 11, e201600260. [Google Scholar] [CrossRef] [PubMed]
  87. You, A.Y.F.; Bergholt, M.S.; St-Pierre, J.P.; Kit-Anan, W.; Pence, I.J.; Chester, A.H.; Yacoub, M.H.; Bertazzo, S.; Stevens, M.M. Raman spectroscopy imaging reveals interplay between atherosclerosis and medial calcification in the human aorta. Sci. Adv. 2017, 3, e1701156. [Google Scholar] [CrossRef] [PubMed]
  88. Bennet, M.; Akiva, A.; Faivre, D.; Malkinson, G.; Yaniv, K.; Abdelilah-Seyfried, S.; Fratzl, P.; Masic, A. Simultaneous Raman microspectroscopy and fluorescence imaging of bone mineralization in living zebrafish larvae. Biophys. J. 2014, 106, L17–L19. [Google Scholar] [CrossRef]
  89. Hogset, H.; Horgan, C.C.; Armstrong, J.P.K.; Bergholt, M.S.; Torraca, V.; Chen, Q.; Keane, T.J.; Bugeon, L.; Dallman, M.J.; Mostowy, S.; et al. In vivo biomolecular imaging of zebrafish embryos using confocal Raman spectroscopy. Nat. Commun. 2020, 11, 6172. [Google Scholar] [CrossRef]
  90. Boyce, B.F. Advances in osteoclast biology reveal potential new drug targets and new roles for osteoclasts. J. Bone Miner. Res. 2013, 28, 711–722. [Google Scholar] [CrossRef]
  91. Zurick, K.M.; Qin, C.; Bernards, M.T. Mineralization induction effects of osteopontin, bone sialoprotein, and dentin phosphoprotein on a biomimetic collagen substrate. J. Biomed. Mater. Res. A 2013, 101, 1571–1581. [Google Scholar] [CrossRef]
  92. Viegas, C.S.; Cavaco, S.; Neves, P.L.; Ferreira, A.; Joao, A.; Williamson, M.K.; Price, P.A.; Cancela, M.L.; Simes, D.C. Gla-rich protein is a novel vitamin K-dependent protein present in serum that accumulates at sites of pathological calcifications. Am. J. Pathol. 2009, 175, 2288–2298. [Google Scholar] [CrossRef]
  93. Viegas, C.S.; Simes, D.C.; Laizé, V.; Williamson, M.K.; Price, P.A.; Cancela, M.L. Gla-rich protein (GRP), a new vitamin K-dependent protein identified from sturgeon cartilage and highly conserved in vertebrates. J. Biol. Chem. 2008, 283, 36655–36664. [Google Scholar] [CrossRef] [Green Version]
  94. Cancela, M.L.; Laizé, V.; Conceição, N. Matrix Gla protein and osteocalcin: From gene duplication to neofunctionalization. Arch. Biochem. Biophys. 2014, 561, 56–63. [Google Scholar] [CrossRef]
  95. Wardrop, D.; Keeling, D. The story of the discovery of heparin and warfarin. Br. J. Haematol. 2008, 141, 757–763. [Google Scholar] [CrossRef]
  96. Danziger, J. Vitamin K-dependent proteins, warfarin, and vascular calcification. Clin. J. Am. Soc. Nephrol. 2008, 3, 1504–1510. [Google Scholar] [CrossRef]
  97. Schurgers, L.J.; Shearer, M.J.; Hamulyak, K.; Stocklin, E.; Vermeer, C. Effect of vitamin K intake on the stability of oral anticoagulant treatment: Dose-response relationships in healthy subjects. Blood 2004, 104, 2682–2689. [Google Scholar] [CrossRef]
  98. Koos, R.; Krueger, T.; Westenfeld, R.; Kuhl, H.P.; Brandenburg, V.; Mahnken, A.H.; Stanzel, S.; Vermeer, C.; Cranenburg, E.C.; Floege, J.; et al. Relation of circulating Matrix Gla-Protein and anticoagulation status in patients with aortic valve calcification. Thromb. Haemost. 2009, 101, 706–713. [Google Scholar]
  99. Mehndiratta, S.; Suneja, A.; Gupta, B.; Bhatt, S. Fetotoxicity of warfarin anticoagulation. Arch. Gynecol. Obstet. 2010, 282, 335–337. [Google Scholar] [CrossRef]
  100. Kruger, T.; Oelenberg, S.; Kaesler, N.; Schurgers, L.J.; van de Sandt, A.M.; Boor, P.; Schlieper, G.; Brandenburg, V.M.; Fekete, B.C.; Veulemans, V.; et al. Warfarin induces cardiovascular damage in mice. Arterioscler. Thromb. Vasc. Biol. 2013, 33, 2618–2624. [Google Scholar] [CrossRef]
  101. Granadeiro, L.; Dirks, R.P.; Ortiz-Delgado, J.B.; Gavaia, P.J.; Sarasquete, C.; Laizé, V.; Cancela, M.L.; Fernandez, I. Warfarin-exposed zebrafish embryos resembles human warfarin embryopathy in a dose and developmental-time dependent manner—From molecular mechanisms to environmental concerns. Ecotoxicol. Environ. Saf. 2019, 181, 559–571. [Google Scholar] [CrossRef]
  102. Hanumanthaiah, R.; Thankavel, B.; Day, K.; Gregory, M.; Jagadeeswaran, P. Developmental expression of vitamin K-dependent gamma-carboxylase activity in zebrafish embryos: Effect of warfarin. Blood Cells Mol. Dis. 2001, 27, 992–999. [Google Scholar] [CrossRef]
  103. Fernandez, I.; Santos, A.; Cancela, M.L.; Laizé, V.; Gavaia, P.J. Warfarin, a potential pollutant in aquatic environment acting through Pxr signaling pathway and gamma-glutamyl carboxylation of vitamin K-dependent proteins. Environ. Pollut. 2014, 194, 86–95. [Google Scholar] [CrossRef]
  104. Fernandez, I.; Vijayakumar, P.; Marques, C.; Cancela, M.L.; Gavaia, P.J.; Laizé, V. Zebrafish vitamin K epoxide reductases: Expression in vivo, along extracellular matrix mineralization and under phylloquinone and warfarin in vitro exposure. Fish Physiol. Biochem. 2015, 41, 745–759. [Google Scholar] [CrossRef] [PubMed]
  105. Li, Q.; Kingman, J.; Sundberg, J.P.; Levine, M.A.; Uitto, J. Etidronate prevents, but does not reverse, ectopic mineralization in a mouse model of pseudoxanthoma elasticum (Abcc6(-/-)). Oncotarget 2018, 9, 30721–30730. [Google Scholar] [CrossRef] [PubMed]
  106. Adirekkiat, S.; Sumethkul, V.; Ingsathit, A.; Domrongkitchaiporn, S.; Phakdeekitcharoen, B.; Kantachuvesiri, S.; Kitiyakara, C.; Klyprayong, P.; Disthabanchong, S. Sodium thiosulfate delays the progression of coronary artery calcification in haemodialysis patients. Nephrol. Dial. Transplant. 2010, 25, 1923–1929. [Google Scholar] [CrossRef] [PubMed]
  107. Van Gils, M.; Willaert, A.; Coucke, P.J.; Vanakker, O.M. The Abcc6a Knockout Zebrafish Model as a Novel Tool for Drug Screening for Pseudoxanthoma Elasticum. Front. Pharmacol. 2022, 13, 822143. [Google Scholar] [CrossRef]
Figure 1. Vascular calcification in six-month-old zebrafish exposed for fifteen days to 25 mg/L of sodium warfarin. (A) Alizarin red S in vivo staining of calcified caudal fin blood vessels (white arrows) and (B) von Kossa’s staining of a calcified artery counterstained with toluidine blue (white arrow). Bar is 500 µm in (A) and is 50 µm in (B).
Figure 1. Vascular calcification in six-month-old zebrafish exposed for fifteen days to 25 mg/L of sodium warfarin. (A) Alizarin red S in vivo staining of calcified caudal fin blood vessels (white arrows) and (B) von Kossa’s staining of a calcified artery counterstained with toluidine blue (white arrow). Bar is 500 µm in (A) and is 50 µm in (B).
Ijms 24 03366 g001
Figure 2. Current models that are available to study ectopic and soft tissue calcification in zebrafish. Information reported here includes target genes, target tissues, and the onset of ectopic calcification observed in zebrafish mutants. Representative image of a zebrafish larvae at 5 dpf. dpf, days post-fertilization; mpf, months post-fertilization; MO, morpholino oligomer. * Morpholino-injected.
Figure 2. Current models that are available to study ectopic and soft tissue calcification in zebrafish. Information reported here includes target genes, target tissues, and the onset of ectopic calcification observed in zebrafish mutants. Representative image of a zebrafish larvae at 5 dpf. dpf, days post-fertilization; mpf, months post-fertilization; MO, morpholino oligomer. * Morpholino-injected.
Ijms 24 03366 g002
Table 1. Zebrafish mutants to study ectopic mineralization disorders.
Table 1. Zebrafish mutants to study ectopic mineralization disorders.
NameGene *DescriptionZFIN IDHuman Disease Reference
enpp1hu4581/hu4581
(dragonfly, dgf)
enpp1Pyrophosphatase, role in the
regulation of bone mineralization
ZDB-GENE-040724-172Generalized arterial calcification of infancy; Pseudoxanthoma elasticum;
Autosomal recessive hypophosphatemic rickets
[30]
abcc6ahu4958/hu4958
(gräte, grt)
abcc6aTransmembrane transporter, role in the regulation of bone mineralizationZDB-GENE-050517-18Generalized arterial calcification of infancy; Pseudoxanthoma elasticum [31,32,33]
klzf3212/zf3212
sa18644
klAnti-aging hormone, role in the regulation of mineral homeostasisZDB-GENE-110221-1Hyperphosphatemic familial tumoral calcinosis-3 [34,35]
fgf23zf3214/zf3214fgf23Growth factor, role in calcium ion homeostasisZDB-GENE-050201-4Autosomal recessive hypophosphatemic rickets; Hyperphosphatemic familial tumoral calcinosis-2 [34]
golgb1bsl077/bsl077golgb1Membrane trafficking in protein’s secretory pathway ZDB-GENE-030429-9Hyperphosphatemic familial tumoral calcinosis [36,37]
MO2-acvr1l
MO4-acvr1l
zf1073Tg
acvr1lActivin A receptor, type 1ZDB-GENE-990415-9Fibrodysplasia ossificans progressiva[38,39]
myorg-E2I2-MO
myorg-ATG-MO
myorgPutative glycosidaseZDB-GENE-091113-62Primary familial brain calcification[40]
* Gene name and acronym: enpp1, ectonucleotide pyrophosphatase/phosphodiesterase 1; abcc6a, ATP-binding cassette, sub-family C (CFTR/MRP), member 6a; kl, klotho; fgf23, fibroblast growth factor 23; golgb1, golgin B1; acvr1l, activin A receptor, type 1 like; myorg, myogenesis regulating glycosidase (putative).
Table 2. Zebrafish mutants yet to be investigated.
Table 2. Zebrafish mutants yet to be investigated.
NameGene *DescriptionZFIN IDHuman Disease
la028295Tg
sa8734
nt5e5′-nucleotidase, role in hereditary arterial/articular calcification syndromeZDB-GENE-040426-1261Calcification of joints and arteries
sa37832ankhaInorganic pyrophosphate transport regulatorZDB-GENE-050913-33Chondrocalcinosis 2
sa12038
sa32626
slc34a2aHigh-affinity inorganic phosphate:sodium symporterZDB-GENE-000524-1Pulmonary alveolar microlithiasis
la022442Tg
sa37585
slc34a2bHigh-affinity inorganic phosphate:sodium symporterZDB-GENE-030709-1Pulmonary alveolar microlithiasis
sa9319gnasGTPaseZDB-GENE-090417-2Osseous heteroplasia, progressive
sa39971mgpInhibitor of vascular mineralizationZDB-GENE-060928Keutel syndrome
sa12462samd9lInflammatory response and the control of extra-osseous calcificationZDB-GENE-130530-738Normophosphatemic familial tumoral calcinosis
sa41932 fam20aGolgi-associated secretory pathway pseudokinaseZDB-GENE-081022-117Enamel renal gingival syndrome
sa20589casrParathyroid hormone secretion and renal tubular calcium re-absorption regulatorZDB-GENE-050119-8Familial hypocalciuric hypercalcemia syndrome
* Gene name and acronym: nt5e, 5′-nucleotidase, ecto (CD73); ankha, ANKH inorganic pyrophosphate transport regulator a; slc34a2a/b, solute carrier family 34 member 2a/b; gnas, GNAS complex locus; mgp, matrix gla protein; samd9l, sterile α motif domain containing 9 like; fam20a, FAM20A golgi associated secretory pathway pseudokinase; casr, calcium-sensing receptor.
Table 3. Compounds to rescue ectopic mineralization in zebrafish.
Table 3. Compounds to rescue ectopic mineralization in zebrafish.
CompoundMutantRescue Effect Reference
Etidronate (100 µM)enpp1−/−Rescues aspects of the dgf phenotype[30]
abcc6a−/−Reduced spinal mineralization[107]
Vitamin K1 (80 µM)enpp1−/−Reduces hypermineralization[43]
abcc6a−/−[43,107]
Sodium thiosulfate (20 µM)abcc6a−/−Reduced spinal mineralization[107]
Magnesium citrate (10 mM)abcc6a−/−Reduced spinal mineralization[107]
Minocycline (3 µM)abcc6a−/−Reduced aberrant mineralization[45]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Santos, J.M.A.; Laizé, V.; Gavaia, P.J.; Conceição, N.; Cancela, M.L. Zebrafish Models to Study Ectopic Calcification and Calcium-Associated Pathologies. Int. J. Mol. Sci. 2023, 24, 3366. https://doi.org/10.3390/ijms24043366

AMA Style

Santos JMA, Laizé V, Gavaia PJ, Conceição N, Cancela ML. Zebrafish Models to Study Ectopic Calcification and Calcium-Associated Pathologies. International Journal of Molecular Sciences. 2023; 24(4):3366. https://doi.org/10.3390/ijms24043366

Chicago/Turabian Style

Santos, João M. A., Vincent Laizé, Paulo J. Gavaia, Natércia Conceição, and M. Leonor Cancela. 2023. "Zebrafish Models to Study Ectopic Calcification and Calcium-Associated Pathologies" International Journal of Molecular Sciences 24, no. 4: 3366. https://doi.org/10.3390/ijms24043366

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop