Next Article in Journal
An Immuno-Fragile Profile Is Associated with Mortality Risk in Patients with Chronic Kidney Disease
Previous Article in Journal
Supra-Sartorial Subcutaneous Infiltration (SSSI) for Anterior Femoral Cutaneous Nerve Coverage in Total Knee Arthroplasty: A Preliminary Clinical Study
Previous Article in Special Issue
Experimental Models of Type 2 Diabetes Mellitus Induced by Combining Hyperlipidemic Diet (HFD) and Streptozotocin Administration in Rats: An Integrative Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Deciphering the Enigma of Calcific Aortic Valve Disease: The Pivotal Role of Animal Models in Unraveling Pathogenesis and Advancing Therapeutic Strategies

1
Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
2
Key Laboratory of Molecular Biological Targeted Therapies of the Ministry of Education, Huazhong University of Science and Technology, Wuhan 430022, China
3
Department of Structure Heart Center, FuWai Yunnan Cardiovascular Hospital, Kunming 650021, China
4
Department of Cardiac Surgery, Children’s Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou 310058, China
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Biomedicines 2025, 13(10), 2369; https://doi.org/10.3390/biomedicines13102369
Submission received: 6 August 2025 / Revised: 16 September 2025 / Accepted: 25 September 2025 / Published: 27 September 2025

Abstract

Calcific aortic valve disease (CAVD) is a prevalent cardiovascular condition and is the most common heart valve disease globally. Hyperlipidemia and aging are key risk factors; consequently, with the aging global population, CAVD incidence continues to rise. Despite extensive research, the pathogenesis of CAVD remains unclear, leading to a lack of effective pharmacological therapies. Consequently, valve replacement surgery persists as the primary treatment option. Establishing suitable animal models is crucial for investigating the complex pathophysiological mechanisms of CAVD in vivo, although an optimal model has yet to be identified. This review provides a concise overview of CAVD pathogenesis and summarizes the application of common animal models—including mice, rats, rabbits, and pigs—in studying valve calcification. We specifically detail the construction of various models and their associated calcific aortic valve phenotypes. Furthermore, we outline common detection methods for assessing aortic valve calcification in these models and suggest future directions for developing improved animal models relevant to CAVD research.

Graphical Abstract

1. Introduction

Calcific aortic valve disease (CAVD) is a prevalent cardiovascular condition and globally the most common heart valve disease [1]. Recent epidemiological studies estimate that CAVD affects approximately 12.6 million individuals worldwide, resulting in 121,700 annual deaths [2]. CAVD development is associated with multiple risk factors, including age, hypertension, diabetes mellitus, obesity, elevated plasma lipoprotein levels, and bicuspid aortic valve [1]. Age is the primary risk factor, with prevalence doubling every decade [3]. Consequently, rising life expectancy is projected to increase the incidence and prevalence of CAVD.
The pathologic changes of CAVD are a continuous process. Initially, CAVD advances to aortic stenosis (AS), characterized histopathologically by increased fibrous matrix deposition and calcified nodule formation [4]. These alterations disrupt the normal leaflet architecture, compromise the aortic valve’s biomechanical properties, and ultimately lead to AS. Severe AS is frequently associated with life-threatening symptoms such as dyspnea, angina pectoris, and syncope [5]. Currently, prosthetic aortic valve replacement is the primary effective treatment for AS [6]. However, surgical risks, alongside structural deterioration or thrombosis of valve prostheses, limit its therapeutic applicability [7]. Consequently, the development of effective pharmaceutical interventions for CAVD has become a major research focus.
Elucidating the pathogenesis of CAVD is crucial for identifying novel therapeutic targets and exploring promising research avenues. CAVD arises from diverse stimuli, including lipid deposition, inflammation, oxidative stress, and abnormal hemodynamic forces, involving valve endothelial cells (VECs), valve interstitial cells (VICs), macrophages, and other immune cells [8,9,10]. The differentiation of VICs into osteoblast-like cells is now recognized as a pivotal event in CAVD progression [11]. CAVD research typically employs both in vitro and in vivo approaches. In vitro VIC culture serves as a widely used model for investigating calcification mechanisms at cellular and molecular levels, whereas in vivo animal models are essential for exploring pathogenesis within the complex pathophysiological environment [12,13]. Although no single animal model fully recapitulates human CAVD, ongoing research focuses on enhancing their translational relevance [14,15].
This review comprehensively outlines the current understanding of CAVD pathogenesis, the development and application of pertinent animal models, and the methodologies for assessing disease progression within these models. Finally, we highlight the existing challenges and future directions for utilizing animal models in CAVD research, providing guidance to advance this critical field.

2. Overview of the Mechanisms of CAVD Development

Although the pathogenesis of CAVD remains incompletely understood, current evidence indicates its development involves several key processes: impaired endothelial barrier function, lipid deposition, immune cell infiltration, and dysregulation of phosphorus and calcium metabolism [3,16,17]. These processes collectively establish a pro-inflammatory and osteogenic microenvironment within the valve. Within this milieu, VICs progressively acquire myofibroblastic and osteoblast-like phenotypes. This phenotypic shift drives fibrocalcific remodeling of the leaflets, ultimately culminating in AS and consequent left ventricular outflow obstruction [1,18] (Figure 1).

2.1. Initiation Phase

The pathological initiation phase of CAVD is characterized by lipid deposition, endothelial damage, and inflammatory infiltration. Maintaining the integrity of the aortic valve endothelial layer is essential for normal valvular homeostasis and physiological function [19]. Abnormal hemodynamic forces, particularly high-velocity blood flow generating shear stress, can mechanically disrupt this endothelial layer, compromising its integrity and barrier function. Conversely, oscillatory shear stress on the aortic-facing side can initiate endothelial–mesenchymal transition (Endo-MT) [20,21,22]. Collectively, this endothelial dysfunction establishes a permissive environment for subsequent lipid deposition and immune cell infiltration [23,24].
Lipoprotein deposition represents a significant inducer of chronic inflammation in CAVD. This is supported by fluorodeoxyglucose positron emission tomography–computed tomography (FDG-PET/CT) imaging demonstrating inflammation in aortic valves during the pre-calcification phase [25]. The primary lipoprotein components deposited within valves include ApoB, ApoA, and Apo(a) [26]. Concurrently prevalent in CAVD is dysfunction of the endothelial nitric oxide synthase (eNOS) pathway, alongside valvular deposition of oxidized nicotinamide adenine dinucleotide (NAD) [27,28]. eNOS dysregulation, coupled with elevated nicotinamide adenine dinucleotide phosphate (NADPH) oxidase levels, drives increased production of reactive oxygen species (ROS). The resulting oxidative stress promotes substantial formation of oxidized low-density lipoprotein (ox-LDL) [29]. Ox-LDL deposition subsequently induces an inflammatory response in the valve. This response is characterized by the upregulation of cell adhesion molecules, including intercellular adhesion molecule 1 (ICAM-1) and vascular cell adhesion molecule 1 (VCAM-1) [26,30,31,32], which promotes the adhesion of immune cells. Additionally, Ox-LDL enhances calcification through upregulation of PiT-1 and promotes pro-inflammatory polarization of macrophages (M1 macrophages) [33].
Following phagocytosis by infiltrating macrophages, these lipids stimulate macrophage secretion of tumor necrosis factor-alpha (TNF-α). TNF-α activates nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), interleukin-1 beta (IL-1β), and interleukin-6 (IL-6), thereby promoting the osteogenic differentiation of VICs [34,35,36,37,38]. Concurrently, endothelial dysfunction and inflammatory responses induce local cell death, releasing apoptotic vesicles and initiating the formation of localized microcalcifications within the valve. Macrophages and VICs release extracellular vesicles (EVs), which are rich in calcium and inorganic phosphorus. The subsequent formation of hydroxyapatite (HA) crystals within these EVs leads to their rupture [39,40,41,42]. The released HA crystals interact with the extracellular matrix (ECM), exacerbating microcalcification and establishing nucleation sites for further mineralization [39,40]. These mineralization foci, in turn, amplify the inflammatory response, creating a self-perpetuating cycle that drives CAVD progression.

2.2. Propagation Phase

Osteogenic differentiation and calcification represent the hallmark features of the propagation phase in CAVD [43]. Following the initiation phase—characterized by chronic inflammatory stimulation and lipid deposition—CAVD progression undergoes a shift, with calcification becoming the predominant pathological alteration [18]. At this stage, calcification is stimulated not only by immune cells but also regulated through multiple signaling pathways. Anatomically, the aortic valve comprises three distinct layers: the fibrosa (aortic-facing), the spongiosa (middle), and the ventricularis (ventricular-facing) [13,44]. A comprehensive proteomic analysis of all three layers by Aikawa et al. revealed heightened signaling activity associated with collagen biosynthesis, modification, and extracellular matrix degradation in VICs residing within the fibrosa layer [45], indicating a heightened propensity for calcification in this region.
The differentiation of VICs into myofibroblasts and osteoblast-like cells represents a critical mechanism in the calcification process during CAVD progression. This process is accompanied by the upregulation of osteogenic markers, including Osteopontin (OPN), Bone morphogenetic protein-2 (BMP2), Bone morphogenetic protein-4 (BMP4), and alkaline phosphatase (ALP) [46,47,48,49]. This process is widely regulated by multiple factors, including cytokines from immune cells [11]. In vitro, transforming growth factor-beta 1 (TGF-β1) stimulates VIC differentiation toward myofibroblasts, whereas fibroblast growth factor-2 (FGF-2) inhibits this process [50]. Myofibroblast differentiation is characterized by increased expression of α-smooth muscle actin (α-SMA), vimentin, and smooth muscle myosin [51]. Subsequently, activation of the mitogen-activated protein kinase (MAPK) pathway promotes the association of low-density lipoprotein (LDL) with glycosaminoglycans, facilitating nodule formation [52,53]. Valvular calcium deposition occurs primarily via two mechanisms: osteogenic differentiation and dystrophic calcification. VIC osteogenic differentiation plays a significant role in the osteogenic pathway [45]. Several key molecular signaling pathways are highly associated with this osteogenic differentiation, including the RANK/RANKL/OPG, Wnt/β-catenin, and Notch1 pathways [6]. The RANK/RANKL/OPG pathway promotes VIC differentiation toward osteoblasts, critically contributing to aortic valve calcification [54]. Binding of Wnt ligands to LRP5 activates the canonical Wnt/β-catenin pathway, which positively regulates osteoblastic differentiation of valve cells and promotes calcification [55]. BMP-2 is a potent inducer of osteoblast differentiation, while Runt-related transcription factor 2 (Runx2) serves as a key transcriptional regulator of osteogenesis. Functional Notch1 signaling inhibits both BMP-2 expression and Runx2 transcriptional activity, thereby suppressing osteoblastic differentiation of valvular mesenchymal stromal cells [56]. Additionally, monocyte-derived osteoclast-like cells are capable of resorbing calcified deposits through the RANK/RANKL/OPG pathway. However, inflammatory cytokines such as TNF-α and IFN-γ impair this osteoclast function, disrupting calcification balance and promoting further deposition [57].
However, osteogenic differentiation contributes to only approximately 13% of calcification in tricuspid aortic valves with advanced aortic stenosis [58]. In contrast, the majority (approximately 83%) of calcific deposits in advanced calcific aortic valves are dystrophic in origin [59]. Some studies suggest this predominance may result from diffuse calcium precipitation on cellular debris following apoptosis of VICs [60]. It should be noted, however, that certain variability exists across studies, and the reported proportions may differ depending on sample characteristics, detection methods, and definition criteria.
Despite substantial advances in related research, the mechanisms driving valvular calcification remain incompletely understood. Consequently, no validated therapeutic target demonstrating clinically meaningful anti-calcification efficacy has yet been identified.

3. CAVD Research Animal Models

Interactions among VECs, VICs, and immune cells play a critical role in aortic valve leaflet remodeling and the pathogenesis of CAVD. This complex pathological process is further modulated by systemic factors, including the host’s metabolic status and immune response [18]. To faithfully recapitulate CAVD progression within a living organism, reliable animal models are indispensable for investigating disease mechanisms, therapeutic efficacy, and toxicity. Although species such as mice, rats, rabbits, and swine are commonly employed in CAVD research, no single model has emerged as optimal. The following sections provide an overview of frequently used animal models and those exhibiting potential for modeling CAVD (Figure 2).

3.1. Mice Model

The mouse model is extensively utilized for in vivo CAVD research, owing to its cost-effectiveness, high reproductive capacity, and amenability to genetic manipulation. C57BL/6 mice, with a typical lifespan of 18–24 months, reach an age considered equivalent to human senescence around 180 days post-birth, thus serving as a suitable model for investigating the role of aging in CAVD [61]. Although CAVD does not develop spontaneously in mice, it can be induced by feeding a high-cholesterol diet for periods exceeding 6 months [62]. While wild-type C57BL/6 mice typically develop only mild to moderate aortic stenosis under these dietary conditions, employing genetically modified mice (e.g., specific knockouts) effectively overcomes this limitation and promotes more robust calcification [63].
Current experimental mouse models for CAVD research predominantly feature single-gene mutations disrupting lipid metabolism, notably low-density lipoprotein receptor-deficient (Ldlr−/−) and apolipoprotein E-deficient (Apoe−/−) mice [64]. These models exhibit markedly elevated plasma levels of very low-density lipoprotein (VLDL) and low-density lipoprotein (LDL). When challenged with a high-cholesterol diet, they readily develop significant aortic valve calcification and initial CAVD manifestations. The Ldlr−/− and Apoe−/− models display distinct phenotypic characteristics. In Ldlr−/− mice, plasma cholesterol is primarily LDL-associated, offering a more human-like lipid profile. Pathological changes in this model are primarily driven by hyperlipidemia, as LDLR deficiency minimally impacts systemic inflammation. However, Ldlr−/− mice on a high-cholesterol diet are susceptible to metabolic syndrome, potentially confounding study endpoints [65]. Conversely, Apoe−/− mice spontaneously develop hypercholesterolemia even on a normal diet and exhibit a propensity for metabolic syndrome. Lipid streaks appear in the proximal aorta by 3 months of age, and diet-induced cardiovascular injury severity correlates with cholesterol intake [66]. Importantly, the plasma lipoprotein profile of Apoe−/− mice diverges from humans: most cholesterol is carried by VLDL and chylomicrons (not LDL), limiting its fidelity to human CAVD progression [67,68]. While both models frequently develop aortic leaflet calcification due to lipid deposition and inflammation, significant hemodynamic stenosis is uncommon [14]. The introduction of the double-mutant hypercholesterolemic model (LDLR−/−/ApoB100/100) effectively addresses this limitation. This model develops significant hemodynamic stenosis and represents a more suitable platform for mechanistic studies [64,69]. Feeding this double-mutant model a high-cholesterol diet for 12 months significantly increases hemodynamic stenosis incidence, enabling exploration of specific mechanisms, such as activated platelets accelerating aortic valve calcification [70].
Rattazzi et al. demonstrated that adding warfarin or rivaroxaban to the Western diet of Apoe−/− mice differentially influenced aortic valve calcification in this model of CAVD with dyslipidemia [71]. Histological and ultrasonographic analyses after 8 weeks revealed significantly increased calcification in the aortic valves of warfarin-treated mice compared to controls, whereas rivaroxaban treatment showed no significant difference. This suggests warfarin may facilitate establishing a CAVD model in this setting. Underlying dyslipidemia provides the foundation for this model. Conversely, in the Ldlr−/− ApoB100APOB100/APOB100 mouse model, dietary supplementation with menaquinone-4 (MK-4) increased plasma lipid levels but did not affect aortic valve morphology, calcification, or fibrosis [72]. Strategically utilizing drugs that promote aortic valve calcification represents a promising approach for developing optimized CAVD animal models.
A significant limitation of Apoe−/− mice is their poor ability to finely modulate plasma cholesterol levels and atherosclerosis extent. More critically, these mice respond poorly to established lipid-lowering and anti-atherosclerotic drugs, restricting their utility for drug efficacy testing [73]. In contrast, transgenic mice expressing both human mutant APOE (APOE3-Leiden) and human cholesteryl ester transfer protein (CETP), designated APOE3-Leiden CETP mice, exhibit moderate, modifiable hyperlipoproteinemic and atherosclerotic phenotypes. These mice demonstrate favorable responses to lipid-lowering and anti-atherosclerotic therapies [74]. Hyperlipidemia-induced lipid deposition constitutes a key pathological feature in early CAVD [75]. Supporting this, Carracedo et al. observed valve thickening after 16 weeks of Western diet feeding; notably, however, alizarin red and von Kossa staining for calcification remained negative [76]. This finding is consistent with early-stage CAVD pathological changes. Consequently, the APOE3-Leiden.CETP model holds promise as a valuable tool for evaluating the therapeutic effects of lipid-lowering and anti-atherosclerotic drugs on early CAVD.
A novel C57BL/6J mouse model for atherosclerosis research has recently been developed through adeno-associated virus serotype 8 (AAV8)-mediated delivery of the gain-of-function mutant Pcsk9D377y [77]. Proprotein convertase subtilisin/kexin type 9 (PCSK9), predominantly expressed in the liver, binds to the LDLR, promoting its lysosomal degradation in hepatocytes [78]. The D377Y mutation (aspartic acid substitution for tyrosine at residue 377) confers a gain-of-function phenotype. AAV8 efficiently transduces hepatocytes, leading to hepatic overexpression of this mutant PCSK9, enhanced LDLR degradation, and consequent hyperlipidemia [79]. This model shares phenotypic similarities with Ldlr−/− mice but enables rapid generation of hyperlipidemic mice without the need for cross-breeding (e.g., with Apoe−/− or Ldlr−/− strains), thereby accelerating atherosclerosis research. Although not yet utilized in CAVD studies, the shared pathophysiological underpinnings of CAVD and atherosclerosis suggest potential applicability of this model for CAVD research. Validating its utility for CAVD modeling will require comprehensive hemodynamic assessment and detailed histopathological analysis of aortic valve tissues.
Beyond lipid-centric models, several transgenic mouse models exhibit key pathological features of CAVD, including aortic valve leaflet calcification, even when maintained on a standard diet. These models provide valuable opportunities to investigate CAVD pathogenesis in vivo independent of high-fat dietary influences, offering a more precise simulation of the disease’s complex initiation characteristics. Notable gene-modified models include mice deficient in chondromodulin-I (Chm-I; Chm1−/−), C-X-C chemokine receptor type 4 (CXCR4; Cxcr4−/−), recombination signal binding protein for immunoglobulin kappa J region (RBPJκ; Rbpj−/−), palmdelphin (Palmd; Palmd−/−), Klotho (Klotho −/−), interleukin-1 receptor antagonist (IL-1Ra; Il1rn−/−), and mitsugumin 53 (MG53; Trim72−/−) [80,81,82,83,84,85,86].
Aged Chm1−/− mice (average ~90 weeks) exhibit typical pathological features of aortic stenosis progression, including early valvular lesions, inflammatory cell infiltration, lipid deposition, calcification, and notably, neoangiogenesis—a feature less common in other murine CAVD models [80]. While most CAVD mouse models require prolonged induction periods (often >10–20 weeks), significant aortic stenosis develops rapidly in male endothelial-specific Cxcr4 knockout mice (Tie2-Cre/Cxcr4f1/f1) fed a normal diet, detectable by echocardiography at 6 weeks; females develop stenosis even faster, within 3 weeks [81]. Aortic valve calcific nodules in Klotho-deficient (Kl−/−) mice closely resemble those in human CAVD [84]. This similarity stems primarily from the development of kidney disease and elevated serum phosphate levels in this model, both established human CAVD risk factors [87]. Furthermore, minimal immune cell infiltration in the valves allows focused investigation of VIC-specific molecular changes. Although echocardiography showed no difference in peak aortic valve velocity between 10-month-old Trim72−/− (MG53-deficient) and wild-type (WT) mice, the mean peak velocity in Trim72−/− mice increased significantly from 1274 ± 66.3 cm/s at 10 months to 2929 ± 1073 cm/s at 24 months (Δv vs. WT, p < 0.0001), providing hemodynamic evidence of progressive aortic valve disease [86]. Crucially, Trim72−/− mice exhibit age-dependent CAVD onset without baseline alterations in ventricular systolic function, establishing this model as significant for CAVD research.
Identified gene mutations in specific mouse models correlate with congenital aortic valve developmental defects in humans, contributing to an elevated incidence of CAVD. As the most prevalent congenital heart defect, bicuspid aortic valve (BAV) affects approximately 1–2% of the population [88]. Notably, 30–50% of patients undergoing surgery for CAVD present with BAV, with disease onset occurring on average a decade earlier than in those with tricuspid aortic valves [89]. Studies indicate that BAV has a heritability as high as 89%, suggesting a strong genetic basis in most cases. Although BAV is widely recognized as heritable, its inheritance does not follow a single-gene model. Key genes implicated in murine BAV development include endothelial nitric oxide synthase (Nos3), notch receptor 1 (Notch1), natriuretic peptide receptor 2 (Npr2), and discoidin, CUB and LCCL domain-containing 2 (Dcbld2) [90,91,92,93]. Phenotypic penetrance and expressivity in these models vary considerably depending on genetic background, reflecting heterogeneity observed in human BAV patients. For instance, Nos3−/− mice on a C57BL/6J background exhibit BAV penetrance ranging from 27% to 42% [90,94,95]. Similarly, Notch1+/− models show a BAV penetrance of 64% [96]. Only approximately 9.4% of Npr2−/− mice develop BAV [92]. Dcbld2−/− mice spontaneously develop aortic valve calcification and hemodynamically significant aortic stenosis (AS), with about 50% exhibiting BAV by one year of age [93]. BAV is the most common congenital cardiac malformation and exhibits a male predominance with a ratio of approximately 3:1 [97]. Consistently, Gata6+/− mice show a similar sex-biased penetrance, affecting 56% of males and 27% of females [98].
While established dietary and transgenic methods exist for modeling cardiovascular diseases, emerging interest focuses on novel animal models utilizing direct mechanical injury to the aortic valve. This technique involves echocardiography-guided insertion of a guidewire via the right common carotid artery into the left ventricle, followed by intentional scratching of the aortic valve leaflets to induce localized injury [99]. Niepmann et al. recently refined this model by modulating the extent of valve damage to recapitulate distinct stages of CAVD pathology [100]. This approach enables stage-specific investigation of CAVD progression, reliably producing key pathological hallmarks—including inflammation, oxidative stress, apoptosis, leaflet thickening, fibrosis, calcification, hemodynamically significant AS, and aortic regurgitation (AR)—thereby mirroring common clinical manifestations. Nevertheless, while the guidewire injury model robustly simulates human CAVD, it inherently models acute injury rather than the decades-long chronic mechanical stress central to spontaneous human disease (Table 1).

3.2. Rat Model

Although mice offer greater ease for genetic manipulation in generating specific knockout or overexpression models, rats exhibit closer physiological, morphological, and genetic similarities to humans, rendering them ideal for biomedical and clinical research [103,104]. In cardiovascular studies, rats display a weaker response to dietary cholesterol than mice. However, atherogenic diets (supplemented with bile acids or thiouracil) effectively induce hyperlipidemia and atherosclerosis in rats, better recapitulating human-like lipid deposition processes [105]. Reaves et al. developed Ldlr−/− hamsters exhibiting impaired clearance of circulating large lipoproteins and accelerated atherosclerosis [106]. Zeng et al. subsequently demonstrated that this model develops significant valvular calcification after 16 weeks on a Western diet, though without concomitant hemodynamic manifestations of AS. Beyond lipid metabolism gene editing, rat CAVD models can also be established by targeting microRNAs (miRNAs), a well-established class of non-coding RNAs. Xu et al. successfully generated miR-214 knockout rats. When challenged with a Western diet and intraperitoneal vitamin D injections for 16 weeks, these miR-214−/− rats exhibited aortic valve thickening, calcification, and increased transvalvular velocity [107,108].
Similar to murine CAVD models, rats can develop CAVD through pharmacological induction, though surgical approaches are also utilized. In rat-based CAVD research, warfarin administration—either via diet or subcutaneous injection—is the primary method for inducing aortic valve calcification [109]. To prevent life-threatening coagulopathy, concurrent vitamin K1 supplementation is essential. Notably, studies by Husseini et al. indicate that warfarin-induced models may develop AS [110]. Alternatively, vitamin D supplementation for 10 weeks induces characteristic CAVD clinical signs and hemodynamic alterations in rats. However, this approach exclusively promotes aortic valve and vascular calcification without concurrent AS development [111].
Metabolic disorders associated with chronic kidney disease (CKD) constitute significant risk factors for CAVD development [87]. To investigate CKD-mediated pathology, Shuvy et al. established a Sprague-Dawley rat model using a phosphate-enriched, uremia-inducing diet. This model recapitulated aortic valve calcification linked to inflammation and osteoblastic activity, as validated by histology, echocardiography, and computed tomography. Notably, calcification regressed upon dietary cessation and mineral balance restoration, demonstrating its dynamic reversibility [112]. Complementing this approach, Messaoudi et al. developed a surgical CKD model in rats involving bilateral renal artery ligation and renal tissue electrocautery, combined with a high-phosphorus diet—a feasible strategy given rodents’ surgical accessibility [113]. This model provides a robust platform for delineating the pathogenesis of CKD-associated CAVD.
Advancing gene editing technologies and increasingly comprehensive rat genomic mapping are poised to drive the development of numerous rat-specific gene-targeted models in the near future. This progress is expected to yield tailored rat models of CAVD, capable of catering to the diverse research demands of the field (Table 2).

3.3. Rabbit Model

The rabbit model is widely utilized in CAVD research owing to its anatomical similarities to humans, including a tri-leaflet aortic valve structure. Furthermore, rabbits exhibit a plasma lipoprotein profile similar to humans, rendering them a valuable model for investigating the role of dyslipidemia in CAVD pathogenesis [114].
New Zealand White rabbits represent the predominant strain for modeling CAVD. These models typically employ a high-cholesterol diet supplemented with vitamin D2 to induce severe AS, detectable within 40 weeks [115]. The critical role of vitamin D2 was definitively established by Doan et al. in a controlled trial [116]; rabbits fed this diet for 12 weeks developed significant aortic valve calcification, reduced valve area, and clinical AS signs, directly linking vitamin D2-induced hypercalcemia to CAVD pathogenesis. However, high-cholesterol diets frequently cause hepatic impairment in rabbits, leading to low survival rates [117]. To circumvent this limitation, Lu et al. generated LDLR−/− rabbits via knockout of the LDL receptor gene’s protein-coding region [118]. This model spontaneously develops hypercholesterolemia without dietary manipulation, eliminating diet-associated confounders.
Although the aforementioned gene-edited rabbits have not yet been applied to CAVD research, Watanabe Heritable Hyperlipidemic (WHHL) rabbits—possessing analogous mutations—are extensively studied in this field. Beyond sharing the advantages of LDLR−/− rabbits, WHHL rabbits fed a standard diet spontaneously develop aortic stenosis, calcification, and upregulation of calcification-associated gene pathways [117]. Moreover, these pathologies exhibit clear age-dependent progression in WHHL rabbits, significantly advancing our understanding of age as a critical risk factor for CAVD development (Table 3).

3.4. Swine Model

The porcine cardiovascular system exhibits significant anatomical and morphological homology with humans, particularly in aortic valve structure. Similarly to human valves, porcine aortic valves possess a tricomposite layered architecture. Unlike murine and rabbit models, swine spontaneously develop valvular sclerosis lesions [120]. Genomic analyses reveal closer evolutionary proximity between swine and humans compared to rodent models, suggesting superior translational relevance at the molecular level, highlighting swine’s potential for CAVD research [121]. Zabirnyk et al. established an ex vivo model using cultured intact porcine valve leaflets, maintaining structural integrity while enabling observation of calcification under pro-calcific conditions [122]. Complementing this, Sider et al. conducted histopathological and immunohistochemical analyses on Yorkshire swine fed high-fat diets. After 5 months, these swine developed proteoglycan-enriched fibrotic lesions in aortic valves—an early hallmark of CAVD pathogenesis [120]. These findings demonstrate remarkable conservation of early CAVD pathological features between swine and humans, positioning porcine models as ideal platforms for investigating initial disease mechanisms.
Early CAVD pathological features—including leaflet thickening, oxidized lipid accumulation, and macrophage infiltration—are evident in Rapacz familial hypercholesterolemic swine [123]. During this initial phase, valvular function remains largely preserved, with advanced aortic stenosis requiring additional pathogenic stimuli. Sider et al. have suggested extending the use of established porcine atherosclerosis models harboring lipid metabolism mutations to CAVD research [124]. Currently, analogous to murine models, swine with ApoE, LDLR, and double-knockout mutations are primarily utilized in atherosclerosis studies [125]. Consequently, dedicated CAVD validation studies are warranted for these porcine models (Table 4).

3.5. Canine Model

Canine models are valuable for cardiovascular disease research due to their physiological similarities with humans, particularly in metabolic processes, cardiovascular structure/function, and other systems. While surgical methods have established canine models of aortic stenosis, these models typically lack pathological changes in the aortic valve [126]. Consequently, canine models have not been employed in studies specifically investigating CAVD.
Canine models can spontaneously develop atherosclerotic lesions similar to those in humans, accompanied by analogous clinical complications [127]. Atherosclerosis in these models can be exacerbated by a high-fat, high-cholesterol diet or by inducing mutations in lipid metabolism genes. Research indicates that adipokines contribute to acute endothelial dysfunction by impairing endothelial vasodilatory mechanisms. This suggests that chronic exposure to circulating adipokines, derived from adipose tissue, may lead to systemic endothelial dysfunction. Given the shared pathogenesis between early atherosclerosis and CAVD, the canine atherosclerosis model might also exhibit pathological changes in the aortic valve; however, this hypothesis requires further validation through in-depth research. Notably, distinct differences exist in the plasma lipid profiles of dogs and humans. Canine blood cholesterol is predominantly composed of high-density lipoprotein (HDL) particles, resembling rodents, whereas human blood cholesterol is primarily made up of LDL particles [128]. Moreover, as companion animals, canines raise unique ethical concerns, and their lower societal acceptance limits their use in CAVD research in many countries [129,130].

3.6. Other Animal Models

Various animal models serve as valuable tools for studying cardiovascular diseases, each offering distinct advantages for research. For instance, zebrafish, owing to their similarity to mammals in heart development, are frequently employed to investigate cardiac development and disease mechanisms [131]. Notably, Gollmann et al. even identified a molecular pathway associated with CAVD through studies of zebrafish mandibular formation [132]. Conversely, sheep share significant similarities with humans in cardiovascular anatomy and physiology, making them a preferred model for surgical procedures [133,134]. Consequently, sheep are extensively used in preclinical evaluations of artificial heart valve replacements, providing a model that closely recapitulates the pathophysiological changes observed in human cardiovascular diseases.

4. Histologic and Imaging Assessment of the Aortic Valve in Animal Models of CAVD

Animal models are crucial for investigating CAVD mechanisms, enabling comprehensive analysis of aortic valves using reliable assays. While histopathology provides a straightforward and reliable assessment, it does not allow for longitudinal monitoring. Consequently, multimodal imaging approaches have become widely used to evaluate aortic valve characteristics and calcification in vivo.

4.1. Histological Examination of Aortic Valve in Animal Models

Histologic examination of the aortic valve is essential for accurately assessing valvular morphology and quantifying calcium deposition in animal models [135]. Commonly used techniques for detecting valve calcification include Alizarin red staining, which binds calcium ions, and von Kossa staining, which reacts with carbonates and phosphates [135,136]. While both assays provide insight into the degree of calcification, Alizarin red staining has been shown to be more effective than von Kossa staining for detecting calcification in C57BL/6 mouse aortic valves, where pigmented deposits can interfere with interpretation due to similar coloration. Overall, von Kossa staining exhibits greater sensitivity for calcified lesions, whereas Alizarin red offers higher specificity. Therefore, their combined use is recommended for more accurate assessment of calcification. Beyond calcification, analysis of other valve tissue components is crucial. Collagen can be assessed using Masson’s trichrome or Picrosirius red staining, while Movat’s pentachrome stain allows simultaneous visualization of multiple components, including collagen, elastin, and proteoglycans [137,138]. Furthermore, attention should be paid to the valve commissures, as calcification commonly initiates at these attachment points.

4.2. Echocardiography

Echocardiography, a non-invasive, safe, and cost-effective modality, is a key tool for assessing heart valve function and is commonly employed in CAVD animal models. Key indicators for evaluating aortic stenosis include aortic valve area, mean pressure gradient, and peak flow velocity, which effectively reflect valvular function and hemodynamic alterations. Although echocardiography cannot precisely quantify calcium levels, it remains the gold standard for assessing valve function, degenerative changes (including fibrotic leaflet thickening and calcification), and overall stenosis severity [139]. For murine cardiac ultrasound imaging, studies have recommended the use of 30 MHz probes (e.g., VisualSonics Vevo 770) for examination. Prior to imaging, the chest hair should be thoroughly removed to minimize acoustic interference. Throughout the procedure, heart rate and core body temperature must be continuously monitored to reduce the impact of anesthesia on cardiac function. It is generally advised to maintain the heart rate between 400 and 500 beats per minute and body temperature at 37 °C [140]. Aortic valve peak velocity is measured in the suprasternal view using pulse-wave Doppler with angle correction set between 45° and 55° [141]. Currently, there is no universally accepted criterion for grading aortic stenosis in mice based on this parameter. Some studies define mild, moderate, and severe stenosis as a peak velocity increase of 15–50%, 50–75%, and greater than 75% above baseline, respectively [100]. In contrast, other researchers consider an absolute aortic valve peak velocity exceeding 2000 mm/s as indicative of stenosis [142]. Compared to transthoracic echocardiography (TTE), transesophageal echocardiography (TEE) provides superior image clarity in large-animal models, particularly for differentiating between tricuspid and bicuspid aortic valves [143].

4.3. Computed Tomography

Computed Tomography (CT) is widely used in clinical practice to identify and quantify aortic valve calcification. The calcium score, calculated using the Agatston method based on the density and volume of calcified plaques detected by CT, shows a strong correlation with the weight of excised calcified valves [144]. This score reflects the progression of CAVD and correlates strongly with the severity of aortic stenosis as assessed by echocardiography. Therefore, CT serves as a valuable tool for the quantitative evaluation of aortic valve calcification in large animal models. However, CT has several limitations. For example, in young patients with early-stage (diastolic) CAVD, the mean transaortic pressure gradient may not correlate with calcium density, reducing the accuracy of CT-based calcification assessment in this population [145]. Studies also indicate that CT-assessed calcium burden in CAVD may differ by sex [146]. Furthermore, CT exhibits limited sensitivity in detecting calcification, often failing to visualize early microcalcifications and only revealing larger, confluent calcified areas. Nevertheless, recent research has demonstrated that contrast-enhanced CT can not only evaluate the extent of calcification but also detect the non-calcified (fibrotic) component of the aortic valve, which is characteristic of early CAVD [147]. Thus, CT represents a promising technique for assessing valvular calcification in large animal models.

4.4. Micro-Computed Tomography

Micro-Computed Tomography (Micro-CT) enables high-resolution analysis of the morphology and mineral density within excised aortic valves. With a spatial resolution reaching 1 µm, Micro-CT is ideally suited for quantifying structural parameters such as porosity, bone thickness, density, particle diameter, and fiber orientation [148]. The technique relies on X-ray attenuation, which varies with material density and thickness, allowing differentiation between calcified and soft tissues. This capability facilitates the generation of contrast-enhanced images depicting calcification distribution and enables precise volumetric quantification of calcified tissue, which correlates with aortic stenosis severity [149]. Compared to the Agatston score, this method offers superior reproducibility, greater stability, and improved sensitivity for detecting small calcified lesions, making it particularly suitable for use in small animal models [150,151]. Therefore, Micro-CT can be applied for in vivo imaging in small animal models. For instance, Gollmann et al. utilized Micro-CT to analyze the relationship between aortic valve calcification and bone formation, demonstrating its utility through concurrent assessment of femur alterations in mice [132]. Spelsberg et al. employed a Scanco Micro-CT 40 system to evaluate the presence and extent of calcification in male New Zealand white rabbits. Scanning parameters included 45 kV voltage, approximately 8 μm voxel size, and optimized sensitivity (1000 projections, 2048 samples, 0.3 s integration time per projection) [152]. The high resolution of micro-CT also allows detailed imaging and characterization of pathological changes in aortic valves in small animal models; however, care should be taken to differentiate such changes from calcification occurring in the mouse aortic arch [153].

4.5. Magnetic Resonance Imaging

Compared to CT, Magnetic Resonance Imaging (MRI) is less commonly employed for diagnosing and managing CAVD. However, it serves as a valuable complementary imaging modality, particularly in aortic stenosis [154]. MRI enables assessment of valve anatomy, flow quantification, and left ventricular volume, mass, remodeling, and function. Its precise anatomical analysis contributes to a growing role in planning aortic valve replacement surgery. The latest consensus document from the European Society of Cardiovascular Radiology highlights potential advantages of MRI, including comprehensive measurements of the valve and aortic root, assessment of ventricular function, and evaluation of the aorta [155]. MRI offers benefits over invasive tests such as TEE and cardiac catheterization and avoids contrast agent use in cases of severe renal impairment [156]. Despite this limitation, MRI cannot assess the degree of aortic valve calcification. In recent years, 4D Flow MRI has emerged as a novel vascular imaging technique. Unlike conventional 3D vascular imaging, it incorporates temporal resolution (time-resolved), allowing visualization of hemodynamic changes throughout the cardiac cycle [157]. Moreover, 4D Flow MRI serves as a key technique linking valvular morphology and hemodynamic abnormalities to pathological changes in the aortic wall. It not only provides deeper insights into the mechanisms underlying aortic pathologies but also offers promising imaging-based evidence for hemodynamic evaluation in large animal models of CAVD [158].

4.6. Positron Emission Tomography (PET)

Positron emission tomography (PET) utilizes radioactive tracers that accumulate in areas of high metabolic activity, enabling the detection of molecular and cellular changes before observable anatomical alterations occur. Currently, two primary tracers are used in PET imaging: 18F-fluorodeoxyglucose (18F-FDG) and 18F-sodium fluoride (18F-NaF) [159]. 18F-FDG, a glucose analog, accumulates in metabolically active cells and serves as a sensitive marker for valvular inflammation. It is taken up by cells via glucose transporter proteins (GLUTs), reflecting the glucose uptake process in macrophages and other inflammatory cells. The enhanced glycolytic activity in these cells makes 18F-FDG a suitable target for inflammation imaging with PET [160,161]. Multiple studies have confirmed a correlation between 18F-FDG signal and atherosclerotic plaques, supporting its role as an early indicator of CAVD. This is further emphasized by the colocalization of macrophage-rich regions with 18F-FDG–positive areas [160,162,163]. In contrast, 18F-NaF is a bone-seeking tracer that binds to hydroxyapatite, a key component of valvular calcification, reflecting its extent. Notably, some studies suggest that 18F-NaF PET/CT may predict hemodynamic progression in calcific AS, whereas 18F-FDG PET/CT does not significantly predict the development of severe AS [164]. This discrepancy likely arises because hemodynamic progression in AS is primarily driven by valve calcification. Supporting this, Sadeghi et al. employed 18F-NaF PET in a mouse model of CAVD, demonstrating through both PET and autoradiography an age-dependent increase in aortic valve microcalcification, indicating aging as a significant risk factor [93]. Collectively, PET holds significant potential to enhance our understanding of CAVD pathogenesis by elucidating its molecular mechanisms and enabling dynamic in vivo tracking of pathological processes.

5. Conclusions and Prospects

Effective treatment of CAVD requires a comprehensive understanding of its etiology and pathogenesis. Despite significant progress over the past two decades in elucidating various pathogenic mechanisms, our knowledge of CAVD pathogenesis remains incomplete. Animal models offer a valuable platform for validating novel CAVD mechanisms and evaluating the efficacy of pharmacological interventions. Current CAVD animal models primarily induce rapid valve calcification through high-cholesterol diets and genetic manipulation of lipid metabolism. Future model development should focus on achieving greater precision by leveraging advanced gene editing technologies and insights from epigenetic regulation. This will enable higher-fidelity recapitulation of human CAVD, including accurate modeling of distinct disease progression stages. For instance, models mimicking the early asymptomatic phase of CAVD would facilitate research into early diagnosis and therapeutic strategies. Similarly, developmental programming models could elucidate how early-life environmental factors influence cardiovascular disease susceptibility, informing preventive approaches. Furthermore, given the multifactorial nature of CAVD, future animal models must emphasize simulating the complex interplay among genetic, environmental, and lifestyle factors, as well as interactions between multiple systems (e.g., endocrine and immune). Developing such integrated models aims to foster a more comprehensive and accurate understanding of CAVD’s intricate pathogenesis.
Animal models provide distinct advantages for research, including the ability to simulate human disease states, shorter lifespans, controllable environments, ethical permissibility, rapid reproductive cycles, and well-defined genetic backgrounds. However, they face inherent challenges due to metabolic, physiological, and anatomical differences compared to humans. Addressing these limitations necessitates a deeper understanding of the underlying pathophysiology and enhancing the translational value of animal models. Strategies include developing humanized models (e.g., humanized mice) and employing genetic engineering to create models with cardiovascular characteristics more closely resembling humans, thereby improving the fidelity of drug screening and evaluation. Moreover, monitoring based on biomarkers or hemodynamic parameters will enable real-time tracking of disease progression, enhance model sensitivity, and provide a more detailed understanding of CAVD formation and developmental stages. This approach allows for a more nuanced understanding of disease dynamics and improves the accuracy and applicability of these therapeutic testing models. Concurrently, the timely integration of novel multi-omics technologies, such as spatial transcriptomics, proteomics, and single-cell genomics, will facilitate the further refinement of CAVD animal models, enabling more precise experimental design.
Ultimately, validating disease mechanisms and bridging preclinical findings to clinical applications depend critically on human patient data and large-scale population studies. Consequently, the future development of animal models for CAVD research will increasingly prioritize precision modeling, simulation of multifactorial interactions and distinct disease stages, and enhancing translational relevance. These advancements aim to provide a stronger scientific foundation for the prevention, diagnosis, and treatment of CAVD.

Author Contributions

P.F.: Data curation, Roles/Writing—original draft, Writing—review and editing. Y.L.: Roles/Writing—original draft, Writing—review and editing. X.Q.: Data curation, Writing—review and editing. F.T.: Data curation. Y.Z.: Visualization. Z.F.: Data curation. M.C.: Software. Z.C.: Methodology. H.Z.: Data curation. T.Z.: Data curation. N.D.: Supervision, Project administration. F.L.: Conceptualization, Supervision, Project administration, Funding acquisition. X.S.: Conceptualization, Supervision, Writing—review and editing. L.X.: Conceptualization, Supervision, Project administration. All authors have read and agreed to the published version of the manuscript.

Funding

The authors declare that financial support was received for the research, authorship, and/or publication of this article. This work was supported by National Natural Science Foundation of China (Grant No. 82322007, Grant No. 82170377). The National Key Project of Research and Development Plan during the fourteenth Five-year Plan Period (Grant No. 2022YFC2503400). The Research Grant of Key Laboratory of Molecular Biological Targeted Therapies of the Ministry of Education (Huazhong University of Science and Technology) [Grant No. 2024SWBS014], and the Open Foundation of Hubei Key Laboratory of Regenerative Medicine and Multi-disciplinary Translational Research [Grant No. 2024zsyx12].

Institutional Review Board Statement

Not applicable.

Conflicts of Interest

The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

ALPalkaline phosphatase
Apoapolipoprotein
ASaortic stenosis
BAVbicuspid aortic valve
BMPbone morphogenetic protein
CAVDcalcific aortic valve disease
CKDchronic kidney disease
CTcomputed tomography
ECMextracellular matrix
Endo-MTendothelial–mesenchymal transition
eNOSendothelial nitric oxide synthase
EVextracellular vesicle
FDG-PET/CTfluorodeoxyglucose positron emission tomography-computed tomography
ICAM-1intercellular adhesion molecule 1
IFN-γinterferon-γ
ILinterleukin
MAPKmitogen-activated protein kinase
MRIMagnetic Resonance Imaging
NADnicotinamide adenine dinucleotide
NADPHnicotinamide adenine dinucleotide phosphate
NF-κBnuclear factor kappa-light-chain-enhancer of activated B cells
OPGosteoprotegerin
OPNosteopontin
ox-LDLoxidized low-density lipoprotein
PCSK9Proprotein convertase subtilisin/kexin type 9
PiT-1inorganic phosphate transporter-1
RANKLreceptor activator of nuclear factor κB ligand
ROSreactive oxygen species
Runx2Runt-related transcription factor 2
TNFtumor necrosis factor-alpha
VCAM-1vascular cell adhesion molecule 1
VECsvalve endothelial cells
VICsvalve interstitial cells

References

  1. Moncla, L.M.; Briend, M.; Bossé, Y.; Mathieu, P. Calcific aortic valve disease: Mechanisms, prevention and treatment. Nat. Rev. Cardiol. 2023, 20, 546–559. [Google Scholar] [CrossRef] [PubMed]
  2. Yadgir, S.; Johnson, C.O.; Aboyans, V.; Adebayo, O.M.; Adedoyin, R.A.; Afarideh, M.; Alahdab, F.; Alashi, A.; Alipour, V.; Arabloo, J.; et al. Global, Regional, and National Burden of Calcific Aortic Valve and Degenerative Mitral Valve Diseases, 1990–2017. Circulation 2020, 141, 1670–1680. [Google Scholar] [CrossRef]
  3. Stewart, B.F.; Siscovick, D.; Lind, B.K.; Gardin, J.M.; Gottdiener, J.S.; Smith, V.E.; Kitzman, D.W.; Otto, C.M. Clinical factors associated with calcific aortic valve disease. Cardiovascular Health Study. J. Am. Coll. Cardiol. 1997, 29, 630–634. [Google Scholar] [CrossRef] [PubMed]
  4. Rajamannan, N.M.; Evans, F.J.; Aikawa, E.; Grande-Allen, K.J.; Demer, L.L.; Heistad, D.D.; Simmons, C.A.; Masters, K.S.; Mathieu, P.; O’Brien, K.D.; et al. Calcific aortic valve disease: Not simply a degenerative process: A review and agenda for research from the National Heart and Lung and Blood Institute Aortic Stenosis Working Group. Executive summary: Calcific aortic valve disease-2011 update. Circulation 2011, 124, 1783–1791. [Google Scholar] [CrossRef]
  5. Pujari, S.H.; Agasthi, P. Aortic stenosis. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2023. [Google Scholar]
  6. Lindman, B.R.; Clavel, M.A.; Mathieu, P.; Iung, B.; Lancellotti, P.; Otto, C.M.; Pibarot, P. Calcific aortic stenosis. Nat. Rev. Dis. Primers 2016, 2, 16006. [Google Scholar] [CrossRef]
  7. Dangas, G.D.; Weitz, J.I.; Giustino, G.; Makkar, R.; Mehran, R. Prosthetic Heart Valve Thrombosis. J. Am. Coll. Cardiol. 2016, 68, 2670–2689. [Google Scholar] [CrossRef]
  8. Freeman, R.V.; Otto, C.M. Spectrum of calcific aortic valve disease: Pathogenesis, disease progression, and treatment strategies. Circulation 2005, 111, 3316–3326. [Google Scholar] [CrossRef]
  9. Perrot, N.; Thériault, S.; Dina, C.; Chen, H.Y.; Boekholdt, S.M.; Rigade, S.; Després, A.A.; Poulin, A.; Capoulade, R.; Le Tourneau, T.; et al. Genetic Variation in LPA, Calcific Aortic Valve Stenosis in Patients Undergoing Cardiac Surgery, and Familial Risk of Aortic Valve Microcalcification. JAMA Cardiol. 2019, 4, 620–627. [Google Scholar] [CrossRef]
  10. Thériault, S.; Dina, C.; Messika-Zeitoun, D.; Le Scouarnec, S.; Capoulade, R.; Gaudreault, N.; Rigade, S.; Li, Z.; Simonet, F.; Lamontagne, M.; et al. Genetic Association Analyses Highlight IL6, ALPL, and NAV1 As 3 New Susceptibility Genes Underlying Calcific Aortic Valve Stenosis. Circ. Genom. Precis Med. 2019, 12, e002617. [Google Scholar] [CrossRef] [PubMed]
  11. New, S.E.; Aikawa, E. Cardiovascular calcification: An inflammatory disease. Circ. J. 2011, 75, 1305–1313. [Google Scholar] [CrossRef]
  12. Mathieu, P.; Boulanger, M.C.; Bouchareb, R. Molecular biology of calcific aortic valve disease: Towards new pharmacological therapies. Expert. Rev. Cardiovasc Ther. 2014, 12, 851–862. [Google Scholar] [CrossRef] [PubMed]
  13. Rutkovskiy, A.; Malashicheva, A.; Sullivan, G.; Bogdanova, M.; Kostareva, A.; Stensløkken, K.O.; Fiane, A.; Vaage, J. Valve Interstitial Cells: The Key to Understanding the Pathophysiology of Heart Valve Calcification. J Am Heart Assoc 2017, 6, e006339. [Google Scholar] [CrossRef]
  14. Sider, K.L.; Blaser, M.C.; Simmons, C.A. Animal models of calcific aortic valve disease. Int. J. Inflam. 2011, 2011, 364310. [Google Scholar] [CrossRef]
  15. Gomez Stallons, M.V.; Wirrig-Schwendeman, E.E.; Fang, M.; Cheek, J.D.; Alfieri, C.M.; Hinton, R.B.; Yutzey, K.E. Molecular Mechanisms of Heart Valve Development and Disease. In Etiology and Morphogenesis of Congenital Heart Disease: From Gene Function and Cellular Interaction to Morphology; Nakanishi, T., Markwald, R.R., Baldwin, H.S., Keller, B.B., Srivastava, D., Yamagishi, H., Eds.; Springer: Tokyo, Janpan, 2016; pp. 145–151. [Google Scholar]
  16. Larsson, S.C.; Wolk, A.; Håkansson, N.; Bäck, M. Overall and abdominal obesity and incident aortic valve stenosis: Two prospective cohort studies. Eur. Heart J. 2017, 38, 2192–2197. [Google Scholar] [CrossRef]
  17. Ljungberg, J.; Johansson, B.; Engström, K.G.; Albertsson, E.; Holmer, P.; Norberg, M.; Bergdahl, I.A.; Söderberg, S. Traditional Cardiovascular Risk Factors and Their Relation to Future Surgery for Valvular Heart Disease or Ascending Aortic Disease: A Case-Referent Study. J. Am. Heart Assoc. 2017, 6, e005133. [Google Scholar] [CrossRef]
  18. Goody, P.R.; Hosen, M.R.; Christmann, D.; Niepmann, S.T.; Zietzer, A.; Adam, M.; Bönner, F.; Zimmer, S.; Nickenig, G.; Jansen, F. Aortic Valve Stenosis: From Basic Mechanisms to Novel Therapeutic Targets. Arter. Thromb. Vasc. Biol. 2020, 40, 885–900. [Google Scholar] [CrossRef]
  19. Fernández Esmerats, J.; Heath, J.; Jo, H. Shear-Sensitive Genes in Aortic Valve Endothelium. Antioxid Redox Signal 2016, 25, 401–414. [Google Scholar] [CrossRef] [PubMed]
  20. Wirrig, E.E.; Yutzey, K.E. Conserved transcriptional regulatory mechanisms in aortic valve development and disease. Arter. Thromb. Vasc. Biol. 2014, 34, 737–741. [Google Scholar] [CrossRef] [PubMed]
  21. Eisenberg, L.M.; Markwald, R.R. Molecular regulation of atrioventricular valvuloseptal morphogenesis. Circ. Res. 1995, 77, 1–6. [Google Scholar] [CrossRef]
  22. Aikawa, E.; Whittaker, P.; Farber, M.; Mendelson, K.; Padera, R.F.; Aikawa, M.; Schoen, F.J. Human semilunar cardiac valve remodeling by activated cells from fetus to adult: Implications for postnatal adaptation, pathology, and tissue engineering. Circulation 2006, 113, 1344–1352. [Google Scholar] [CrossRef]
  23. Otto, C.M.; Kuusisto, J.; Reichenbach, D.D.; Gown, A.M.; O’Brien, K.D. Characterization of the early lesion of ‘degenerative’ valvular aortic stenosis. Histological and immunohistochemical studies. Circulation 1994, 90, 844–853. [Google Scholar] [CrossRef]
  24. Yearwood, T.L.; Misbach, G.A.; Chandran, K.B. Experimental fluid dynamics of aortic stenosis in a model of the human aorta. Clin. Phys. Physiol. Meas. 1989, 10, 11–24. [Google Scholar] [CrossRef]
  25. Abdelbaky, A.; Corsini, E.; Figueroa, A.L.; Subramanian, S.; Fontanez, S.; Emami, H.; Hoffmann, U.; Narula, J.; Tawakol, A. Early aortic valve inflammation precedes calcification: A longitudinal FDG-PET/CT study. Atherosclerosis 2015, 238, 165–172. [Google Scholar] [CrossRef] [PubMed]
  26. O’Brien, K.D.; Reichenbach, D.D.; Marcovina, S.M.; Kuusisto, J.; Alpers, C.E.; Otto, C.M. Apolipoproteins B, (a), and E accumulate in the morphologically early lesion of ‘degenerative’ valvular aortic stenosis. Arter. Thromb. Vasc. Biol. 1996, 16, 523–532. [Google Scholar] [CrossRef]
  27. Miller, J.D.; Chu, Y.; Brooks, R.M.; Richenbacher, W.E.; Peña-Silva, R.; Heistad, D.D. Dysregulation of antioxidant mechanisms contributes to increased oxidative stress in calcific aortic valvular stenosis in humans. J. Am. Coll. Cardiol. 2008, 52, 843–850. [Google Scholar] [CrossRef]
  28. Liberman, M.; Bassi, E.; Martinatti, M.K.; Lario, F.C.; Wosniak, J., Jr.; Pomerantzeff, P.M.; Laurindo, F.R. Oxidant generation predominates around calcifying foci and enhances progression of aortic valve calcification. Arter. Thromb. Vasc. Biol. 2008, 28, 463–470. [Google Scholar] [CrossRef] [PubMed]
  29. Parhami, F.; Morrow, A.D.; Balucan, J.; Leitinger, N.; Watson, A.D.; Tintut, Y.; Berliner, J.A.; Demer, L.L. Lipid oxidation products have opposite effects on calcifying vascular cell and bone cell differentiation. A possible explanation for the paradox of arterial calcification in osteoporotic patients. Arter. Thromb. Vasc. Biol. 1997, 17, 680–687. [Google Scholar] [CrossRef]
  30. Olsson, M.; Thyberg, J.; Nilsson, J. Presence of oxidized low density lipoprotein in nonrheumatic stenotic aortic valves. Arter. Thromb. Vasc. Biol. 1999, 19, 1218–1222. [Google Scholar] [CrossRef] [PubMed]
  31. Mohty, D.; Pibarot, P.; Després, J.P.; Côté, C.; Arsenault, B.; Cartier, A.; Cosnay, P.; Couture, C.; Mathieu, P. Association between plasma LDL particle size, valvular accumulation of oxidized LDL, and inflammation in patients with aortic stenosis. Arter. Thromb. Vasc. Biol. 2008, 28, 187–193. [Google Scholar] [CrossRef]
  32. Sucosky, P.; Balachandran, K.; Elhammali, A.; Jo, H.; Yoganathan, A.P. Altered shear stress stimulates upregulation of endothelial VCAM-1 and ICAM-1 in a BMP-4- and TGF-beta1-dependent pathway. Arter. Thromb. Vasc. Biol. 2009, 29, 254–260. [Google Scholar] [CrossRef]
  33. Lau, W.L.; Festing, M.H.; Giachelli, C.M. Phosphate and vascular calcification: Emerging role of the sodium-dependent phosphate co-transporter PiT-1. Thromb. Haemost. 2010, 104, 464–470. [Google Scholar] [CrossRef] [PubMed]
  34. Mathieu, P.; Bouchareb, R.; Boulanger, M.C. Innate and Adaptive Immunity in Calcific Aortic Valve Disease. J. Immunol. Res. 2015, 2015, 851945. [Google Scholar] [CrossRef]
  35. Hsu, H.; Shu, H.B.; Pan, M.G.; Goeddel, D.V. TRADD-TRAF2 and TRADD-FADD interactions define two distinct TNF receptor 1 signal transduction pathways. Cell 1996, 84, 299–308. [Google Scholar] [CrossRef] [PubMed]
  36. Lee, H.L.; Woo, K.M.; Ryoo, H.M.; Baek, J.H. Tumor necrosis factor-alpha increases alkaline phosphatase expression in vascular smooth muscle cells via MSX2 induction. Biochem. Biophys. Res. Commun. 2010, 391, 1087–1092. [Google Scholar] [CrossRef]
  37. Isoda, K.; Matsuki, T.; Kondo, H.; Iwakura, Y.; Ohsuzu, F. Deficiency of interleukin-1 receptor antagonist induces aortic valve disease in BALB/c mice. Arter. Thromb. Vasc. Biol. 2010, 30, 708–715. [Google Scholar] [CrossRef] [PubMed]
  38. Galeone, A.; Brunetti, G.; Oranger, A.; Greco, G.; Di Benedetto, A.; Mori, G.; Colucci, S.; Zallone, A.; Paparella, D.; Grano, M. Aortic valvular interstitial cells apoptosis and calcification are mediated by TNF-related apoptosis-inducing ligand. Int. J. Cardiol. 2013, 169, 296–304. [Google Scholar] [CrossRef]
  39. Kim, K.M. Calcification of matrix vesicles in human aortic valve and aortic media. Fed Proc 1976, 35, 156–162. [Google Scholar]
  40. Proudfoot, D.; Skepper, J.N.; Hegyi, L.; Bennett, M.R.; Shanahan, C.M.; Weissberg, P.L. Apoptosis regulates human vascular calcification in vitro: Evidence for initiation of vascular calcification by apoptotic bodies. Circ. Res. 2000, 87, 1055–1062. [Google Scholar] [CrossRef]
  41. New, S.E.; Goettsch, C.; Aikawa, M.; Marchini, J.F.; Shibasaki, M.; Yabusaki, K.; Libby, P.; Shanahan, C.M.; Croce, K.; Aikawa, E. Macrophage-derived matrix vesicles: An alternative novel mechanism for microcalcification in atherosclerotic plaques. Circ. Res. 2013, 113, 72–77. [Google Scholar] [CrossRef]
  42. Hjortnaes, J.; New, S.E.; Aikawa, E. Visualizing novel concepts of cardiovascular calcification. Trends Cardiovasc. Med. 2013, 23, 71–79. [Google Scholar] [CrossRef]
  43. New, S.E.; Aikawa, E. Molecular imaging insights into early inflammatory stages of arterial and aortic valve calcification. Circ. Res. 2011, 108, 1381–1391. [Google Scholar] [CrossRef]
  44. Aikawa, E.; Libby, P. A Rock and a Hard Place: Chiseling Away at the Multiple Mechanisms of Aortic Stenosis. Circulation 2017, 135, 1951–1955. [Google Scholar] [CrossRef]
  45. Decano, J.L.; Iwamoto, Y.; Goto, S.; Lee, J.Y.; Matamalas, J.T.; Halu, A.; Blaser, M.; Lee, L.H.; Pieper, B.; Chelvanambi, S.; et al. A disease-driver population within interstitial cells of human calcific aortic valves identified via single-cell and proteomic profiling. Cell Rep. 2022, 39, 110685. [Google Scholar] [CrossRef]
  46. Wang, G.; Qin, M.; Zhang, B.; Yan, Y.; Yang, F.; Chen, Q.; Liu, Y.; Qiao, F.; Ni, Y. Decreased expression of RPL15 and RPL18 exacerbated the calcification of valve interstitial cells during aortic valve calcification. Cell Biol. Int. 2023, 47, 1749–1759. [Google Scholar] [CrossRef]
  47. Chen, L.; Liu, H.; Sun, C.; Pei, J.; Li, J.; Li, Y.; Wei, K.; Wang, X.; Wang, P.; Li, F.; et al. A Novel LncRNA SNHG3 Promotes Osteoblast Differentiation Through BMP2 Upregulation in Aortic Valve Calcification. JACC Basic Transl. Sci. 2022, 7, 899–914. [Google Scholar] [CrossRef]
  48. Cai, Z.; Zhu, M.; Xu, L.; Wang, Y.; Xu, Y.; Yim, W.Y.; Cao, H.; Guo, R.; Qiu, X.; He, X.; et al. Directed Differentiation of Human Induced Pluripotent Stem Cells to Heart Valve Cells. Circulation 2024, 149, 1435–1456. [Google Scholar] [CrossRef]
  49. Huang, Y.; Jiang, C.; Chen, L.; Han, J.; Liu, M.; Zhou, T.; Dong, N.; Xu, K. Gli1 promotes the phenotypic transformation of valve interstitial cells through Hedgehog pathway activation exacerbating calcific aortic valve disease. Int. J. Biol. Sci. 2023, 19, 2053–2066. [Google Scholar] [CrossRef]
  50. Latif, N.; Quillon, A.; Sarathchandra, P.; McCormack, A.; Lozanoski, A.; Yacoub, M.H.; Chester, A.H. Modulation of human valve interstitial cell phenotype and function using a fibroblast growth factor 2 formulation. PLoS ONE 2015, 10, e0127844. [Google Scholar] [CrossRef] [PubMed]
  51. Witt, W.; Jannasch, A.; Burkhard, D.; Christ, T.; Ravens, U.; Brunssen, C.; Leuner, A.; Morawietz, H.; Matschke, K.; Waldow, T. Sphingosine-1-phosphate induces contraction of valvular interstitial cells from porcine aortic valves. Cardiovasc Res. 2012, 93, 490–497. [Google Scholar] [CrossRef] [PubMed]
  52. Osman, N.; Grande-Allen, K.J.; Ballinger, M.L.; Getachew, R.; Marasco, S.; O’Brien, K.D.; Little, P.J. Smad2-dependent glycosaminoglycan elongation in aortic valve interstitial cells enhances binding of LDL to proteoglycans. Cardiovasc Pathol. 2013, 22, 146–155. [Google Scholar] [CrossRef] [PubMed]
  53. Hutcheson, J.D.; Ryzhova, L.M.; Setola, V.; Merryman, W.D. 5-HT(2B) antagonism arrests non-canonical TGF-β1-induced valvular myofibroblast differentiation. J. Mol. Cell Cardiol. 2012, 53, 707–714. [Google Scholar] [CrossRef]
  54. Panizo, S.; Cardus, A.; Encinas, M.; Parisi, E.; Valcheva, P.; López-Ongil, S.; Coll, B.; Fernandez, E.; Valdivielso, J.M. RANKL increases vascular smooth muscle cell calcification through a RANK-BMP4-dependent pathway. Circ. Res. 2009, 104, 1041–1048. [Google Scholar] [CrossRef] [PubMed]
  55. Caira, F.C.; Stock, S.R.; Gleason, T.G.; McGee, E.C.; Huang, J.; Bonow, R.O.; Spelsberg, T.C.; McCarthy, P.M.; Rahimtoola, S.H.; Rajamannan, N.M. Human degenerative valve disease is associated with up-regulation of low-density lipoprotein receptor-related protein 5 receptor-mediated bone formation. J. Am. Coll Cardiol. 2006, 47, 1707–1712. [Google Scholar] [CrossRef] [PubMed]
  56. Garg, V.; Muth, A.N.; Ransom, J.F.; Schluterman, M.K.; Barnes, R.; King, I.N.; Grossfeld, P.D.; Srivastava, D. Mutations in NOTCH1 cause aortic valve disease. Nature 2005, 437, 270–274. [Google Scholar] [CrossRef] [PubMed]
  57. Miller, J.D.; Weiss, R.M.; Heistad, D.D. Calcific aortic valve stenosis: Methods, models, and mechanisms. Circ. Res. 2011, 108, 1392–1412. [Google Scholar] [CrossRef]
  58. Torre, M.; Hwang, D.H.; Padera, R.F.; Mitchell, R.N.; VanderLaan, P.A. Osseous and chondromatous metaplasia in calcific aortic valve stenosis. Cardiovasc. Pathol. 2016, 25, 18–24. [Google Scholar] [CrossRef]
  59. Mohler, E.R., 3rd; Gannon, F.; Reynolds, C.; Zimmerman, R.; Keane, M.G.; Kaplan, F.S. Bone formation and inflammation in cardiac valves. Circulation 2001, 103, 1522–1528. [Google Scholar] [CrossRef]
  60. Hutcheson, J.D.; Aikawa, E.; Merryman, W.D. Potential drug targets for calcific aortic valve disease. Nat. Rev. Cardiol. 2014, 11, 218–231. [Google Scholar] [CrossRef]
  61. Yanai, S.; Endo, S. Functional Aging in Male C57BL/6J Mice Across the Life-Span: A Systematic Behavioral Analysis of Motor, Emotional, and Memory Function to Define an Aging Phenotype. Front. Aging Neurosci. 2021, 13, 697621. [Google Scholar] [CrossRef]
  62. Aikawa, E.; Aikawa, M.; Libby, P.; Figueiredo, J.L.; Rusanescu, G.; Iwamoto, Y.; Fukuda, D.; Kohler, R.H.; Shi, G.P.; Jaffer, F.A.; et al. Arterial and aortic valve calcification abolished by elastolytic cathepsin S deficiency in chronic renal disease. Circulation 2009, 119, 1785–1794. [Google Scholar] [CrossRef]
  63. Drolet, M.C.; Roussel, E.; Deshaies, Y.; Couet, J.; Arsenault, M. A high fat/high carbohydrate diet induces aortic valve disease in C57BL/6J mice. J. Am. Coll. Cardiol. 2006, 47, 850–855. [Google Scholar] [CrossRef]
  64. Weiss, R.M.; Ohashi, M.; Miller, J.D.; Young, S.G.; Heistad, D.D. Calcific aortic valve stenosis in old hypercholesterolemic mice. Circulation 2006, 114, 2065–2069. [Google Scholar] [CrossRef] [PubMed]
  65. Lee, Y.T.; Lin, H.Y.; Chan, Y.W.; Li, K.H.; To, O.T.; Yan, B.P.; Liu, T.; Li, G.; Wong, W.T.; Keung, W.; et al. Mouse models of atherosclerosis: A historical perspective and recent advances. Lipids Health Dis. 2017, 16, 12. [Google Scholar] [CrossRef]
  66. Golforoush, P.; Yellon, D.M.; Davidson, S.M. Mouse models of atherosclerosis and their suitability for the study of myocardial infarction. Basic Res. Cardiol. 2020, 115, 73. [Google Scholar] [CrossRef]
  67. Getz, G.S.; Reardon, C.A. Apoprotein E and Reverse Cholesterol Transport. Int. J. Mol. Sci. 2018, 19, 3479. [Google Scholar] [CrossRef]
  68. Ilyas, I.; Little, P.J.; Liu, Z.; Xu, Y.; Kamato, D.; Berk, B.C.; Weng, J.; Xu, S. Mouse models of atherosclerosis in translational research. Trends Pharmacol. Sci. 2022, 43, 920–939. [Google Scholar] [CrossRef] [PubMed]
  69. Miller, J.D.; Weiss, R.M.; Serrano, K.M.; Brooks, R.M., 2nd; Berry, C.J.; Zimmerman, K.; Young, S.G.; Heistad, D.D. Lowering plasma cholesterol levels halts progression of aortic valve disease in mice. Circulation 2009, 119, 2693–2701. [Google Scholar] [CrossRef] [PubMed]
  70. Miller, J.D.; Weiss, R.M.; Serrano, K.M.; Castaneda, L.E.; Brooks, R.M.; Zimmerman, K.; Heistad, D.D. Evidence for active regulation of pro-osteogenic signaling in advanced aortic valve disease. Arterioscler. Thromb. Vasc. Biol. 2010, 30, 2482–2486. [Google Scholar] [CrossRef]
  71. Rattazzi, M.; Faggin, E.; Bertacco, E.; Nardin, C.; Pagliani, L.; Plebani, M.; Cinetto, F.; Guidolin, D.; Puato, M.; Pauletto, P. Warfarin, but not rivaroxaban, promotes the calcification of the aortic valve in ApoE−/− mice. Cardiovasc. Ther. 2018, 36, e12438. [Google Scholar] [CrossRef]
  72. Weisell, J.; Ruotsalainen, A.K.; Näpänkangas, J.; Jauhiainen, M.; Rysä, J. Menaquinone 4 increases plasma lipid levels in hypercholesterolemic mice. Sci. Rep. 2021, 11, 3014. [Google Scholar] [CrossRef]
  73. Zadelaar, S.; Kleemann, R.; Verschuren, L.; de Vries-Van der Weij, J.; van der Hoorn, J.; Princen, H.M.; Kooistra, T. Mouse models for atherosclerosis and pharmaceutical modifiers. Arter. Thromb. Vasc. Biol. 2007, 27, 1706–1721. [Google Scholar] [CrossRef] [PubMed]
  74. Jongejan, Y.K.; Eikenboom, J.C.J.; Gijbels, M.J.J.; Berbée, J.F.P.; van Vlijmen, B.J.M. Atherothrombosis model by silencing of protein C in APOE*3-Leiden.CETP transgenic mice. J. Thromb. Thrombolysis 2021, 52, 715–719. [Google Scholar] [CrossRef] [PubMed]
  75. Thanassoulis, G. Lipoprotein (a) in calcific aortic valve disease: From genomics to novel drug target for aortic stenosis. J. Lipid Res. 2016, 57, 917–924. [Google Scholar] [CrossRef]
  76. Carracedo, M.; Pawelzik, S.C.; Artiach, G.; Pouwer, M.G.; Plunde, O.; Saliba-Gustafsson, P.; Ehrenborg, E.; Eriksson, P.; Pieterman, E.; Stenke, L.; et al. The tyrosine kinase inhibitor nilotinib targets the discoidin domain receptor DDR2 in calcific aortic valve stenosis. Br. J. Pharmacol. 2022, 179, 4709–4721. [Google Scholar] [CrossRef]
  77. Xu, M.; Wu, X.; Liu, Z.; Ding, Y.; Kong, W.; Little, P.J.; Xu, S.; Weng, J. A novel mouse model of diabetes, atherosclerosis and fatty liver disease using an AAV8-PCSK9-D377Y injection and dietary manipulation in db/db mice. Biochem. Biophys. Res. Commun. 2022, 622, 163–169. [Google Scholar] [CrossRef]
  78. Guarnieri, F.; Kulp, J.L., Jr.; Kulp, J.L., 3rd; Cloudsdale, I.S. Fragment-based design of small molecule PCSK9 inhibitors using simulated annealing of chemical potential simulations. PLoS ONE 2019, 14, e0225780. [Google Scholar] [CrossRef]
  79. Xia, X.D.; Peng, Z.S.; Gu, H.M.; Wang, M.; Wang, G.Q.; Zhang, D.W. Regulation of PCSK9 Expression and Function: Mechanisms and Therapeutic Implications. Front. Cardiovasc. Med. 2021, 8, 764038. [Google Scholar] [CrossRef]
  80. Yoshioka, M.; Yuasa, S.; Matsumura, K.; Kimura, K.; Shiomi, T.; Kimura, N.; Shukunami, C.; Okada, Y.; Mukai, M.; Shin, H.; et al. Chondromodulin-I maintains cardiac valvular function by preventing angiogenesis. Nat. Med. 2006, 12, 1151–1159. [Google Scholar] [CrossRef]
  81. Winnicki, A.; Gadd, J.; Ohanyan, V.; Hernandez, G.; Wang, Y.; Enrick, M.; McKillen, H.; Kiedrowski, M.; Kundu, D.; Kegecik, K.; et al. Role of endothelial CXCR4 in the development of aortic valve stenosis. Front. Cardiovasc. Med. 2022, 9, 971321. [Google Scholar] [CrossRef]
  82. Nus, M.; MacGrogan, D.; Martínez-Poveda, B.; Benito, Y.; Casanova, J.C.; Fernández-Avilés, F.; Bermejo, J.; de la Pompa, J.L. Diet-induced aortic valve disease in mice haploinsufficient for the Notch pathway effector RBPJK/CSL. Arter. Thromb. Vasc. Biol. 2011, 31, 1580–1588. [Google Scholar] [CrossRef] [PubMed]
  83. Han, Y.; Zhang, J.; Yang, Z.; Jian, W.; Zhu, Y.; Gao, S.; Liu, Y.; Li, Y.; He, S.; Zhang, C.; et al. Palmdelphin Deficiency Evokes NF-κB Signaling in Valvular Endothelial Cells and Aggravates Aortic Valvular Remodeling. JACC Basic Transl. Sci. 2023, 8, 1457–1472. [Google Scholar] [CrossRef]
  84. Cheek, J.D.; Wirrig, E.E.; Alfieri, C.M.; James, J.F.; Yutzey, K.E. Differential activation of valvulogenic, chondrogenic, and osteogenic pathways in mouse models of myxomatous and calcific aortic valve disease. J. Mol. Cell. Cardiol. 2012, 52, 689–700. [Google Scholar] [CrossRef]
  85. Yoon, D.; Choi, B.; Kim, J.E.; Kim, E.Y.; Chung, S.H.; Min, H.J.; Sung, Y.; Chang, E.J.; Song, J.K. Autotaxin inhibition attenuates the aortic valve calcification by suppressing inflammation-driven fibro-calcific remodeling of valvular interstitial cells. BMC Med. 2024, 22, 122. [Google Scholar] [CrossRef]
  86. Adesanya, T.M.A.; Russell, M.; Park, K.H.; Zhou, X.; Sermersheim, M.A.; Gumpper, K.; Koenig, S.N.; Tan, T.; Whitson, B.A.; Janssen, P.M.L.; et al. MG 53 Protein Protects Aortic Valve Interstitial Cells From Membrane Injury and Fibrocalcific Remodeling. J. Am. Heart Assoc. 2019, 8, e009960. [Google Scholar] [CrossRef]
  87. Rattazzi, M.; Bertacco, E.; Del Vecchio, A.; Puato, M.; Faggin, E.; Pauletto, P. Aortic valve calcification in chronic kidney disease. Nephrol. Dial Transpl. 2013, 28, 2968–2976. [Google Scholar] [CrossRef] [PubMed]
  88. Tripathi, A.; Wang, Y.; Jerrell, J.M. Population-based treated prevalence, risk factors, and outcomes of bicuspid aortic valve in a pediatric Medicaid cohort. Ann. Pediatr. Cardiol. 2018, 11, 119–124. [Google Scholar] [CrossRef] [PubMed]
  89. Roberts, W.C.; Ko, J.M. Frequency by decades of unicuspid, bicuspid, and tricuspid aortic valves in adults having isolated aortic valve replacement for aortic stenosis, with or without associated aortic regurgitation. Circulation 2005, 111, 920–925. [Google Scholar] [CrossRef]
  90. Lee, T.C.; Zhao, Y.D.; Courtman, D.W.; Stewart, D.J. Abnormal aortic valve development in mice lacking endothelial nitric oxide synthase. Circulation 2000, 101, 2345–2348. [Google Scholar] [CrossRef]
  91. Nigam, V.; Srivastava, D. Notch1 represses osteogenic pathways in aortic valve cells. J. Mol. Cell. Cardiol. 2009, 47, 828–834. [Google Scholar] [CrossRef] [PubMed]
  92. Blaser, M.C.; Wei, K.; Adams, R.L.E.; Zhou, Y.Q.; Caruso, L.L.; Mirzaei, Z.; Lam, A.Y.; Tam, R.K.K.; Zhang, H.; Heximer, S.P.; et al. Deficiency of Natriuretic Peptide Receptor 2 Promotes Bicuspid Aortic Valves, Aortic Valve Disease, Left Ventricular Dysfunction, and Ascending Aortic Dilatations in Mice. Circ. Res. 2018, 122, 405–416. [Google Scholar] [CrossRef]
  93. Ahmad, A.A.; Ghim, M.; Toczek, J.; Neishabouri, A.; Ojha, D.; Zhang, Z.; Gona, K.; Raza, M.Z.; Jung, J.J.; Kukreja, G.; et al. Multimodality Imaging of Aortic Valve Calcification and Function in a Murine Model of Calcific Aortic Valve Disease and Bicuspid Aortic Valve. J. Nucl. Med. Off. Publ. Soc. Nucl. Med. 2023, 64, 1487–1494. [Google Scholar] [CrossRef]
  94. Peterson, J.C.; Chughtai, M.; Wisse, L.J.; Gittenberger-de Groot, A.C.; Feng, Q.; Goumans, M.T.H.; VanMunsteren, J.C.; Jongbloed, M.R.M.; DeRuiter, M.C. Bicuspid aortic valve formation: Nos3 mutation leads to abnormal lineage patterning of neural crest cells and the second heart field. Dis. Models Mech. 2018, 11, dmm034637. [Google Scholar] [CrossRef]
  95. El Accaoui, R.N.; Gould, S.T.; Hajj, G.P.; Chu, Y.; Davis, M.K.; Kraft, D.C.; Lund, D.D.; Brooks, R.M.; Doshi, H.; Zimmerman, K.A.; et al. Aortic valve sclerosis in mice deficient in endothelial nitric oxide synthase. Am. J. Physiol. Heart Circ. Physiol. 2014, 306, H1302–H1313. [Google Scholar] [CrossRef]
  96. Lin, X.; Liu, X.; Wang, L.; Jiang, J.; Sun, Y.; Zhu, Q.; Chen, Z.; He, Y.; Hu, P.; Xu, Q.; et al. Targeted next-generation sequencing identified ADAMTS5 as novel genetic substrate in patients with bicuspid aortic valve. Int. J. Cardiol. 2018, 252, 150–155. [Google Scholar] [CrossRef]
  97. Siu, S.C.; Silversides, C.K. Bicuspid aortic valve disease. J. Am. Coll. Cardiol. 2010, 55, 2789–2800. [Google Scholar] [CrossRef] [PubMed]
  98. Dutta, P.; James, J.F.; Kazik, H.; Lincoln, J. Genetic and Developmental Contributors to Aortic Stenosis. Circ Res 2021, 128, 1330–1343. [Google Scholar] [CrossRef]
  99. Honda, S.; Miyamoto, T.; Watanabe, T.; Narumi, T.; Kadowaki, S.; Honda, Y.; Otaki, Y.; Hasegawa, H.; Netsu, S.; Funayama, A.; et al. A novel mouse model of aortic valve stenosis induced by direct wire injury. Arterioscler. Thromb. Vasc. Biol. 2014, 34, 270–278. [Google Scholar] [CrossRef] [PubMed]
  100. Niepmann, S.T.; Steffen, E.; Zietzer, A.; Adam, M.; Nordsiek, J.; Gyamfi-Poku, I.; Piayda, K.; Sinning, J.M.; Baldus, S.; Kelm, M.; et al. Graded murine wire-induced aortic valve stenosis model mimics human functional and morphological disease phenotype. Clin. Res. Cardiol. Off. J. Ger. Card. Soc. 2019, 108, 847–856. [Google Scholar] [CrossRef] [PubMed]
  101. Tanaka, K.; Sata, M.; Fukuda, D.; Suematsu, Y.; Motomura, N.; Takamoto, S.; Hirata, Y.; Nagai, R. Age-associated aortic stenosis in apolipoprotein E-deficient mice. J. Am. Coll. Cardiol. 2005, 46, 134–141. [Google Scholar] [CrossRef]
  102. Aikawa, E.; Nahrendorf, M.; Sosnovik, D.; Lok, V.M.; Jaffer, F.A.; Aikawa, M.; Weissleder, R. Multimodality molecular imaging identifies proteolytic and osteogenic activities in early aortic valve disease. Circulation 2007, 115, 377–386. [Google Scholar] [CrossRef]
  103. Dwinell, M.R. Online tools for understanding rat physiology. Brief. Bioinform. 2010, 11, 431–439. [Google Scholar] [CrossRef] [PubMed]
  104. Aitman, T.J.; Critser, J.K.; Cuppen, E.; Dominiczak, A.; Fernandez-Suarez, X.M.; Flint, J.; Gauguier, D.; Geurts, A.M.; Gould, M.; Harris, P.C.; et al. Progress and prospects in rat genetics: A community view. Nat. Genet. 2008, 40, 516–522. [Google Scholar] [CrossRef] [PubMed]
  105. Cunha, L.F.; Ongaratto, M.A.; Endres, M.; Barschak, A.G. Modelling hypercholesterolaemia in rats using high cholesterol diet. Int. J. Exp. Pathol. 2021, 102, 74–79. [Google Scholar] [CrossRef]
  106. Reaves, S.K.; Wu, J.Y.; Wu, Y.; Fanzo, J.C.; Wang, Y.R.; Lei, P.P.; Lei, K.Y. Regulation of intestinal apolipoprotein B mRNA editing levels by a zinc-deficient diet and cDNA cloning of editing protein in hamsters. J. Nutr. 2000, 130, 2166–2173. [Google Scholar] [CrossRef]
  107. Yang, X.; Zeng, J.; Xie, K.; Su, S.; Guo, Y.; Zhang, H.; Chen, J.; Ma, Z.; Xiao, Z.; Zhu, P.; et al. Advanced glycation end product-modified low-density lipoprotein promotes pro-osteogenic reprogramming via RAGE/NF-κB pathway and exaggerates aortic valve calcification in hamsters. Mol. Med. 2024, 30, 76. [Google Scholar] [CrossRef]
  108. Li, N.; Bai, Y.; Zhou, G.; Ma, Y.; Tan, M.; Qiao, F.; Li, X.; Shen, M.; Song, X.; Zhao, X.; et al. miR-214 Attenuates Aortic Valve Calcification by Regulating Osteogenic Differentiation of Valvular Interstitial Cells. Mol. Ther. Nucleic Acids 2020, 22, 971–980. [Google Scholar] [CrossRef]
  109. Price, P.A.; Faus, S.A.; Williamson, M.K. Warfarin causes rapid calcification of the elastic lamellae in rat arteries and heart valves. Arter. Thromb. Vasc. Biol. 1998, 18, 1400–1407. [Google Scholar] [CrossRef]
  110. Côté, N.; El Husseini, D.; Pépin, A.; Bouvet, C.; Gilbert, L.A.; Audet, A.; Fournier, D.; Pibarot, P.; Moreau, P.; Mathieu, P. Inhibition of ectonucleotidase with ARL67156 prevents the development of calcific aortic valve disease in warfarin-treated rats. Eur. J. Pharmacol. 2012, 689, 139–146. [Google Scholar] [CrossRef]
  111. Roosens, B.; Droogmans, S.; Hostens, J.; Somja, J.; Delvenne, E.; Hernot, S.; Bala, G.; Degaillier, C.; Caveliers, V.; Delvenne, P.; et al. Integrated backscatter for the in vivo quantification of supraphysiological vitamin D(3)-induced cardiovascular calcifications in rats. Cardiovasc. Toxicol. 2011, 11, 244–252. [Google Scholar] [CrossRef] [PubMed]
  112. Shuvy, M.; Abedat, S.; Beeri, R.; Danenberg, H.D.; Planer, D.; Ben-Dov, I.Z.; Meir, K.; Sosna, J.; Lotan, C. Uraemic hyperparathyroidism causes a reversible inflammatory process of aortic valve calcification in rats. Cardiovasc. Res. 2008, 79, 492–499. [Google Scholar] [CrossRef]
  113. Bonnard, B.; Pieronne-Deperrois, M.; Djerada, Z.; Elmoghrabi, S.; Kolkhof, P.; Ouvrard-Pascaud, A.; Mulder, P.; Jaisser, F.; Messaoudi, S. Mineralocorticoid receptor antagonism improves diastolic dysfunction in chronic kidney disease in mice. J. Mol. Cell. Cardiol. 2018, 121, 124–133. [Google Scholar] [CrossRef]
  114. Wu, J.; Li, W.H.; Wang, W.R.; Jin, X.Q.; Liu, E.Q. Proteomics Analysis of Lipid Metabolism and Inflammatory Response in the Liver of Rabbits fed on a High Cholesterol Diet. Cell Biochem. Biophys. 2023, 81, 231–242. [Google Scholar] [CrossRef]
  115. Cimini, M.; Boughner, D.R.; Ronald, J.A.; Aldington, L.; Rogers, K.A. Development of aortic valve sclerosis in a rabbit model of atherosclerosis: An immunohistochemical and histological study. J. Heart. Valve Dis. 2005, 14, 365–375. [Google Scholar]
  116. Ngo, D.T.; Stafford, I.; Kelly, D.J.; Sverdlov, A.L.; Wuttke, R.D.; Weedon, H.; Nightingale, A.K.; Rosenkranz, A.C.; Smith, M.D.; Chirkov, Y.Y.; et al. Vitamin D(2) supplementation induces the development of aortic stenosis in rabbits: Interactions with endothelial function and thioredoxin-interacting protein. Eur. J. Pharmacol. 2008, 590, 290–296. [Google Scholar] [CrossRef] [PubMed]
  117. Hara, T.; Tsukada, N.; Okano, M.; Ishida, T.; Hirata, K.I.; Shiomi, M. Progression of calcific aortic valve sclerosis in WHHLMI rabbits. Atherosclerosis 2018, 273, 8–14. [Google Scholar] [CrossRef]
  118. Lu, R.; Yuan, T.; Wang, Y.; Zhang, T.; Yuan, Y.; Wu, D.; Zhou, M.; He, Z.; Lu, Y.; Chen, Y.; et al. Spontaneous severe hypercholesterolemia and atherosclerosis lesions in rabbits with deficiency of low-density lipoprotein receptor (LDLR) on exon 7. eBioMedicine 2018, 36, 29–38. [Google Scholar] [CrossRef] [PubMed]
  119. Rajamannan, N.M.; Subramaniam, M.; Caira, F.; Stock, S.R.; Spelsberg, T.C. Atorvastatin inhibits hypercholesterolemia-induced calcification in the aortic valves via the Lrp5 receptor pathway. Circulation 2005, 112, I229–I234. [Google Scholar] [CrossRef]
  120. Sider, K.L.; Zhu, C.; Kwong, A.V.; Mirzaei, Z.; de Langé, C.F.; Simmons, C.A. Evaluation of a porcine model of early aortic valve sclerosis. Cardiovasc. Pathol. Off. J. Soc. Cardiovasc. Pathol. 2014, 23, 289–297. [Google Scholar] [CrossRef] [PubMed]
  121. Zhu, J.; Chen, C.; Yang, B.; Guo, Y.; Ai, H.; Ren, J.; Peng, Z.; Tu, Z.; Yang, X.; Meng, Q.; et al. A systems genetics study of swine illustrates mechanisms underlying human phenotypic traits. BMC Genom. 2015, 16, 88. [Google Scholar] [CrossRef]
  122. Zabirnyk, A.; Perez, M.D.M.; Blasco, M.; Stensløkken, K.O.; Ferrer, M.D.; Salcedo, C.; Vaage, J. A Novel Ex Vivo Model of Aortic Valve Calcification. A Preliminary Report. Front. Pharmacol. 2020, 11, 568764. [Google Scholar] [CrossRef]
  123. Porras, A.M.; Shanmuganayagam, D.; Meudt, J.J.; Krueger, C.G.; Hacker, T.A.; Rahko, P.S.; Reed, J.D.; Masters, K.S. Development of Aortic Valve Disease in Familial Hypercholesterolemic Swine: Implications for Elucidating Disease Etiology. J. Am. Heart. Assoc. 2015, 4, e002254, Erratum in J. Am. Heart Assoc. 2016, 5, 10. [Google Scholar] [CrossRef]
  124. Chen, J.H.; Chen, W.L.; Sider, K.L.; Yip, C.Y.; Simmons, C.A. β-catenin mediates mechanically regulated, transforming growth factor-β1-induced myofibroblast differentiation of aortic valve interstitial cells. Arter. Thromb. Vasc. Biol. 2011, 31, 590–597. [Google Scholar] [CrossRef] [PubMed]
  125. Huang, L.; Hua, Z.; Xiao, H.; Cheng, Y.; Xu, K.; Gao, Q.; Xia, Y.; Liu, Y.; Zhang, X.; Zheng, X.; et al. CRISPR/Cas9-mediated ApoE−/− and LDLR−/− double gene knockout in pigs elevates serum LDL-C and TC levels. Oncotarget 2017, 8, 37751–37760. [Google Scholar] [CrossRef]
  126. Wang, K.; Zhou, X.; Zhou, Z.; Tarakji, K.; Carneiro, M.; Penn, M.S.; Murray, D.; Klein, A.; Humphries, R.G.; Turner, J.; et al. Blockade of the platelet P2Y12 receptor by AR-C69931MX sustains coronary artery recanalization and improves the myocardial tissue perfusion in a canine thrombosis model. Arter. Thromb. Vasc. Biol. 2003, 23, 357–362. [Google Scholar] [CrossRef] [PubMed]
  127. Zhao, H.; Liu, E.; Zhang, Y.Q. Dog models of human atherosclerotic cardiovascular diseases. Mamm. Genome 2023, 34, 262–269. [Google Scholar] [CrossRef]
  128. Maldonado, E.N.; Romero, J.R.; Ochoa, B.; Aveldaño, M.I. Lipid and fatty acid composition of canine lipoproteins. Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2001, 128, 719–729. [Google Scholar] [CrossRef]
  129. Hasiwa, N.; Bailey, J.; Clausing, P.; Daneshian, M.; Eileraas, M.; Farkas, S.; Gyertyán, I.; Hubrecht, R.; Kobel, W.; Krummenacher, G.; et al. Critical evaluation of the use of dogs in biomedical research and testing in Europe. Altex 2011, 28, 326–340. [Google Scholar] [CrossRef] [PubMed]
  130. Hamlin, R.L. Animal models of ventricular arrhythmias. Pharmacol. Ther. 2007, 113, 276–295. [Google Scholar] [CrossRef]
  131. Bowley, G.; Kugler, E.; Wilkinson, R.; Lawrie, A.; van Eeden, F.; Chico, T.J.A.; Evans, P.C.; Noël, E.S.; Serbanovic-Canic, J. Zebrafish as a tractable model of human cardiovascular disease. Br. J. Pharmacol. 2022, 179, 900–917. [Google Scholar] [CrossRef]
  132. Gollmann-Tepeköylü, C.; Graber, M.; Hirsch, J.; Mair, S.; Naschberger, A.; Pölzl, L.; Nägele, F.; Kirchmair, E.; Degenhart, G.; Demetz, E.; et al. Toll-Like Receptor 3 Mediates Aortic Stenosis Through a Conserved Mechanism of Calcification. Circulation 2023, 147, 1518–1533. [Google Scholar] [CrossRef]
  133. Flameng, W.; Meuris, B.; De Visscher, G.; Cunanan, C.; Lane, E.; Verbeken, E.; Herijgers, P.; Herregods, M.C. Trilogy pericardial valve: Hemodynamic performance and calcification in adolescent sheep. Ann. Thorac. Surg. 2008, 85, 587–592. [Google Scholar] [CrossRef] [PubMed]
  134. Goecke, T.; Theodoridis, K.; Tudorache, I.; Ciubotaru, A.; Cebotari, S.; Ramm, R.; Höffler, K.; Sarikouch, S.; Vásquez Rivera, A.; Haverich, A.; et al. In vivo performance of freeze-dried decellularized pulmonary heart valve allo- and xenografts orthotopically implanted into juvenile sheep. Acta Biomater. 2018, 68, 41–52. [Google Scholar] [CrossRef] [PubMed]
  135. Cottignoli, V.; Cavarretta, E.; Salvador, L.; Valfré, C.; Maras, A. Morphological and chemical study of pathological deposits in human aortic and mitral valve stenosis: A biomineralogical contribution. Pathol. Res. Int. 2015, 2015, 342984. [Google Scholar] [CrossRef]
  136. Bowler, M.A.; Merryman, W.D. In vitro models of aortic valve calcification: Solidifying a system. Cardiovasc. Pathol. 2015, 24, 1–10. [Google Scholar] [CrossRef]
  137. Schoen, F.J.; Levy, R.J. Calcification of tissue heart valve substitutes: Progress toward understanding and prevention. Ann. Thorac. Surg. 2005, 79, 1072–1080. [Google Scholar] [CrossRef]
  138. Doello, K. A new pentachrome method for the simultaneous staining of collagen and sulfated mucopolysaccharides. Yale J. Biol. Med. 2014, 87, 341–347. [Google Scholar]
  139. Yousry, M.; Rickenlund, A.; Petrini, J.; Gustavsson, T.; Prahl, U.; Liska, J.; Eriksson, P.; Franco-Cereceda, A.; Eriksson, M.J.; Caidahl, K. Real-time imaging required for optimal echocardiographic assessment of aortic valve calcification. Clin. Physiol. Funct. Imaging 2012, 32, 470–475. [Google Scholar] [CrossRef]
  140. Gao, S.; Ho, D.; Vatner, D.E.; Vatner, S.F. Echocardiography in Mice. Curr. Protoc. Mouse Biol. 2011, 1, 71–83. [Google Scholar] [CrossRef]
  141. Becker, K. Wire Injury-Induced Moderate Aortic Valve Stenosis in Mice Is Accompanied by a Chronic Systemic Inflammatory Reaction. Cells 2025, 14, 883. [Google Scholar] [CrossRef]
  142. Billig, H.; Schmitt, J.; Singer, L.; Bourauel, C.; Schildberg, F.A.; Masson, W.; Lübbering, N.; Vogel, W.; Perner, S.; Stei, M.; et al. Aortic valve stenosis and osteoporosis: Insights from a mouse model. BMC Cardiovasc. Disord. 2025, 25, 562. [Google Scholar] [CrossRef] [PubMed]
  143. de Bruijn, S.F.; Agema, W.R.; Lammers, G.J.; van der Wall, E.E.; Wolterbeek, R.; Holman, E.R.; Bollen, E.L.; Bax, J.J. Transesophageal echocardiography is superior to transthoracic echocardiography in management of patients of any age with transient ischemic attack or stroke. Stroke 2006, 37, 2531–2534. [Google Scholar] [CrossRef]
  144. Kang, D.H.; Park, S.J.; Rim, J.H.; Yun, S.C.; Kim, D.H.; Song, J.M.; Choo, S.J.; Park, S.W.; Song, J.K.; Lee, J.W.; et al. Early surgery versus conventional treatment in asymptomatic very severe aortic stenosis. Circulation 2010, 121, 1502–1509. [Google Scholar] [CrossRef]
  145. Shen, M.; Tastet, L.; Capoulade, R.; Larose, É.; Bédard, É.; Arsenault, M.; Chetaille, P.; Dumesnil, J.G.; Mathieu, P.; Clavel, M.A.; et al. Effect of age and aortic valve anatomy on calcification and haemodynamic severity of aortic stenosis. Heart 2017, 103, 32–39. [Google Scholar] [CrossRef] [PubMed]
  146. Aggarwal, S.R.; Clavel, M.A.; Messika-Zeitoun, D.; Cueff, C.; Malouf, J.; Araoz, P.A.; Mankad, R.; Michelena, H.; Vahanian, A.; Enriquez-Sarano, M. Sex differences in aortic valve calcification measured by multidetector computed tomography in aortic stenosis. Circ. Cardiovasc Imaging 2013, 6, 40–47. [Google Scholar] [CrossRef] [PubMed]
  147. Cartlidge, T.R.; Bing, R.; Kwiecinski, J.; Guzzetti, E.; Pawade, T.A.; Doris, M.K.; Adamson, P.D.; Massera, D.; Lembo, M.; Peeters, F.; et al. Contrast-enhanced computed tomography assessment of aortic stenosis. Heart 2021, 107, 1905–1911. [Google Scholar] [CrossRef]
  148. Orzechowska, S.; Wróbel, A.; Goncerz, G.; Podolec, P.; Rokita, E. Physicochemical and micro-tomographic characterization of inorganic deposits associated with aortic stenosis. J. Heart. Valve Dis. 2014, 23, 40–47. [Google Scholar] [PubMed]
  149. Chitsaz, S.; Gundiah, N.; Blackshear, C.; Tegegn, N.; Yan, K.S.; Azadani, A.N.; Hope, M.; Tseng, E.E. Correlation of calcification on excised aortic valves by micro-computed tomography with severity of aortic stenosis. J. Heart. Valve Dis. 2012, 21, 320–327. [Google Scholar]
  150. Thomas, I.C.; Forbang, N.I.; Criqui, M.H. The evolving view of coronary artery calcium and cardiovascular disease risk. Clin. Cardiol. 2018, 41, 144–150. [Google Scholar] [CrossRef]
  151. Lembo, M.; Joshi, S.S.; Geers, J.; Bing, R.; Carnevale, L.; Pawade, T.A.; Doris, M.K.; Tzolos, E.; Grodecki, K.; Cadet, S.; et al. Quantitative Computed Tomography Angiography for the Evaluation of Valvular Fibrocalcific Volume in Aortic Stenosis. JACC Cardiovasc. Imaging 2024, 17, 1351–1362. [Google Scholar] [CrossRef]
  152. Rajamannan, N.M.; Subramaniam, M.; Stock, S.R.; Stone, N.J.; Springett, M.; Ignatiev, K.I.; McConnell, J.P.; Singh, R.J.; Bonow, R.O.; Spelsberg, T.C. Atorvastatin inhibits calcification and enhances nitric oxide synthase production in the hypercholesterolaemic aortic valve. Heart 2005, 91, 806–810. [Google Scholar] [CrossRef]
  153. Wait, J.M.; Tomita, H.; Burk, L.M.; Lu, J.; Zhou, O.Z.; Maeda, N.; Lee, Y.Z. Detection of aortic arch calcification in apolipoprotein E-null mice using carbon nanotube-based micro-CT system. J. Am. Heart Assoc. 2013, 2, e003358. [Google Scholar] [CrossRef]
  154. Mittal, T.K.; Marcus, N. Imaging diagnosis of aortic stenosis. Clin. Radiol. 2021, 76, 3–14. [Google Scholar] [CrossRef] [PubMed]
  155. Francone, M.; Budde, R.P.J.; Bremerich, J.; Dacher, J.N.; Loewe, C.; Wolf, F.; Natale, L.; Pontone, G.; Redheuil, A.; Vliegenthart, R.; et al. CT and MR imaging prior to transcatheter aortic valve implantation: Standardisation of scanning protocols, measurements and reporting-a consensus document by the European Society of Cardiovascular Radiology (ESCR). Eur. Radiol. 2020, 30, 2627–2650. [Google Scholar] [CrossRef] [PubMed]
  156. Wink, M.H.; Wijkstra, H.; De La Rosette, J.J.; Grimbergen, C.A. Ultrasound imaging and contrast agents: A safe alternative to MRI? Minim. Invasive Ther. Allied Technol. 2006, 15, 93–100. [Google Scholar] [CrossRef] [PubMed]
  157. Soulat, G.; McCarthy, P.; Markl, M. 4D Flow with MRI. Annu. Rev. Biomed. Eng. 2020, 22, 103–126. [Google Scholar] [CrossRef]
  158. Lo Presti, F.; Guzzardi, D.G.; Bancone, C.; Fedak, P.W.M.; Della Corte, A. The science of BAV aortopathy. Prog. Cardiovasc. Dis. 2020, 63, 465–474. [Google Scholar] [CrossRef]
  159. Taralli, S.; Caldarella, C.; Lorusso, M.; Scolozzi, V.; Altini, C.; Rubini, G.; Calcagni, M.L. Comparison between 18F-FDG and 18F-NaF PET imaging for assessing bone metastases in breast cancer patients: A literature review. Clin. Transl. Imaging 2020, 8, 65–78. [Google Scholar] [CrossRef]
  160. Rudd, J.H.; Narula, J.; Strauss, H.W.; Virmani, R.; Machac, J.; Klimas, M.; Tahara, N.; Fuster, V.; Warburton, E.A.; Fayad, Z.A.; et al. Imaging atherosclerotic plaque inflammation by fluorodeoxyglucose with positron emission tomography: Ready for prime time? J. Am. Coll. Cardiol. 2010, 55, 2527–2535. [Google Scholar] [CrossRef] [PubMed]
  161. Leppänen, O.; Björnheden, T.; Evaldsson, M.; Borén, J.; Wiklund, O.; Levin, M. ATP depletion in macrophages in the core of advanced rabbit atherosclerotic plaques in vivo. Atherosclerosis 2006, 188, 323–330. [Google Scholar] [CrossRef]
  162. Weiss, R.M.; Miller, J.D.; Heistad, D.D. Fibrocalcific aortic valve disease: Opportunity to understand disease mechanisms using mouse models. Circ. Res. 2013, 113, 209–222. [Google Scholar] [CrossRef]
  163. Zhang, Z.; Machac, J.; Helft, G.; Worthley, S.G.; Tang, C.; Zaman, A.G.; Rodriguez, O.J.; Buchsbaum, M.S.; Fuster, V.; Badimon, J.J. Non-invasive imaging of atherosclerotic plaque macrophage in a rabbit model with F-18 FDG PET: A histopathological correlation. BMC Nucl. Med. 2006, 6, 3. [Google Scholar] [CrossRef] [PubMed]
  164. Dweck, M.R.; Jenkins, W.S.; Vesey, A.T.; Pringle, M.A.; Chin, C.W.; Malley, T.S.; Cowie, W.J.; Tsampasian, V.; Richardson, H.; Fletcher, A.; et al. 18F-sodium fluoride uptake is a marker of active calcification and disease progression in patients with aortic stenosis. Circ. Cardiovasc. Imaging 2014, 7, 371–378. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Phases and Key Mechanisms of CAVD Pathogenesis. Initiation Phase: High-velocity blood flow induces endothelial damage, promoting inflammation through increased expression of adhesion molecules, lipid deposition, and macrophage recruitment. Oxidized lipids (Ox-LDL) enhance calcification via upregulation of PiT-1 and pro-inflammatory polarization of macrophages. Endothelial dysfunction leads to reduced eNOS activity. Inflammatory cytokines (TNF-α, IL-6) drive endothelial-to-mesenchymal transition (Endo-MT) via NF-κB signaling, generating valve interstitial cells (VICs). Propagation Phase: Factors derived from M1 macrophages (M1-polarized macrophages) promote osteogenic differentiation of VICs, upregulating osteogenic markers such as OPN, BMP2, BMP4, and ALP. Valve thickening results from calcification and fibrosis. Activated VICs—differentiated into myofibroblast-like and osteoblast-like phenotypes—release calcifying vesicles or undergo apoptosis/necrosis, forming hydroxyapatite nuclei that develop into calcified nodules. Although monocyte-derived osteoclast-like cells can resorb calcification via OPG/RANKL/RANK signaling, their function is impaired by inflammatory cytokines (TNF-α, IFN-γ), disrupting calcification balance and exacerbating mineral deposition.
Figure 1. Phases and Key Mechanisms of CAVD Pathogenesis. Initiation Phase: High-velocity blood flow induces endothelial damage, promoting inflammation through increased expression of adhesion molecules, lipid deposition, and macrophage recruitment. Oxidized lipids (Ox-LDL) enhance calcification via upregulation of PiT-1 and pro-inflammatory polarization of macrophages. Endothelial dysfunction leads to reduced eNOS activity. Inflammatory cytokines (TNF-α, IL-6) drive endothelial-to-mesenchymal transition (Endo-MT) via NF-κB signaling, generating valve interstitial cells (VICs). Propagation Phase: Factors derived from M1 macrophages (M1-polarized macrophages) promote osteogenic differentiation of VICs, upregulating osteogenic markers such as OPN, BMP2, BMP4, and ALP. Valve thickening results from calcification and fibrosis. Activated VICs—differentiated into myofibroblast-like and osteoblast-like phenotypes—release calcifying vesicles or undergo apoptosis/necrosis, forming hydroxyapatite nuclei that develop into calcified nodules. Although monocyte-derived osteoclast-like cells can resorb calcification via OPG/RANKL/RANK signaling, their function is impaired by inflammatory cytokines (TNF-α, IFN-γ), disrupting calcification balance and exacerbating mineral deposition.
Biomedicines 13 02369 g001
Figure 2. In Vivo Animal Models for CAVD: Pathogenesis, Research Applications, and Detection Techniques. Diverse large and small animal models of CAVD—including spontaneous, induced, and genetically modified models targeting risk factors—have been utilized to elucidate the pathophysiological mechanisms underlying CAVD. Specific models show significant promise for future CAVD research and the evaluation of novel detection technologies.
Figure 2. In Vivo Animal Models for CAVD: Pathogenesis, Research Applications, and Detection Techniques. Diverse large and small animal models of CAVD—including spontaneous, induced, and genetically modified models targeting risk factors—have been utilized to elucidate the pathophysiological mechanisms underlying CAVD. Specific models show significant promise for future CAVD research and the evaluation of novel detection technologies.
Biomedicines 13 02369 g002
Table 1. Strengths and Limitations of Mouse Models for CAVD. HF: high fat. HC: high cholesterol. Hemodynamically significant stenosis was defined as a ≥50% increase in the peak velocity relative to the baseline value.
Table 1. Strengths and Limitations of Mouse Models for CAVD. HF: high fat. HC: high cholesterol. Hemodynamically significant stenosis was defined as a ≥50% increase in the peak velocity relative to the baseline value.
SpeciesDietHistopathological Changes in Aortic ValveAdvantagesLimitationsHemodynamically Significant Stenosis?Respond to Interventions?
Mice
ApoE−/−Chow
  • Lipid deposition
  • Calcification
  • Monocyte/inflammatory cell infiltration
  • Develop hyperlipidemia and complex lesions on regular chow diet
  • Absence of a human-like lipid profile
<2% [101]No
HF/HC
  • Lipid deposition
  • Fibrosis
  • Calcification
  • Monocyte/inflammatory cell infiltration
  • Higher total cholesterol levels than Ldlr−/− mice
<2% [62,102]Yes
LDLR−/−HF/HC
  • Lipid deposition
  • Calcification
  • Monocyte/inflammatory cell infiltration
  • Lipoprotein pattern similar to humans
  • Develop site-specific lesions in a time-dependent manner
  • Ldlr mutations are common in patients with familial hypercholesterolemia
  • Require HF/HC Diet for CAVD induction
NoYes
LDLR−/−/ApoB100/100Chow
  • Lipid deposition
  • Calcification
  • Monocyte/inflammatory cell infiltration
  • Myofibroblast activation
  • Develop CAVD on a regular chow diet
  • Develop more human-like hypercholesterolemia
  • Expensive and time-consuming
Yes, ~30% [64]No
HF/HC
  • Lipid deposition
  • Calcification
  • Monocyte/inflammatory cell infiltration
  • Myofibroblast activation
  • Fibrosis
Yes, ~50% [70]Yes
APOE*3-Leiden.CETPHF/HC
  • Lipid deposition
  • Fibrosis
  • Thickness
  • Develop more human-like hypercholesterolemia
  • Favorable response to lipid-lowering and anti-atherosclerotic medications
  • Absence of positive alizarin red or von Kossa staining
Not knownYes
Chm1 −/−Chow
  • Lipid deposition
  • Thickness
  • Neoangiogenesis
  • Calcification
  • Monocyte/inflammatory cell infiltration
  • Develop CAVD on a regular chow diet
  • Associated with neoangiogenesis
  • Expensive and time-consuming
Yes [80]No
Tie2-Cre/CXCR4fl/flChow
  • Fibrosis
  • Thickness
  • Calcification
  • Time-saving
  • Develop CAVD on a regular chow diet
  • Lack of early pathological characterization of CAVD
Yes, 50–90% [81]No
Klotho−/−Chow
  • Calcification
  • Develop CAVD on a regular chow diet
  • Non-Monocyte/inflammatory cell infiltration
  • Develop robust nodular aortic valve calcification similar to the human
  • Develop kidney disease and elevated serum phosphate levels
  • Expensive and time-consuming
  • Limited scope
Not knownYes
MG-53−/−Chow
  • Fibrosis
  • Thickness
  • Calcification
  • Develop CAVD on a regular chow diet
  • Displayed an age-dependent phenotype of CAVD
  • Time-consuming
Yes [86]No
eNOS−/−Chow
  • Calcification
  • Bicuspid aortic valves in ~40% of mice
  • Limited scope
Not knownYes
NOTCH-1+/−HF/HC
  • Thickness
  • Calcification
  • Develop bicuspid aortic valves
  • Limited scope
NoYes
NPR-2+/−HF/HC
  • Fibrosis
  • Thickness
  • Calcification
  • Bicuspid aortic valves in 9.4% of mice
  • Limited scope
Yes [92]No
DCBLD2−/−Chow
  • Calcification
  • Bicuspid aortic valves in ~50% of mice
  • Limited scope
Yes [93]Yes
C57BL/6J Chow+
Mechanical injury
  • Thickness
  • Calcification
  • Controlled injury
  • Time-saving
  • Limited relevance to human pathophysiology
  • High mortality rate.
  • Standardization to be improved.
Yes [99]Yes
Table 2. Strengths and Limitations of Rats Models for CAVD. WD: Western diet. Hemodynamically significant stenosis was defined as a ≥50% increase in the peak velocity relative to the baseline value.
Table 2. Strengths and Limitations of Rats Models for CAVD. WD: Western diet. Hemodynamically significant stenosis was defined as a ≥50% increase in the peak velocity relative to the baseline value.
SpeciesDietHistopathological Changes in Aortic ValveAdvantagesLimitationsHemodynamically Significant Stenosis?Respond to Interventions?
Rat
LDLR−/−HF/HC
  • Thickness
  • Calcification
  • Time-saving
  • Similar to the process of lipid deposition induction in humans
  • Expensive and time-consuming
No [106]No
miR-214−/−WD +
Vitamin D2
  • Calcification
  • Thickness
  • Monocyte/inflammatory cell infiltration
  • Time-saving
  • Expensive and time-consuming
Yes [108]No
Sprague-DawleyRodent diet 5001 + Warfarin + Vitamin K1
  • Calcification
  • Time-saving
  • Impaired coagulation.
Not knownNo
Chow + Vitamin D3
  • Calcification
  • Cheap and easy to operate
  • Limited relevance to human pathophysiology
No [111]No
High-phosphate Diet
  • Calcification
  • Monocyte/inflammatory cell infiltration
  • Cheap and easy to operate
  • Associated with chronic renal disease
  • Limited scope
  • Reversible valve calcification
No [112]No
Table 3. Strengths and Limitations of Rabbits Models for CAVD. HF: high fat. HC: high cholesterol. Hemodynamically significant stenosis was defined as a ≥50% increase in the peak velocity relative to the baseline value.
Table 3. Strengths and Limitations of Rabbits Models for CAVD. HF: high fat. HC: high cholesterol. Hemodynamically significant stenosis was defined as a ≥50% increase in the peak velocity relative to the baseline value.
SpeciesDietHistopathological Changes in Aortic ValveAdvantagesLimitationsHemodynamically Significant Stenosis?Respond to Interventions
Rabbit
New Zealand White HF/HC + Vitamin D3
  • Lipid deposition
  • Calcification
  • Inflammatory cell infiltration
  • Time-saving
  • Similar to a plasma lipid protein distribution of humans
  • Liver dysfunction
Yes [116]Yes
WatanabeHF/HC
  • Lipid deposition
  • Fibrosis
  • Calcification
  • Inflammatory cell infiltration
  • Time-saving
  • Require HF/HC Diet for CAVD induction
No [119]No
Chow
  • Thickness
  • Calcification
  • Inflammatory cell infiltration
  • Develop CAVD on a regular chow diet
  • Age-dependent
  • Spontaneously develop CAVD
  • Avoid liver dysfunction
  • Time-consuming
Yes [117]No
Table 4. Strengths and Limitations of Swine Models for CAVD. HF: high fat. HC: high cholesterol. Hemodynamically significant stenosis was defined as a ≥50% increase in the peak velocity relative to the baseline value.
Table 4. Strengths and Limitations of Swine Models for CAVD. HF: high fat. HC: high cholesterol. Hemodynamically significant stenosis was defined as a ≥50% increase in the peak velocity relative to the baseline value.
SpeciesDietHistopathological Changes in Aortic ValveAdvantagesLimitationsHemodynamically Significant Stenosis?Respond to Interventions
Swine
YorkshireHF/HC
  • Lipid deposition
  • Develop valvular sclerosis lesions spontaneously
  • Expensive and time-consuming
  • Lack of late pathological changes in CAVD.
No [120]No
Rapacz familial hypercholesterolemicChow
  • Thickness
  • Lipid deposition
  • Inflammatory cell infiltration
  • Few complications
  • Show numerous critical features of early CAVD
  • Expensive and time-consuming
  • Lack of late pathological changes in CAVD.
No [123]No
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

Fan, P.; Liu, Y.; Qian, X.; Tong, F.; Zheng, Y.; Fan, Z.; Chen, M.; Chen, Z.; Zhai, H.; Zeng, T.; et al. Deciphering the Enigma of Calcific Aortic Valve Disease: The Pivotal Role of Animal Models in Unraveling Pathogenesis and Advancing Therapeutic Strategies. Biomedicines 2025, 13, 2369. https://doi.org/10.3390/biomedicines13102369

AMA Style

Fan P, Liu Y, Qian X, Tong F, Zheng Y, Fan Z, Chen M, Chen Z, Zhai H, Zeng T, et al. Deciphering the Enigma of Calcific Aortic Valve Disease: The Pivotal Role of Animal Models in Unraveling Pathogenesis and Advancing Therapeutic Strategies. Biomedicines. 2025; 13(10):2369. https://doi.org/10.3390/biomedicines13102369

Chicago/Turabian Style

Fan, Pengning, Yuqi Liu, Xingyu Qian, Fuqiang Tong, Yidan Zheng, Zhengfeng Fan, Ming Chen, Zhe Chen, Haoyang Zhai, Teng Zeng, and et al. 2025. "Deciphering the Enigma of Calcific Aortic Valve Disease: The Pivotal Role of Animal Models in Unraveling Pathogenesis and Advancing Therapeutic Strategies" Biomedicines 13, no. 10: 2369. https://doi.org/10.3390/biomedicines13102369

APA Style

Fan, P., Liu, Y., Qian, X., Tong, F., Zheng, Y., Fan, Z., Chen, M., Chen, Z., Zhai, H., Zeng, T., Dong, N., Li, F., Shi, X., & Xu, L. (2025). Deciphering the Enigma of Calcific Aortic Valve Disease: The Pivotal Role of Animal Models in Unraveling Pathogenesis and Advancing Therapeutic Strategies. Biomedicines, 13(10), 2369. https://doi.org/10.3390/biomedicines13102369

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