Open access peer-reviewed chapter - ONLINE FIRST

Valvular Interstitial Cells: Physiology, Isolation, and Culture

Written By

Marcus Ground, Karen Callon, Rob Walker, Paget Milsom and Jillian Cornish

Submitted: 18 May 2023 Reviewed: 24 July 2023 Published: 18 September 2023

DOI: 10.5772/intechopen.112649

Technologies in Cell Culture - A Journey From Basics to Advanced Applications IntechOpen
Technologies in Cell Culture - A Journey From Basics to Advanced ... Edited by Soumya Basu

From the Edited Volume

Technologies in Cell Culture - A Journey From Basics to Advanced Applications [Working Title]

Prof. Soumya Basu, Dr. Amit Ranjan and Dr. Subhayan Sur

Chapter metrics overview

88 Chapter Downloads

View Full Metrics

Abstract

Valvular interstitial cells (VICs) are the primary cellular component of the heart valve. Their function is to maintain the structure of the valve leaflets as they endure some three billion beats in the course of a human lifespan. Valvular pathology is becoming ever more prevalent in our ageing world, and there has never been a greater need for understanding of the pathological processes that underpin these diseases. Despite this, our knowledge of VIC pathology is limited. The scientific enquiry of valve disease necessitates stable populations of VICs in the laboratory. Such populations are commonly isolated from porcine and human tissue. This is achieved by digesting valve tissue from healthy or diseased sources. Understanding of the many VIC phenotypes, and the biochemical cues that govern the transition between phenotypes is essential for experimental integrity. Here we present an overview of VIC physiology, and a tried-and-true method for their isolation and culture. We make mention of several biochemical cues that the researcher may use in their culture media to ensure high quality and stable VIC populations.

Keywords

  • valvular interstitial cell
  • heart valve
  • cell culture
  • myofibroblasts
  • aortic stenosis

1. Introduction

The four valves of the mammalian heart are delicate, complex structures that ensure unidirectional flow of blood through the heart’s four chambers. Two atrioventricular valves, the tricuspid and mitral, sit between the atria and ventricles. During systole, these valves are closed to prevent the backflow of blood into the atria. The semilunar valves—so named for their half-moon shaped leaflets—sit between the ventricles are their outflows: the pulmonary trunk and the aorta. During systole, these valves are open, allowing blood to exit the heart. During diastole, they slam shut, preventing the blood from flowing backwards. The valves themselves comprise two main structural features: the valve leaflets (also called ‘cusps’) and the fibrocartilaginous ring onto which they are mounted. The leaflets are beautifully thin sheets of tissue organised into three distinct layers each with a definite function. The upstream layer, the ‘ventricularis’ of the semilunar valves and the ‘atrialis’ of the atrioventricular valves is rich in radially oriented elastin, allowing the leaflet to hinge open and closed. The middle layer, the ‘spongiosa’, is rich in glycosaminoglycans, forming the bulk of the matrix and structurally linking the superficial layers. Finally, the downstream surface, the ‘fibrosa’, is rich in circumferentially oriented collagen that confers strength against the mechanical load of the blood pressure when the valve is closed [1, 2].

Two key cell types are responsible for maintaining the structure and function of the heart valve leaflet. Valvular endothelial cells (VECs) form a confluent monolayer on the surface of the valve. These cells are phenotypically distinct from endothelial cells elsewhere in the vasculature.

The second key cell type, and the object of this chapter, is the valvular interstitial cell (VIC). These are effectively fibroblasts that populate the extracellular matrix of the valve leaflets.

Functionally, VICs are responsible for maintaining the matrix of heart valve over the course of growth and development. This is no mean feat—the heart valves must endure some three billion beats over their lifetime.

Advertisement

2. Valvular interstitial phenotypes and their physiology

There are five recognised phenotypes of valvular interstitial cell: embryonic, quiescent, progenitor, activated, and osteoblastic.

2.1 Embryonic VICs

Embryonic VICs are responsible for valvulogenesis—the formation of the valves from the endocardial cushion during fetal development. The precise detail of how this occurs in not well understood, but several key features of these embryonic VICs are known. Firstly, they are highly proliferative, a feature which distinguishes them from other VIC phenotypes [3]. Second, is that these cells are endothelial in origin, having undergone the endothelial-to-mesenchymal transition coordinated by a complex sequence of cell signallers. Notch and TGF family ligands (such as BMP-2 and VEGF) induce TGF-β and Wnt/β-catenin signalling which supresses endothelial-type gene expression in the endothelial cells, and instead encourages the cell to become embryonic VICs, a mesenchymal cell type [4, 5, 6]. Once this transition is complete, the maturing valve and its embryonic VICs ‘striate’ the matrix into the three aforementioned layers. Precisely how this occurs is unknown, but recent evidence suggests that the differentiation is aided by the haemodynamic forces of the fetal circulation [7].

2.2 Progenitor VICs

The VIC population is not a static one. Rather, a subpopulation of VICs present in the valve tissue in adulthood express surface markers of progenitor cells. Studies on healthy porcine valves show that approximately 5% of VICs express ABCG2, NG2, or SSEA-4 [8]. The origin of these progenitor cells in not entirely clear. Evidence suggests that some of these progenitor cells migrate to the valve from the blood stream—studies in mice have shown that bone marrow progenitor cells marked with green fluorescent protein appear in the valve, and assume the role of qVICs by secreting collagen 1 [9]. Other researchers suggest that another source of pVICs are VICs already residing within the valve, and are ‘activated’ in response to injury [3].

2.3 Quiescent VICs and activated VICs

Quiescent VICs (qVICs) maintain the matrix throughout adult life [3]. They are neither very metabolically nor mitotically active. Instead, their role in maintaining the valve matrix is by sensing mechanical forces, and responding by differentiating into activated VICs (aVICs). The activated VIC is a myofibroblastic phenotype—characterised by the synthesis of ECM proteins and α-smooth muscle actin (a contractile protein) [10]. The differentiation from qVIC to aVIC is the key feature of mechano-regulated growth of this tissue from fetus to adult. As the heart grows, the pressures across the valve leaflet increase, introducing undue stress to the ECM. qVICs resident in the ECM can sense this stress, and differentiate into aVICs that then secrete matrix proteins in order to strengthen the matrix, until such a time when the matrix is able to support the increased pressure. At this time the ‘stress’ stimulus is lost and the aVIC then dedifferentiates back into a qVIC. It should be no surprise that the fetal heart valve contains a high proportion of aVICs, and that the healthy adult valve contains nearly no aVICs [11].

This mechano-regulation of heart valve growth and development relies on the ability of the qVIC’s mechano-sensing ability. This is achieved through three surface protein classes: integrins, cadherins, and mechano-sensitive ion channels [12]. Integrins bind the VIC to the matrix, forming a continuation of the cytoskeleton intracellularly with the ECM extracellularly. Cadherins are transmembrane proteins that bind other cells—in the case of VICs, cadherin 2 is upregulated in qVICs, and cadherin 11 in aVICs [12]. Mechano-sensitive ion channels, particularly calcium channels, also enable mechano-sensation by altering ion concentration within the cell in response to stress-induced deformation [13]. Collectively, these three mechano-sensing modalities induce a transcriptional change within a qVIC in response to stress, which causes aVIC differentiation, upregulation of ECM component synthesis, and the production of α-SMA [10].

2.4 Osteoblastic VICs

Osteoblastic VICs (obVICs) are, as the name suggests, bone-producing cells that arise from VICs in the context of heart valve calcification. Up to 13% of the cellular content of end-stage diseased valve leaflets are positive for osteoblast markers RUNX2 and ALP, and histology of diseased leaflets shows evidence of lamellar bone [14]. Exactly where the obVICs come from is not certain. Some authors suggest that obVICs arise exclusively from qVICs in the context of age-related valve disease [10], while others suggest that obVICs come from a restricted range of susceptible VICs or perhaps pVICs [14].

Advertisement

3. Valvular interstitial cells in health and disease

3.1 From qVIC to aVIC and back again: dynamic reversibility of VIC activation

The key feature of VIC physiology that underpins normal growth and development, and the pathogenesis of heart valve disease, is the reversibility of the transition between qVIC and aVIC [15]. This reversibility (depicted in Figure 1), or the failure thereof, is perhaps best illustrated by two examples: growth and development by fetal heart valves, and the development of calcific aortic valve disease. Each involves substantial activation of VICs and the subsequent production of matrix.

Figure 1.

Shows the ‘dynamic reversibility’ of VICs as they transition between their quiescent and activated states, as well as the key protein expression markers associated with each phenotype.

The fetal heart valve is an immature structure—not yet required to bear the forces of systolic blood pressure while the fetal circulation, with its shunts, remains active. Once a baby is born, however, those shunts close, and the semilunar valves must immediately assume their role in preventing blood from flowing backwards into the ventricles from their respective outflows. The transition from immature fetal valve to fully competent valve requires several histological changes. First, the immature, loosely organised ECM network of the early valve must become more structured and load-bearing. Secondly, the resident VICs must proliferate in order to adequately populate the valve matrix, and will need to change their phenotype to meet the increased mechanical needs of the growing valve [11].

The immature valve matrix has a lower proportion of elastin than the adult valve, and a much greater proportion of glycosaminoglycan [11]. Interestingly, the proportion of collagen—the primary ECM component of the valve by weight—appears to be fairly stable from the first trimester through to adulthood. What does change markedly, however, is the degree of ‘organisation’, assessed by various authors using methods such as picrosirius red staining viewed under polarised light [11], or by quantifying the degree of hydroxyproline cross-linking [16]. In the first instance, collagen fibre thickness and alignment is significantly increased from fetus to child, and in the second, hydroxyproline crosslinking is significantly elevated. The net effect is substantial increase in the strength of the valve matrix.

How do the resident VICs achieve this? The answer lies in the presence of aVICs. The cell population of the fetal valve is up to 30% activated VIC, as determined by α-SMA staining [11]. These cells contribute to the changes in valve ECM makeup by simultaneous upregulation of matrix proteins (collagen, for example) and matrix metalloproteases (MMPs) [17]. The combination of upregulated ECM structural proteins and the enzymes that break them down is ‘remodelling’—i.e., the overall proportion of collagen remains static, but the maturation into strong, organised bundles proceeds.

In the fetal valve, the laydown of collagen in the correct orientation is driven by the mechano-sensing ability of the resident VICs. But as we age, there are situations where the dynamic reversibility of qVIC and aVIC phenotypes is upset, and the resulting products of VIC activation lead to disease. The prescient example is calcific aortic valve disease (CAVD)—the most common valvular pathology of the developed world, with prevalence rates of up to 15% in those over 75 years old [18]. In the early phase of this disease, aVIC activation and the accumulation of structural ECM components mimics the changes seen in fetal valves, only in this circumstance, the interruption of the ‘dynamic reversibility’ of aVICs is lost, and the valves become stiff and non-compliant [19, 20]. Valves affected by CAVD are characterised by the overproduction of collagen [21] and the dysregulation of MMP expression [22].

The ‘dynamic reversibility’ of VIC phenotypes and the mechanical properties of the valve matrix are a true ‘chicken and egg’ problem. VIC activation invariably leads to stiffening and matrix remodelling (either physiological in the case of the fetal valve, or pathological in the case of CAVD), which, in turn, results in a stiffer matrix, which then serves a signal for qVICs to activate [23].

3.2 VIC calcification: murky origins

The origin if the calcification in heart valve disease is not perfectly understood. What is known is that VICs can calcify via two distinct mechanisms: calcific nodule formation and obVIC differentiation (depicted in Figure 2) [10]. The relative contributions of these two mechanisms to heart valve disease, or if the two mechanisms are independent of one another is not known.

Figure 2.

Shows the two phenotypes of VIC calcification: the dystrophic calcific nodules that occur after aVIC apoptosis, and the ossification of obVICs. What is unclear is if the obVIC phenotype arises directly from qVICs or aVICs.

Current evidence suggests that calcific nodules arise from aVICs, and are in fact calcified apoptotic bodies left after the cell dies. This type of calcification is termed ‘dystrophic’ to distinguish it from the matrix calcification caused by obVICs [24]. Where the obVICs arise from, is not clear. Some have suggested that obVICs arise from the qVIC population independently, and in response to changes in matrix mechanics or the accumulation of calcium [25], while others suggest that obVICs arise from aVICs in a manner that depends ‘activating’ signalling [26].

Support for the idea that the obVIC and aVIC calcification are distinct pathways is seen in the apparent changes in α-SMA expression, which is markedly reduced in presence of osteogenic medium and markedly increased in the presence of TGF-β. Conversely, qVICs cultured in osteogenic medium will demonstrate upregulation in key osteoblast markers: ALP and OCN [25]. To further confuse the true origin of obVICs, the same pattern of gene expression occurs when valvular endothelial cells are cultured in osteogenic media [27].

3.3 VIC signalling molecules

The various signalling molecules that induced change in VICs has been extensively reviewed elsewhere [10]. There are, however, three molecules that commonly feature in culture medium preparations, and the physiology that underpins their use is included here.

3.3.1 TGF-β

Perhaps the most well characterised stimulus for VIC activation is transforming growth factor β (TGF-β) [28, 29, 30, 31, 32, 33, 34]. TGF-β, or more specifically the isoform TGF-β1 is one member of a much larger ‘TGF-β superfamily’, which includes 30 species such as bone morphogenic proteins (BMPs), growth and differentiation factors, and various other peptides key to growth and development [35]. In adulthood, TGF-β1 is involved in a myriad of cell functions, from cell fate determination, cell migration, response to injury, and ECM regulation [34, 36, 37]. In the context of VICs, the key function of TGF-β is its ability to regulate activation of VICs and maintain homeostasis of the valve ECM by induction of the aVIC phenotype. As well as structural proteins, ECM contains large amounts of ‘latent’ signalling molecules—signallers bound up with latency-associated peptides. These peptide complexes are hidden from cells, and so do not enact their functions until they are cleaved from the matrix during proteolysis following injury [36, 38]. TGF-β, as a promoter of ECM production, serves as a ‘switch’ in heart valves: when the valve is unable to cope with a tension force, microscopic injury releases TGF-β from the matrix, which in turn signals to cells to secrete more ECM, until such time that the matrix can withhold the tension and the TGF-β switch is turned off. It is through delicate control of TGF-β stimulated ECM secretion that a valve grows and remodels throughout the changing haemodynamic environment from fetal development to adulthood [6, 39].

TGF-β acts on cells via several biochemical pathways. The most well characterised pathway with respect to VIC physiology is the ‘canonical TGF-β’ pathway, or Smad pathway [34, 38, 40]. TGF-β forms a homodimer in the extracellular space and binds two type 2 TGF-β receptors (Tgfbr2), which then recruit two type 1 TGF-β receptors (Tgfbr1) forming an activated receptor-ligand complex [39]. The activated Tgfbr1 is then able to phosphorylate intracellular messenger proteins Smad2 and Smad3, which form a complex with Smad4 and move to the nucleus as transcription factors to activate the associated genes [39, 41]. It is this process that is responsible for the activation of qVICs to myofibroblasts and for the development of dystrophic nodules [3].

Non-Smad pathways initiated by TGF-β are termed ‘non canonical’. These include the ERK/MAPK pathway [41], which has been shown to regulate key aspects of aVIC expression in a manner independent of the canonical TGF-β pathway [42].

3.3.2 FGFs

The fibroblast growth factor (FGF) family comprises 23 distinct proteins, 18 of which are ligands to various FGF receptors, effecting a wide range of cellular functions: from migration to proliferation to cell survival [43, 44]. Of interest to the VIC researcher is FGF-2, also called basic FGF (bFGF), so named for its discovery in the alkaline fraction of pituitary extracts [45]. FGF-2 has been implicated in numerous biological processes, from cell migration and fate determination to pathological signalling in cancer metastasis [46]. Most interesting is that while this molecule is present and active throughout the body, a distinct physiological role has not been identified. Homozygous FGF-2 knockout mice survive perfectly well and give birth to healthy young.

FGF-2’s signalling pathway first involves heparin binding [46]. Two heparin-FGF2 complexes then bind two FGF receptors (FGFR) to form a stable dimer, which then activates intracellular tyrosine kinase domains and effects changes in gene expression through a number of signalling cascades, among them MAPK and AKT pathways [43].

FGF-2 has been shown to be particularly important in regulating the quiescent VIC phenotype [34, 47, 48, 49]. The mechanism of FGF-2’s effect appears to be multifaceted: both repressing the canonical TGF-β pathway by blocking Smad nuclear translocation [48], and by activating of the Akt/mTOR pathway [34, 49]. The downstream effects of this activation have been shown to oppose the effect of TGF-β. FGF-2 appears to promote the qVIC phenotype: VICs treated with FGF-2 have a spindle-shaped morphology, and express more vimentin and less α-SMA [17].

3.3.3 Nitric oxide

The function of nitric oxide (NO) in the vasculature is understood to be a local control mechanism for maintaining blood pressure and perfusion. First, NO is produced in endothelial cells by endothelial nitric oxide synthase (eNOS) in response to stretch or biochemical stimuli [50]. Then, NO is able to rapidly diffuse into the surrounding tissue by virtue of its small size, where it binds guanylyl cyclase (a cytoplasmic receptor) [51] and leads to a myriad of downstream effects including relaxation of the vascular smooth muscle [50]. With respect to heart valves, NO production by VECs and its subsequent action of VICs is an important paracrine regulator of valve cell function [3, 10]. More importantly, dysregulation of NO signalling is implicated in the development of heart valve disease [52, 53, 54, 55]. In the initial stages of CAVD, eNOS is upregulated on the aortic-side endothelium [56], where it contributes to pathological neovascularisation of the valve tissue [57]. The early eNOS response is a protective mechanism to temper the proliferation of VICs, but as overexpression of eNOS continues, resistance to NO develops—a phenomenon termed ‘NOS decoupling’ [58].

With respect to VIC activation, exogenous NO inhibits the formation of calcific nodules in porcine VIC cultures under TGF-β stimulation [59]. Culturing VICs in media supplemented with DETA-NONOate—a biologically inert molecule that spontaneously releases two NO molecules in a predictable manner [60]—limits the formation of new nodules.

This effect is not purely induced by exogenous NO, rather it can be replicated by co-culturing VICs with NO-producing VECs [61]. Under these conditions, VECs can inhibit the formation of aVIC-related calcific nodules in a manner that is reversed by addition of a NO scavenger. This suggests that of the multitude of paracrine signals that travel between the endothelium and the interstitium, NO is paramount in maintaining the valve in its quiescent state.

Advertisement

4. Sources of valvular interstitial cells

There are a number of VIC sources for a researcher’s consideration. No one option is without issue. Perhaps the easiest source of VICs is from animals. Pigs are by far the most common source of VICs for research [61, 62, 63, 64, 65, 66, 67, 68, 69, 70, 71, 72, 73, 74, 75, 76, 77, 78, 79, 80, 81], as they are very easily obtainable from meat processing facilities. As a biproduct of industry, obtaining and using porcine VICs requires little or no ethical oversight [82]. Another notable benefit is the anatomical similarity between the pig and human heart. This is of particular importance as the mechanical and structural forces regulate VIC physiology and gene expression [23, 68], and so choosing a structurally similar heart approximates a physiologically similar VIC. In a similar vein, pigs housed for research can be induced into diseased states for investigation of a particular pathology. For example, pigs fed on high-cholesterol diets experience atherosclerotic lesions, though no porcine model of calcific valve disease has been trialled to date [52, 83]. It should be noted that VICs derived from meat industry pigs can be considered healthy qVICs by definition—their valves are not experiencing rapid growth, nor has age-related disease begun to influence their phenotype. This fact is either a boon or an issue for the cell culturist. Those interested in studying the development of valve disease might be eager to use a healthy population of qVICs, while those searching for therapeutic options for persistent aVIC or obVIC presence will likely have to choose another source. In our experience, porcine VICs are easily obtained and fairly forgiving when it comes to cell culture. Cultures derived from digested porcine valves are adequately proliferative and less sensitive to aVIC formation when cultured on hard plastic.

Sheep may represent a promising animal alternative for obtaining VICs liable to calcify. Sheep exhibit an elevated calcium metabolism compared to humans, and this has led to their use as an experimental model of heart valve calcification [84]. Several authors have made use ovine VICs for experimentation [85, 86].

Human VICs are the best option for studying heart valve disease, as none of the animal models approximate our valvular pathologies perfectly. The most ideal source is a young, healthy adult human heart valve. Cells from this source, in theory, are almost entirely qVICs. However, there are a number of constraints that make sourcing healthy human tissue difficult.

First is availability—unlike other cell types, VICs are not sourced from a healthy tissue that is routinely removed from people undergoing surgery (as it the case in, for example, adipose tissue removed during breast cancer surgery), and suitable heart valve tissue from younger donors is contained within a heart that is likely more suitable for heart transplantation. To complicate matters further, in the instance where a young donor heart is not suitable for transplant, the heart valve may be harvested as a ‘homograft’ (a heart valve transplant).

The second constraint is time. While VICs are relatively robust in the absence of nutrient supply [10], good laboratory practice dictates that cells should be isolated and incubated as quickly as possible. Very rarely are healthy heart valves available within temporal or spatial proximity to a laboratory. Despite these constraints, various researchers have made use of healthy human VICs, typically from donor hearts not suitable for either transplantation or homograft use [47, 87].

A much more commonplace source of human VICs is those isolated from patients undergoing valve replacement surgery. Valve replacement surgeries are very common—our experience at Auckland City Hospital saw an average of 5 valves available per week. The obvious problem with this source is the inherent disease state of the cells within the valve. Though our protocol above recommends removal of the more calcified portion of the valve, the pathology is not localised at the calcification, and altered stiffness characteristics across the whole leaflet ensures that all cells present will have some degree of pathological programming [53].

The diseased state of these cells may actually be useful for researchers concerned with the study of aortic calcification—the most common valvular pathology in the developed world. Many examples of diseased valves used for this purpose are available in the literature [47, 87, 88, 89, 90, 91, 92, 93, 94, 95, 96, 97, 98, 99, 100, 101, 102].

The key consideration to address with diseased human VICs is ethics. Patients must consent to their tissue being harvested and used in research, and protection of the tissue and any data generated must be ensured by the researcher. In addition, many cultures have unique requirements for the disposal or return of human tissue [103].

Advertisement

5. Isolation protocol

This isolation protocol was developed by our laboratory for the isolation of human VICs from patients undergoing valve replacement surgery [104]. An abbreviated flow chart is included in Figure 3.

Figure 3.

Shows and abbreviated flow chart of steps 7–18.

5.1 Consumables

  • Collagenase II (Sigma, #C6885)

  • Sterile phosphate buffered saline

  • Sterile DMEM (Sigma, #D5649) supplemented with sodium bicarbonate per manufacturer’s instructions

  • Fetal bovine serum (FBS)

  • 50 mL containers

  • 15 mL centrifuge tubes

  • 50 mL centrifuge tube

  • 90 mm petri dishes

  • Sterile guards

  • #10 Scalpel blades and handle

  • Sterile forceps

  • 70 μm cell strainer (Bio-Strategy, #BDAA352350)

  • 0.2 μm sterile filter (Bio-Strategy, #SART16534-K)

  • 30 mL syringe

  • T25 cell culture flasks (Sigma, #C6356)

  • 25 mL serological pipettes

  • Disposable ‘squeezy’ pipettes

5.2 Hardware

  • Tissue culture hood

  • Incubator, 37°C at 5% CO2

  • Orbital shaker-incubator, 37°C

  • Sterilisation services (i.e., autoclaving service, sterile filtration capabilities)

  • Balance

  • Centrifuge

  • Electric pipette filler

  • Microscope with haemocytometer slide

  • Tube rack

5.3 Method

Prior to tissue collection:

  1. In the tissue culture hood, prepare at least 100 mL of DMEM supplemented with 10% FBS, and warm to 37°C

  2. Prepare a > 20 mL aliquot of sterile PBS for tissue collection, and warm to 37°C

  3. Turn on the shaker-incubator to equilibrate to 37°C

  4. Weigh out 30kU collagenase II. Note that the mass of the collagenase is lot-number specific - refer to the specification sheet for the number of units per milligramme. Add the collagenase II to a 50 mL container containing 30 mL PBS, gently agitate to dissolve

  5. In the tissue culture hood, aspirate the collagenase II into the 30 mL syringe. Attach the 0.2 μL filter to the syringe and gently push the collagenase through the filter into a sterile container. Collagenase II loses its activity within hours, and should be prepared as close to the time of digest as possible. Divide the collagenase into 2 portions of 15 mL each in 50 mL containers. Label them ‘Digest 1’ and ‘Digest 2’

  6. Once sealed, remove ‘Digest 2’ from the hood and weigh the container, recording the mass to the nearest milligramme. This will allow weighing of the usable valve tissue in a sterile manner

    In the operating room:

  7. Make sure to bring the warmed aliquot of PBS for tissue collection. When the surgeon removes the valves, ensure the scrub nurse or assisting surgeon offers the container in a sterile manner, and that the tissue is added to the container and sealed shut all while in the sterile field. Return to the laboratory as quickly as possible

    In the laboratory:

  8. Using sterile forceps, add the leaflets to the ‘Digest 1’ container

  9. Add to the shaker-incubator for 10 min at 37°C with gentle agitation

  10. Once complete, arrange the leaflets on a 90 mm petri dish, and scrape down the outside using a scalpel blade. If the calcifications make this too difficult, then consider removing the calcified sections before scraping. Rinse the valves using a squeezy pipette and sterile PBS

  11. If hVECs are desired, collect the rinsing media and pool this with suspension in ‘Digest 1’, centrifuge, and resuspend the cell pellet in 1 mL of DMEM +10% FBS. Note that the expected pellet is small, and the suspension should be plated in a 6-well plate or smaller

  12. If not already done so, remove calcified sections of the valve. Finely mince the tissue using the scalpel, and transfer to the tissue to ‘Digest 2’

  13. Weigh ‘Digest 2’ and calculate the difference from recorded mass in step 7. If the mass is less than 500 mg, then there is a low likelihood that the culture will generate enough cells to succeed. We recommend proceeding only if the tissue mass exceeds 500 mg

  14. Add ‘Digest 2’ to the shaker-incubator at 37°C for 2 h with gentle agitation

  15. In a tube rack, assemble the 50 mL tube with the 70 μm cell strainer. Pour the digestate through the strainer, and agitate the contents using a squeezy pipette

  16. Centrifuge at 1200 RPM for 2 min

  17. Discard the supernatant, and, without disturbing the pellet, rinse the tube with 15 mL of DMEM +10% FBS to remove any residual collagenase. Spin briefly to re-pellet any cells that may have been disturbed

  18. Discard the supernatant. Resuspend the cell pellet in 2 mL of DMEM +10% FBS. Take and 30 μL sample and pipette directly onto the haemocytometer. Do not perform cell count immediately - instead, get the cells into the incubator as quickly as possible: immediately add the suspension to T25 culture flask and make up to final volume with 8 mL of DMEM +10% FBS. Add to the incubator at 37°C

  19. Perform a cell count, and record. Note that digests that yield <100,000 cells are unlikely to be successful

  20. After the digest:

  21. Monitor the culture daily for confluency and signs of myofibroblastic activation. hVIC cultures generally reach 80% confluency in 8 days. Monitor for signs of myofibroblast differentiation, including pleiomorphism and non-fusiform morphologies

  22. Replace entire medium every four days

  23. When using hVICs in experiments, make sure to seed at a density of 10,000 cells/cm2, and anticipate that growth will plateau within 5 days

Advertisement

6. Quality control considerations

Regardless of cell source, the goal for VIC culture is a population of phenotypically stable cells (i.e. few aVICs) with as little endothelial contamination as possible.

To assess phenotypic stability, we encourage researchers to plate out a sample of their VIC culture and assess it for features of myofibroblast activation in aVICs. Typically, we >5000 cells from our digestate and plated these in a 24-well plate, and left to grow for up to 6 days. We then assessed for the presence of aVICs first by visual inspection on phase microscopy, and then by α-SMA staining. aVICs differ from pVICs in their morphology. On phase microscopy, qVICs are slender, spindle-shaped cells with a central body surrounded by two or three thin processes. Their area rarely exceeds 5000μm2, and their aspect ratio (the ratio between their major and minor axes) should be greater than 3 [17]. aVICs on the other hand are much larger, with areas often exceeding 10,000μm2. Their shape is much more circular, with a large cell body and loss of the thin processes. Similar cell area measurements were found by Porras and colleagues, who used FIB media to prevent (or reverse) aVIC formation in porcine cultures [72].

We suggest staining for α-SMA and vimentin to assess the degree of aVIC presence (an example of which can be seen in Figure 4). aVICs have dense bands of α-SMA running across the cell, terminating in adhesion complexes that link the intracellular force generated by this protein to the ECM [105]. True qVICs will have very little α-SMA staining, if at all.

Figure 4.

Shows two cultures of VICs: one rich in qVICs on the left, and another rich in aVICs. Cells are stained for vimentin (green) and α-SMA (red). Cells were cultured for four days on plastic. Scale bars represent 100 μm.

Many researchers will quantify aVIC numbers by using α-SMA positivity, either by fluorescent intensity measurement or thresholding numbers of positive cells [4748]. In our experience, there is little utility in attempting to quantify the degree of VIC activation using α-SMA staining or phase microscopy, as all VICs will show some α-SMA expression and variation in morphology. More intensive methods for quantifying α-SMA include western blotting and real time PCR [47, 106]. These are comparative methods (i.e., they require two groups), and so are of little use in the ‘quality control’ stage. Experimentally, we have found that expression of ACTA2 (α-SMA’s gene) and a number of other aVIC-associated genes correlate with the presence of ‘aVIC-looking’ cells on phase microscopy and α-SMA staining. We suggest that researchers subjectively assess the appearance of their VIC cultures on day 6 post-isolation.

The second main feature of VIC quality control is endothelial contamination (Figure 5). Despite the initial digest and scraping we suggest in the protocol, it is impossible to remove all VECs. Endothelial contamination can run as high as 10% [10]. In our experience, VIC cultures generated by the means outlined above will produce cultures with less than 1% von Willebrand factor (vWF) positivity. To assess VEC contamination, we suggest first saving and plating the digestate from digest 1 in a 6-well plate or smaller (see step 11) for use as a positive control. In the same plate, we suggest reserving a small volume of the VIC suspension from digest 2, and culturing both for up to 6 days. We then suggest staining the two wells for an endothelial marker. The choice of marker is not especially important: VECs express vWF, CD31 (PECAM) and CD34, and are negative for α-SMA [74, 107].

Figure 5.

Shows a quality control procedure performed on a VIC culture (left). The yellow arrow indicates VEC contamination. This was performed alongside a VEC culture as a positive control. This culture was grown from ‘Digest 1’ aspirate. Both cultures were grown for four days on plastic. Scale bars represent 100 μm.

Advertisement

7. Culture conditions

The majority of the literature has VICs cultured on flat plastic in Dublecco’s modified Eagle medium (DMEM) supplemented with 10% fetal bovine serum (FBS). We have had good results with this formulation. There are, however, a number of other culture media that should be considered. Alternatives to DMEM include M199 medium [29, 108] and DMEM-F12 [73, 109].

Good laboratory practice stipulates that isolated cells should be cultured in such a way that preserves their in vivo phenotype as faithfully as possible. And so, it befalls the VIC researcher to consider the inherent aVIC promoting properties of chosen media. The supplementation of media with 10% FBS is standard practice for the culture of any fibroblast [110]. However, careful thought should be put into the inherent content of peptides that stimulate VIC activation. TGF-β, for example, is present in FBS in concentrations ranging from 10 to 20 ng/mL. TGF-β is a strong promoter of VIC activation, and several authors have mitigated its effect by using a 2% FBS medium supplemented with insulin as a mitotic agent. This so-called ‘FIB’ media, first used by Latif and colleagues [47, 72], is also supplemented with 10 ng/mL FGF-2—an agent shown to reduce VIC activation in favour of the quiescent phenotype. We have demonstrated that VICs cultured in FIB media show significantly downregulated expression of aVIC-associated genes (α-SMA in particular), and a morphology more typical of qVICs (greater aspect ratio, and reduced total cell area) compared to VICs cultured in the standard DMEM preparation [17]. The inherent ability of FIB medium to suppress aVIC formation can be potentiated further by culturing VICs on collagen-coated plates [72].

Induction of the obVIC phenotype requires osteogenic medium. This invariably contains ascorbic acid, dexamethasone, and β-glycerophosphate [28, 109, 111], supplemented in some instances with TGF-β or BMP-2 [89].

Seeding densities vary widely in the literature, and will differ depending on the surface on which the VICs are grown. On hard plastic or glass, we have found VICs to be rather sensitive to contact inhibition and myofibroblastic activation. We suggest that VICs are seeded at a density of 10,000 cells/cm2 for experiments running for less than five days. At this density, cell growth reaches a plateau on day 4 [104].

Advertisement

8. Conclusion

VICs are the specialised fibroblasts that populate the human valve and maintain its structure and function throughout our lives. The increasing incidence of valvular pathology means that there is no better time to gain understanding about the physiological and pathological processes that affect these cells. Despite this, our understanding of VIC health and disease is in its infancy. Culture of VICs is an important part of the scientific exploration of heart valve disease. Several key methodological considerations need to be addressed for the jobbing scientist:

  • VICs come in a variety of phenotypes. Some are implicated in embryogenesis, while other in disease

  • Biochemical signals govern the phenotypic transition of VICs between these phenotypes

  • A normal, physiological dynamism exists between the qVIC and the aVIC. This is the key process that underpins normal valve growth and development

  • Upsetting this dynamic transition is implicated in the development of valvular disease. Chief among them is calcific aortic valve disease

  • Phenotypically stable VIC cultures can come from pigs and humans

  • Diseased VICs are the most readily attainable human cell source, and with careful consideration of culture medium preparations, stable cultures of qVICs can be readily and easily obtained

  • qVICs are liable to become aVICs in culture. The researcher is therefore tasked with ensuring the culture conditions avoid errant VIC activation

Advertisement

Acknowledgments

This work was supported by the Green Lane Research and Educational Fund and the Health Research Council of New Zealand.

References

  1. 1. Chester AH, El-Hamamsy I, Butcher JT, Latif N, Bertazzo S, Yacoub MH. The living aortic valve: From molecules to function. Global Cardiology Science Practise. 2014;2014(1):11
  2. 2. Loukas M, Bilinsky E, Bilinsky S, Blaak C, Tubbs RS, Anderson RH. The anatomy of the aortic root. Clinical Anatomy. 2014;27(5):748-756
  3. 3. Liu AC, Joag VR, Gotlieb AI. The emerging role of valve interstitial cell phenotypes in regulating heart valve pathobiology. American Journal of Pathology [Internet]. 2007;171(5):1407-1418. DOI: 10.2353/ajpath.2007.070251
  4. 4. Bosada FM, Devasthali V, Jones KA, Stankunas K. Wnt/β-catenin signaling enables developmental transitions during valvulogenesis. Development. 2016;143(6):1041-1054
  5. 5. Neri T, Hiriart E, van Vliet PP, Faure E, Norris RA, Farhat B, et al. Human pre-valvular endocardial cells derived from pluripotent stem cells recapitulate cardiac pathophysiological valvulogenesis. Nature Communication [Internet]. 2019;10(1):1-14. DOI: 10.1038/s41467-019-09459-5
  6. 6. Butcher JT, Markwald RR. Valvulogenesis: The moving target. Philosophical Transaction on Royal Society B Biology Science. 2007;362(1484):1489-1503
  7. 7. Lindsey SE, Butcher JT. The cycle of form and function in cardiac valvulogenesis. Aswan Heart Center Science Practise Series. 2011;2011(2)
  8. 8. Wang H, Sridhar B, Leinwand LA, Anseth KS. Characterization of cell subpopulations expressing progenitor cell markers in porcine cardiac valves. PLoS One. 2013;8(7):1-11
  9. 9. Hajdu Z, Romeo SJ, Fleming PA, Markwald RR, Visconti RP, Drake CJ. Recruitment of bone marrow-derived valve interstitial cells is a normal homeostatic process. Journal of Molecular Cell Cardiology [Internet]. 2011;51(6):955-965. DOI: 10.1016/j.yjmcc.2011.08.006
  10. 10. Rutkovskiy A, Malashicheva A, Sullivan G, Bogdanova M, Kostareva A, Stensløkken KO, et al. Valve interstitial cells: The key to understanding the pathophysiology of heart valve calcification. Journal of American Heart Association [Internet]. 2017;6(9). Available from: http://www.ncbi.nlm.nih.gov/pubmed/28912209
  11. 11. Aikawa E, Whittaker P, Farber M, Mendelson K, Padera RF, Aikawa M, et al. Human semilunar cardiac valve remodeling by activated cells from fetus to adult: Implications for postnatal adaptation, pathology, and tissue engineering. Circulation. 2006;113(10):1344-1352
  12. 12. Howsmon DP, Sacks MS. On valve interstitial cell signaling: The link between multiscale mechanics and mechanobiology. Cardiovascular Engineering and Technology. 2021;12(1):15-27
  13. 13. Simard C, Aize M, Chaigne S, Mpweme Bangando H, Guinamard R. Ion channels in the development and remodeling of the aortic valve. International Journal of Molecular Sciences. 2023;24(6):5860
  14. 14. Liu X, Xu Z. Osteogenesis in calcified aortic valve disease: From histopathological observation towards molecular understanding. Progress in Biophysics Molecular Biology [Internet]. 2016;122(2):156-161. DOI: 10.1016/j.pbiomolbio.2016.02.002
  15. 15. Rabkin-Aikawa E, Farber M, Aikawa M, Schoen FJ. Dynamic and reversible changes of interstitial cell phenotype during remodeling of cardiac valves. The Journal of Heart Valve Disease. 2004;13(5):841-847
  16. 16. Van Geemen D, Soares ALF, Oomen PJA, Driessen-Mol A, Janssen-Van Den Broek MWJT, Van Den Bogaerdt AJ, et al. Age-dependent changes in geometry, tissue composition and mechanical properties of fetal to adult cryopreserved human heart valves. PLoS One. 2016;11(2)
  17. 17. Ground M, Ground M, Yutzey K, Waqanivavalagi S, Park YE, Callon K, et al. Fibroblast growth factor 2 inhibits myofibroblastic activation of valvular interstitial cells. PLoS One [Internet]. 2022;17(6):e0270227. DOI: 10.1371/journal.pone.0270227
  18. 18. Coffey S, Cairns BJ, Iung B. The modern epidemiology of heart valve disease. Heart [Internet]. 2016;102(1):75-85. Available from: http://heart.bmj.com/lookup/doi/10.1136/heartjnl-2014-307020
  19. 19. Mrsic Z, Hopkins SP, Antevil JL, Mullenix PS. Valvular heart disease. Primary Care – Clinical Official Practise [Internet]. 2018;45(1):81-94. DOI: 10.1016/j.pop.2017.10.002
  20. 20. Helms AS, Bach DS. Valvular heart disease. Primary Care – Clinical Official Practise [Internet]. 2013;40(1):91-108. DOI: 10.1016/j.pop.2012.11.005
  21. 21. Gomez-Stallons MV, Tretter JT, Hassel K, Gonzalez-Ramos O, Amofa D, Ollberding NJ, et al. Calcification and extracellular matrix dysregulation in human postmortem and surgical aortic valves. Heart. 2019;105(21):1616-1621
  22. 22. Ohukainen P, Ruskoaho H, Rysa J. Cellular mechanisms of valvular thickening in early and intermediate calcific aortic valve disease. Current Cardiology Reviews. 2018;14(4):264-271
  23. 23. Butcher JT, Nerem RM. Valvular endothelial cells and the mechanoregulation of valvular pathology. Philosophical Transaction Royal Society B Biological Science. 2007;362(1484):1445-1457
  24. 24. Merryman WD, Schoen FJ. Mechanisms of calcification in aortic valve disease: Role of mechanokinetics and mechanodynamics. Current Cardiology Reports. 2013;15(5):355
  25. 25. Monzack EL, Masters KS. Can valvular interstitial cells become true osteoblasts? A side-by-side comparison. Journal of Heart Valve Diseases [Internet]. 2011;20(4):449-463. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3624763/pdf/nihms412728.pdf
  26. 26. Galeone A, Paparella D, Colucci S, Grano M, Brunetti G. The role of TNF- α and TNF superfamily members in the pathogenesis of calcific aortic valvular disease. Scientific World Journal. 2013;2013(Ldl):1-10
  27. 27. Hjortnaes J, Shapero K, Goettsch C, Hutcheson JD, Keegan J, Kluin J, et al. Valvular interstitial cells suppress calcification of valvular endothelial cells. Atherosclerosis [Internet]. 2015;242(1):251-260. DOI: 10.1016/j.atherosclerosis.2015.07.008
  28. 28. Cloyd KL, El-Hamamsy I, Boonrungsiman S, Hedegaard M, Gentleman E, Sarathchandra P, et al. Characterization of porcine aortic valvular interstitial cell “calcified” nodules. PLoS One. 2012;7(10):1-9
  29. 29. Song R, Fullerton DA, Ao L, Zheng D, Zhao K, seng, Meng X. BMP-2 and TGF-β1 mediate biglycan-induced pro-osteogenic reprogramming in aortic valve interstitial cells. Journal of Molecular Medicine. 2015;93(4):403-412
  30. 30. Das D, Holmes A, Murphy GA, Mishra K, Rosenkranz AC, Horowitz JD, et al. TGF-beta1-induced MAPK activation promotes collagen synthesis, nodule formation, redox stress and cellular senescence in porcine aortic valve interstitial cells. Journal of Heart Valve Disease [Internet]. 2013;22(5):621-630. Available from: http://www.ncbi.nlm.nih.gov/pubmed/24383372
  31. 31. Walker GA, Masters KS, Shah DN, Anseth KS, Leinwand LA. Valvular myofibroblast activation by transforming growth factor-β: Implications for pathological extracellular matrix remodeling in heart valve disease. Circulation Research. 2004;95(3):253-260
  32. 32. Cushing MC, Liao JT, Anseth KS. Activation of valvular interstitial cells is mediated by transforming growth factor-β1 interactions with matrix molecules. Matrix Biology. 2005;24(6):428-437
  33. 33. Wang F, Zhang C, Kwagh J, Strassle B, Li J, Huang M, et al. TGFβ2 and TGFβ3 mediate appropriate context-dependent phenotype of rat valvular interstitial cells. iScience. 2021;24(3):102133
  34. 34. Han L, Gotlieb AI. Fibroblast growth factor-2 promotes in vitro heart valve interstitial cell repair through the Akt1 pathway. Cardiovascular Pathology [Internet]. 2012;21(5):382-389. DOI: 10.1016/j.carpath.2011.12.001
  35. 35. Weiss A, Attisano L. The TGFbeta superfamily signaling pathway. Wiley Interdisciplinary Review Developmental Biology [Internet]. 2013;2(1):47-63. Available from: https://onlinelibrary.wiley.com/doi/10.1002/wdev.86
  36. 36. Miyazono K. Positive and negative regulation of TGF-β signaling. Journal of Cell Science. 2000;113(7):1101-1109
  37. 37. Meng X, Nikolic-Paterson DJ, Lan HY. TGF-β: The master regulator of fibrosis. Nature Review Nephrology [Internet]. 2016;12(6):325-338. DOI: 10.1038/nrneph.2016.48
  38. 38. Piersma B, Bank RA, Boersema M. Signaling in fibrosis: TGF-β, WNT, and YAP/TAZ converge. Frontiers in Medicine. 2015;2(SEP):1-14
  39. 39. Saitoh M. Regulation of EMT by TGF-$β$ signaling in cancer cells. In: Shinomiya N, Kataoka H, Xie Q , editors. Regulation of Signal Transduction in Human Cell Research [Internet]. Singapore: Springer Singapore; 2018. pp. 71-84. DOI: 10.1007/978-981-10-7296-3_4
  40. 40. Leopold JA. Cellular mechanisms of aortic valve calcification. Circulation. Cardiovascular Interventions. 2012;5(4):605-614
  41. 41. Clayton SW, Ban GI, Liu C, Serra R. Canonical and noncanonical TGF-β signaling regulate fibrous tissue differentiation in the axial skeleton. Scientific Report [Internet]. 2020;10(1):1-11. DOI: 10.1038/s41598-020-78206-4
  42. 42. Hutcheson JD, Chen J, Sewell-Loftin MK, Ryzhova LM, Fisher CI, Su YR, et al. Cadherin-11 regulates cell-cell tension necessary for calcific nodule formation by valvular myofibroblasts. Arteriosclerosis, Thrombosis, and Vascular Biology. 2013;33(1):114-120
  43. 43. Turner N, Grose R. Fibroblast growth factor signalling: From development to cancer. Nature Reviews. Cancer. 2010;10(February):116-129
  44. 44. Li X. Design and discovery of FGF/FGFR inhibitors. In: Li X, editor. Fibroblast Growth Factors [Internet]. Amsterdam: Elsevier; 2018. pp. 339-383. Available from: https://www.sciencedirect.com/science/article/pii/B9780128161425000060
  45. 45. Gospodarowicz D. Localisation of a fibroblast growth factor and its effect alone and with hydrocortisone on 3T3 cell growth. Nature [Internet]. 1974;249(5453):123-127. DOI: 10.1038/249123a0
  46. 46. Beenken A, Mohammadi M. The FGF family: Biology, pathophysiology and therapy. Nature Reviews. Drug Discovery. 2009;8(3):235-253
  47. 47. Latif N, Quillon A, Sarathchandra P, McCormack A, Lozanoski A, Yacoub MH, et al. Modulation of human valve interstitial cell phenotype and function using a fibroblast growth factor 2 formulation. PLoS One [Internet]. 2015;10(6):e0127844. Available from: https://dx.plos.org/10.1371/journal.pone.0127844
  48. 48. Cushing MC, Mariner PD, Liao J, Sims EA, Anseth KS. Fibroblast growth factor represses Smad-mediated myofibroblast activation in aortic valvular interstitial cells. FASEB Journal [Internet]. 2008;22(6):1769-1777. Available from: https://onlinelibrary.wiley.com/doi/10.1096/fj.07-087627
  49. 49. Lam NT, Tandon I, Balachandran K. The role of fibroblast growth factor 1 and 2 on the pathological behavior of valve interstitial cells in a three-dimensional mechanically-conditioned model. Journal of Biological Engineering. 2019;13(1):1-14
  50. 50. Knowles RG, Moncada S. Nitric oxide as a signal in blood vessels. Trends in Biochemical Science [Internet]. 1992;17(10):399-402. Available from: https://linkinghub.elsevier.com/retrieve/pii/096800049290008W
  51. 51. Martin E, Berka V, Tsai AL, Murad F. Soluble guanylyl cyclase: The nitric oxide receptor. Methods in Enzymology [Internet]. 2005;396:478-492. Available from: http://www.ncbi.nlm.nih.gov/pubmed/16291255
  52. 52. Miller JD, Weiss RM, Heistad DD. Calcific aortic valve stenosis: Methods, models, and mechanisms. Circulation Research. 2011;108(11):1392-1412
  53. 53. Sverdlov AL, Ngo DT, Chapman MJ, Ali OA, Chirkov YY, Horowitz JD. Pathogenesis of aortic stenosis: Not just a matter of wear and tear. American Journal of Cardiovascular Diseases [Internet]. 2011;1(2):185-199. Available from: http://www.ncbi.nlm.nih.gov/pubmed/22254198%0Ahttp://www.pubmedcentral.nih.gov/articlerender.fcgi?artid=PMC3253493
  54. 54. Nightingale AK, Horowitz JD. Aortic sclerosis: Not an innocent murmur but a marker of increased cardiovascular risk. Heart. 2005;91(11):1389-1393
  55. 55. Lindman BR, Clavel MA, Mathieu P, Iung B, Lancellotti P, Otto CM, et al. Calcific aortic stenosis. Nature Review in Disease Primer. 2016;2:16006
  56. 56. Guerraty MA, Grant GR, Karanian JW, Chiesa OA, Pritchard WF, Davies PF. Hypercholesterolemia induces side-specific phenotypic changes and peroxisome proliferator-activated receptor-gamma pathway activation in swine aortic valve endothelium. Arteriosclerosis Thrombosis Vascular Biology [Internet]. 2010;30(2):225-231. Available from: http://www.ncbi.nlm.nih.gov/pubmed/19926833
  57. 57. Soini Y, Salo T, Satta J. Angiogenesis is involved in the pathogenesis of nonrheumatic aortic valve stenosis. Human Pathology. 2003;34(8):756-763
  58. 58. Miller JD, Chu Y, Brooks RM, Richenbacher WE, Peña-Silva R, Heistad DD. Dysregulation of antioxidant mechanisms contributes to increased oxidative stress in calcific aortic valvular stenosis in humans. Journal of American College Cardiology [Internet]. 2008;52(10):843-850. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0735109708021165
  59. 59. Kennedy JA, Hua X, Mishra K, Murphy GA, Rosenkranz AC, Horowitz JD. Inhibition of calcifying nodule formation in cultured porcine aortic valve cells by nitric oxide donors. European Journal of Pharmacology [Internet]. 2009;602(1):28-35. DOI: 10.1016/j.ejphar.2008.11.029
  60. 60. Pulfer SK. Nitric Oxide Releasing Polymers and their Application to Vascular Devices [Internet]. Ann Arbor: The University of Akron; 1995. Available from: http://ezproxy.auckland.ac.nz/login?url=https://www.proquest.com/dissertations-theses/nitric-oxide-releasing-polymers-their-application/docview/304166110/se-2?accountid=8424
  61. 61. Gould ST, Matherly EE, Smith JN, Heistad DD, Anseth KS. The role of valvular endothelial cell paracrine signaling and matrix elasticity on valvular interstitial cell activation. Biomaterials [Internet]. 2014;35(11):3596-3606. DOI: 10.1016/j.biomaterials.2014.01.005
  62. 62. Blevins TL, Carroll JL, Raza AM, Grande-Allen KJ. Phenotypic characterization of isolated valvular interstitial cell subpopulations. Journal of Heart Valve Disease [Internet]. 2006;15(6):815-822. Available from: http://europepmc.org/abstract/MED/17152790
  63. 63. Benton JA, Fairbanks BD, Anseth KS. Characterization of valvular interstitial cell function in three dimensional matrix metalloproteinase degradable PEG hydrogels. Biomaterials [Internet]. 2009;30(34):6593-6603. DOI: 10.1016/j.biomaterials.2009.08.031
  64. 64. Benton JA, Deforest CA, Vivekanandan V, Anseth KS. Photocrosslinking of gelatin macromers to synthesize porous hydrogels that promote valvular interstitial cell function. Tissue Engineering – Part A. 2009;15(11):3221-3230
  65. 65. Gu X, Masters KS. Role of the MAPK/ERK pathway in valvular interstitial cell calcification. American Journal of Physiology. 2009;296(6):1748-1757
  66. 66. Gu X, Masters KS. Role of the Rho pathway in regulating valvular interstitial cell phenotype and nodule formation. American Journal of Physiology. 2011;300(2):448-458
  67. 67. Coombs KE, Leonard AT, Rush MN, Santistevan DA, Hedberg-Dirk EL. Isolated effect of material stiffness on valvular interstitial cell differentiation. Journal of Biomedical Materials Research – Part A. 2017;105(1):51-61
  68. 68. Kural MH, Billiar KL. Mechanoregulation of valvular interstitial cell phenotype in the third dimension. Biomaterials [Internet]. 2014;35(4):1128-1137. Available from: https://linkinghub.elsevier.com/retrieve/pii/S0142961213012775
  69. 69. Stephens EH, Huynh TN, Cieluch JD, Grande-Allen KJ. Fibronectin-based isolation of valve interstitial cell subpopulations: Relevance to valve disease. Journal of Biomedical Materials Research Part A [Internet]. 2010;92A(1):340-349. Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3624763/pdf/nihms412728.pdf
  70. 70. Quinlan AMT, Billiar KL. Investigating the role of substrate stiffness in the persistence of valvular interstitial cell activation. Journal of Biomedical Materials Research Part A [Internet]. 2012;23(1). Available from: https://www.ncbi.nlm.nih.gov/pmc/articles/PMC3624763/pdf/nihms412728.pdf
  71. 71. Gu X, Masters KS. Regulation of valvular interstitial cell calcification by adhesive peptide sequences. Journal of Biomedical Materials Research Part A [Internet]. 2010;9999(1). Available from: https://onlinelibrary.wiley.com/doi/10.1002/jbm.a.32660
  72. 72. Porras AM, van Engeland NCA, Marchbanks E, McCormack A, Bouten CVC, Yacoub MH, et al. Robust generation of quiescent porcine valvular interstitial cell cultures. Journal of the American Heart Association. 2017;6(3):1-14
  73. 73. Stephens EH, Carroll JL, Grande-Allen KJ. The use of collagenase III for the isolation of porcine aortic valvular interstitial cells: Rationale and optimization. The Journal of Heart Valve Disease. 2007;16(2):175-183
  74. 74. Gould RA, Butcher JT. Isolation of valvular endothelial cells. Journal of Visualized Experiments. 2010;46:7-12
  75. 75. Cheung WY, Young EWK, Simmons CA. Techniques for isolating and purifying porcine aortic valve endothelial cells. The Journal of Heart Valve Disease. 2008;17(6):674-681
  76. 76. Cimini M, Rogers KA, Boughner DR. Aortic valve interstitial cells: An evaluation of cell viability and cell phenotype over time. The Journal of Heart Valve Disease. 2002;11(6):881-887
  77. 77. Benton JA, Kern HB, Anseth KS. Substrate properties influence calcification in valvular interstitial cell culture. The Journal of Heart Valve Disease. 2008;17(6):689-699
  78. 78. Masjedi S, Amarnath A, Baily KM, Ferdous Z. Comparison of calcification potential of valvular interstitial cells isolated from individual aortic valve cusps. Cardiovascular Pathology [Internet]. 2016;25(3):185-194. DOI: 10.1016/j.carpath.2015.12.002
  79. 79. Ma H, Macdougall LJ, GonzalezRodriguez A, Schroeder ME, Batan D, Weiss RM, et al. Calcium signaling regulates valvular interstitial cell alignment and myofibroblast activation in fast-relaxing boronate hydrogels. Macromolecular Bioscience. 2020;20(12):1-14
  80. 80. Mabry KM, Lawrence RL, Anseth KS. Dynamic stiffening of poly(ethylene glycol)-based hydrogels to direct valvular interstitial cell phenotype in a three-dimensional environment. Biomaterials [Internet]. 2015;49:47-56. DOI: 10.1016/j.biomaterials.2015.01.047
  81. 81. Witt W, Büttner P, Jannasch A, Matschke K, Waldow T. Reversal of myofibroblastic activation by polyunsaturated fatty acids in valvular interstitial cells from aortic valves. Role of RhoA/G-actin/MRTF signalling. Journal of Molecular Cell Cardiology [Internet]. 2014;74:127-138. DOI: 10.1016/j.yjmcc.2014.05.008
  82. 82. Kheradvar A, Zareian R, Kawauchi S, Goodwin RL, Rugonyi S. Animal models for heart valve research and development. Drug Discovery Today Disease Model [Internet]. 2017;24:55-62. DOI: 10.1016/j.ddmod.2018.04.001
  83. 83. Sider KL, Blaser MC, Simmons CA. Animal models of calcific aortic valve disease. International Journal f Inflammation. 2011;2011(Ldl):1-18
  84. 84. Tsang HG, Clark EL, Markby GR, Bush SJ, Hume DA, Corcoran BM, et al. Expression of calcification and extracellular matrix genes in the cardiovascular system of the healthy domestic sheep (Ovis aries). Frontiers in Genetics. 2020;11:1-22
  85. 85. Merryman WD, Youn I, Lukoff HD, Krueger PM, Guilak F, Hopkins RA, et al. Correlation between heart valve interstitial cell stiffness and transvalvular pressure: Implications for collagen biosynthesis. American Journal of Physiology. 2006;290(1):224-231
  86. 86. Jian B, Narula N, Li Q , Mohler ER, Levy RJ. Progression of aortic valve stenosis: TGF-β1 is present in calcified aortic valve cusps and promotes aortic valve interstitial cell calcification via apoptosis. Annals of Thoracic Surgery [Internet]. 2003;75(2):457-465. Available from: https://www.sciencedirect.com/science/article/pii/S0003497502043126
  87. 87. Xue Y, Hilaire C, Hortells L, Phillippi JA, Sant V, Sant S. Shape-specific nanoceria mitigate oxidative stress-induced calcification in primary human valvular interstitial cell culture. Cellular and Molecular Bioengineering. 2017;10(5):483-500
  88. 88. Fang M, Wang CG, Zheng C, Luo J, Hou S, Liu K, et al. Mir-29b promotes human aortic valve interstitial cell calcification via inhibiting TGF-β3 through activation of wnt3/β-catenin/Smad3 signaling. Journal of Cellular Biochemistry. 2018;119(7):5175-5185
  89. 89. Zhang M, Liu X, Zhang X, Song Z, Han L, He Y, et al. MicroRNA-30b is a multifunctional regulator of aortic valve interstitial cells. Journal of Thoracic Cardiovascular Surgery [Internet]. 2014;147(3):1073-1080.e2. DOI: 10.1016/j.jtcvs.2013.05.011
  90. 90. Masoumi N, Larson BL, Annabi N, Kharaziha M, Zamanian B, Shapero KS, et al. Electrospun PGS: PCL microfibers align human Valvular interstitial cells and provide tunable scaffold anisotropy. Advanced Healthcare Materials. 2014;3(6):929-939
  91. 91. Li XF, Wang Y, Zheng DD, Xu HX, Wang T, Pan M, et al. M1 macrophages promote aortic valve calcification mediated by microRNA-214/TWIST1 pathway in valvular interstitial cells. American Journal of Translational Research. 2016;8(12):5773-5783
  92. 92. Venardos N, Nadlonek NA, Zhan Q , Weyant MJ, Reece TB, Meng X, et al. Aortic valve calcification is mediated by a differential response of aortic valve interstitial cells to inflammation. Journal of Surgical Research [Internet]. 2014;190(1):1-8. Available from: file:///C:/Users/Carla%0ACarolina/Desktop/Artigos%0Apara%0Aacrescentar%0Ana%0Aqualificação/The%0Aimpact%0Aof%0Abirth%0Aweight%0Aon%0Acardiovascular%0Adisease%0Arisk%0Ain
  93. 93. Sun W, Zhao R, Yang Y, Wang H, Shao Y, Kong X. Comparative study of human aortic and mitral valve interstitial cell gene expression and cellular function. Genomics [Internet]. 2013;101(6):326-335. DOI: 10.1016/j.ygeno.2013.03.004
  94. 94. Sainger R, Grau JB, Branchetti E, Poggio P, Seefried WF, Field BC, et al. Human myxomatous mitral valve prolapse: Role of bone morphogenetic protein 4 in valvular interstitial cell activation. Journal of Cell Physiology [Internet]. 2012;227(6):2595-2604. Available from: https://onlinelibrary.wiley.com/doi/10.1002/jcp.22999
  95. 95. Artiach G, Carracedo M, Seime T, Plunde O, Laguna-Fernandez A, Matic L, et al. Proteoglycan 4 is increased in human calcified aortic valves and enhances valvular interstitial cell calcification. Cells [Internet]. 2020;9(3):684. Available from: https://www.mdpi.com/2073-4409/9/3/684
  96. 96. Albanese I, Yu B, Al-Kindi H, Barratt B, Ott L, Al-Refai M, et al. Role of noncanonical Wnt signaling pathway in human aortic valve calcification. Arteriosclerosis, Thrombosis, and Vascular Biology. 2017;37(3):543-552
  97. 97. Latif N, Sarathchandra P, Chester AH, Yacoub MH. Expression of smoothmuscle cellmarkers and co-activators in calcified aortic valves. European Heart Journal. 2015;36(21):1335-1345
  98. 98. Galeone A, Brunetti G, Oranger A, Greco G, Di Benedetto A, Mori G, et al. Aortic valvular interstitial cells apoptosis and calcification are mediated by TNF-related apoptosis-inducing ligand. International Journal of Cardiology [Internet]. 2013;169(4):296-304. DOI: 10.1016/j.ijcard.2013.09.012
  99. 99. Poggio P, Branchetti E, Grau JB, Lai EK, Gorman RC, Gorman JH, et al. Osteopontin-CD44v6 interaction mediates calcium deposition via phospho-akt in valve interstitial cells from patients with noncalcified aortic valve sclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2014;34(9):2086-2094
  100. 100. Vadana M, Cecoltan S, Ciortan L, Macarie RD, Tucureanu MM, Mihaila AC, et al. Molecular mechanisms involved in high glucose-induced valve calcification in a 3D valve model with human valvular cells. Journal of Cellular and Molecular Medicine. 2020;24(11):6350-6361
  101. 101. Dittfeld C, Reimann G, Mieting A, Büttner P, Jannasch A, Plötze K, et al. Treatment with XAV-939 prevents in vitro calcification of human valvular interstitial cells. PLoS One. 2018;13(12):1-21
  102. 102. Branchetti E, Sainger R, Poggio P, Grau JB, Patterson-Fortin J, Bavaria JE, et al. Antioxidant enzymes reduce DNA damage and early activation of valvular interstitial cells in aortic valve sclerosis. Arteriosclerosis, Thrombosis, and Vascular Biology. 2013;33(2):66-74
  103. 103. Lucassen A, Wheeler R. Legal implications of tissue. Annals of the Royal College of Surgeons of England. 2010 Apr;92(3):189-192
  104. 104. Ground M, Park YE, Waqanivavalagi S, Callon K, Walker R, Milsom P, et al. Generating robust human valvular interstitial cell cultures: Protocol and considerations. Journal of Molecular Cell Cardiology [Internet]. 2022;173(October):118-126. DOI: 10.1016/j.yjmcc.2022.10.008
  105. 105. Cherng S, Young J, Ma H. Alpha-smooth muscle actin (α -SMA). Journal of American Science [Internet]. 2008;4(4):7-9. Available from: http://sciencepub.net/american/0404/02_0405_mahongbao-sma_am.pdf
  106. 106. Werner S, Lützkendorf J, Müller T, Müller LP, Posern G. MRTF-A controls myofibroblastic differentiation of human multipotent stromal cells and their tumour-supporting function in xenograft models. Scientific Reports. 2019;9(1):1-12
  107. 107. Chalajour F, Treede H, Ebrahimnejad A, Lauke H, Reichenspurner H, Ergun S. Angiogenic activation of valvular endothelial cells in aortic valve stenosis. Experimental Cell Research. 2004;298(2):455-464
  108. 108. Babu AN, Meng X, Zou N, Yang X, Wang M, Song Y, et al. Lipopolysaccharide stimulation of human aortic valve interstitial cells activates inflammation and osteogenesis. The Annals of Thoracic Surgery. 2008;86(1):71-76
  109. 109. Poggio P, Sainger R, Branchetti E, Grau JB, Lai EK, Gorman RC, et al. Noggin attenuates the osteogenic activation of human valve interstitial cells in aortic valve sclerosis. Cardiovascular Research. 2013;98(3):402-410
  110. 110. Rittié L, Fisher GJ. Isolation and culture of skin fibroblasts. In: Fibrosis Research [Internet]. Totowa, NJ: Humana Press; 2005. pp. 83-98. Available from: http://link.springer.com/10.1385/1-59259-940-0:083
  111. 111. Gao Y, Li N, Xue Q , Fan X, Liu X, Han L. Basic fibroblast growth factor inhibits aortic valvular interstitial cells calcification via Notch1 pathway. Journal of Investigative Medicine. 2022;2021:002132

Written By

Marcus Ground, Karen Callon, Rob Walker, Paget Milsom and Jillian Cornish

Submitted: 18 May 2023 Reviewed: 24 July 2023 Published: 18 September 2023