Open access

Pericardial Processing: Challenges, Outcomes and Future Prospects

Written By

Escande Rémi, Nizar Khelil, Isabelle Di Centa, Caroline Roques, Maguette Ba, Fatima Medjahed-Hamidi, Frederic Chaubet, Didier Letourneur, Emmanuel Lansac and Anne Meddahi-Pelle

Submitted: 16 December 2010 Published: 15 September 2011

DOI: 10.5772/24949

From the Edited Volume

Biomaterials Science and Engineering

Edited by Rosario Pignatello

Chapter metrics overview

6,669 Chapter Downloads

View Full Metrics

1. Introduction

The pericardium is a biological tissue widely used as a biomaterial for tissue engineering applications, including the construction of a variety of bioprostheses such as vascular grafts, patches for abdominal or vaginal wall reparation and, more frequently, heart valves.

However, despite significant advances, some drawbacks have been found in these bioprostheses such as biological matrix deterioration and tissue degeneration associated with calcifications, even though xenopericardium or glutaraldehyde-treated autologous pericardium were used.

In non-autologous pericardial processing, the pericardium must be decellularized in order to remove cellular antigens and procalcific remnants while preserving extracellular matrix integrity. A large variety of decellularization protocols exist, such as chemical, physical or enzymatic methods. Additional cross-linking processing must be carried out to render the tissue non-antigenic and mechanically strong.

So far, almost all bioprosthetic materials made of pericardium,and used in clinical practice, are glutaraldehyde-treated bovine or porcine xenopericardium. However, long-term reports are raising issues concerning their durability, especially highlightingthe high risk of calcification. Regarding heart valves, calcification currently represents the major drawback leading to potential failure of the bioprosthesis.

The aim of this review is to present current issues, challenges, outcomes and future prospects of pericardial processing, including decellularization and cross-linking steps. Understanding current issues and improving pericardial processing will allow refining bioprosthesis conception and patients’ safety.

Advertisement

2. Characteristics of the pericardium

2.1. Localization and composition

The pericardium is a connective tissue sac surrounding the heart. It is composed by two layers: a deeper layer closely adherent to the heart, the visceral serous pericardium, orepicardium, and an upper layer: the parietal pericardium. The two layers are separated by the pericardial cavity. The parietal pericardium can be excised and easily tested without causing major complications such as contracture or ischemia (Fomovsky et al., 2010).

Figure 1.

Histology ofovine pericardium showing the collagen organization (arrows). Hematoxylin-Eosin staining.

The pericardium is composed of a simple squamous epithelium and connective tissue. It is a collagen-rich biological tissue containing mostly type I collagen, as well as glycoproteins and glycosaminoglycans (GAGs) in addition to its constitutive cells (Figure 1). Collagenis structured intodifferentlevelsoforganizationranging from fibrils to laminates, fibers and fiber bundles (Allen & Didio, 1984; Lee & Boughner, 1981). This organization determines the mechanical properties of the pericardial tissue (Sacks, 2003; Liao et al., 2005; Wiegner & Bing, 1981; Lee & Boughner, 1985) and provides an anisotropic and non-linear mechanical behaviour (Zioupos & Barbenel, 1994). Interestingly, depending on the location on the pericardium, the thickness and mechanical properties vary (Hiester & Sacks 1998a, 1998b). Thus, the location of the sample that will be harvested should be carefully selected when designing a tissue engineering protocol.

2.2. Sources of pericardium

Currently marketed heart valve bioprostheses are prepared from bovine or porcine pericardium (Vesely, 2005). Other pericardial tissues from different species have been assessed or are currently used in clinical practice such as equine (DeCarbo et al., 2010; Yamamoto et al., 2009; Sato et al.; 2008.), canine (Lee & Boughner ; 1981; Wiegner & Bing, 1981; 1985), or, even more unusually, ostrich (Maestro et al., 2006) or kangaroo pericardium (Neethling et al., 2000; 2002). However, those exogenous grafts raise several issues, and especially the immune response against the bioprosthesis as well as the viral status of the graft.

Human autologous pericardium is thus an interesting option, presenting several advantages over allografts since it is free of donor-derived pathogens and does not induce any immune response (Mirsadraee et al., 2007), is easily available, easily handled and of low cost.Ultimately, these characteristics allow for shorter and less aggressive pericardial processing before implantation of the bioprosthesis. However, because of intermittent reports of its tendency to retract or become aneurysmal, the general opinion has been negative (Edwards et al., 1969, Bahnson et al., 1970). For cusp tissue replacement or valve tissue replacement, stabilization of pericardium is performed with a solution of 0,2% to 0,6% glutaraldehyde in order to prevent secondary shrinkage (Duran et al., 1998; Al-Halees et al., 1998, 2005; Goetz et al., 2002).

Advertisement

3. Processing of pericardium

As allografts have been the main source for pericardial bioprostheses currently in use, significant processing steps have to be performed prior to clinical use. In particular, as xenogeneic cellular antigens induce an immune response or an immune-mediated rejection of the tissue, decellularization protocols are widely used to reduce the host tissue response (Gilbert et al., 2006.). Once decellularized, the free-cell pericardial tissue is composed of extracellular matrix proteins which are generally conserved among species, and thus can be easily used as a scaffold for the host cell attachment, migration and proliferation (Schmidt & Baier, 2000). This scaffold considerably accelerates tissue regeneration. Overall, tissue decellularization aims at reducing tissue antigenicity and host response while preserving the mechanical integrity, biological activity and composition of the ECM (Simon et al., 2006; Gilbert et al., 2006).

3.1. Extracellular matrix decellularization methods

Most decellularization protocolsinclude a combination of various methods, such as physical, enzymatic or chemical treatments (Gilbert et al., 2006; Crapo et al., 2011). Physical methods can either rely on snap freezing (Jackson et al., 1988; Roberts et al., 1991), mechanical force (Freytes et al., 2004) or mechanical agitation (Schenke-Layland et al., 2003), whereas enzymatic protocols employ nucleases, calcium chelating agents or protease digestion (Teebken et al., 2000; Bader et al., 1998; McFetridge et al., 2004; Gamba et al., 2002).

Regarding physical decellularization processes, sonication, based on the use of ultrasounds to disrupt the cell membrane, has been investigated. Such treatment considerably affects the pericardial architecture and full decellularization cannot be achieved. Thus sonication has to be carried out simultaneously with chemical treatments in order to fully decellularize the pericardial tissue and remove cellular debris. However, this combination leads to alterations of the extracellular matrix (ECM) architecture.

For the enzymatic procedure, the main enzyme employed is trypsin, cleaving peptide bonds on the C-side of arginine and lysine and thus allowing separation of the cells from the ECM.

Chemical protocols involve use of alkaline and acid treatments (Freytes et al., 2004), ionic detergents, sodium dodecyl sulfate (SDS), sodium deoxycholate and Triton X-200 (Rieder et al. 2004; Hudson et al., 2004), non-ionic detergents, such as Triton X-100 (Grauss et al., 2003), zwitterionic detergents (Dahl et al., 2003), tri(n-butyl)phosphate (Woods & Gratzer, 2005) as well as hypertonic or hypotonic solutions (Goissis et al., 2000; Woods & Gratzer, 2005; Vyavahareet al., 1997; Dahl et al., 2003). These modalities will either mediate lysis of the cells or solubilization of the cellular components.

Overall, standard decellularization protocols for allografts consist of a multimodal process starting with the lysis of the cell membrane using either ionic solutions or physical treatments. This initial step is then followed by enzymatic treatments to separate any cellular components from the ECM. Subsequently, detergents are used tosolubilize the nuclear and cytoplasmic cellular components. At the end of the procedure, all residual cell debris is removed from the remaining ECM. A washing step must also be carried out following the decellularization protocol to remove residual chemicals, thus avoiding any host tissue response (Gilbert et al., 2006). The efficiency of the decellularization protocol and the preservation of the ECM have to be assessed using histological tools.

Concerning pericardial decellularization, several protocols, which have provided interesting results, can be found in the literature. (Courtman et al., 2004; Liang et al., 2004; Wei et al., 2005; Chang et al., 2005; Mendoza-Novelo et al.,2010, Ariganello et al.,2011 ). Courtman et al. proposed the use of a non-ionic detergent, Triton X-100 and an enzymatic extraction process.After this treatment, the acellular matrix was shown to be composed of collagen, elastin and glycosaminoglycans (GAG). Microscopy revealed that all cellular components were removed and that matrix ultrastructure was intact. More recently, Mendoza-Novelo et al. compared the surfactant tridecyl alcohol ethoxylate (ATE) and the reversible alkaline swelling (RAS) treatments to Triton X-100(Mendoza-Novelo et al., 2010 ). Histological results indicated a significant reduction of cellular antigens with these three decellularization processes. Nevertheless, the native GAG content varied significantly. It decreased from 88.6 ± 0.2% to 62.7 ± 1.1% and 61.6 ± 0.6% for RAS treatment, ATE and Triton X-100 respectively.

On human pericardial tissue, Mirsadraee et al. used a protocol employing hypotonic buffer, SDS, protease inhibitors and nuclease solution. Following decellularization, the tissue is decontaminated using a peracetic acid solution (Mirsadraee et al., 2006; 2007). With this process, glycosaminoglycans and structural proteins, such as collagen, remained intact.

Finally, when dealing with autologous pericardium grafting, full decellularization might not be necessary and thus, simpler protocols can be used. For instance, surgeons commonly prepare autologous pericardium for heart valve replacement by mechanical friction. This allows removing sub-pericardial fat before implantation whilebetter preserving the pericardial architecture stability. This mechanical treatment mainly removes superficial cells, thus allowing 50% of viable pericardial cells to remain in the graft (personal data). The preservation of the pericardial architecture as well as part of the pericardial cells, should maintain a better integrity of the graft, while allowing re-cellularization of the superficial layers.

3.2. Effects of decellularization

Depending on the protocol, decellularization may have an impact on the structural and mechanical properties of the treated tissue (Gilbert et al., 2006). According to Zhou et al., decellularization protocols differ significantly in terms of alteration of ECM histoarchitecture (Zhou et al., 2010). For instance, decellularization protocols have a strong impact on the amount of GAGs remaining in a tissue (Badylak et al., 2009; Mendoza-Novelo et al., 2010). Removing GAGs from a tissue leads to adverse effects on pericardial viscoelastic properties. This can be easily understood since water retention is an important function of GAGs in tissues (Lovekamp et al., 2006). Moreover, GAG content plays a key biological role in cellular signaling and communication. Thus, decreasing GAG content leads to an impaired tissue response and repair.Therefore, the decellularization protocol has to be carefully chosen depending on the tissue type as well as the targeted application.Ideally, the process shouldremove all cellular antigens without compromising the structure and mechanical properties of the tissue.

Liao et al.(Liao et al., 2008) investigated the effect of three decellularization protocols on the mechanical and structural properties on porcine aortic valve leaflets. These protocols were based on the use of SDS, Trypsin and Triton X-100. They showed that decellularization resulted in collagen network disruption, and that the ECM pore size varied as a function of the protocol used. For example, leaflets treated with SDS displayed a dense ECM network and small pore sizes,characteristics that may have an impact on the recolonization of interstitial cells.

It has been demonstrated that decellularization of bovine pericardium with SDS causes irreversible denaturation, swelling and a decrease in tensile strength compared to native tissue (Courtman et al. 1994; García-Paéz et al., 2000; Mendoza-Novelo et al., 2009). Because of these deleterious effects on pericardial tissue, non-ionic detergents are preferred for decellularization processes (Mendoza-Novelo et al., 2010 ). Nevertheless, some issues may be encountered with the use of non-ionic detergents. Indeed, toxic effects (Argese et al., 1994) and estrogenic effects (Soto et al., 1991; Jobling et al., 1993) have been reported after the use of non-ionic detergents such as alkylphenol ethoxylates.

Decellularization mediates alterations of the structural and mechanical properties of the tissue, but this impact varies depending on the protocol used. For instance, Mirsadraee et al. (Mirsadraee et al., 2006) did not observe any significant changes using an SDS-based decellularization protocol in the ultimate tensile strength compared to native tissue on human pericardial tissue. They also observed an increased extensibility of the tissue when cut parallel to collagen bundles.

Tissue decellularization reduces the cellular and humoral immune response targeted against the bioprosthesis (Meyer et al., 2005). However, removing cells does not ensure adequate removal of xenoantigens, nor mitigation of the immune response (Goncalves et al., 2005; Kasimir et al., 2006; Simon et al., 2003; Vesely et al., 1995). For this reason, decellularization protocols have turned to antigen removal protocols (Ueda et al., 2006; Kasimir et al., 2005). The presence of cell membrane antigens, such as oligosaccharide beta-Gal has been reported to lead to an immune response that can be prevented by effective decellularization (Badylak et al., 2008). Interestingly, Griffiths et al. (Griffiths et al., 2008) used an immunoproteomic approach to study the ability of bovine pericardium to generate a humoral immune response. They identified thirty one putative protein antigens. Some of them, such as albumin, hemoglobin chain A and beta hemoglobin have been identified as xenoantigens. Recently, Ariganello et al. provided evidence that decellularized bovine pericardium induced less differentiation of the monocytes to macrophages compared to polydimethylsiloxane or polystyrene surfaces (Ariganello et al., 2010; 2011). Nevertheless, the effects of the host immune response to acellular pericardium remain to be fully characterized. Understanding this phenomenon is necessary to develop new pericardium preparations and thus improve biological scaffold integration and clinical safety (Badylak & Gilbert, 2008).

Overall, no optimal decellularization treatment has been identified so far, but depending on the target tissue as well as the implantation site, the protocol can be adapted to provide the best decellularization efficiency / functional characteristics ratio. Moreover, some additional treatment can be performed following the decellularization step in order to improve the mechanical and biological features of the graft.

Advertisement

4. Pericardial extracellular matrix treatment

The decellularization process will lead to important alterations of the biomaterial. Its mechanical strength will be diminished and after implantation it will undergo rapid resorption. Hence, approximately 60% of the mass of the ECM isdegraded and resorbed between one and three months after in vivografting (Badylak & Gilbert, 2008).It has also been noted that acellular pericardial tissue, mostly made of type I collagen, is highly thrombogenic (Keuren et al., 2004). Finally, preventing calcification of the graft is also a priority to ensure the long-term benefit of the implantation.

To optimize the features of the bioprosthesis before its clinical grafting, several treatments have been developed and are summarized in Table 1.

ReagentsReferences
Cross-linking treatmentAcyl azide(Petite et al., 1990)
Carbodiimides(Sung et al., 2003)
Cyanimide(Pereira et al., 1990)
Dye-mediated photooxidation(Moore et al., 1994)
Epoxy compound(Sung et al., 1997)
Formaldehyde(Nimni et al, 1988)
Genepin(Sung et al., 1999, 2003; Wei et al., 2005)
Glutaraldehyde(Huang-Lee et al, 1990; Jayakrishnan et al., 1996; Thubrikar et al., 1983)
Glutaraldehyde acetals(Yoshioka et al., 2008)
Penta-golloyl glucose(Tedder et al., 2008)
Phytate(Grases et al., 2006, 2008)
Proanthocyanidin(Han et al., 2003)
Reuterin(Chen et al., 2002)
Tannic acid(Cwalina et al., 2005; Jastrzebska et al., 2006;
Wang et al., 2008)
Coating
treatment
Chitosan(Nogueira et al., 2010)
RGD polypeptides(Dong et al., 2009)
Silk fibroin(Nogueira et al., 2010)
Heparin sodium(Lee et al., 2000)
Titanium(Guldner et al., 2009)
Post-fixative treatmentAmino acids(Jorge-Herrero et al., 1996; Moritz et al., 1991)
Glycine(Lee et al., 2010)
Heparin(Lee et al., 2000, 2001)
Hyaluronic acid(Ohri et al., 2004)
L-arginine(Jee et al., 2003)
L-glutamic(Grimm et al., 1991; Leukauf et al., 1993)
Lyophilization(Santibáñez-Salgado et al., 2010)
Sulphonated poly(ethylene oxide)(Lee et al., 2001)

Table 1.

Pericardial processing.

4.1. Cross-linking treatment of pericardial tissue

Cross-linking processing must be carried out to render the tissue non-antigenic, mechanically strong and to minimize xenogeneic tissue degradation (Eliezer et al., 2005; Love, 1997). Nevertheless, degradation should not only be considered as a negative phenomenon, as low molecular weight peptides formed during ECM degradation may have a chemo-attractant potential for several cell types (Badylak & Gilbert, 2008). It is thus the degradation rate of the scaffold that should be primarily considered and evaluated. Depending on the application and cells involved, the degradation rate has to be investigated to ensure proper host cell recruitment and tissue remodelling. The pathways of the immune response involved in this process remain to be fully described (Badylak & Gilbert, 2008).

Introducing cross-links between the polypeptide chains of the ECM has been shown to reduce immunogenicity of the pericardium (Mirsadrae et al., 2007) as well as its biodegradability (Taylor et al., 2006) by increasing its resistance to enzymatic degradation.

Until now, glutaraldehyde (GA)-fixed bovine pericardium has been preferred as a substitute to autologous human pericardium. GA was first introduced by Carpentier et al. (Carpentier et al., 1969) as a cross-linking reagent to chemically modify the collagen andrender the tissue immunologically acceptable in the human host. Fixation was shown to increase stability and strength of the pericardium (Jayakrishnan & Jameela, 1996). GA remains the gold standard as a cross-linking reagent despite its well-known drawbacks. Indeed, GA has been reported to accelerate the calcification process, which considerably limits its application.Calcification is thus the main cause oflong-term failure of GA-fixed pericardial valves (Gallo et al., 1985; Grabenwogeret al., 1996). Furthermore, a GA-treated pericardium has a poor ability to regenerate in vivo due to the cross-linking of the tissue. Moreover GA residues display cytotoxic effects preventing hostcell attachment, migration and proliferation (Huang-Lee et al., 1990).

It is now accepted that GA cross-linking increases tissue stiffness (Thubrikar et al., 1983) with the possibility of tissue buckling (Vesely et al., 1988). Standard use of GA cross-linking leads to a high risk of calcific degeneration as well as tissue fatigue (Grabenwoger et al., 1992). This is mostly due to inflammatory and cytotoxicity changes (Huang Lee et al., 1990), and continuous wear and tear leading to collagen fiber fragmentation.

Besides glutaraldehyde, several cross-linking compounds have been reported in the literature such as genipin (Wei et al., 2005) or epoxy compound (Sung et al., 1997). These alternative methods are used to bridge hydroxylysine residues of different polypeptide chains or amino groups of lysine by oligomeric or monomeric crosslinks (Sung et al., 2003). Because of the adverse effects of cross-linking with glutaraldehyde or other aldehyde treatments such as formaldehyde (Nimni et al., 1988) or dialdehyde starch (Rosenberg, 1978), numerous non-aldehyde treatments have been proposed, such as carbodiimides (Sung et al., 2003), glycerol (Ferrans et al., 1991), glycidal ethers (Thyagarajanet al., 1992) including poly(glycidylether) (Noishiki et al., 1986), acyl azide (Petite et al., 1990), cyanimide (Pereira et al., 1990), genipin (Wei et al., 2005), or dye-mediated photo-oxidation, phytate (Grases et al.,2008).

Genipin, obtained from the fruits of Gardenia jasminoides ELLIS (Fujikawa et al., 1987; Tsai et al., 1994), exhibited better results than glutaraldehyde regarding its cytotoxicity (Sung et al., 1999), inflammatory response, ability to prevent calcification and tissue-induced mechanical properties (Wei et al., 2005). Epoxy compound, initially proposed by Noishiki et al. (Noishiki et al., 1989), was shown to be less cytotoxic, superior in pliability and to better inhibit calcification than glutaraldehyde (Sung et al., 1997).

Carbodiimides generate amide-type crosslinks via direct cross-linking of the polypeptide chains. Use of carbodiimide cross-linking leads to the activation of the carboxylic acid groups of glutamic or aspartic acid residues to obtain O-acylisourea groups. Hydroxyline residues or free amino groups of lysine generate a nucleophilic attack which allows cross-link formation (Timkovich, 1977). It was noted that adding N-hydroxysuccinimides to carbodiimides considerably increases cross-link number (Olde Damink et al., 1996). In addition, the use of carbodiimides displayed increased stability towards enzymatic degradation on collagen-based tissue such as pericardium (Sung et al., 2003).

Glutaraldehyde acetal cross-linking reagent has been developed with glutaraldehyde in acid ethanolic solution (Yoshioka & Goissis, 2008), protecting free aldehydic reactive groups and minimizing the polymeric formation of glutaraldehyde. This reduces superficial effects with glutaraldehyde cross-linking on pericardial tissue.

Crosslinking of the pericardial tissue with a dye-mediated photo-oxydation process provides chemical, enzymatic and in vivo stability as well as biomechanical integrity of the treated tissue (Moore et al., 1994). Penta-golloyl glucose, a collagen-binding polyphenol, stabilizes collagen, preventing its degradation, and allows progressive host cell infiltration as well as ECM remodeling. An invivo study has shown that porcine pericardium does not calcify with such treatment at 6 weeks when implanted subdermally in rats (Tedder et al., 2008). Reuterin, an antimicotic and antibacterial compound obtained from Lactobacillus reuteri(Axelsson et al., 1989), has been studied as a crosslinking reagent (Chen et al., 2002). It is a three-carbon aldehydereacting, as formaldehyde, with free amino groups. Reuterin cross-linked pericardiumexhibits comparable results to glutaraldehyde in terms of resistance against enzymatic degradation, denaturation temperature and free amino group content, while decreasing cytotoxic effects (Chen et al., 2002). Tannic acid has been studied on pericardial tissue and was shown to crosslink proteins by creating multiple hydrogen bonds due to its hydroxyl groups (Cwalina et al., 2005; Jastrzebska et al., 2006). It exerts an anti-inflammatory effect, especially on macrophages, as well as an anti-calcification effect on glutaraldehyde-fixed bovine pericardium (Wang et al., 2008). Proanthocyanidin, a natural crosslinking reagent with polyphenolic structures, has the potential to create a stable hydrogen-bonded structure and to increase collagen synthesis, generating nonbiodegradable collagen matrices (Han et al., 2003). Proanthocyanidin-treated pericardial tissues are non-cytotoxic and resist against enzyme digestion, and have been shown to be compatible with cell attachment and proliferation. Phytate has been suggested as an anti-calcification reagent (Grases et al., 2006, 2008) and has achieved promising results, to be validated by further studies. Other amide-type crosslinks, based on the activation of carboxyl groups, have been studied, such as diphenylphosphorylazide or ethyldimethylaminopropyl carbodiimide. It appears, according to Jorge-Herrero et al., that these two chemical treatments are not a good alternative compared to glutaraldehyde. Indeed, pericardial tissues treated with those reagents are less resistant to calcifications and proteolytic attacks (Jorge-Herrero et al., 1999).

Numerous alternative treatments to glutaraldehyde cross-linking have been developed and investigated over the years. However, most of them were mainlyevaluatedin vitro and compared only to glutaraldehyde. A comprehensive comparative study of the different reagents remains to be conducted in terms of benefits regarding the tissue properties as well as their potential toxicity or deleterious effects.

4.2. Coating of the pericardium

Another possible post-decellularization treatment resides in the coating of the bioprosthesis. This procedure should allow improvement of graft integration at the site of implantation as well as decreasing degradation of the pericardial tissue.

Coating bovine pericardium with biopolymeric films, either chitosan or silk fibroin, has been investigated by Nogueira et al. (Nogueira et al., 2010). These methods are interesting approaches and both treatments appear to be non-cytotoxic. Nevertheless, chitosan does not allow endothelialisation and silk fibroin-coated bovine pericardium calcifies in vivo. Further investigation has to be performed to tackle these major concerns.

In their study, Dong et al. suggested treating bovine pericardium with acetic acid coupled with RGD polypeptides (Dong et al., 2009). Acetic acid increases pericardial scaffold pore size and porosity while RGD peptides is meant to improve cell adhesion and growth. Hence, RGD polypeptides have been identified in fibronectin (Pierschbacher & Ruoslahti, 1984), collagen, vitronectin and membrane proteins (Ruoslahti & Pierschbacher, 1987). These sequences have an impact on integrins, which display cell adhesion receptor roles controlling cell signaling pathways.

4.3. Pericardium anti-calcification treatments

The mechanism of calcification on glutaraldehyde-treated pericardium is not well understood because of its complexity. Nevertheless, there is evidence that pericardial tissue residual antigens, free aldhehyde groups of glutaraldehyde and phospholipids are involved in this mechanism.

Thus, circulating antibodies can contribute to pericardial calcification due to a possible immune response. Free aldehyde groups of glutaraldehyde can attract host plasma calcium, increasing tissue calcification. Phospholipids may bind calcium and play an important role in the calcium phosphate crystal formation. Several strategies have been investigated to tackle these major issues.

Suppression of residual antigenicity has been proposed to prevent calcification and it has been shown to be effective. This was performed by fixation treatments using a broad rangeof high concentrations of glutaraldehyde (Trantina-Yates et al., 2003; Zilla et al., 2000). To remove free aldehyde groups, a large number of amino acids or amino compounds were studied. Post-fixation treatments with aminoacids displayedan improved spontaneous endothelialisation in vivo of glutaraldehyde-fixed bovine pericardium (Moritz et al., 1991; Jorge-Herrero et al., 1996).The use of L-glutamic acid did reduce residual and unbound aldehyde groups, on glutaraldehyde-fixed bovine pericardium and significantly decreased the risk of calcification (Grimm et al., 1991; Leukauf et al., 1993). Post-treatment with L-arginine also resulted in decreased calcium deposition (Jee et al., 2003). Recently, Lee et al. proposed a post-fixation treatment with glycine (Lee et al., 2010). Early results are promising but require further investigation on larger studies.

Alcohol solutions, including ethanol, have been investigated as a treatment to remove tissue phospholipids, thus preventing calcification (Pathak et al., 2004; Vyavahare et al., 1998). Besides, other techniques have been proposed to minimize the side effects of glutaraldehyde residues on GA-treated pericardium. Lyophilization has been shown to decrease aldehyde residues, decreasing the risk of calcification and cytotoxicity (Santibáñez-Salgado et al., 2010).

Moreover, treatments with heparin or sulphonated poly(ethylene oxide) following glutaraldehyde pre-treatment have been proposed (Lee et al., 2000, 2001). Both methods blockside effects of GA residues and thus prevent calcification of the pericardium. Finally,a modified adipic dihydrazide hyaluronic acid has been proposed to be grafted on to glutaraldehyde-treated bovine pericardium (Ohri et al., 2004). Calcifications decreased considerably with this post-treatment compared to the control group at two weeks following a subcutaneous implantation in mice.

Advertisement

5.Applications of the pericardium as a biomaterial

So far, the pericardium has been mostly used for cardio-vascular applications, i.e. vascular grafts (Schmidt & Baier, 2000; Chvapil et al., 1970; Matsagas et al., 2006; Menasche et al., 1984

Pericardium
source
SurgicalfieldsProductCompany
Bovine orporcineSoft tissue repair
Hernia repair
Abdominal & thoracic wall defects
-Peripatch Implantable surgical tissue
-TutoMesh
Neovasc,
Maverick Biosciences PTY Limited,
Tutogen medical GmbH, RTI Biologics,
Med&Care,
Biovascular Inc,
Novomedics
Strip reinforcement-Veritas Peristrips DrySynovis Life Technology
Orbital repair-Tutopatch
-Ocugard
Tutogen medical GmbH, RTI Biologics,
Med&Care,
Biovascular Inc,
Novomedics
Dural repair-Lyolem r All BPNational tissue Bank Malaysia
Perivascular Patch-Peripatch biologic vascular patchNeovac
Cardiac reconstruction and repair-Peripatch Implantable Surgical TissueNeovasc,Maverick Biosciences PTY Limited
Heart valve replacement-PercevalS aortic valve
-Mitroflow pericardial aortic valve
-Freedom solo
-Carpentier-Edwards PERIMOUNT Magna EaseAortic Heart Valve
-Carpentier-Edwards PERIMOUNTMagna Mitral Ease Heart Valve
-Carpentier-Edwards PERIMOUNT TheonAortic Heart Valve
-Carpentier-Edwards PERIMOUNT TheonMitral Replacement System
Sorin group


Edwards Life Sciences







EquineTendon repair-OrthADAPTSynovis Life Technologies Inc
HumanValvuloplasty
Heart valve
-Xeno or (tissue bank) or autologous graftsLausberg et al, 2006
Mirsadaee et al, 2006

Table 2.

Applications of pericardium as medical devices.

; Moon & West; 2008), and heart valves (Ishihara et al., 1981; Schoen & Levy, 1999; Flanagan & Pandit, 2003; Vesely, 2005). Pericardial bioprostheseshave also been described for the treatment of acquired cardiac pathologies, including postinfarction septal defects (David et al., 1995), reconstruction of mitral valve annulus (David et al., 1995a, 1995b) or outflow obstruction (Sommers & David, 1997).

Additionally, pericardiumhas also been used for the construction of bioprostheses in non-cardiac treatments such as patches for vaginal (Lazarou et al., 2005) or abdominal wall reparation (Limpert et al., 2009), dural repair (Cantore et al., 1987) or tracheoplasty (Dunham et al., 1994).

Advertisement

6. Conclusion

For clinical application, pericardial tissue has to be decellularized to prevent an immune responses or immune-mediated rejection of the pericardium. Various decellularization protocols have been largely reviewed here. The choice of the decellularization strategy has an impact on the mechanical properties, the scaffold pore size, the scaffold tissue integration and the development of long-term calcification. All these considerations should be carefully taken into account when designing new pericardial-based biomaterials. Currently, glutaraldehyde is the gold standard for pericardial treatment used in clinical practice. Nevertheless, it has important drawbacks including cytotoxic effects, prevention of host cell attachment, migration and proliferation (Huang-Lee et al., 1990), and a high propensity to calcify. Alternative treatments to replace or complement glutaraldehyde crosslinking of the pericardium have been investigated using other crosslinking reagents, decellularization, lyophilisation or coating with biopolymers (Nogueira et al., 2010). Despite many studies, it is still difficult to know which strategy to adopt regarding pericardial treatment. First, we do not have enough follow-up to permit evaluation of most of these alternatives and treatments. Second, every new treatment proposed is generally compared only to glutaraldehyde. It is thus not possible to classify these treatments by efficiency. Finally, the protocol for an optimal treatment depends largely on the final application targeted. In addition, there have been recent advances in tissue regeneration with the emergence of cell therapy and new pericardial treatments with cellular growth factors promoting recellularization (Chang et al., 2007). However, further improvements need to be achieved to transform these techniques into clinical applications. The use of autologous pericardium in cardiac valvular therapy is also a challenging alternative. Nevertheless, it still currently requires the development of local pericardial treatments aiming to favor the valvular remodelling. The understanding of current issues and the improvement of pericardial processing may have a huge impact for bioprothesis conception and patient safety.

References

  1. 1. Al-HaleesZ.GometzaB.DuranC. M.Aortic valve repair with bovine pericardium. (1998Ann Thorac Surg, 65601602
  2. 2. Al-HaleesZ.AlShahid. M.AlSanei. A.SallehuddinA.DuranC.Upto. .yearsfollow-up.ofaortic.valvereconstruction.withpericardium. a.stentlessreadily.availablecheap.valve?2005Eur J Cardiothorac Surg. Aug; 28200205
  3. 3. Allen DJ, Didio LJA.1984The structure of native human, bovine and porcine parietal pericardium. Anatomical Record, 20837A7A
  4. 4. ArgeseE.MarcominiA.BettiolC.PerinG.MianaP.1994Submitochondrial particle response to linear alkylbenzene sulfonates, nonylphenol polyethoxylates and their biodegradation derivates. Environ Toxicol Chem, 13737742
  5. 5. Ariganello MB, Labow RS, Lee JM.2010In vitro response of monocyte-derived macrophages to a decellularized pericardial biomaterial. J Biomed Mater Res, 931280288
  6. 6. Ariganello MB, Simionescu DT, Labow RS, Lee JM.2011Macrophage differentiation and polarization on a decellularized pericardial biomaterial. Biomaterials, 322439449
  7. 7. AxelssonL.ChungT. C.DobrogoszW. J.LindgrenL. E.1989Production of a broad spectrum antimicrobial substance by Lactobacillusreuteri. Microbial Ecol Health Disease, 2131136
  8. 8. BaderA.SchillingT.TeebkenO. E.BrandesG.HerdenT.SteinhoffG.HaverichA.1998Tissue engineering of heart valves-human endothelial cell seeding of detergent acellularized porcine valves. Eur J Cardiothorac Surg, 143279284
  9. 9. Badylak SF, Freytes DO, Gilbert TW.2009Extracellular matrix as a biological scaffold material: structure and function. Acta Biomater, 51113
  10. 10. Badylak SF, Gilbert TW.2008Immune response to biologic scaffold materials. Seminars in Immunology, 202109116
  11. 11. BahnsonH. T.HardestyR. L.BakerL. D.BrookesD. I. I.GallD. A.1970Fabrication and evaluation of tissue leafletsfor aortic and mitral valve replacement. Ann Surg, 171939947
  12. 12. CantoreG.GuidettiB.DelfiniR.1987Neurosurgical use of human dura mater sterilized by gamma rays and stored in alcohol: long-term results. J Neurosurg, 6619395
  13. 13. CarpentierA.LemaigreG.RobertL.CarpentierS.DubostC.1969Biological factors affecting long-term results in valvular heterografts. J Thorac Cardiovasc Surg, 584467483
  14. 14. ChangY.ChenS. C.WeiH. J.WuT. J.LiangH. C.LaiP. H.YangH. H.SungH. W.2005Tissue regeneration observed in a porous acellular bovine pericardium used to repair a myocardial defect in the right ventricle of a rat model. J Thorac Cardiovasc Surg, 1305705711
  15. 15. ChangY.LaiP. H.WeiH. J.LinW. W.ChenC. H.HwangS. M.ChenS. C.SungH. W.2007Tissue regeneration observed in a basic fibroblast growth factor-loaded porous acellular bovine pericardium populated with mesenchymal stem cells. J Thorac Cardiovasc Surg, 13416573
  16. 16. Chen CN, Sung HW, Liang HF, Chang WH.2002Feasibility study using a natural compound (reuterin) produced by Lactobacillus reuteri in sterilizing and crosslinking biological tissues. J Biomed Mater Res, 613360369
  17. 17. ChvapilM.KronenthalR. L.van Winkle JrW.1970Medical and surgical applications of collagen, In: International review of connective tissue research, Hall DA, Jackson DS. 6161Chapter 1], Academic Press, NY.
  18. 18. CourtmanD. W.CAPereiraKashef. V.Mc CombD.LeeJ. M.WilsonG. J.1994Development of a pericardial acellular matrix biomaterial: biochemical and mechanical effects of cell extraction. J Biomed Mater Res, 286655666
  19. 19. Crapo PM, Gilbert TW, Badylak SF.2011An overview of tissue and whole organ decellularization processes. Biomaterials, 321232333243
  20. 20. CwalinaB.TurekA.NozynskiJ.JastrzebskaM.NawratZ.2005Structural changes in pericardium tissue modified with tannic acid. Int J Artif Organs, 286648653
  21. 21. DahlS. L.KohJ.PrabhakarV.NiklasonL. E.2003Decellularized native and engineered arterial scaffolds for transplantation. Cell Transplant, 126659666
  22. 22. DavidT. E.DaleL.SunZ.1995Postinfarction ventricular septal rupture: Repair by endocardial patch with infarct exclusion. J Thorac Cardiovasc Surg, 110513151322
  23. 23. DavidT. E.FeindelC. M.ArmstrongS.et al.1995Reconstruction of the mitral annulus. A ten-year experience. J Thorac Cardiovas Surg, 1101323x1328
  24. 24. DeCarbo WT, Feldner BM, Hyer CF.2010Inflammatory Reaction to Implanted Equine Pericardium Xenograft. J Foot Ankle Surg, 492155158
  25. 25. DongX.WeiX.YiW.GuC.KangX.LiuY.LiQ.YiD.2009RGD-modified acellular bovine pericardium as a bioprosthetic scaffold for tissue engineering. J Mater Sci Mater Med, 2023272336
  26. 26. MEDunhamHolinger. L. D.BackerC. L.MavroudisC.1994Management of severe congenital tracheal stenosis. Ann Otol Rhinol Laryngol, 103351356
  27. 27. DuranC. M.GometzaB.ShahidM.Al-HaleesZ.Treated bovine and autologous pericardium for aortic valve reconstruction. (1998Ann Thorac Surg. Dec; 66S166S199
  28. 28. Edwards WS.1969Aortic valve replacement with autogenous tissue. Ann Thorac Surg 8126132
  29. 29. Eliezer MA, Lydia MM, Virna VR, Carlos FR.2005Mechanics of biomaterials: vascular graft prostheses. Proceedings of Application of Engineering Mechanics in Medicine, GED, University of Puerto Rico, Mayaguez, Group A: A1A25, May 2005.
  30. 30. FerransV. J.MileiJ.IshiharaT.StorinoR.1991Structural changes in implanted cardiac valvular bioprostheses constructed of glycerol-treated dura mater. Eur J Cardiothorac Surg, 53144154
  31. 31. FlanaganT. C.PanditA.2003Living artificial heart valve alternatives: a review. Eur Cell Mater, 62845
  32. 32. FomovskyG. M.ThomopoulosS.HolmesJ. W.2010Contribution of extracellular matrix to the mechanical properties of the heart. J Mol Cell Cardiol, 483490496
  33. 33. Freytes DO, Badylak SF, Webster TJ, Geddes LA, Rundell AE.2004Biaxial strength of multilaminated extracellular matrix scaffolds. Biomaterials, 251223532361
  34. 34. FujikawaS.YokotaT.KogaK.KumadaJ.1987The continuous hydrolysis of geniposide to genipin using immobilized β-glucosidase on calcium alginate gel. J Biotechnol Lett, 9697702
  35. 35. GalloI.ArtinanoE.NistalF.1985Four- to seven-year follow-up of patients undergoing Carpentier-Edwards porcine heart valve replacement. Thorac Cardiovasc Surg, 336347351
  36. 36. GambaP. G.ConconiM. T.LoPiccolo. R.ZaraG.SpinazziR.ParnigottoP. P.2002Experimental abdominal wall defect repaired with acellular matrix. Pediatr Surg Int, 185-6327331
  37. 37. García-PaézJ. M.HerreroJ.Carrera-SanMartín. A.García-SestafeJ. V.TéllezG.MillánI.SalvadorJ.CordonA.Castillo-OlivaresJ. L.2000The influence of chemical treatment and suture on the elastic behavior of calf pericardium utilized in the construction of cardiac bioprostheses. J Mater Sci Mater Med, 115273277
  38. 38. Gilbert TW, Sellaro TL, Badylak SF.2006Decellularization of tissues and organs. Biomaterials, 271936753683
  39. 39. GoetzW. A.LimH. S.LansacE.WeberP. A.DuranC. M. A.temporarilystented.autologouspericardial.aorticvalve.prosthesis2002J Heart Valve Dis.Sep;11(5):(696-702).
  40. 40. GoissisG.SuziganS.ParreiraD. R.ManigliaJ. V.BraileD. M.RaymundoS.2000Preparation and characterization of collagen-elastin matrices from blood vessels intended as small diameter vascular grafts. Artif Organs, 243217223
  41. 41. Goncalves AC, Griffiths LG, Anthony RV, Orton EC.2005Decellularization of bovine pericardium for tissue-engineering by targeted removal of xenoantigens. J Heart Valve Dis, 142212217
  42. 42. GrabenwogerM.GrimmM.EyblE.LeukaufC.MMMullerPlenck.Jr BöckH.P.1992Decreased tissue reaction to bioprosthetic heart valve material after glutamic acid treatment. A morphological study. J Biomed Mater Res, 26912311240
  43. 43. GrabenwogerM.SiderJ.FitzalF.ZelenkaC.WindbergerU.GrimmM.MoritzA.BöckA.WolnerE.1996Impact of glutaraldehyde on calcification of pericardial bioprosthetic heart valve material. Ann Thorac Surg, 623772777
  44. 44. GrasesF.SanchisP.Costa-BauzáA.BonninO.IsernB.PerellóJ.PrietoR. M.2008Phytate inhibits bovine pericardium calcification in vitro. Cardiovasc Pathol, 173139145
  45. 45. GrasesF.SanchisP.PerelloJ.IsernB.PrietoR. M.Fernandez-PalomequeC.FiolM.BonninO.TorresJ. J.2006Phytate (myo-inositol hexakisphosphate) inhibits cardiovascular calcifications in rats. Front Biosci, 11136142
  46. 46. GraussR. W.HazekampM. G.van VlietS.Gittenberger-deGroot. A. C.De RuiterM. C.2003Decellularization of rat aortic valve allografts reduces leaflet destruction and extracellular matrix remodeling. J Thorac Cardiovasc Surg, 126620032010
  47. 47. Griffiths LG, Choe LH, Reardon KF, Dow SW, Orton EC.2008Immunoproteomic identification of bovine pericardium xenoantigens. Biomaterials, 292635143520
  48. 48. GrimmM.EyblE.GrabenwogerM.GriesmacherA.LosertU.BockP.MMMullerWolner. E.1991Biocompatibility of aldehyde-fixed bovine pericardium. An in vitro and in vivo approach toward improvement of bioprosthetic heart valves. J Thorac Cardiovasc Surg, 1022195201
  49. 49. GuldnerN. W.JasmundI.ZimmermannH.HeinleinM.GirndtB.MeierV.FlüßF.RohdeD.GebertA.SieversH. H.2009Detoxification and Endothelialization of Glutaraldehyde-Fixed Bovine Pericardium With Titanium Coating: A New Technology for Cardiovascular Tissue Engineering. Circulation, 1191216531660
  50. 50. HanB.JaurequiJ.WeiTang. B.MENimni2003Proanthocyanidin: A natural crosslinking reagent for stabilizing collagen matrices. J Biomed Mater Res A, 651118124
  51. 51. Hiester ED, Sacks MS.1998aOptimal bovine pericardial tissue selection sites. I. Fiber architecture and tissue thickness measurements. J Biomed Mater Res, 392207214
  52. 52. Hiester ED, Sacks MS.1998bOptimal bovine pericardial tissue selection sites. II. Cartographic analysis. J Biomed Mater Res, 392215221
  53. 53. Huang Lee LLH, Cheung DT, Nimni ME.1990Biochemical changes and cytotoxicity associated with the degradation of polymeric glutaraldehyde derived cross-links. J Biomed Mater Res, 24911851201
  54. 54. HudsonT. W.ZawkoS.DeisterC.LundyS.HuC. Y.LeeK.CESchmidt2004Optimized acellular nerve graft is immunologically tolerated and supports regeneration. Tissue Eng, 1011-1216411651
  55. 55. IshiharaT.FeransV. J.JonesM.BoyceS. W.RobertsW. C.1981Structure of bovine parietal pericardium and of unimplanted Ionescu shiley pericardial valvular bioprostheses. J Thorac Cardiovasc Surg, 815747757
  56. 56. JacksonD. W.GroodE. S.WilcoxP.ButlerD. L.SimonT. M.HoldenJ. P.1988The effects of processing techniques on the mechanical properties of bone-anterior cruciate ligament-bone allografts. An experimental study in goats. Am J Sports Med, 162101105
  57. 57. JastrzebskaM.Zalewska-RejdakJ.WrzalikR.KocotA.MrozI.BarwinskiB.TurekA.CwalinaB.2006Tannic acid-stabilized pericardium tissue: IR spectroscopy, atomic force microscopy, and dielectric spectroscopy investigations. J Biomed Mater Res A, 781148156
  58. 58. JayakrishnanA.JameelaS. R.1996Glutaraldehyde as a fixative in bioprostheses and drug delivery matrices. Biomaterials, 175471484
  59. 59. Jee KS, Kim YS, Park KD, Kim YH.2003A novel chemical modification of bioprosthetic tissues using L-arginine. Biomaterials, 242034093416
  60. 60. JoblingS.SumpterJ. P.1993Detergent components in sewage effluent are weakly estrogenic to fish- an in vitro study using rainbow-trout. Aquat Toxicol, 27361372
  61. 61. Jorge-HerreroE.FernandezP.EscuderoC.Garcia-PaezJ. M.Castillo-OlivaresJ. L.1996Calcification of pericardial tissue pretreated with different amino acids. Biomaterials, 176571575
  62. 62. Jorge-HerreroE.FernandezP.TurnayJ.OlmoN.CaleroP.GarciaR.FreileI.Castillo-OlivaresJ. L.1999Influence of different chemical cross-linking treatments on the properties of bovine pericardium and collagen. Biomaterials, 206539545
  63. 63. KasimirM. T.RiederE.SeebacherG.NigischA.DekanB.WolnerE.WeigelG.SimonP.2006Decellularization does not eliminate thrombogenicity and inflammatory stimulation in tissue-engineered porcine heart valves. J Heart Valve Dis, 152278286discussion 286].
  64. 64. KasimirM. T.RiederE.SeebacherG.WolnerE.WeigelG.SimonP.2005Presence and elimination of the xenoantigen gal (alpha1, 3) gal in tissue-engineered heart valves. Tissue Eng, 117-812741280
  65. 65. KeurenJ. F. W.WieldersS. J. H.DriessenA.VerhoevenM.HendriksM.LindhoutT.2004Covalently-bound heparin makes collagen thromboresistant. Arterioscler Thromb Vasc Biol, 243613617
  66. 66. LazarouG.PowersK.PenaC.BruckL.MSMikhail2005Inflammatory reaction following bovine pericardium graft augmentation for posterior vaginal wall defect repair. Int Urogynecol J Pelvic Floor Dysfunct, 163242244
  67. 67. LeeC.KimS. H.ChoiS. H.KimY. J.2011High-concentration glutaraldehyde fixation of bovine pericardium in organic solvent and post-fixation glycine treatment: in vitro material assessment and in vivo anticalcification effect. Eur J Cardiothorac Surg, 393381387
  68. 68. Lee JM, Boughner DR.1981Tissue mechanics of canine pericardium in different test environment. Circ Res, 492533544
  69. 69. Lee JM, Boughner DR.1985Mechanical properties of human pericardium. Differences in viscoelastic response when compared with canine pericardium. Circ Res, 573475481
  70. 70. LeeW. K.ParkK. D.HanD. K.SuhH.ParkJ. C.KimY. H.2000Heparinized bovine pericardium as a novel cardiovascular bioprosthesis. Biomaterials, 212223232330
  71. 71. LeeW. K.ParkK. D.KimY. H.SuhH.ParkJ. C.LeeJ. E.SunK.MJBaekKim. H. M.KimS. H.2001Improved Calcification Resistance and Biocompatibility of Tissue Patch Grafted with Sulfonated PEO or Heparin after Glutaraldehyde Fixation. J Biomed Mater Res, 5812735
  72. 72. LeukaufC.SzelesC.SalaymehL.GrimmM.GrabenwogerM.LosertU.MoritzA.WolnerE.1993In vitro and in vivo endothelialization of glutaraldehyde treated bovine pericardium. J Heart Valve Dis, 22230235
  73. 73. LiangH. C.ChangY.HsuC. K.LeeM. H.SungH. W.2004Effects of crosslinking degree of an acellular biological tissue on its tissue regeneration pattern. Biomaterials, 251735413552
  74. 74. LiaoJ.JoyceE. M.MSSacks2008Effects of decellularization on the mechanical and structural properties of the porcine aortic valve leaflet. Biomaterials, 29810651074
  75. 75. LiaoJ.YangL.GrashowJ.MSSacks2005Molecular orientation of collagen in intact planar connective tissues under biaxial stretch. Acta Biomater, 114554
  76. 76. Limpert JN, Desai AR, Kumpf AL, Fallucco MA, Aridge DL.2009Repair of abdominal wall defects with bovine pericardium. Am J Surg, 198e60e65
  77. 77. Love JW.1997Cardiac prostheses, In: Principles of tissue engineering, Lanza R, Langer R, Chick W, 365378R.G. Landes Company, New York.
  78. 78. LovekampJ. J.SimionescuD. T.MercuriJ. J.ZubiateB.MSSacksVyavahare. N. R.2006Stability and function of glycosaminoglycans in porcine bioprosthetic heart valves. Biomaterials, 27815071518
  79. 79. MMMaestroTurnay. J.OlmoN.FernándezP.SuárezD.GarcíaPáez. J. M.UrilloS.MALizarbe-HerreroJorge.E.2006Biochemical and mechanical behavior of ostrich pericardium as a new biomaterial. Acta Biomater,22213219
  80. 80. MatsagasM. I.BaliC.ArnaoutoglouE.PapakostasJ. C.NassisC.PapadopoulosG.KappasA. M.2006Carotid endarterectomy with bovine pericardium patch angioplasty: mid-term results. Ann Vasc Surg, 205614619
  81. 81. Mc FetridgeP. S.DanielJ. W.BodamyaliT.HorrocksM.ChaudhuriJ. B.2004Preparation of porcine carotid arteries for vascular tissue engineering applications. J Biomed Mater Res A, 702224234
  82. 82. MenascheP.FlaudP.HucCo. A.PiwnicaA.1984Collagen vascular grafts: a step towards improved compliance in small-caliber by-pass surgery: preliminary report. Life Support Syst, 24233237
  83. 83. Mendoza-NoveloB.EEAvila-RodríguezCauich.Jorge-HerreroJ. V.RojoE.GuineaF. J.Mata-MataG. V.J. L.2011Decellularization of pericardial tissue and its impact on tensile viscoelasticity and glycosaminoglycan content. Acta Biomater, 7312411248
  84. 84. Mendoza-NoveloB.Cauich-RodriguezJ. V.2009The effect of surfactants, crosslinking agents and L-cysteine on the stabilization and mechanical properties of bovine pericardium. J Appl Biomater Biomech, 72123131
  85. 85. MeyerS. R.NagendranJ.DesaiL. S.RayatG. R.ChurchillT. A.AndersonC. C.RajotteR. V.LakeyJ. R. T.RossD. B.2005Decellularization reduces the immune response to aortic valve allografts in the rat. J Thorac Cardiovasc Surg, 1302469476
  86. 86. MirsadraeeS.WilcoxH. E.KorossisS.KearneyJ. N.WattersonK. G.FisherJ.InghamE.2006Development and characterization of an acellular human pericardial matrix. Tissue Eng, 124763773
  87. 87. MirsadraeeS.WilcoxH. E.WattersonK. G.KearneyJ. N.HuntJ.FisherJ.InghamE.2007Biocompatibility of Acellular Human Pericardium. J Surg Res, 1432407414
  88. 88. Moon JJ, West JL.2008Vascularization of engineered tissues: approaches to promote angiogenesis in biomaterials. Curr Top Med Chem, 84300310
  89. 89. MAMooreBohachevsky. I. K.CheungD. T.BDBoyanChen. W.BickersR. R.Mc IlroyB. K.1994Stabilization of pericardial tissue by dye-mediated photooxidation. J Biomed Mater Res, 285611618
  90. 90. MoritzA.GrimmM.EyblE.GrabenwogerM.GrabenwogerF.BockP.WolnerE.1991Improved spontaneous endothelialization by postfixation treatment of bovine pericardium. Eur J Cardiothorac Surg, 53155159discussion 160].
  91. 91. NeethlingW. M.CooperS.Van DenHeever. J. J.HoughJ.HodgeA. J.2002Evaluation of kangaroo pericardium as an alternative substitute for reconstructive cardiac surgery. J Cardiovasc Surg (Torino), 433301306
  92. 92. NeethlingW. M.PapadimitriouJ. M.SwartsE.HodgeA. J.2000Kangaroo versus porcine aortic valve tissue-valve geometry morphology, tensile strength and calcification potential. J Cardiovasc Surg (Torino), 413341348
  93. 93. MENimniCheung. D.StratesB.KodamaM.SheikhK.1988Bioprosthesis derived from cross-linked and chemically modified collagenous tissues, In: Collagen, Vol. III, Nimni ME, 138CRC Press, Boca Raton, FL.
  94. 94. NogueiraG. M.RodasA. C. D.WeskaR. F.AimoliC. G.HigaO. Z.MaizatoM.AALeinerPitombo. R. N. M.PolakiewiczB.MMBeppu2010Bovine pericardium coated with biopolymeric films as an alternative to prevent calcification: In vitro calcification and cytotoxicity results. Materials Science and Engineering C, 30575582
  95. 95. NoishikiY.KodairaK.FuruseM.MiyataT.1989Method of preparing antithrombogenic medical materials, U.S. Patent 4
  96. 96. NoishikiY.MiyataT.KodairaK.1986Development of a small caliber vascular graft by a new crosslinking method incorporating slow heparin release collagen and natural tissue compliance. ASAIO Trans, 321114119
  97. 97. OhriR.HahnS. K.ASHoffmanStayton. P. S.GiachelliC. M.2004Hyaluronic acid grafting mitigates calcification of glutaraldehyde-fixed bovine pericardium. J Biomed Mater Res A, 702328334
  98. 98. OldeDamink. L. H.DijkstraP. J.MJvan Luyn vanWachem. P. B.NieuwenhuisP.FeijenJ.1996Cross-linking of dermal sheep collagen using a water-soluble carbodiimide. Biomaterials, 178765773
  99. 99. CPPathakAdams. A. K.SimpsonT.PhillipsR. E.MAMoore2004Treatment of bioprosthetic heart valve tissue with long chain alcohol solution to lower calcification potential. J Biomed Mater Res A, 691140144
  100. 100. CAPereiraLee. J. M.HabererS.1990Effect of alternative crosslinking methods on the strain rate viscoelastic properties of bovine pericardial bioprosthetic material. J Biomed Mater Res, 243345361
  101. 101. PetiteH.RaultI.HucA.MenascheP.HerbageD.1990Use of the acyl azide method for cross-linking collagen-rich tissues such as pericardium. J Biomed Mater Res, 242179187
  102. 102. MDPierschbacherRuoslahti. E.1984Cell attachment activity of fibronectin can be duplicated by small synthetic fragments of the molecule. Nature, 30959633033
  103. 103. RiederE.KasimirM. T.SilberhumerG.SeebacherG.WolnerE.SimonP.WeigelG.2004Decellularization protocols of porcine heart valves differ importantly in efficiency of cell removal and susceptibility of the matrix to recellularization with human vascular cells. J Thorac Cardiovasc Surg, 1272399405
  104. 104. RobertsT. S.Drez JrD.Mc CarthyW.PaineR.1991Anterior cruciate ligament reconstruction using freeze-dried, ethylene oxide-sterilized, bone-patellar tendon-bone allografts. Two year results in thirty-six patients. Am J Sports Med, 1913541
  105. 105. RosenbergD.1978Dialdehyde starch tanned bovine heterografts: Development, In: Vascular grafts, Sawyer PN, Kaplitt MJ, 261270Appleton Century-Crofts, New York.
  106. 106. RuoslahtiE.MDPierschbacher1987New perspectives in cell adhesion: RGD and integrins. Science, 2384826491497
  107. 107. Sacks MS.2003Incorporation of experimentally-derived fiber orientation into a structural constitutive model for planar collagenous tissues. J Biomech Eng, 1252280287
  108. 108. Santibáñez-SalgadoJ. A.Olmos-ZúñigaJ. R.Pérez-LópezM.Aboitiz-RiveraC.Gaxiola-GaxiolaM.Jasso-VictoriaR.Sotres-VegaA.Baltazares-LippM.Pérez-CovarrubiasD.Villalba-CalocaJ.2010Lyophilized Glutaraldehyde-Preserved Bovine Pericardium for Experimental Atrial Septal Defect Closure. Eur Cell Mater, 19158165
  109. 109. SatoH.SuzukiN.BabaT.UedaT.MawatariT.IzumiyamaO.MorishitaK.HasegawaT.2008Repair of ventricular septal perforation with asymmetrical conical patch exclusion. Ann Thorac Cardiovasc Surg, 143192195
  110. 110. Schenke-LaylandK.VasilevskiO.OpitzF.KonigK.RiemannI.HalbhuberK. J.WahlersT.StockU. A.2003Impact of decellularization of xenogeneic tissue on extracellular matrix integrity for tissue engineering of heart valves. J Struct Biol, 1433201208
  111. 111. Schmidt CE, Baier JM.2000Acellular vascular tissues: natural biomaterials for tissue repair and tissue engineering. Biomaterials, 212222152231
  112. 112. Schoen FJ, Levy RJ.1999Tissue heart valves: current challenges and future research perspectives. J Biomed Mater Res, 474439465
  113. 113. SimonP.KasimirM. T.RiederE.WeigelG.2006Tissue Engineering of heart valves-Immunologic and inflammatory challenges of the allograft scaffold. Progress in Pediatric Cardiology, 21161165
  114. 114. SimonP.KasimirM. T.SeebacherG.WeigelG.UllrichR.Salzer-MuharU.RiederE.WolnerE.2003Early failure of the tissue engineered porcine heart valve SYNERGRAFT in pediatric patients. Eur J Cardiothorac Surg, 23610021006discussion 1006].
  115. 115. Sommers KE, David TE.1997Aortic valve replacement with patch enlargement of the aortic annulus. Ann Thorac Surg, 63616081612
  116. 116. SotoA. M.JusticiaH.WrayJ. M.SonnenscheinC.1991P-nonylphenol, an estrogenic xenobiotic released from modified polystyrene. Environ Health Perspect, 92167173
  117. 117. Sung HW, Chang WH, Ma CY, Lee MH.2003Crosslinking of biological tissues using genipin and/or carbodiimide. J Biomed Mater Res A, 643427438
  118. 118. Sung HW, Hsu CS, Wang SP, Hsu HL.1997Degradation potential of biological tissues fixed with various fixatives: An in vitro study. J Biomed Mater Res, 352147155
  119. 119. Sung HW, Huang RN, Huang LLH, Tsai CC.1999In vitro evaluation of cytotoxicity of a naturally occurring crosslinking reagent for biological tissue fixation. J Biomater Sci Polymer, 106378
  120. 120. Taylor PM, Cass AEG, Yacoub MH.2006Extracellular matrix scaffolds for tissue engineering heart valves. Progress in Pediatric Cardiology, 21219225
  121. 121. METedderLiao. J.WeedB.StablerC.ZhangH.SimionescuA.SimionescuD. T.2009Stabilized Collagen Scaffolds for Heart Valve Tissue Engineering. Tissue Eng Part A, 15612571268
  122. 122. TeebkenO. E.BaderA.SteinhoffG.HaverichA.2000Tissue engineering of vascular grafts: human cell seeding of decellularised porcine matrix. Eur J Vasc Endovasc Surg, 194381386
  123. 123. MJThubrikarDeck.JDAouadJ.NolanS. P.1983Role of mechanical stress in calcification of aortic bioprosthetic valves. J Thorac Cardiovasc Surg, 861115125
  124. 124. ThyagarajanK.NguyenH.TuR.LohreJ.GuidaS.SagartzJ. W.QuijanoR. C.1992Preliminary evaluation of calcification potential of Denacolt treated small diameter biological vascular grafts. Trans Soc Biomater, 15686688
  125. 125. TimkovichR.1977Detection of the stable addition of carbodiimide to proteins. Anal Biochem, 791-2135143
  126. 126. Trantina-YatesA. E.HumanP.ZillaP.2003Detoxification on top of enhanced, diamine-extended glutaraldehyde fixation significantly reduces bioprosthetic root calcification in the sheep model. J Heart Valve Dis, 12193101discussion 100-1].
  127. 127. TsaiT. H.WestlyJ.LeeT. F.ChenC. F.1994Identification and determination of geniposide, genipin, gardenoside, and geniposidic acid from herbs by HPLC/photodiode-array detection. J Liq Chromatography, 1721992205
  128. 128. UedaY.TorrianniM. W.CoppinC. M.IwaiS.SawaY.MatsudaH.2006Antigen clearing from porcine heart valves with preservation of structural integrity. Int J Artif Organs, 298781789
  129. 129. VeselyI.BoughnerD.SongT.1988Tissue buckling as a mechanism of bioprosthetic valve failure. Ann Thorac Surg, 463302308
  130. 130. VeselyI.NoseworthyR.PringleG.1995The hybrid xenograft/autograft bioprosthetic heart valve: in vivo evaluation of tissue extraction. Ann Thorac Surg, 60No. Suppl. 2, S359S364
  131. 131. VeselyI.2005Heart Valve Tissue Engineering. Circ Res, 978743755
  132. 132. VyavahareN.HirschD.LernerE.BaskinJ. Z.SchoenF. J.BiancoR.KruthH. S.ZandR.LevyR. J.1997Prevention of bioprosthetic heart valve calcification by ethanol preincubation. Efficacy and mechanisms. Circulation, 952479488
  133. 133. VyavahareN. R.HirschD.LernerE.BaskinJ. Z.ZandR.SchoenF. J.LevyR. J.1998Prevention of calcification of glutaraldehyde-crosslinked porcine aortic cusps by ethanol preincubation: Mechanistic studies of protein structure and water-biomaterial relationships. J Biomed Mater Res, 404577585
  134. 134. WangD.JiangH.LiJ.ZhouJ. Y.HuS. S.2008Mitigated calcification of glutaraldehyde-fixed bovine pericardium by tannic acid in rats. Chin Med J (Engl), 1211716751679
  135. 135. WeiH. J.LiangH. C.LeeM. H.HuangY. C.ChangY.SungH. W.2005Construction of varying porous structures in acellular bovine pericardia as a tissue-engineering extracellular matrix. Biomaterials, 261419051913
  136. 136. Wiegner AW, Bing OH, Borg TK, Caulfield JB.1981Mechanical and structural of canine pericardium. Circ Res, 493807814
  137. 137. WoodsT.GratzerP. F.2005Effectiveness of three extraction techniques in the development of a decellularized bone-anterior cruciate ligament-bone graft. Biomaterials, 263573397349
  138. 138. YamamotoH.YamamotoF.IshibashiK.MotokawaM.2009In situ replacement with equine pericardial roll grafts for ruptured infected aneurysms of the abdominal aorta. J Vasc Surg, 49410411045
  139. 139. YoshiokaS. A.GoissisG.2008Thermal and spectrophotometric studies of new crosslinking method for collagen matrix with glutaraldehyde acetals. J Mater Sci Mater Med, 19312151223
  140. 140. ZhouJ.FritzeO.SchleicherM.WendelH. P.Schenke-LaylandK.HarasztosiC.HuS.StockU. A.2010Impact of heart valve decellularization on 3-D ultrastructure, immunogenicity and thrombogenicity. Biomaterials, 31925492554
  141. 141. ZillaP.WeissensteinC.HumanP.DowerT.vonOppell. U. O.2000High glutaraldehyde concentrations mitigate bioprosthetic root calcification in the sheep model. Ann Thorac Surg, 70620912095
  142. 142. ZiouposP.BarbenelJ. C.1994Mechanics of native bovine pericardium. I. The multiangular behavior of strength and stiffness of the tissue. Biomaterials, 155366373

Written By

Escande Rémi, Nizar Khelil, Isabelle Di Centa, Caroline Roques, Maguette Ba, Fatima Medjahed-Hamidi, Frederic Chaubet, Didier Letourneur, Emmanuel Lansac and Anne Meddahi-Pelle

Submitted: 16 December 2010 Published: 15 September 2011