Cellular biology of cryopreserved allograft valves
Tetsuya Kitagawa, Yutaka Masuda, Takashi Tominaga and Masashi Kano

Department of Cardiovascular Surgery, The University of Tokushima School of Medicine, Tokushima, Japan

Abstract: Although analyzing the precise mechanisms of cryopreserved allograft valve failure may be difficult due to a number of crucial reasons and the interrelationships between the overlapping mechanisms, there is some evidence that cryopreservation is currently the best method of storing allograft valves. The present review shows the basic cellular biology of cryopreserved allograft valves for long-term durability, particularly relevant to allograft valve cellular viability, the immune response mainly caused by viable donor cells, and the preservation and regeneration of the intrinsic extracellular matrix. The present findings are as follows. First, cryopreservation produces serious damage to cytosolic and mitochondrial functions of both endothelial cells and fibroblasts, which may cause valve failure after implantation. Second, although the collagen synthesis of cryopreserved valves was relatively maintained, total protein synthesis was highly diminished and the collagenolytic ability was activated immediately after the thawing process. These findings imply that the cryopreservation itself may cause the collagen metabolism to become degradable, which will lead to valve failure. Further examination of collagen metabolism and modulation of the collagenolytic activity will be necessary to improve the tissue preservation for improved clinical use. J. Med. Invest. 48:123-132, 2001

Keywords:cryopreservation;allograft valve;cellular viability;collagen synthesis;collagenolysis;matrix metalloproteinase

INTRODUCTION
Since the first orthotopic implantation of an allograft aortic valve more than 30 years ago (1-4), allograft aortic and pulmonary valves have been increasingly used in the surgical management of aortic valve and aortic root disease (5-8), as well as for the correction of complex congenital heart disease (9-15), and their usefulness is not deniable. Although early allograft valves were variably sterilized in ethylene oxide, preserved in β-propriolactone, irradiated, or freeze-dried, subsequent developments in preservation techniques including antibiotic use (16), improved culture media, and freezing have led to the use of cryopreserved allograft valves. The most common cryopreservation method involves freezing in dimethyl sulfoxide and storage with liquid nitrogen (16).
Although aortic valve replacement with cryopreserved allograft valves achieves excellent early results, eventual failure of these tissues is common (17-20). Despite long-standing and widespread use of cryopreserved allograft valves, the basic cellular biology of these tissues is unknown. Ongoing particular interests and controversies concern allograft valve cellular viability (21-27), the contribution of the immune response mainly caused by viable donor cells (28, 29), and the preservation and regeneration of intrinsic extracellular matrix. It is unclear how may these factors may be associated with valve durability and competence. The present study was undertaken to review the biology of cryopreserved valves relevant to long-term durability and "better" preservation techniques.

The most common cryopreservation method
Cryopreservation techniques include four processes as follows : first, sterilization ; second, freezing ; third, storage;and fourth, thawing.

1) Sterilization:Presently, the most commonly used and widely accepted method for allograft tissues is broad-spectrum antibiotics in a nutrient medium (Fig. 1).

2) Freezing : Freezing rate of -1°C per minute would be best to maximize fibroblast viability (30-32) (Fig. 2). Freezing is done kinetically in the presence of a cryoprotectant such as glycerol or dimethyl sulfoxide. As the allograft temperature approaches -20°C, most of extracellular water has frozen, and the remaining freezing medium has begun to solidify. The release of heat associated with water crystallization rapidly diminishes at this point. To maintain a consistent -1°C per minute freezing rate, the chamber must be rewarmed to temperatures just below that of the allografts. From this point, the temperature, which declines within the chamber, is directly reflected in parallel with the temperature decline of the allograft tissue.

3) Storage : A convenient storage system is the vapor phase of a liquid nitrogen freezer (-196°C).

4) Thawing : Fast warming is usually desirable and has been recommended with heart valves. The actual achievable warming rate is dependent upon the total volume of the graft and solution, the insulating qualities of the packaging, and temperature of the thawing medium. Thawing time is commonly 12-14 min ( - 15°C/min) for aortic and pulmonary valued conduits with a total combined volume of 100 ml for valve and medium.

Cellular viability of cryopreserved valves
The rarity of valve tissue degeneration of pulmonary autografts and the demonstration that these autografts can grow does suggest that autogenous valve tissue with intact endothelial and fibroblast cells has distinct advantages over allograft valve tissue (33). Several studies suggested that allograft valve viability associated with cryopreservation techniques was recognized as one of the most influencing factors on the long-term durability of cryopreserved allograft valves (34, 35).
Questions of cryopreservation techniques are particularly relevant in addressing allograft valve viability and the ability to repair leaflet wear and tear injury. It is unclear whether viable donor cells, including intrinsic cuspal interstitial tissue cells, mainly consisting of fibroblasts, and endothelial cells, are present at the time of implantation of cryopreserved allograft valves and whether they persist over the long term. In addition, it is unknown if long-term hemodynamic competence of implanted cryopreserved allograft valves requires donor cell viability and the regeneration of the intrinsic extracellular matrix.
Hillbert et al. reported that apoptosis occurred in endothelial cells and cuspal interstitial tissue cells of implanted cryopreserved allograft valves, and this might lead to their loss of cellularity (36). This apoptosis may be related to various factors, including immunologic and chemical injury, and hypoxia during valve processing and reperfusion injury at the time of implantation. Whether the apoptosis and acellularity or patchy cellularity commonly observed in clinically explanted allograft valves is due to cryopreservation techniques themselves remains to be determined (37).

1) Fibroblasts viability
If the cell viability in allograft aortic valves is crucial in determining the long-term fate of the valves, then the specific cell population of the greatest importance is likely to be the fibroblasts. These cells are by far the most abundant in aortic valves and are responsible for protein synthesis and structural integrity. Prolonged valve competence and durability of cryopreserved valves have been suggested to be derived from the viability of the donor fibroblasts, which can remodel the matrix assembly (28, 29).
VanderKamp et al. (30-32) demonstrated that a constant -1°C per minute freezing would be the best rate to maximize the fibroblast viability, and this theory was adopted and has been used as a freezing standard by the clinical tissue bank (e.g. : Cryolife, Inc., Marietta, Ga.). For heart valves, the presence of a high percentage of the fibroblasts which are capable of resynthesizing the collagenous matrix of the valve, as well as maintaining mechanical integrity, is the primary consideration (31, 38, 39).
The viability of any tissue after cryopreservation was suggested to be dependent, in part, upon handling during procurement and prefreezing storage (22-25). Any exposure to non-physiological conditions, such as ischemia, hypoxia, or anoxia causes direct toxicity to most cell types, with subsequent stresses of freezing and thawing (27). Although previous examinations of allograft aortic valves have successfully demonstrated the presence or absence of endothelial cell viability in vivo, viability of the fibroblast population has not been demonstrated except in an in vitro model (37). Unlike the endothelial cells, whose relatively rapid rate of replication allows convenient methods of assessing their viability, the fibroblast cells replicate slowly and less predictably. Consequently, the evaluation of in vivo fibroblast viability requires a method of study that is based on normal metabolic activities of the cell.
There are some experimental studies that favor persistence of donor fibroblast viability after implantation (21-25). Niwaya et al. reported that fibroblast viability of the cryopreserved human allograft valve was well preserved (>70%) with a warm ischemic time of less than 520 minutes by flow cytometry (24). Fibroblasts exist within the noncellular matrix, which is composed primarily of collagen, elastin, and proteoglycans (40). Collagen represents the largest portion of the extracellular matrix in the aortic valve. Maintenance of the normal collagen synthesis may be important for the preservation of normal tissue structure and function. If the cellular viability of the fibroblasts helps to determine the capacity of the graft to resist deterioration, this may be mediated by collagen production. Lupinetti et al. demonstrated that allografts retain a persistent capacity for procollagen synthesis of fibroblasts similar to that of the native aortic valve (41, 42). However, some clinical studies did not favor donor fibroblast viability after implantation (37). Although persistence of donor fibroblasts has been demonstrated, the histological finding of explanted cryopreserved allograft valves, including allograft valves explanted for reasons other than degeneration, showed acellularity or rare cellularity or patchy cellularity of leaflets (37). The demonstration that fibroblasts of homogenized recently cryopreserved aortic valve tissue can incorporate tritiated glycine into collagen after short-term implantation (21, 43) does not necessarily translate into the long-term postimplantation ability to repair and regenerate leaflet structure, which is implied by the term "viable".
Our previous study demonstrated that the cytosolic function of cusps, mainly consisting of fibroblasts, was comparatively preserved ; however, the mitochondrial function was still more diminished during these processes (44). These findings imply that cryopreservation causes a latent injury even in fibroblasts, which may cause valve failure after implantation. Lu et al. also reported that the mitochondrial dehydrogenase activity of the porcine valve was significantly diminished after cryopreservation processing (45).

2) Endothelial cell viability and immunogenicity
The importance of endothelial cells to the long-term fate of allograft valves is unknown. Functions of the vascular endothelium include resistance to thrombosis, maintenance of hemostasis, and modulation of vascular smooth muscle activity. Mediation of immunologic and inflammatory responses is another function of endothelium that is particularly important for allograft valves (46). The vascular endothelium constitutively expresses Class I antigen and is induced to express Class II antigens when it is exposed to an allogenic milieu (47-49). Accordingly, vascular endothelium is considered to be the most immunostimulatory component of the whole organ allografts. Whether this property of vascular endothelium is applicable to the endothelium of dynamic valves and can be markedly altered by cryopreservation remains to be determined.
First, several studies have reported on the viability and function of donor endothelial cells of cryopreserved allograft valves (6, 50-53). Yankah et al. described 70% to 80% endothelial cell viability in cryopreserved human allograft valves (54, 55). However, Lupinetti et al. demonstrated that viable endothelial cells were only observed on 21 of 131 (16%) cryopreserved allograft specimens (51).
Most previously published studies examining the cellular viability of allograft valves used specimens obtained immediately after harvest, disinfection, or thawing, but not after subsequent implantation. Thus, influences of implantation, including immunologic consequences, were not considered. Such studies may not be able to predict long-term cellular viability in implanted valves (44, 50, 51). Previous studies demonstrated that early endothelial viability was well maintained in fresh grafts, but completely abolished in cryopreserved grafts. A previous study showed a sharp divergence between the pathologic fate of endothelium and that of fibroblast cells of cryopreserved valves and emphasized the importance of analyzing the cell populations independently. Our previous study (44) revealed the cytosolic esterase activity of cryopreserved human umbilical vein endothelial cells fell to 28%±9.0% of fresh specimens, and mitochondrial dehydrogenase activity fell to 44%±6.1%. These findings suggest that cryopreservation appeared to produce serious damage to cytosolic and mitochondrial functions of endothelial cells. We presumed that the cell membrane is easily damaged soon after harvest. Mitochondria are the center of the intracellular energy source. The more the mitochondrial function is aggravated due to cryopreservation process, the more the cell membrane deteriorates due to energy depletion.
Loss of the endothelium may contribute to enhanced graft longevity by reducing the host immune response, which may contribute to valve degeneration. However, loss of the endothelium may increase the capacity for thrombus formation and adversely affect the underlying fibroblasts, thereby accelerating graft deterioration. It is unknown to what extent valve degeneration is attributed to immune responses.
To elucidate the morphology, mechanisms of deterioration, cellular viability, extracellular matrix integrity, and the role of immune responses in the dysfunction of cryopreserved allograft valves, Mitchell et al. evaluated explanted cryopreserved allograft valves and compared them with aortic valves removed from short-term and long-term transplanted hearts (18). Cryopreserved allograft valves are morphologically nonviable;their collagen is flattened but largely preserved (18). They are unlikely to grow, remodel, or exhibit active metabolic functions, and their usual degeneration cannot be attributed to immunologic responses. In contrast aortic valves of transplanted hearts maintain near-normal overall architecture and cellularity and do not show apparent immunologic injury, even in the setting of fatal myocardial parenchymal rejection or graft arteriosclerosis. Allografts implanted for more than one day showed progressive collagen hyalinization and loss of normal structural complexity and cellularity including endothelium and deep connective tissue cells. Inflammatory cells were generally minimal or absent in the allografts. Cryopreserved allograft valves have minimal viable cells, but largely retain the original collagen network ; preservation of autolysis-resistant collagenous skeleton probably provides the structural basis of function. Although, in some studies, short-course immunosuppression has been recommended to prevent early failure of allograft valves (56), in another study, the usual degeneration was not derived from immunological responses. These findings suggest that the immunogenicity of cryopreserved allograft valves can be markedly altered and disappears by cryopreservation process.
However, Mohan et al. insisted that after "better" cryopreservation, the endothelial cell function is similar to that of normal endothelium and that "a lack of viable endothelium" suggests that "preservation of the remaining valve may be the suspect" (22). Cryopreservation must aim to preserve a viable endothelial lining, because its loss could be the main reason behind the accelerated thrombosis, calcification, and infection of commercial bioprosthetic valves. Unfortunately, there is currently no clinical information to support this position. Using the occurrence of "accelerated thrombosis, calcification, and infection of commercial bioprosthetic valves", that is without endothelial lining as justification for preserving the endothelium of allografts is not sufficient.
It cannot yet be determined whether rare endothelial cells or a viable endothelial lining has a positive, negative, or neutral effect on the long-term structure and function of cryopreserved allograft aortic and pulmonary valves, respectively.

Collagen synthesis and collagenolysis
For the long-term performance of cryopreserved allograft valves, a more important factor may be preserving the intactness of the original collagen network (36) that provides the structural basis of the valve matrix (Fig. 3). Despite the widespread use of cryopreserved valves, the balance of collagen synthesis and collagenolysis ability of the valves has not yet been clearly revealed (57-59). Controlled collagenolysis ability was suggested to play an important role in the first step of the regeneration process (18), and consequent collagen synthesis is a second step.
Lupinetti et al. (41, 42) demonstrated the presence of procollagen and the α1-(I) procollagen gene expression in allograft aortic valves after implantation for 3 days, which suggested donor fibroblast viability. Fibroblasts have been known to exist within the matrix (40) and to participate in the valve remodeling through collagen metabolism. In our previous study, the collagen content in the cryopreserved cusps was kept at the same degree as that of the fresh cusps, and the collagen synthesis ability in the cryopreserved cusps was also relatively preserved (43). However, the protein synthesis ability, including collagen synthesis, in the cryopreserved cusps decreased significantly (43). Although the collagen synthesis ability was preserved, like the findings of Lupinetti et al. (41, 42), such a marked reduction in protein synthesis ability may have had a significant impact on the long-term cellular viability.
The matrix metalloproteinases (MMPs) are a family of zinc-dependent enzymes that are able to digest a wide range of extracellular matrix proteins (60-62). The expression of the MMPs from the endothelial cells (63, 64), the fibroblast cells (65, 66), and ischemic myocardium (67, 68) was consecutively revealed (63, 64). MMP-1, an interstitial collagenase, is mainly produced from fibroblasts, and is known to degrade structural type I collagen in the matrix. MMP-2 and MMP-9 are mainly produced from the endothelium, and are known to be involved in the degradation of type IV collagen, which is a major component of the subendothelial basement membrane (Fig. 4).
Our previous study demonstrated that the cytosolic and mitochondrial function of endothelial cells were seriously damaged during the cryopreservation and thawing processes (44), and those processes may cause a latent cytosolic and mitochondrial injury even in the fibroblasts. Despite the anticipated reduced cellular viability, particularly endothelial cells, in the cryopreserved cusps, the activity of the MMP-1 and MMP-2 in the supernatant of the cryopreserved cusps was observed at the quantitatively same degree as those of the fresh cusps (43). These findings imply that the cryopreservation and thawing process causes MMP-1 and MMP-2 release from endothelial and fibroblast cells of allograft cusps, and activates them before the implantation (Fig. 5). There is a possibility that the activated MMP-2 will degrade the basement membrane consisting of type IV collagen and activated MMP-1 will destruct the cusp matrix. Most cryopreserved valves may have the potential to degrade and autolyze the matrix extensively during processing, intraoperative preparation, and possibly after implantation.
From these apparently discrepant results, it is suggested that the collagen synthesis ability is preserved even in the slightly injured fibroblast cells of cryopreserved valves, however, the protein synthesis ability might be extremely reduced because of limited endothelial collaboration (43, 69, 70) and the decrease in the protein production relevant to the intracellular organellar function (44), which maintains the cell viability, and that the collagenolytic activation occurs just before implantation (43). With respect to those findings, the injured fibroblasts are going to be non-viable, collagen synthesis is suggested to deteriorate, and the original collagenous skeleton degrades in the near future. The collagenolysis dominant state was suspected in the cryopreserved valves.
Although it is necessary to elucidate the dynamics of tissue inhibitors of MMPs in the cryopreserved valves, the control of the MMPs activity in the cryopreserved valves may become more effective to obtain the long-term durability rather than the preservation of collagen synthesis ability. Recently, there were some reports that doxycycline, a derivative of tetracycline, suppresses the development of the elastase induced abdominal aortic aneurysms (71, 72), through the direct MMPs-inhibiting activities. Other MMPs inhibitors, marimastat (BB-2516), batimastat (BB-94) and BE16627B, were also developed as agents for inhibiting cancer metastasis (73-75). These agents might improve the allograft preservation in clinical use.
In conclusion, the findings of this review suggest the possibility that the current cryopreservation techniques and management, themselves, may lead a cryopreserved valve to become non-viable acellular tissue by the collagenolytic activation and degradation of protein synthesis, which will cause the destruction of the valve matrix. Further examination of collagen metabolism and modulation of the collagenolytic activity will be necessary to improve the allograft preservation for better clinical use. We believe that the enthusiasm for allograft tissue implantation should be considered in the overall context of allograft failure.

REFERENCES
1. Ross DN : Homograft replacement of the aortic valve. Lancet 2:487, 1962
2. Ross DN : Replacement of the aortic and mitral valves with a pulmonary autograft. Lancet 2:956-958, 1967
3. Eguchi S, Asano K:Homograft of pulmonary artery or ascending aorta with valve as a right ventricular outflow. J Thorac Cardiovasc Surg56:413-420, 1968
4. Barratt-Boyes BG, Roche AH, Whitoloch RM :Six year review of the results of freehand aortic valve replacement using an antibiotic sterilized homograft valve. Circulation 55 : 353-361, 1977
5. Okita Y, Franciosi G, Matsuki O, Robles A, Ross DN : Early and late results of aortic root replacement with antibiotic-sterilized aortic homograft. J Thorac Cardiovasc Surg 95:696-704, 1988
6. Haydock D, Barratt-Boyes B, Macedo T, Kirklin JW, Blackstone E : Aortic valve replacement for active infectious endocarditis in 108 patients:a comparison of freehand allograft valves with mechanical prostheses and bioprostheses. J Thorac Cardiovasc Surg 103:130-139, 1992
7. Chambers JC, Somerville J, Stone S, Ross DN :Pulmonary autograft procedure for aortic valve disease : long-term results of the pioneer series. Circulation 96:2206-2214, 1997
8. Niwaya K, Knott-Craig CJ, Santangelo K, Lane MM, Chandrasekaran K, Elkins RC : Advantage of autograft and homograft valve replacement for complex aortic valve endocarditis. Ann Thorac Surg 67:1603-1608, 1999
9. Gerosa G, McKay R, Davies J, Ross DN : Comparison of the aortic homograft and the pulmonary autograft for aortic valve or root replacement in children. J Thorac Cardiovasc Surg 102:51-61, 1991
10. Michler RE, Chen JM, Quaegebeur JM : Novel technique for extending the use of allografts in cardiac operations. Ann Thorac Surg 57 : 83-87, 1994
11. Stark J:The use of valved conduits in pediatric cardiac surgery. Pediatr Cardiol 19 : 282-288, 1998
12. Perron J, Moran AM, Gauvreau K, del Nido PJ, Mayer Jr JE, Jonas RA : Valved homograft conduit repair of the right heart in early infancy. Ann Thorac Surg 68:542-548, 1999
13. Dittrich S, Alexi-Meskishvili VV, Yankah AC, Dahnert I, Meyer R, Hetzer R, Lange PE : Comparison of porcine xenografts and homografts for pulmonary valve replacement in children. Ann Thorac Surg 70:717-722, 2000
14. Yoshikawa Y, Kitamura S, Taniguchi S, Kameda Y, Niwaya K, Sakaguchi H : Pulmonary ventricular outflow reconstruction with a size-reduced cryopreserved pulmonary valve allograft : mid-term follow up. Jpn Circ J 64:23-26, 2000
15. Matsuki O, Okita Y, Almeida RS, McGoldrick JP, Hooper TL, Robles A, Ross DN:Two decades' experience with aortic valve replacement with pulmonary autograft. J Thorac Cardiovasc Surg 95:705-711, 1988
16. Lange PL, Hopkins RA : Cardiac reconstruction with allograft valves. Springer-Verlag, New York, 1989, pp 46-53
17. Salim KA, DiSessa TG, Alpert BS, Arheart KL, Novick WM, Watson DC : The fate of homograft conduits in children with congenital heart disease : an angiographic study. Ann Thorac Surg 59:67-73, 1995
18. Mitchell RN, Jonas RA, Schoen FJ. Pathology of explanted cryopreserved allograft heart valves :comparison with aortic valves from orthotopic heart transplants. J Thorac Cardiovasc Surg 115 :118-127, 1998
19. LeBlanc JG, Russell JL, Sett SS, Potts JE : Intermediate follow-up right ventricular outflow tract reconstruction with allograft conduits. Ann Thorac Surg 66:S174-178, 1998
20. Stark J, Bull C, Stajevic M, Jothi M, Elliott M, de Leval M:Fate of subpulmonary homograft conduits:determinants of late homograft failure. J Thorac Cardiovasc Surg 115:506-516, 1998
21. Al-Janabi N, Gonzalez-Lavin L, Neirotti R, Ross DN : Viability of fresh aortic valve homografts :a quantitative assessment. Thorax 27 : 83-86, 1972
22. Brockbank KGM : Cell viability in fresh, refrigerated, and cryopreserved human heart valve leaflets. Ann Thorac Surg 49:848-849, 1990
23. Lang SJ, Giordano MS, Carlon-Cardo C, Summers BD, Statiano-Coico L, Hajjar DP:Biochemcal and cellular characterization of cardiac valve tissue after cryopreservation or antibiotic preservation. J Thorac Cardiovasc Surg 108:63-67, 1994
24. Niwaya K, Sakaguchi H, Kawachi K and Kitamura S : Effect of warm ischemia and cryopreservation on cell viability of human allograft valves. Ann Thorac Surg 60:S114-117, 1995
25. Gall KL, Smith SE, Willmette CA, O'Brien MF : Allograft heart valve viability and valve-processing variables. Ann Thorac Surg 65 : 1032-1038, 1998
26. O'Brien MF, Johnston N, Stafford G, Gardner M, Pohlner P : A study of the cells in the explanted viable cryopreserved allograft valve. J Card Surg 3:279-287, 1988
27. Wassenaar C, Wijsmuller EG, Van Herwerden LA, Aghai Z, Van Tricht CLJ, Bos E : Cracks in cryopreserved aortic allografts and rapid thawing. Ann Thorac Surg 60:S165-167, 1995
28. Arminger LC : Postimplantation leaflet cellularity of valve allografts : are donor cells beneficial or detrimental? Ann Thorac Surg 66:S233-235, 1998
29. Koolbergen DR, Hazekamp MG, Kurvers M, de Heer E, Cornelisse CJ, Huysmans HA, Bruijn JA : Tissue chimerism in human cryopreserved homograft valve explants demonstrated demonstrated by hybridization. Ann Thorac Surg 66:S225-232, 1998
30. Van Der Kamp AWM, Nauta J : Fibroblast function and the maintenance of the aortic-valve matrix. Cardiovasc Res 13:167-172, 1979
31. Van Der Kamp AWM, Visser WJ, Dongen JM, Nauta J, Galjaard H : Preservation of aortic heart valves with maintenance of cell viability. J Surg Res 30:47-56, 1981
32. Mochtar B, Van Der Kamp AWM, Jongh EJ, Nauta J : Cell survival in canine aortic heart valves stored in nutrient medium. Cardiovasc Res 18 :497-501, 1984
33. Heacox AE, McNally RT, Brockbank KGM:Factors affecting the viability of cryopreserved allograft heart valves. In : Yankah AC, Hetzer R, Miller DC, Ross DN, Sommerville J, Yacoub MH, eds. Cardiac valve allografts 1962-1987. Springer-Verlag, New York, 1988, pp 37-41
34. O'Brien MF, Stafford EG, Gardner MA, Pohlner PG, McGiffin DC : A Comparison of aortic valve replacement with viable cryopreserved and fresh allograft valves, with a note on chromosomal studies. J Thorac Cardiovasc Surg 94 : 812-823, 1987
35. Angell WW, Oury JH, Lamberti JJ, Koziol J:Durability of the viable aortic allograft. J Thorac Cardiovasc Surg 98:48-55, 1989
36. Hilbert SL, Luna RE, Zhang J, Wang Y, Hopkins RA, Yu Z, Ferrans VJ:Allograft heart valves:the role of apoptosis-mediated cell loss. J Thorac Cardiovasc Surg 117:454-462, 1999
37. Mitchell RN, Jonas RA, Schoen FJ : Structure-function correlations in cryopreserved allograft cardiac valves. Ann Thorac Surg 60:S108-113, 1995
38. Brockbank KGM, Carpenter JF, Dawson P : Effects of storage temperature on viable bioprosthetic heart valves. Cryobiology 29:537-542, 1992
39. Hu JF, Hopkins R, Wolfinbarger I:Effects of antibiotics on cellular viability in porcine heart valve tissue. Cardiovasc Res 23 : 960-964, 1989
40. Broom ND:The observation of collagen and elastin structures in wet whole mounts of pulmonary and aortic leaflets. J Thorac Cardiovasc Surg 75:121-130, 1978
41. Lupinetti FM, Kneebone JM, Rekhter MD, Brockbank KGM, Gordon D : Procollagen production in fresh and cryopreserved aortic valve grafts. J Thorac Cardiovasc Surg 113 : 102-107, 1997
42. Song YC, Yao LY, Kneebone JM, Lupinetti FM :Effect of cryopreservation and histocompatibility on type I procollagen gene expression in aortic valve grafts. J Thorac Cardiovasc Surg 114 : 421-427, 1997
43. Kano M, Masuda Y, Hori T, Yoshizumi M, Kitaichi T, Wakisaka Y, Tominaga T, Yasuta O, Kitagawa T : Collagen synthesis and collagenase activity of cryopreserved heart valve. J Thorac Cardiovasc Surg:2001 in press
44. Tominaga T, Kitagawa T, Masuda Y, Hori T, Kano M, Yasuta O, Katoh I : Viability of cryopreserved semilunar valves. an evaluation of cytosolic and mitochondrial activities. Ann Thorac Surg 70:792-757, 2000
45. Lu JH, Chiu YT, Sung HW, Hwang B, Chong CK, Chen SP, Mao SJ, Yang PZ, Chang Y:XTT-colorimetric assay as a marker viability in cryoprocessed cardiac valve. J Mol Cell Cardiol 29:1189-1194, 1997
46. Rocca FD, Sartore S, Guidolin D, Bertiplaglia B, Gerosa G, Casarotto D, Pauletto P:Cell composition of the human pulmonary valve : a comparative study with the aortic valve-the VESALIO project. Ann Thorac Surg 70 : 1594-1600, 2000
47. Cochran RP, Kunzelman KS : Cryopreservation does not alter antigenic expression of aortic allografts. J Surg Res 46:597-599, 1989
48. Lupinetti FM, Christy JP, King DM, Khatib HE, Thompson SA:Immunogenicity, antigenicity, and endothelial viability of aortic valves preserved 4°C in a nutrient medium. J Cardiac Surg 6:454-461 1991
49. Quigley RL, Switzer DSS, Victor TA, Goldschmidt RA, Salinger MH, Arentzen CE, Alexander JC, Anderson RW:Modulation of alloreactivity in transplant recipients by phenotypic manipulation of donor endothelium. J Thorac Cardiovasc Surg 109:905-909, 1995
50. Killinger Jr WA, Dorofi DB, Tinsley Jr EA, Keagy BA, Johnson G : Flow cytometric analysis of organ preservation-induced endothelial cell membrane damage. Ann Thorac Surg 53:472-476, 1992
51. Lupinetti FM, Tsai TT, Kneebone JM, Bove EL : Effect of cryopreservation on the presence of endothelial cells on human valve allografts. J Thorac Cardiovasc Surg 106:912-917, 1993
52. Mohan R, Feng XJ, Walter P, Herman A:Cryopreserved heart valve allografts can have a normal endothelium. J Thorac Cardiovasc Surg 106:912-917, 1993
53. Pompilio G, Polvani GL, Rossoni G, Porqueddu M, Berti F, Barajon I, Petruccioli MG, Guarion A, Aguggini G, Biglioli P, Sala A:Effects of warm ishcemia on valve endothelium. Ann Thorac Surg 63:656-662, 1997
54. Yankah AC, Randzio G, Wottge HU, Bernhard A : Factors influencing endothelial-cell viability during procurement and preservation of valve allografts. In : Thiede A, Doltz E, Engemann R, Hamelmann H, eds. Microsurgical models in rats for transplantation research. Springer-Verlag Heidelberg, 1985, pp107-111
55. Yankah AC, Dreyer W, Wottge HU, Muller-Rucholtz W, Bernhard A : Kinetics of endothelial cells preserved aortic valve allografts used for heterotopic transplantation in inbred rat strains. In : Bodnar E, Yacoub MH, eds. Biologic and bioprosthetic valves. Yorke Medical Books, New York, 1986, pp 73-84
56. Yankah AC, Wottge HU, Muller-Ruchholtz W. Short-course cyclosporin A therapy for definite allograft valves survival immunosuppression in allograft valve operations. Ann Thorac Surg 60 :S146-150, 1995
57. Lester WM, Damji AA, Tanaka M, and Gedeon I:Bovine mitral valve organ culture:role of interstitial cells in repair of valvular injury. J Mol Cell Cardiol 24:43-53, 1992
58. Mcgregor CGA, Bradley JF, Mcgee JO, Wheatley DJ:Tissue culture, protein and collagen synthesis in antibiotic sterilized canine heart valves. Cardiovascular Research 10:389-393, 1976
59. Henney AM, Parker DJ, and Davies MJ:Collagen biosynthesis in normal and abnormal human heart valves. Cardiovascular Research 16 :624-630, 1982
60. Simionescu A, Simionescu D, Deac R : Biochemical pathways of tissue degeneration in bioprosthetic cardiac valves. ASAIO Journal 43 : M561, 1996
61. Woessner JF Jr : Matrix metalloproteinases and their inhibitors in connective tissue remodeling. FASEB J 5:2145-2154, 1991
62. Gogly B, Groult N, Hornebeck W, Godeau G, Pellat B:Collagen zymography as a sensitive and specific technique for the determination of subpicogram levels of interstitial collagenase. Anal Biochem 255:211-216, 1998
63. Cagliero E, Roth T, Roy S, Maiello M, Lorenzi M : Expression of genes related to the extracellular matrix in human endothelial cells:differential modulation by elevated glucose concentrations, phorbol esters, and cAMP. J Biol Chem 266:14244-14250, 1991
64. Belkhiri A, Richards C, Whaley M, McQueen SA, Orr FW : Increased expression of activated matrix metalloproteinase-2 by human endothelial cells after sublethal H2O2 exposure. Lab Invest 77:533-539, 1997
65. Bendeck MP, Zempo N, Clowes AW, Galardy RE, Reidy MA:Smooth muscle cell migration and matrix metalloproteinase expression after arterial injury in the rat. Circ Res. 75 : 539-545, 1994
66. Tyagi SC, Kumar S, Katwa L : Differential regulation of extracellular matrix metalloproteinase and tissue inhibitor by heparin and cholesterol in fibroblast cells. J Mol Cell Cardiol 29:391-404, 1997
67. Danielsen CC, Wiggers H, Andersen HR:Increased amounts of collagenase and gelatinase in porcine myocardium following ischemia and reperfusion. J Moll Cell Cardiol 30 : 1431-1442, 1998
68. Cleutjens JP, Kandala JC, Guarda E, Guntaka RV, Weber KT:Regulation of collagen degradation in the rat myocardium after infarction. J Mol Cell Cardiol. 27:1281-1292, 1995
69. Guarda E, Myers PR, Brilla CG, Tyagi SC, Weber KT : Endothelial cell induced modulation of cardiac fibroblast collagen metabolism. Cardiovasc Res 27:1004-1008, 1993
70. Villanueva AG, Farber HW, Rounds S, Goldstein RH : Stimulation of fibroblast collagen and total protein formation by an endothelial cell-derived factor. Circ Res 69:134-141, 1991
71. Curci JA, Petrinec D, Liao S, Golub LM, Thompson RW : Pharmacologic suppression of experimental abdominal aortic aneurysms : a comparison of doxycycline and four chemically modified tetracycline. J Vasc Surg 28:1082-1093, 1998
72. Boyle JR, McDermott E, Growther M, Wills AD, Bell PRF, Thompson MM : Doxycycline inhibits elastin degradation and reduces metalloproteinase activity in a model of aneurysmal disease. J Vasc Surg 27:354-361, 1998
73. Porter KE, Loftus IM, Peterson M, Bell PR, London NJ, Thompson MM : Marimastat inhibits neoinitimal thickening in a model of human vein graft stenosis. Br J of Surg 85 : 1373-1377, 1998
74. De Smet BJ, de Kleijn D, Hanemaaijer R, Verheijen JH, Robertus L, van Der Helm YJ, Post MJ. Metalloproteinase inhibition reduces constrictive arterial remodeling after balloon angioplasty :a study in the atherosclerotic Yucatan micropig. Circulation 101:2962-2967, 2000
75. Bigatel DA, Elmore JR, Carey DJ, Cizmeci-Smith G, Franklin DP, Youkey JR : The matrix metalloproteinase inhibitor BB-94 limits expansion of experimental abdominal aortic aneurysms. J Vasc Surg 29:130-139, 1999

Received for publication May 18, 2001;accepted June 20, 2001.

Address correspondence and reprint requests to Tetsuya Kitagawa, M.D., PhD., Department of Cardiovascular Surgery, The University of Tokushima School of Medicine, Kuramoto-cho, Tokushima770-8503, Japan and Fax:+81-88-633-7152.