Top Banner

of 19

The Roles of Tissue Engineering and Vascularisation in the Development of Micro-Vascular Networks- A Review

Oct 12, 2015

Download

Documents

Adela Cezara

tissue engineering
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
  • pital Hampstead NHS Trust, London NW3 2QG, UK

    Received 1 April 2004; accepted 5 July 2004

    Available online 20 August 2004

    Keywords: Micro-vascular; Tissue engineering; Capillary beds; Tissue vascularisation

    3.1.2. Conguration of vessels . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1863

    ARTICLE IN PRESS3.2. Extracellular matrix scaffolds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1863

    3.2.1. Biomaterial substitutes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1864

    3.2.2. Superstructure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1867

    Corresponding author. Tel.: +44-20-7830-2901; fax: +44-20-7472-6444.E-mail address: [email protected] (A.M. Seifalian).0142-9612/$ - se

    doi:10.1016/j.biContents

    1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1858

    1.1. Reconstructive surgery . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1858

    1.2. Tissue engineering and its limited success . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1858

    1.3. Microvessel development. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1858

    1.4. Tissue perfusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1859

    2. Search methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1860

    3. Tissue-engineered microvessel beds . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1860

    3.1. Micro-vascular tissue engineering. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1860

    3.1.1. Types of prostheses . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1861Abstract

    The construction of tissue-engineered devices for medical applications is now possible in vitro using cell culture and bioreactors.

    Although methods of incorporating them back into the host are available, current constructs depend purely on diffusion which

    limits their potential. The absence of a vascular network capable of distributing oxygen and other nutrients within the tissue-

    engineered device is a major limiting factor in creating vascularised articial tissues. Though bio-hybrid prostheses such as vascular

    bypass grafts and skin substitutes have already been developed and are being used clinically, the absence of a capillary bed linking

    the two systems remains the missing link. In this review, the different approaches currently being or that have been applied to

    vascularise tissues are identied and discussed.

    r 2004 Elsevier Ltd. All rights reserved.bDepartment of Plastic Surgery, Royal Free HosBiomaterials 26 (2005) 18571875

    Review

    The roles of tissue engineering and vascularisation in the developmentof micro-vascular networks: a review

    Ruben Y. Kannana, Henryk J. Salacinskia, Kevin Salesa, Peter Butlerb,Alexander M. Seifaliana,

    aBiomaterials & Tissue Engineering Centre (BTEC), University Department of Surgery, Royal Free and University College Medical School,

    University College London, Rowland Hill Street, London NW3 2PF, UK

    www.elsevier.com/locate/biomaterialse front matter r 2004 Elsevier Ltd. All rights reserved.

    omaterials.2004.07.006

  • . . . .

    . . . .

    . . . .

    . . . .

    4. Conclusion . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1870

    . . . .

    . . . .

    . . . .

    more case-specic [4]. More recently, the introduction of was seeded with cultured keratinocytes. The CSR was

    INateriperforator aps has improved the utility of aps as areconstructive option [5]. Nevertheless, free tissuetransfer confers morbidity to the patient as one functionis sacriced for another [6]. Therefore, alternativesources of tissue for reconstruction are needed.

    1.2. Tissue engineering and its limited success

    More than 21 million patients per year in the USalone rely on biomedical implants [7]. These include

    used in 12 pediatric burn wounds and by 14 days45.7714.2% had vascularised as compared to 9871%in split skin grafts [15]. However, no clinical studies onscaffolds with an inherent vascular network or incorpo-rated angiogenic factors have been done.

    1.3. Microvessel development

    Human microvasculature begins with arteries dividingconsecutively into smaller branches like meta-arterioles4.1. Future perspectives. . . . . . . . . . . . . . . . . . . . . . .

    References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

    Further-reading . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

    1. Introduction

    1.1. Reconstructive surgery

    In current practice, reconstructive surgery is essentialin order to achieve wound closure following majorcancer resections and trauma. Traditionally, reconstruc-tive surgeons have employed the reconstructive ladderconcept; a hierarchical system by which the simplestoption, usually skin grafting or primary closure, ischosen rst. If this fails, then a slightly more complexprocedure is selected. With the increasing demand forquality wound closure, maximal aesthetics with mini-mum morbidity, the concept of the reconstructiveelevator [1] is applicable. As opposed to the reconstruc-tive ladder, the best option, which in most cases is a ap,is chosen initially.A ap may be dened as a segment of tissue with an

    independent blood supply. These may be classied intolocal, regional and free aps. Free aps are based on theconcept of angiosome [2]; an area of tissue with aninherent vascular network supplied by a single vascularpedicle. Buncke and McLean performed the rst micro-vascular tissue transplant in 1969 [3]. Since then, freeaps have revolutionised reconstructive surgery as theyare more versatile and reliable than other aps. Flapsare now being pre-fabricated prior to surgery to become3.2.3. Cells seeded onto matrices . . . . . . . . . . .

    3.3. Vascularisation . . . . . . . . . . . . . . . . . . . . . . . . .

    3.3.1. Inosculation. . . . . . . . . . . . . . . . . . . . . .

    3.3.2. Angioinduction . . . . . . . . . . . . . . . . . . .

    ARTICLER.Y. Kannan et al. / Biom1858bypass grafts, dental implants and articial dermalsubstitutes like Integras. Current technology limits thesurvival of these implants as they depend initially upondiffusion and in the later stages on neovascularisation.Overdependence on the former limits the thickness of. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1870

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1870

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1875

    these implants, while depending on neovascularisationcan cause excessive brovascular ingrowth and hencescarring. There are no tissue-engineered constructspresently available which have an inherent vasculaturesuch as a capillary bed ready to be connected to the hostvascular system. This represents a major limitation asthe driving force of living tissue is vascularity. Althoughattempts to develop small-diameter vascular grafts havebeen successful in vitro [8,9], it remains to be seenthrough clinical studies whether they are reliable enoughto sustain ow in vivo [1012]. Even so, they are limitedby the absence of a viable capillary network for nutrientexchange. This missing link represents the bridgebetween the host and the tissue-engineered implant.Most clinical studies in articial tissue so far have

    utilised dermal substitutes like Integras in the treatmentof burns. After application, it undergoes imbibition,broblast migration, neovascularisation and nallyvessel maturation. This process takes months, afterwhich skin grafts or cultured keratinocytes may beapplied. One centre used it in 30 patients with goodresults [13], while others have successfully seededcultured Fb and keratinocytes onto it prior to clinicalapplication. They found superior results when it wasdone with concurrent skin grafting [14]. Sheridan andco-workers developed a composite skin replacement(CSR) composed of acellular allogeneic dermis, which. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1867

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1868

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1868

    . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1869

    PRESSals 26 (2005) 18571875(80100 mm) until nally forming capillaries (1015 mm).These vessels serve to redistribute blood and its nutrientswhilst lowering the pressure head. This allows blood toperfuse the tissue, allowing more efcient exchange ofmetabolites. Eventually, the capillaries unite into

  • post-capillary venules (PCVs), venules and nally veinsfor the return of waste products. The venular componentof vasculature also serves as capacitance vessels [16].The vascular tree is formed during the early gestation.

    Angiogenic cells form clusters which coalesce to formsolid tubes, which eventually canalise to form bloodvessels [17]. The outer ring consists of angioblasts whichform the vessel walls. The subsequent differentiation ofthese precursor angioblasts into endothelial cells (EC)and the denovo formation of a vascular network aretermed vasculogenesis. These vessels are capillary-liketo begin with and eventually differentiate into eitherarteries or veins [18]. The creation of this frameworkdepends on guidance molecules within the matrix suchas ephrins at the arterio-venous interface [19] andreversion-inducing cysteine-rich (RECK) protein forparacellular proteolysis [20].The adult vascular network remodels itself by

    arteriogenesis with the opening up and then thesubsequent enlargement of existing collaterals (so calledcollateral enlargement), as well as the formation of

    Remodelling is mediated by monocytes and endothe-lial progenitor cells (EPCs) as shown in Fig. 1 and is a

    ability of the membrane (P) and surface area (S) as

    ARTICLE IN PRESSR.Y. Kannan et al. / Biomaterials 26 (2005) 18571875 1859completely new vessels from the already existing vessels(so-called arterialisation) [21]. Micro-vascular remodel-ling is a mechanistic process delineated to specic tissuetype and specic stimuli. Therefore, with the exceptionsof skeletal muscle responding to exercise and the femalemenstrual cycle itself, micro-vascular remodelling islimited solely to pathological situations, in particularinammation, wound healing, ischemia, and that ofhypoxia, together with a few other rare circumstances.Fig. 1. Series of events illustrating the dual nature of vessel

    development: angiogenesis and arteriogenesis. Keys: EPC, endothelial

    progenitor cells; BM, basement membrane; SM, smooth muscle cells.represented below:

    D PSC:P in turn, depends on effective pore area (A) and porelength (L). Capillary exchange is a mixture of diffusion-and ow-limited exchange, with the latter beingimportant for the transfer of water-soluble moleculesand the former for gas transport.Plasma is a two-phase uid consisting of solvent and

    solute phases. While solvents pass unimpeded into theextracellular matrix (ECM), solutes are transported bymeans of convection (bulk transport) and diffusion [28],represented by the formula

    F QD;mechanism that is dependent on changes in shear stress.Studies have shown that turbulent ow at low ow ratesof 1.5 dynes/cm2 itself activates ECs while a normallaminar shear stress of 815 dynes/cm2 does not [22]. Ina rabbit femoral artery occlusion model, collateralrecruitment occurred within a week, with removal ofunwanted vessels by 3 weeks [23]. Angiogenesis is thecoordinated migration and proliferation of EC andpericytes from the existing vascular bed [24] and theirsubsequent maturation and stabilisation by envelopingsmooth muscle cells (SMCs) [25]. Stimulated byhypoxia, these vessels proliferate by either capillarysprouting [21] or intussusception [26], particularly intovenules. Vasculogenesis occurs from migrating dediffer-entiated EPCs which form tubules [27].

    1.4. Tissue perfusion

    Microvessels divide into numerous smaller branchesover a given volume of tissue, thus maximising theavailable area for nutrient exchange. In this microenvir-onment, stasis of ow is prevented by the Fahraeus-Lindqvuist effect, repulsive charges between blood cellsand vessel wall, as well as the thin glycocalyx lm on theendothelial layer (Fig. 2) [16].As blood slows down in the meta-arterioles, intra-

    vascular transit time increases. As this happens, nutrientexchange shifts from ow-limited to diffusion-limitedexchange within the capillaries (Fig. 3). Diffusion (D) isdependent on the concentration gradient (C), perme-Fig. 2. Factors preventing thrombogenicity in low-ow states.

  • 3. Tissue-engineered microvessel beds

    3.1. Micro-vascular tissue engineering

    Microvessel beds provide the vascular infrastructurefor living tissue [6]. The development of independentlyvascularised articial tissue requires a source of vascu-larisation (intrinsic existing blood vessels) and a matrixthrough which exuding nutrients can perfuse cells [30].The matrix functions as a reservoir of growth factors toinduce incoming blood vessel (angioinduction) as well asa scaffold to seed cells like EPCs, which participate inthe formation of newer blood vessels by the process ofarteriogenesis (Fig. 4). Microvessel networks, articialmatrices and neovascularisation constitute the triad oftissue vascularisation (Fig. 5).A successful micro-vascular system depends on the

    graft material and how it is arranged. The materialwould have to be non-thrombogenic and must havesimilar compliance to native vessels to avoid intimal

    IN PRESSaterials 26 (2005) 18571875where F is solute ow and Q is convective ow. Soluteefux is dependent on the rate of solvent transfer withinthe medium. Solutes like glucose are transported viaconvection with water (solvent), whilst gaseous ex-change is based on Kroghs diffusion model.This is augmented by the 5mmHg negative pressure

    as a result of ECM hydraulic conductivity, the ability oftissue to conduct water across it. Hydraulic conductivity(H) of ECM is proportional to the hydraulic radius ofthe gel (the ratio of its porosity, P, to the net surfacearea of the glycosaminoglycans/GAGs):

    Har=S:

    Fig. 3. Flow- and diffusion-limited exchange.ARTICLER.Y. Kannan et al. / Biom1860The interstitial conductivity of ECM is proportional toits hydration and is usually in the range of10101013 cm3/s/dyne. This force is balanced by theintrinsic hydraulic resistivity of the ECs lining themicrovessels. Studies have shown this to be in thevicinity of 108 cm3/s/dyne [29]. Nutrient exchangedepends on the balance between hydrostatic, osmoticand interstitial pressures [16].

    2. Search methods

    All the studies were identied by following databases:Pubmed; Ovid online; Infotrieve; Proquest; Sciencedirect; Isi web of science; Biomed central; Ingenitaselect; Elsevier texts and Blackwell-synergy searchesbetween years 1966 and 2003, with the followingkeywords: Tissue-engineered blood vessels; Vascularisa-tion of acellular tissue; Vascularisation of tissue;Angiogenesis; Vascular tissue engineering; Microvascu-lature; Tissue-engineered aps; Pre-fabricated aps; Softtissue engineering; Polymers in vascular tissue engineer-ing; Capillary beds; Microvasculature; Polymers intissue engineering; Collage; Dacron; PTFE; Alginate.hyperplasia [31] at its arterial end and must besufciently porous to allow nutrient exchange at thecapillary level. An articial capillary network wouldinclude small arteries (12mm) conducting blood intoan arteriolar network (1001000 mm), which wouldeventually end in capillary-like vessels of 1015 mm.These end capillaries would need to be within150200 mm of every target cell [30] before converginginto a venous collecting system. Therefore, botharteriolar and venular components of the scaffold wouldhave to be in close proximity.Fig. 4. Inter-relationships between various methods of vessel devel-

    opment. Keys: EC, endothelial cell; EPC, endothelial progenitor cells.

  • 3.1.1. Types of prostheses

    The following section has been subdivided intomacro- and micro-vascular grafts, the reason being thatmicro-vessels are being developed from previous tech-

    nology as used in the development of larger macro-vessels, and only rarely are completely new material-based approaches used to date. This discussion isnecessary to deal with the issues of potential overlapsand discrepancies during the transition between thehigh-ow arterial system and the lower-ow capillarysystem and how, in light of these, tissue engineering ofmicro-vessels differs. The various graft options aresummarised in Table 1.

    3.1.1.1. Macrovascular grafts. The current resultswith PTFE and Dacront are clinically acceptable inperipheral bypass grafts [32], where as patency issignicantly lower in grafts 6mm or smaller [3335].The primary problem at lower ow rates is thrombo-genicity, especially with Dacron-based grafts, and theincreased susceptibility of these vessels to intimalhyperplasia. Luminal modication of these grafts usingirradiation, presealing and antiplatelet/anticoagulant/growth factor incorporation [30,47,48] to reducethrombogenicity had been employed [46], but endothe-lialisation is signicantly superior [49,50]. Endothelia-lisation of vascular prostheses is dependent on thebiomaterial used and pore morphology. Matsuda andcolleagues reported that pores between 18 and 50 mm in

    ARTICLE IN PRESS

    Table 1

    Current status of macro- and microvascular grafts

    Authors Graft type Subjects ID (mm)

    Harris et al. (2002) [37] PTFE Rats o1

    Demiri et al. (1999) [36] PTFE Rats 1

    Seifalian et al. (2003) [41] CPU Dogs 5

    Shinoka et al. (1998) [176] Polyglactin-

    PGA

    Sheep 15

    Shum-Tim et al. (1999) [177] PHA-PGA Sheep 7

    Meinhart et al. (2001) [32] EC-seeded

    ePTFE

    Humans 6 to 7

    Lambert et al. (1999) [178] Heparin-

    coated

    Dacron

    Humans 6 to 7

    Devine et al. (2001) [179] Heparin-

    bonded

    Dacron

    Humans 7 to 9

    Fig. 5. The triad of tissue vascularisation.

    R.Y. Kannan et al. / Biomaterials 26 (2005) 18571875 1861Weinberg et al. (1986) [180] Collagen-Fb-

    SMC

    In vitro Unknown

    LHeureux et al. (1998) [65] Collagen-EC-

    SMC-Fb

    In vitro 4.6

    Wilson et al. (1995) [63] AAM Dogs 3 to 4Teebken et al. (2001) [35] AAM Pigs 5

    Keys: AAM, allogeneic acellularised matrix; ePTFE, expanded polytetra

    polyurethane graft; ID, internal diameter; PHA, polyhydroxyalkanoate; PGVessels Patency rates Comments

    Supercial

    epigastric

    20% at 4

    weeks

    Vein grafts had 100% patency

    Femoral 25% at 4

    weeks

    Vein grafts had 100% patency

    Aorto-iliac 100% at 3.5

    years

    Human trials under way

    Pulmonary 100% at 7

    weeks

    Good tissue ingrowth but increase

    in vessel ID

    Aorta 100% at 21

    weeks

    Outer PHA ring prevented

    increase in vessel ID

    Infra-popliteal 74% at 7

    years

    Comparable to vein grafts

    Infra-popliteal 58% at 2.5

    years

    Better results than plain Dacron

    Femoro-

    popliteal

    55% at 3.5

    years

    This trial shows better results than

    PTFE

    92% EC coverage but weak

    bursting strength

    Comparable bursting strengths to

    human vessels

    Coronary 44% at 6 All failures due to acute graftmonths rejection

    Carotid 38% at 4

    months

    Disappointing results due to graft

    rejection

    uoroethylene; Dacron, polyethylene tetraphthlate; CPU, compliant

    A, polyglycolic acid.

  • An in vivo study in a rat femoral artery model showeda 2025% patency rate at 1 month with PTFEmicrovessels (o1mm), while all vein grafts in similarsettings remained patent [36,37]. In order to improvepatency rates, luminal endothelialisation has beenattempted. Kidd and colleagues, through 1mm inter-position ePTFE vascular grafts in rat aortas, coatedwith ECM (such as matragel) and incorporated withnon-tumorigenic human keratinocyte cell line (HaCaT)or EpsteinBarr Virus (EBV)-transformed human B-celllymphocyte (TOU II-4) cell lines, showed ablumenalvascularisation with the formation of an EC lining 5weeks post-implantation [10]. Correlation of theseresults to large animal models is hindered by the factthat rat animal models rapidly endothelialise, whilst thelatter do not to the same extent, this warranting furtherresearch in this particular eld [52,9].An in vitro biological micro-vascular model has been

    developed by Neumann and co-workers, wherein 80 mmdiameter nylon strands were used as templates onto

    IN PRESSaterials 26 (2005) 18571875diameter are optimal for endothelialisation [42].Smaller pores would elicit an inammatory reaction[43], while increasing pore sizes and porosities improvethe radial compliance of these grafts [44] and reduce thelikelihood of intimal hyperplasia.More compliant biomaterials such as poly(carbo-

    nate-urea)urethane (CPU) are now being introduced[53]. CPU is currently undergoing Phase I clinical trialsas a lower limb graft and has been found to retainsimilar compliance to native vessels after implantationand exhibited little intimal hyperplasia [39,40]. There-fore, it has the potential to be used to construct thearterial conducting component of articial capillarybeds. Synthetic protein polymers cross-linked by g-irradiation represent a new generation of biopolymers.Preliminary reports suggest that they have similarelasticity as arteries, with a controllable rate ofdegradation [45]. This recent trend illustrates thelimitations of PTFE and Dacront at lower ow ratesand stresses the need for alternative biomaterials.Materials such as PTFE and Dacron have littlepotential for micro-vessel grafts due to their poorhaemodynamics caused by lack of arterial compliance.This results in thrombogenicity to the extent that inlow-ow applications (as is the care for microvessels)below 4mm their usage is predelicted by their proprietyfor blockage. Polyurethanes such as CPU due to theirsuperior haemodynamics (a result of their arterial likevisco-elasticity and honeycomb structure) have highapplicability for microvessel development and haveshown little blockage in 2mm applications (unpub-lished data).

    3.1.1.2. Micro-vascular grafts. Micro-vascular graftsare those with internal diameters of 1mm or less, andmay be classied into conducting arterial and distribut-ing capillary vessels. The construction of arterialconduits is based on small-calibre vascular grafttechnology. These new grafts may be constructed byusing (1) newer polymers such as CPU, (2) by coatinggraft lumen [5457] with anti-platelet agents or cells(most commonly endothelial cells, but also possiblybroblasts or smooth muscle cells) and (3) constructingbiological or bio-hybrid grafts in vitro prior to re-implantation [38, 5961]. As these vessels divide further,they need to branch into a distributing capillary networkwith larger pore sizes and thinner vessel walls forenhanced nutrient exchange. Although autologous veingrafts, the current gold standard for micro-vascularrepairs, are compliant and non-thrombogenic, they arelimited by the need for additional vein-harvesting

    ARTICLER.Y. Kannan et al. / Biom1862procedures. Furthermore, the construction of articialvascularised tissue requires an inherent vascular net-work [58]. Vein grafts are not suitable for this purpose asit is technically impossible to dissect out a capillary bedin its entirety.which SMCs in a culture medium could attach to. Aftera week, this strand was physically removed and thegrafts were perfused in a pulsatile ow chambersimulating the arterial system (Fig. 6). After 28 days inculture, these vessels were shown to have 3040 layers ofSMCs evenly distributed throughout the lumen andstrong enough to withstand its own weight. Non-absorbable nylon was preferred to absorbable cellulosesince constructs based on cellulose had irregular, unevenluminal surfaces [43]. This model has the potential toform arterioles and capillaries in vitro, which could thenFig. 6. A viable microvessel perfusion system. Modied from Ref. [66].

    Key: SM, smooth muscle cell.

  • regeneration with predominant sprouting from the venousend. They concluded that while an AV loop quantitativelyincreased capillary and new tissue formation, ligating theloop ensured better tissue organisation [72].

    3.2. Extracellular matrix scaffolds

    ECM is composed of collagen, bronectin, laminin,GAGs and water [73]. Fibroblasts synthesise non-cellular components of the matrix, as shown in Table2. Using acellular dermal grafts in SpragueDawley ratgroins, matrices were pre-fabricated by vascularisingthem with the supercial epigastric vessels. Histological

    IN

    Fig. 7. The effect of vessel conguration on tissue vascularisation

    (modied from Ref. [72]. Group Icontrol (acellular dermis); Group

    IIarterio-venous loop embedded in the matrix Group IIIarterio-

    venous bundle ligated within the matrix; Group IVow-through

    graft).

    Table 2

    Non-cellular components of ECM and their function [73]

    Component Function

    Collagen type I Structural framework

    Collagen type IV Basement membrane for EC attachment

    Collagen type VII Keratinocyte attachment

    Fibronectin Cell adhesion

    Laminin Protein adhesion

    GAGs Cytokine and growth factor binding

    ateribe transplanted into living tissue. Further results usingECs in a similar model are being undertaken [66].Bio-hybrid microvessels have also been developed in

    vitro using co-cultures of EC and SMC. These cells werepassed through polypropylene capillaries at 3 dynes/cm2

    laminar shear stress. The cells lined the lumen of thesetubes while SMCs managed to migrate outside these150mm diameter microvessels through pores (0.5mm) toform a two separate monolayers as in normal vessels [64].

    3.1.2. Configuration of vessels

    The limiting factor in metabolite exchange is theextra-vascular distance needed to be traversed. Thephysiological response to this is to pair up both thearteriolar and venular components together. Betweenthem, numerous small capillaries run across tissue at across-sectional density of 1300 permm2, with an inter-capillary distance of 34 mm to form a rich perfusingnetwork. This is well within the maximum diffusingdistance of oxygen, glucose, carbon dioxide and otherwaste metabolites [16]. In studies on rat mesentery, itwas found that arterioles and venules travelling togetherformed a counter-current system responsible for auto-regulation and improved nutrient exchange [67]. There-fore, arteries and veins should be paired for greaterefcacy.A study of the temporal relationship of pre-fabricated

    aps was performed by inserting a ap of tissue underrat abdominal skin. The femoral arterio-venous pediclesinduced ap neovascularisation as early as 2 weeks post-surgery, but it took up to 8 weeks for the vascularpedicle to sustain the ap alone [68]. However, in asimilar study, the ap was found to survive after itspedicle was divided after 34 weeks [69]. Studies onhybrid aps have also shown that the pedicle can bedivided by 4 weeks, provided a highly porous biomater-ial like polyethylene is used along with living tissue as aap [70,71].Tanaka and associates used the rat femoral vessel

    system to determine whether loop, ligated bundles orow-through pedicles were most effective in initiatingand sustaining tissue perfusion and neovascularisation.Using a collagen I dermal implant in the rat groin, theyformed four groups. In the rst group, the implant wasplaced on a recipient bed with no vascular pedicle. In thesecond group, a vascular loop formed of either anarterial or venous graft was employed. In the thirdgroup, an arteriovenous (AV) bundle ligated at one endwas embedded within the matrix, and in the fourthgroup an AV ow-through graft was placed across thematrix (Fig. 7). Four weeks later, the matrices were

    ARTICLER.Y. Kannan et al. / Biomsectioned and stained. Matrices from group II showedthe greatest increase in volume with ow rates of up to20 times normal. However, compared to the third group,it had less tissue organisation. AV ligation (group III)showed the densest vascularity and maximal tissuePRESSals 26 (2005) 18571875 1863analysis showed inltration by mononuclear cells withinthe rst 72 h with neo-vascularisation occurring betweendays 3 and 14. Simultaneously, SMC and EC were laidalong the lines of stress. Devascularised matrices(control group) had a 90% hernia rate, while those with

  • an intact vascular pedicle within had no evidence of ahernia (po0.01). Therefore, it was concluded thatvascularised scaffolds were structurally and functionallysuperior to non-vascularised ones [74].

    3.2.1. Biomaterial substitutes

    The ideal matrix should be biocompatible, porous,supportive, readily available, permeable to incomingvessels or chemicals, non-immunogenic [62], biodegrad-able, inert, have a slow-release mechanism and, lastly, beeasily administered as an injectable. Tissue-engineeredconstructs act as temporary scaffolds, which maintaincells in situ and induce neovascularisation. Eventually,the existing matrix will be replaced by the products ofnewer cells such as collagen, laminin, elastin, GAGs orbronectin. These undergo continuous remodelling [75]as depicted in Fig. 8, and ultimately reorganise intonear-normal tissue [76]. Current scaffolds can beclassied into natural and synthetic types, each with itsown advantages and disadvantages. Natural biomater-ials are characteristically biodegradable, non-immuno-genic and bio-compatible. A list of natural ECMavailable is summarised in Table 3.

    3.2.1.1. Natural. Alginate is an anionic polysaccharidecomposed of b-D-mannuronic acid and a-L-glucuronicacid cross-linked in a gel by non-toxic levels of calciumions. It has been incorporated with various growthfactors like basic broblast growth factor (bFGF),vascular endothelial growth factors (VEGF) and hepar-in within microspheres [77]. Alginate gel is a hydrogelsystem that is independent of temperature changes [78]but tends to release its chemicals within 4 days [79]. Thiscan be overcome by incorporating microspheres andpeptides [80] or cross-linking with heparin [81]. Alginategels modied with arginine-glycine-aspartic acid (RGD)peptide sequences [82] and syngeneic broblasts formedstrong gels and were more tissue-like [83]. An agarosegel cross-linked with heparin has been used to deliverbasic broblast growth factor (FGF) beads to the

    ARTICLE IN PRESS

    ent

    Agarose Agarose Solid gel

    R.Y. Kannan et al. / Biomaterials 26 (2005) 185718751864Hyaluronic acid

    (HA)

    Glucuronic acid+N-

    acetyl-D-glucosamine

    (linear GAG)

    Biodegradable gel,

    biocompatible,

    7photopolymerisable

    Fibrin glue Fibrin monomers Injectable gelaFig. 8. The effect of cell traction on tissue patterns.

    Table 3

    Natural materials used in ECM constructs

    Biomaterials Composition Characteristics

    Alginate b-D-mannuronicacid+a-L-glucuronicacid

    Temperature-independ

    gelCollagen/gelatine Type I collagen

    (atelocollagen)

    Porous and permeable

    Matrigel Collagen type

    IV+laminin+entactin+proteoglycansInert, permeable, injectable gelStructuporcine myocardium, but delivery characteristics arestill sub-optimal [84].Hyaluronic acid (HA), a linear glycosaminoglycan

    (glucuronic acid N-acetyl D-glucosamine), has goodbiocompatibility and is degraded by hyaluronidase.However, it has poor adherence and inhibits ECproliferation, while its degradation products are pro-angiogenic [85]. To improve this, chitosan, a glycosa-minoglycan (N-acetyl D-glucosamine), has been addedto HA to form a composite material in the synthesis ofendotheliased human skin equivalent [86]. Chitosanattracts neutrophils and activates macrophages [87].Microspheres lled with FGF within similar constructswere used to improve neovascularisation in rat tissue[88]. The addition of methacrylate groups to HA,followed by exposure to light of 480520 nm wave-lengths, could photopolymerise composite HA gels intoany given shape with minimal gel swelling. RGDsequences could also be added to the composite to

    Comments Modications References

    Burst release Microsphere/bead

    incorporation, heparin

    cross-linkage (controlled

    release), RGD-peptide

    sequence incorporation

    [78,8082]

    Sub-optimal release Heparin addition (increase

    factor load)

    [84]

    Angio-inhibitory,

    degradation products are

    angiogenic, non-adhesive

    to cells

    Chitosan cross-linkage

    (improves angiogenesis),

    RGD sequences (improve

    cell adherence),

    methacrylate addition

    [85]

    Structurally weak Heparin cross-linkage [90](controlled release)

    Low factor-loading

    capacity

    Heparin cross-linkage [91]

    rally weakNone[94]aIdeal for local delivery.

  • INateriimprove cell adherence. In vitro studies on thiscomposite showed improved broblast viability andproliferation compared to standard HA gels [89].Fibrin glue derived from blood clots is useful as an

    injectable medium for the local delivery of growthfactors like VEGF. Burst release is a problem that isovercome by the incorporation of heparin for bettercontrol [90]. Collagen type I is highly porous (pore sizesof 50150 mm) and has been seeded with Fb or pre-adipocytes [91]. Chondrocytes embedded within three-dimensional (3-D) collagen gels were found to be viableand capable of synthesising proteoglycans (PGs) [92].Unfortunately, loading factors onto it is difcult, butthis can be overcome by cross-linking collagen gels withheparin. This has been shown to increase the incorpora-tion of angiogenic factors sufciently to culture humanumbilical vein cells (HUVEC) in vitro [93]. ECM gel(Matrigelt) that is composed of basement membraneproteins has been used in vitro as scaffolds [94], but hasso far few clinical uses [95]. Other natural substitutesused are porcine intestinal sub-mucosa [96], acellulardermis, cadaveric fascia and amniotic membrane [97].The most signicant drawback of these ECM substitutesis that they are structurally weak with unmodulatablehydraulic conductivity.

    3.2.1.2. Synthetic. Numerous polymers have beenemployed in tissue engineering, including poly(propy-lene fumarates)/PPF, poly anhydrides (PA), polyglycolicacid (PGA), polylactic acid (PLA), polycarbonates (PC),polylactone (PL), polyurethanes (PU), poly(orthoesters)(POE), polyphosphazenes (PPZ) and poly(ethylene-terephthalate) (PET) [98] and the commonest polymerin use, polyester (PE). These are being used withincreasing frequency in biomedical devices.PGA is an inelastic polyester with high crystallinity

    (4650%). It is degraded by the diffusion of waterfollowed by hydrolysis of the crystalline elements of thepolymer into glycolic acid [99]. PLA is less crystallinethan PGA but more hydrophobic, making it lesssusceptible to hydrolysis. Poly(lactide-co-glycolide)(PLGA) co-polymer in the amorphous form is a high-porosity, open-pore system which degrades in 26months by means of hydrolysis of the ester bonds[100]. A signicant drawback is that angiogenic factorsincorporated within it tend to leach out. This can berectied using microsphere, beads or bioactive foams[101] for slower release of chemicals. Experiments usingmicrospheres containing recombinant VEGF withinPLGA foams, which were then implanted into ratfascial aps, showed a more controlled release of growth

    ARTICLER.Y. Kannan et al. / Biomfactors [102].1,3-trimethylene carbonate (TMC) and D,L-lactide

    (DLLA) (19:81, molecular weight ratio) are amorphousco-polymers of PLA. Salt-leaching these polymersproduced foams of high pore interconnectivity andporosity with an average pore size of 100 mm. Cardio-myocytes grown on it were able to adhere andproliferate, indicating that TMC-DLLA co-polymersare not cytotoxic. In vitro signs of degradation occurredby 4 months via hydrolysis [103]. When these polymerswere implanted subcutaneously in rats, degradationstarted at 3 weeks and by one year 96% of its mass waslost. These co-polymers also elicited a sterile and acuteinammatory reaction, resulting in the formation of acapsule in vivo [104]. Degradation was far more rapid inpoly TMC polymers (3 weeks). Silica-based poly(di-methylsiloxane) (PDMS) has been polymerised withPLA-PGA composites to form PLGA/TEGOMERS,which have shown to be biocompatible, supportive tocell growth and non-toxic [105].Experiments with micro-vascular cell cultures mainly

    involve PLA, PGA or their composite scaffolds. Theseare summarised in Table 4. PLs like poly CL arebiocompatible with prolonged degradation times of upto 3 years in vitro [106]. Combining CL with TMC slowsdown degradation of the primary TMC polymer [107].The mechanism of degradation is hydrolysis withcaproic acid, the by-product. Its disadvantage is that itis structurally weak.PPFs are polyesters based on fumarates. They can be

    injected into any space and further photo cross-linkedby adding methacrylates [108]. PPFs are structurallystrong and capable of withstanding great loads. In vitrodegradation (up to 200 days) occurs following hydro-lysis of its ester bonds to form fumaric acid, a naturalmetabolite, propylene glycol and poly(acrylic acid-co-fumarate) [109]. Apart from being biodegradable, PPFhas also been found to be non-cytotoxic with minimalinammatory reaction in vivo [108].PA forms soft tissue constructs being biocompatible,

    thermoplastic, with controlled degradation characteris-tics (surface erosion). It is a weak polymer limiting itsuse in load-bearing devices. It may be strengthened bythe addition of imide groups [110]. PAs are degraded byhydrolysis of its anhydride bonds by 12 months in vitrointo dicarboxylic acid [111]. In vivo implantation ofthese polymers shows evidence of good vascularisationof itself by 4 weeks [112].PCs are thermoplastic and strong enough to sustain

    heavy compressive loads. Their advantages are that theyare biodegradable, biocompatible and have very lowrate of degradation. However, hydrolysis of its carbo-nate groups culminates in the formation of acidic by-products. This is an undesirable property for an ECMsubstitute [113].Bioelastic materials, monomeric protein polymers

    PRESSals 26 (2005) 18571875 1865which convert heat or chemical energy into mechanicalenergy, represent the next generation of polymers. Heatexchange, hydrophobicity or hydrophilicity induces amechanical tensile force within the polymer that ismediated by competition between the molecules for

  • ARTIC

    LEIN

    PRESS

    Table 4

    Synthetic materials used for ECM construction in various tissues

    Polymer Compressive

    strength

    Synthesis DT (months) By-products Advantages Disadvantages References

    PGA 7.5GPa E/M/C 612 Glycolic acid Porous, biocompatible, non-

    toxic, non-inammatory,

    natural waste metabolite

    Not pliable [99]

    PLA 2.7GPa E/M/C 424 L-lactic acid Porous, biocompatible, non-toxic, slower degradation,

    natural waste metabolite

    Hydrophobic [113]

    PLGA 1.9GPa E/M/C 1216 D,L-lactic acid Porous, biocompatible, non-

    toxic, non-inammatory

    Excessive leakage [100]

    PL 0.4GPa E/M/C 424 Caproic acid Non-toxic, biocompatible Weak structure [106]PPF 230MPa Injection E30 Fumarate, polyethylene

    glycol, poly(acrylic-co-

    fumaric) acid

    Non-toxic, biocompatible,

    minimal inammation, photo-

    cross-linkage, strong

    Not signicant [108-109]

    PA 1.3MPa Thermoplastic 12 (in vitro) Dicarboxylic acids Biocompatible, thermoplastic,

    surface erosion, angioinductive

    Weak [111]

    PC Sufcient for load-

    bearing

    Thermoplastic Very slow Tyrosine, CO2 and alcohols Porous, biocompatible,

    thermoplastic, osteoconductive

    Acidic byproducts [181]

    PU (LDI) 840Mpa Thermoset 12 Lysine, glycolic acid, caproic

    acid

    Biocompatible, strong, pliable Some PUs (TDI) are toxic [113]

    PPZ Sufcient for load-

    bearing

    Phosphates and ammonia Biocompatible, osteoconductive,

    strong

    Not signicant [113]

    POE Sufcient for load-

    bearing

    Regulatable

    with lactide

    Carboxylic acids Biocompatible, osteoconductive,

    strong

    Not signicant [182]

    PEEU Tensile strain

    (929MPa)

    Biocompatible, strong Poor cell adhesion [183]

    Ppy o13 Electrical conduction, minimalinammation

    [184]

    Keys: C, casting ; DT, degradation time; E, extrusion; M, moulding; , unknown.

    R.Y

    .K

    an

    na

    net

    al.

    /B

    iom

    ateria

    ls2

    6(

    20

    05

    )1

    85

    7

    18

    75

    1866

  • hydration. These forces modulate cells within theconstruct to form micropatterns and secrete newECM. This process is called cellular tensegrity ormechanotransduction [114]. An example would bepoly(N-isopropylacrylamide), which can mechanicallyrecongure themselves with changes in hydrophilicityand temperature [98].

    3.2.1.3. Solgel systems. Gels are composed of threecomponents namely solute, solution and voids, with thethree in equilibrium with one another [115]. A typical gelis polyethyleneglycol (PEG). As shown in Fig. 9,polymer molecules are initially functionalised withadhesion factors. These are then cross-linked via theformation of reactive termini. Cells migrating into this

    ARTICLE INR.Y. Kannan et al. / Biomaterimatrix break down the cross-linking peptide leading tolocal degradation and hence release of the incorporatedfactors such as heparin and VEGF [95]. These syntheticgels need to be bio-degradable and cell-responsive [116].Using a PEG-based polymer, by shortening the blocklength from a mean molecular weight of 930 to6090 kDa, biodegradation of the hydrogel could beminimised [117]. The same effect has been shown intriblock co-polymers of poly(L-lactide)/PLLA andpoly(ethylene oxide) (PEO) (PLLA-PEO-PLLA) [115].Photo-polymerisable poly vinyl alcohol (PVA) hydro-

    gels [118] are injectable. As such, it is possible to placethese gels in vivo using minimally invasive proceduresand then allowing cross-linkage to occur. This gel wasfound to be highly elastic and strong. Since celladherence was poor, these constructs were modiedwith the RGD peptide sequence [119]. This principle wasused by Cao and colleagues to immobilise ovalbuminwithin a 3-D hydrogel [120]. De Rosa and co-workersFig. 9. The synthesis of biomimetic gels.proposed a polyelectrolyte gel with a cationic surfacecharge across it. These charges increased the adhesion ofFb to the gel as well as their proliferation, therebyminimising the risk of brous encapsulation [121].

    3.2.2. Superstructure

    Superstructure refers to the integration of 2-D poresor structures into a 3-D conguration, whereby theseoverlap in a regular or irregular pattern. In polymers,the former confers crystallinity and the latter, amor-phous characteristics. The superstructure of a polymerdetermines pore characteristics. Studies have shown thatcells aggregate less within smaller pores and thesesmaller aggregates proliferate to a lesser extent [122].Hence, in scaffolds with smaller pore sizes and volumes,cells spread faster [123].Superstructures exist in three main forms namely

    textile (woven/knitted), open-pore and angiopolar sys-tems. The angiopolar principle involves spatially or-ientating pores within matrices to induce cells,microvessels and nutrients to propagate in a givendirection [124]. Pores within biopolymers may beclassied into macropores (E500 mm), mesopores(2030 mm) and micropores (o10 mm). Macroporesregulate the optimum cell type and seeding densitywithin a construct. Mesopores allow microvessels toinltrate the polymer. It has been observed in early invivo studies on rats that arterioles and venules tend toproliferate on the inner surface of mesopores whilecapillaries lined its outer surface [124]. Lastly, micro-pores function as a regulator of molecular transportwithin a polymer system. Pore characteristics may bemodulated depending on the method of polymersynthesis like extrusion, leaching, casting, moulding orelectrospray.

    3.2.3. Cells seeded onto matrices

    Seeding cells onto a matrix is integral to the formationof a bio-hybrid scaffold as cells like broblasts andchondrocytes secrete ECM. However, long-term cellviability in such articial constructs is a limiting factoras these cells have to remain viable in vivo untilneovascularisation occurs. Fibroblasts are the mostabundant cells within the ECM, but scaffolds seededwith broblasts tend to contract following in vivoimplantation, thus impairing broblast migration. In astudy comparing pure and GAG-chitosan-collagensponges, the latter minimally contracted and showed a300% increase in broblast proliferation at 6 weekscompared to pure collagen sponges [125]. This wasaugmented by incorporating RGD-peptide sequences to

    PRESSals 26 (2005) 18571875 1867improve broblast attachment and proliferation [126].Engineered cartilage using chondrocytes has the poten-tial to act as a viable ECM scaffold (as these cells havelow metabolic rates and require minimal perfusion), intowhich micro-vascular networks can actively proliferate

  • ated ex vivo [127] on PLGA scaffolds up to 2 months

    further improves vascularisation prior to actual cell

    INateriseeding [131].

    3.3. Vascularisation

    While scaffolds and microvessels are essential com-ponents of a capillary bed, it is equally important for thetissue-engineered device to connect to the host tissue.This would entail the induction of incoming micro-vessels into the scaffold (angioinduction) as well asallow the inherent microvessels to grow out and meetthese vessels (inosculation).

    3.3.1. Inosculation

    The presence of an inbuilt micro-vascular networkitself within a matrix is insufcient unless the intra- andperi-vascular regions are seeded with ECs, SMCs andbroblasts. Therefore, for the growth of vessels withinan engineered vascularised tissue construct out into thehost tissue, it is necessary for these to either link orhook-up with the hosts vasculature, this process is aterm described as inosculation. While ECs confer non-thrombogenicity to the graft, circulating haematopoieticstem cells such as EPCs are capable of stimulating theformation of a vascular network (post-natal vasculogen-esis) [134]. Their sources are the bone marrow, mono-following implantation, but viability decreased there-after. However, the study did not mention whether thesecells survived by neovascularisation or diffusion. Pre-adipocytes attached maximally to hyaluronic acidscaffolds with mean pore sizes of 400 mm [128]. Pre-laminating these cells layer onto layer in vitro improvedits vascularisation characteristics [129]. Fatty tissue fromlipoaspirate (LPA) has been shown differentiate intomany cell lines and hence form a source of stem cells[130]. Injecting the cDNA of VEGF into scaffoldsand grow. Embedded chondrocytes within brin glueshowed that these cells can maintain viability in a non-vascularised environment [132] although chondrocytesalone forms rigid tissues. Brown and co-workersdeveloped a hybrid chondrocyte-SMC with a compres-sive modulus between muscle and cartilage [133].Research has demonstrated that fat has potential as a

    bulking agent; however, the difculties associated withthe extraction of adipocytes and their subsequent culture(the propensity for intra-cellular lipid vacuole bursting)has rendered their usage in tissue engineering to beextremely limited. Pre-adipocytes extracted from theepididymal fat pads of SpragueDawley rats prolifer-

    ARTICLER.Y. Kannan et al. / Biom1868nuclear cells and the vessel wall. These cells can beharvested by enrichment following extraction or ex vivocultures [135]. Once these cells are injected into thecirculation, EPCs move on to sites of neovascularisation(therapeutic vasculogenesis) [136].Early studies on vasculogenesis used cells like quailblastodiscs, murine embryonic stem cells, HUVEC andhuman pulmonary micro-vascular endothelial cells(HPMEC) grown on collagen, brin, gelatin andmethylcellulose matrices (Table 5). These cells havebeen shown to grow into capillary-like tubes (CLTs),which may be controlled by altering the growth factorgradient or biomechanical tension across the gel. Whilein culture, tissues can be genetically modied bytransfecting them with viral vectors. Overexpressionand carcinogenicity are a problem. Alternatively, cellslike SMCs could be transduced with multiple genes andthen be allowed to repopulate the microenvironment(transgenesis). SMCs were harvested from tissue en-zymatically and cultured serially until differential celladhesion molecules are removed. Next, the SMCs weremagnetically extracted and incubated with high-titreretroviral supernatants. 99% of these cells were success-fully transfected [137].EC are sourced from arteries, veins, omentum and

    subcutaneous fat [138]. Using enzymatic degradation,EC are harvested from vein grafts by either thecannulation or eversion technique and cultured for 56weeks. Grenier and co-workers have introduced amethod of obtaining EC, SMC and broblasts fromthe same vein biopsy [139]. Isolation of EC from fattytissue requires it to be minced, enzymatically digested,centrifuged in a Percoll gradient before extracting ECusing magnetic beads [138].Prior to in vivo implantation, EC or EPCs would need

    to be seeded onto scaffolds. The seeding density woulddepend on the scaffold material, its porosity and themethod of cell inoculation. The standard mode forpromoting cell attachment is to culture cells onto thepolymer with or without adhesion molecules. Adisadvantage is that cells tend to adhere only to theouter layers of the scaffold. In order to achieve morehomogenous implantation throughout a 3-D scaffold,other methods like the drop on and low-pressurecentrifugation techniques which systematically seed cellsonto scaffolds have been introduced [128].Human adipose stromal cells placed within micro-

    carriers and co-cultured with HUVECs were grown in aserum-free static culture model with the addition ofangiogenic factors. By day 16, stable CLTs with patentlumens were formed by outgrowing ECs. Larger loopscoalesced while smaller ones regressed, similar to thepruning phenomenon that is observed during embry-ological development of the vascular system. In vitro,this is termed guided migration.Clinically, the larger the microvessel, the greater the

    PRESSals 26 (2005) 18571875perfusion [140]. An in vivo model wherein humandermal microvascular endothelial cells (HDMEC)seeded onto biodegradable polymer matrices were putinto immunodecient mice showed the following char-acteristics. On day 1 EC migrated through the matrix;

  • IN

    indu

    bryo

    tokin

    tokin

    ateriARTICLE

    Table 5

    In vitro developmental models of angiogenesis and vasculogenesis

    Cells CLS

    formation

    (day)

    Morphogenesis

    Bovine capillary EC 23 Spontaneous

    Embryonic stem cells 12 Spontaneous em

    body formation

    Muscular tissue fragments adipose

    tissue frag.

    312 Spontaneous

    Bovine aortic EC, adrenal capillary

    EC, HUVEC

    1 Spontaneous

    Bovine capillary EC 515 Cytokines

    Bovine capillary EC 23 Phorbol ester

    Human umbilical vein EC, HDMEC 1 Spontaneous

    Bovine capillary EC 12 Spontaneous cy

    Rat aortic explants 7 Spontaneous

    Bovine aortic EC 1018 Spontaneous

    Human umbilical vein EC 1 Spontaneous

    Rat fat microvessels fragments 46 Spontaneous

    Calf pulmonary aortic EC 27 Spontaneous cy

    Human umbilical vein EC, bovine

    retinal EC

    12 Spontaneous

    R.Y. Kannan et al. / Biomon day 5 they formed CLTs and by 1 week haddifferentiated into functional microvessels. Maturationoccurred by 21 days and these microvessels eventuallylinked up with the host vessels[141,142]. Immunohisto-chemical analysis of a murine myocardial model showedthat monocytes/macrophages produced holes throughthe myocardium using proteolytic enzymes, the lumensof which were eventually colonised by EPCs [143].

    3.3.2. Angioinduction

    The process of inducing incoming microvessel forma-tion depends on the growth factors used and the methodof delivery. Their angiogenic effects were studied usingin vivo models like chorio-allantoic membrane (CAM)[144,145]. FGF and VEGF are mainly involved invasculogenesis. Angiogenic sprouting is mediated bytransforming growth factor-b (TGF-b), while matura-tion of vessels is via angiopoietin-1 and -2 (Ang-tie) andplatelet-derived growth factor (PDGF) pathways [146].Their effects are summarised in Table 6.The primary stimulus and factor causative for the

    process of capillary sprouting is hypoxia.Within 30min of its onset, early growth response

    factors (Egr-1 and -3) are expressed [147]. VEGFmodies the ECM to allow EC and broblast migration[140,30], acting on the venules, it increases their

    Embryonic stem cells 11 Spontaneous embryo

    body formation

    Human placental blood vessels 721 Spontaneous

    Mice microvessel fat pad 14 Spontaneous cytokin

    Bovine aortic EC, human HUVEC 3 Spontaneous

    Human marrow microvascular EC 2150 Spontaneous cytokin

    Keys: EC, endothelial cells; HUVEC, human umbilical vein endothelial

    bronectin.PRESS

    ction Matrix Spatial

    organisation

    References

    Collagen I sandwich 3-D [156]

    id Culture dish 3-D [157]

    Fibrin+Collagen I 3-D [158]

    Fibrin 2-D [159]

    Collagen I 3-D [160]

    Fibrin 3-D [161]

    Matrigel 2-D [162]

    es Fn+Collagen

    IV+Matrigel

    2-D [163]

    Fibrin+Collagen I 3-D [164]

    Collagen I 2-D [165]

    Fibrin I or II sandwich 3-D [166]

    Collagen I 3-D [167]

    es Fibrin in micro-carriers 3-D [168]

    Fibrin 2-D [169]

    als 26 (2005) 18571875 1869permeability. Subtypes A and B are generally involvedin EC migration and proliferation, while C and Dsubtypes are responsible for venous and lymphaticproliferation [25]. FGF is another angiogenic factorused. Derived from SMCs and ECs, FGF is stimulatedby EC regeneration, hypoxia and collateral formation toelicit cell induction and proliferation. In pre-fabricatedaps, Bayati and colleagues showed that tissue viabilitywas enhanced by using FGF [148].Monocyte chemoattracted protein-1 (MCP-1) is

    released following shear stress to the vessel wall. Itattracts circulating and in-situ MCs to home in at sites ofneovascularisation [149]. Granulocyte-Macrophage Col-ony Stimulating Factor (GM-CSF) acts synergisticallywith MCP-1 by promoting arteriogenesis, stimulatingMC/MP release from the bone marrow as well asprolonging their life span [17]. TGF-b described earlieralso serves as a growth factor which, depending on thereceptor, either activin-like kinase type-1 (ALK-1) orALK-5, stimulates or inhibits angiogenesis, respectively[150]. This illustrates the overlapping functions of thesemediators. The slow release of these growth factorswould diminish function, while excessive amountswould induce brovascular growth.Control over spatial and temporal patterns of the

    chemicals release is necessary to form an organised,

    id methylcellulose 3-D [170]

    Fibrin 3-D [171]

    es Collagen gel 3-D [185]

    Fibrin+Collagen I 3-D [186]

    es Fn+Collagen I 2-D [187]

    cells; HDMEC, human dermal microvascular endothelial cells; Fn,

  • bioactive scaffolds [152], preferably surface-erodingscaffolds like poly(glycerol sebacate) (PGS) [153].Other

    IN

    Recep

    FGFr

    VEGFR2/Flk-1 EC formation

    VEGF

    VEGF

    ALK-

    Tie-1

    PDGF

    Tie-1

    Eph B

    wth f

    aterimethods of delivery include viral transduction [154],direct inoculation [155] or genetically modied cells[137].

    4. Conclusion

    The biggest hurdle in the construction of tissue-engineered aps is the inability of existing synthetic ortissue-engineered small-diameter vascular grafts(o6mm) to sustain ow through them and, moreimportantly, the absence of any source of an incorpora-congured and functional micro-vascular network. Invitro, growth factors may be incorporated by co-culturing them with constructs or placing these factorswithin bioarticial organs like microcarriers which allowregulated release of chemicals. In vivo, these factors areintroduced directly into the bloodstream, incorporatedinto cells or packed into defects as beads [151] or

    ARTICLE

    Table 6

    Role of angiogenic growth factors during vascular development [146]

    Event Factor

    Angioblast induction FGF

    Conversion of angioblasts to EC VEGF

    EC formation into tubes VEGF

    Angiogenic sprouting VEGF

    EC activation TGF-b1Vessel thickening Angiopoietin-1

    Smooth muscle recruitment PDGF-bVessel thinning Angiopoietin-2

    Arterio-venous differentiation Ephrin B2

    Keys: FGF, broblast growth factor; VEGF, vascular endothelial gro

    derived growth factor-beta.

    R.Y. Kannan et al. / Biom1870table capillary bed, natural or otherwise. Current trialson EC-seeded infra-popliteal vessel bypass grafts haveonly been proven to be as successful as vein grafts in thesame setting [32], while the development of an articialcapillary bed is limited to in vitro models [156171].However, a better understanding of microuidics andfurther breakthroughs in nanotechnology may makethis more likely.

    4.1. Future perspectives

    Micro-contact printing, printing biological moleculesdirectly onto scaffolds, is spawning a new generation oftissue-engineered constructs. Here, a blueprint is madeusing a computer-aided design and the construct is thenbuilt within 24 h. For instance, gold or silver plates areinitially patterned with alkane-thiolates onto whichbronectin molecules are attached. In turn, bronectinbinds to EC in culture [172]. These individual celldroplets then coalesce due to tissue uidity. Whenapplied in many layers, three-dimensional constructscan then be developed. This technique has been used toform articial liver micro-channels [173]. As the keyissue is tissue or cell perfusion, these constructs arenally placed within a bio-reactor [174]. Recently, aporous endotheliased network based on human pul-monary endothelial cells was successfully developedusing this technology [175]. Once popularised, thistechnique would revolutionise fabrication of precisetissue-engineered constructs.Perhaps, the most exciting tissue engineering advance

    is nanotechnology. Earlier generations of biomaterialshad micro-dimensions while biomolecules are nanos-tructures. As such, physiological processes within thebody could not be modulated by these devices until now.The recent discovery by Moldovan and colleagues thatECs and EPCs in the bloodstream are capable ofliterally drilling holes through matrices has opened up anew perspective. These groups have proposed buildingan angiogenesis assist device or angiochip usingnanotechnology, wherein foci of EC would be spatially

    R2/Flk-1 Primitive vascular plexus

    R-2/Flk-1, VEGFR-1/Flt-1 Angiogenesis

    1, ALK-5 Angiogenic modulation

    Vessel stabilisation

    R-b Vessel stabilisationand Tie-2 Antagonistic to angiopoietin-1

    4 Remodelling

    actor; TGF-b1, transforming growth factor beta-1; PDGF-b, platelet-PRESS

    tor Action

    Angioblast formation

    als 26 (2005) 18571875distributed within a three-dimensional matrix intercon-nected by articially created grooves for proliferationand subsequent tube formation [172]. This device iscurrently being used to develop liver tissue. Similarly,they could be spatially distributed within any tissue andact as angioconductive and angioinductive foci. Ulti-mately, it is the temporal synergism achieved betweenvessel, matrix and cell, which would determine thecreation of a vascular blueprint for tissue engineering.

    References

    [1] Dunn R, Watson S. Why climb a ladder when you can take the

    elevator? Plast Reconstr Surg 2001;107:283.

    [2] Taylor GI, Palmer JH. The vascular territories (angiosomes) of

    the body: experimental study and clinical applications. Br J Plast

    Surg 1987;40:11341.

    [3] McDowell F. Plastic surgery in the twentieth century. Ann Plast

    Surg 1978;1:21724.

    [4] Song JX, Xing X, Chen JP. Reconstruction of digital avulsion

    with pre-fabricated subdermal vascular network skin ap by

  • INateriultrasonic liposuction. Zhongguo Xiu Fu Chong Jian Wai Ke Za

    Zhi 2001;15:1968.

    [5] Kroll SS, Roseneld L. Perforator-based aps for low posterior

    midline defects. Plast Reconstr Surg 1988;81:5616.

    [6] McGregor IA, McGregor AD. Fundamental techniques of

    plastic surgery. Edinburgh: Churchill Livingstone; 1995.

    [7] Vacanti JP, Langer R. Tissue engineering: the design and

    fabrication of living replacement devices for surgical reconstruc-

    tion and transplantation. Lancet 1999;354(Suppl 1):SI324.

    [8] Seifalian AM, Tiwari A, Hamilton G, Salacinski HJ. Improving

    the clinical patency of prosthetic vascular and coronary bypass

    grafts: the role of seeding and tissue engineering. Artif Organs

    2002;26:30720.

    [9] Tiwari A, Salacinski HJ, Punshon G, Hamilton G, Seifalian

    AM. Development of a hybrid cardiovascular graft using a tissue

    engineering approach. FASEB J 2002;16:7916.

    [10] Kidd KR, Patula VB, Williams SK. Accelerated endothelializa-

    tion of interpositional 1-mm vascular grafts. J Surg Res

    2003;113:23442.

    [11] Mitchell SL, Niklason LE. Requirements for growing tissue-

    engineered vascular grafts. Cardiovasc Pathol 2003;12:5964.

    [12] Fields C, Cassano A, Allen C, Meyer A, Pawlowski KJ, Bowlin

    GL, Rittgers SE, Szycher M. Endothelial cell seeding of a 4-mm

    I.D. polyurethane vascular graft. J Biomater Appl

    2002;17:4570.

    [13] Moiemen NS, Staiano JJ, Ojeh NO, Thway Y, Frame JD.

    Reconstructive surgery with a dermal regeneration template:

    clinical and histologic study. Plast Reconstr Surg

    2001;108:93103.

    [14] Wisser D, Steffes J. Skin replacement with a collagen based

    dermal substitute, autologous keratinocytes and broblasts in

    burn trauma. Burns 2003;29:37580.

    [15] Sheridan RL, Morgan JR, Cusick JL, Petras LM, Lydon MM,

    Tompkins RG. Initial experience with a composite autologous

    skin substitute. Burns 2001;27:4214.

    [16] Levick JR. An introduction to cardiovascular physiology.

    London: Arnold; 2000. p. 118238.

    [17] Buschmann I, Schaper W. The pathophysiology of the collateral

    circulation (arteriogenesis). J Pathol 2000;190:33842.

    [18] Murohara T. Therapeutic vasculogenesis using human cord

    blood-derived endothelial progenitors. Trends Cardiovasc Med

    2001;11:3037.

    [19] Adams RH, Klein R. Eph receptors and ephrin ligands. Essential

    mediators of vascular development. Trends Cardiovasc Med

    2000;10:1838.

    [20] Welm B, Mott J, Werb Z. Developmental biology: vasculogen-

    esis is a wreck without RECK. Curr Biol 2002;12:R20911.

    [21] Peirce SM, Skalak TC. Microvascular remodeling: a complex

    continuum spanning angiogenesis to arteriogenesis. Microcircu-

    lation 2003;10:99111.

    [22] Davies PF, Remuzzi A, Gordon EJ, Dewey Jr. CF, Gimbrone Jr.

    MA. Turbulent uid shear stress induces vascular endothelial

    cell turnover in vitro. Proc Natl Acad Sci USA 1986;83:

    21147.

    [23] Hoefer IE, van Royen N, Buschmann IR, Piek JJ, Schaper W.

    Time course of arteriogenesis following femoral artery occlusion

    in the rabbit. Cardiovasc Res 2001;49:60917.

    [24] Lindner V, Maciag T. The putative convergent and divergent

    natures of angiogenesis and arteriogenesis. Circ Res

    2001;89:7479.

    ARTICLER.Y. Kannan et al. / Biom[25] Felmeden DC, Blann AD, Lip GY. Angiogenesis: basic

    pathophysiology and implications for disease. Eur Heart J

    2003;24:586603.

    [26] Burri PH, Djonov V. Intussusceptive angiogenesisthe alter-

    native to capillary sprouting. Mol Aspects Med 2002;23:

    S1S27.[27] Tepper OM, Galiano RD, Kalka C, Gurtner GC. Endothelial

    progenitor cells: the promise of vascular stem cells for plastic

    surgery. Plast Reconstr Surg 2003;111:84654.

    [28] Hughes D. Transvascular uid dynamics. Vet Anaesth Analg

    2000;27:639.

    [29] Secomb TW, Hsu R, Pries AR. A model for red blood cell

    motion in glycocalyx-lined capillaries. Am J Physiol

    1998;274:H101622.

    [30] Cassell OC, Hofer SO, Morrison WA, Knight KR. Vascularisa-

    tion of tissue-engineered grafts: the regulation of angiogenesis in

    reconstructive surgery and in disease states. Br J Plast Surg

    2002;55:60310.

    [31] Salacinski HJ, Goldner S, Giudiceandrea A, Hamilton G,

    Seifalian AM, Edwards A, Carson RJ. The mechanical behavior

    of vascular grafts: a review. J Biomater Appl 2001;15:

    24178.

    [32] Meinhart JG, Deutsch M, Fischlein T, Howanietz N, Froschl A,

    Zilla P. Clinical autologous in vitro endothelialization of 153

    infrainguinal ePTFE grafts. Ann Thorac Surg 2001;71:S32731.

    [33] Budd JS, Allen KE, Hartley G, Bell PR. The effect of preformed

    conuent endothelial cell monolayers on the patency and

    thrombogenicity of small calibre vascular grafts. Eur J Vasc

    Surg 1991;5:397405.

    [34] Schmedlen RH, Elbjeirami WM, Gobin AS, West JL. Tissue

    engineered small-diameter vascular grafts. Clin Plast Surg

    2003;30:50717.

    [35] Teebken OE, Pichlmaier AM, Haverich A. Cell seeded

    decellularised allogeneic matrix grafts and biodegradable poly-

    dioxanone-prostheses compared with arterial autografts in a

    porcine model. Eur J Vasc Endovasc Surg 2001;22:13945.

    [36] Demiri EC, Iordanidis SL, Mantinaos CF. Experimental use of

    prosthetic grafts in microvascular surgery. Handchir Mikrochir

    Plast Chir 1999;31:1026.

    [37] Harris JR, Seikaly H. Evaluation of polytetrauoroethylene

    micrografts in microvascular surgery. J Otolaryngol

    2002;31:8992.

    [38] Bos GW, Poot AA, Beugeling T, van Aken WG, Feijen J. Small-

    diameter vascular graft prostheses: current status. Arch Physiol

    Biochem 1998;106:10015.

    [39] Salacinski HJ, Tai NR, Carson RJ, Edwards A, Hamilton G,

    Seifalian AM. In vitro stability of a novel compliant poly(-

    carbonate-urea)urethane to oxidative and hydrolytic stress. J

    Biomed Mater Res 2002;59:20718.

    [40] Tiwari A, Salacinski H, Seifalian AM, Hamilton G. New

    prostheses for use in bypass grafts with special emphasis on

    polyurethanes. Cardiovasc Surg 2002;10:1917.

    [41] Seifalian AM, Salacinski HJ, Tiwari A, Edwards A, Bowald S,

    Hamilton G. In vivo biostability of a poly(carbonate-urea)ur-

    ethane graft. Biomaterials 2003;24:254957.

    [42] Matsuda T, Nakayama Y. Surface microarchitectural design in

    biomedical applications: in vitro transmural endothelialization

    on microporous segmented polyurethane lms fabricated using

    an excimer laser. J Biomed Mater Res 1996;31:23542.

    [43] Ko IK, Iwata H. An approach to constructing three-dimensional

    tissue. Ann N Y Acad Sci 2001;944:44355.

    [44] Kowligi RR, von Maltzahn WW, Eberhart RC. Fabrication and

    characterization of small-diameter vascular prostheses. J Biomed

    Mater Res 1988;22:24556.

    [45] Urry DW, Pattanaik A. Elastic protein-based materials in tissue

    reconstruction. Ann N Y Acad Sci 1997;831:3246.

    PRESSals 26 (2005) 18571875 1871[46] Salacinski HJ, Tiwari A, Hamilton G, Seifalian AM. Cellular

    engineering of vascular bypass grafts: role of chemical coatings

    for enhancing endothelial cell attachment. Med Biol Eng

    Comput 2001;39:60918.

    [47] Salacinski H, Tiwari A, Hamilton G, Seifalian AM. Performance

    of a polyurethane vascular prosthesis carrying a dipyridamole

  • INateri(Persantin) coating on its lumenal surface. J Biomed Mater Res

    2002;61:3378.

    [48] Salacinski HJ, Hamilton G, Seifalian AM. Surface functionali-

    zation and grafting of heparin and/or RGD by an aqueous-

    based process to a poly(carbonate-urea)urethane cardiovascular

    graft for cellular engineering applications. J Biomed Mater Res

    2003;66A:68897.

    [49] Salacinski HJ, Tai NR, Punshon G, Giudiceandrea A, Hamilton

    G, Seifalian AM. Optimal endothelialisation of a new compliant

    poly(carbonate-urea)urethane vascular graft with effect of

    physiological shear stress. Eur J Vasc Endovasc Surg

    2000;20:34252.

    [50] Salacinski HJ, Punshon G, Krijgsman B, Hamilton G, Seifalian

    AM. A hybrid compliant vascular graft seeded with micro-

    vascular endothelial cells extracted from human omentum. Artif

    Organs 2001;25:97482.

    [52] Rashid ST, Salacinski HJ, Hamilton G, Seifalian AM. The use of

    animal models in developing the discipline of cardiovascular

    tissue engineering: a review. Biomaterials 2004;25:162737.

    [53] Xue L, Greisler HP. Biomaterials in the development and future

    of vascular grafts. J Vasc Surg 2003;37:47280.

    [54] Massia SP, Stark J. Immobilized RGD peptides on surface-

    grafted dextran promote biospecic cell attachment. J Biomed

    Mater Res 2001;56:3909.

    [55] Merzkirch C, Davies N, Zilla P. Engineering of vascular

    ingrowth matrices: are protein domains an alternative to

    peptides? Anat Rec 2001;263:37987.

    [56] Tiwari A, Kidane A, Salacinski H, Punshon G, Hamilton G,

    Seifalian AM. Improving endothelial cell retention for single

    stage seeding of prosthetic grafts: use of polymer sequences of

    arginine-glycine-aspartate. Eur J Vasc Endovasc Surg

    2003;25:3259.

    [57] Krijgsman B, Seifalian AM, Salacinski HJ, Tai NR, Punshon G,

    Fuller BJ, Hamilton G. An assessment of covalent grafting of

    RGD peptides to the surface of a compliant poly(carbonate-

    urea)urethane vascular conduit versus conventional biological

    coatings: its role in enhancing cellular retention. Tissue Eng

    2002;8:67380.

    [58] Iurlaro M, Sanders JE, Zhu WH, Scatena M, Mitchell SB,

    Nicosia RF. Use of vascular explants for ex vivo neovascular-

    ization of biomaterials. Microvasc Res 2002;64:398404.

    [59] Matsuda T, Akutsu T, Kira K, Matsumoto H. Development of

    hybrid compliant graft: rapid preparative method for recon-

    struction of a vascular wall. ASAIO Trans 1989;35:5535.

    [60] Thomas AC, Campbell GR, Campbell JH. Advances in vascular

    tissue engineering. Cardiovasc Pathol 2003;12:2716.

    [61] Teebken OE, Bader A, Steinhoff G, Haverich A. A new concept

    for substitutes in vascular surgery. Langenbecks Arch Chir

    Suppl Kongressbd 1998;115:12569.

    [62] Cooper DK. Xenoantigens and xenoantibodies. Xenotransplan-

    tation 1998;5:617.

    [63] Wilson GJ, Courtman DW, Klement P, Lee JM, Yeger H.

    Acellular matrix: a biomaterials approach for coronary artery

    bypass and heart valve replacement. Ann Thorac Surg

    1995;60:S3538.

    [64] Redmond EM, Cahill PA, Sitzmann JV. Perfused transcapillary

    smooth muscle and endothelial cell co-culturea novel in vitro

    model. In Vitro Cell Dev Biol Anim 1995;31:6019.

    [65] LHeureux N, Paquet S, Labbe R, Germain L, Auger FA. A

    ARTICLER.Y. Kannan et al. / Biom1872completely biological tissue-engineered human blood vessel.

    FASEB J 1998;12:4756.

    [66] Neumann T, Nicholson BS, Sanders JE. Tissue engineering of

    perfused microvessels. Microvasc Res 2003;66:5967.

    [67] Harris NR. Arteriovenous pairing: a determinant of capillary

    exchange. News Physiol Sci 2003;18:837.[68] Kostakoglu N, Manek S, Green CJ. The development of

    neovascularisation in ap perfabrication with vascular implanta-

    tion: an experimental study. Br J Plast Surg 1997;50:42834.

    [69] Ouyang T, Guo E, Zhang M. Experimental study on vascular-

    ized area of the prefabricated axial skin ap after implantation of

    vascular bundles. Zhonghua Zheng Xing Shao Shang Wai Ke Za

    Zhi 1996;12:3269.

    [70] Karatas O, Atabey A, Demirdover C, Barutcu A. Delayed

    prefabricated arterial composite venous aps: an experimental

    study in rabbits. Ann Plast Surg 2000;44:4452.

    [71] Can Z, Apaydin I, Ercocen AR, Demirseren ME, Sabuncuoglu

    B. Prefabrication of a high-density porous polyethylene implant

    using a vascular induction technique. Ann Plast Surg

    1998;41:2649.

    [72] Tanaka Y, Sung KC, Tsutsumi A, Ohba S, Ueda K, Morrison

    WA. Tissue engineering skin aps: which vascular carrier,

    arteriovenous shunt loop or arteriovenous bundle, has more

    potential for angiogenesis and tissue generation? Plast Reconstr

    Surg 2003;112:163644.

    [73] Badylak SF. The extracellular matrix as a scaffold for tissue

    reconstruction. Semin Cell Dev Biol 2002;13:37783.

    [74] Chung S, Hazen A, Levine JP, Baux G, Olivier WA, Yee HT,

    Margiotta MS, Karp NS, Gurtner GC. Vascularized acellular

    dermal matrix island aps for the repair of abdominal muscle

    defects. Plast Reconstr Surg 2003;111:22532.

    [75] Driessen NJ, Peters GW, Huyghe JM, Bouten CV, Baaijens FP.

    Remodelling of continuously distributed collagen bres in soft

    connective tissues. J Biomech 2003;36:11518.

    [76] Nor JE, Peters MC, Christensen JB, Sutorik MM, Linn S, Khan

    MK, Addison CL, Mooney DJ, Polverini PJ. Engineering and

    characterization of functional human microvessels in immuno-

    decient mice. Lab Invest 2001;81:45363.

    [77] Kawada A, Hiura N, Tajima S, Takahara H. Alginate

    oligosaccharides stimulate VEGF-mediated growth and migra-

    tion of human endothelial cells. Arch Dermatol Res

    1999;291:5427.

    [78] Draget KI, Skjak-Braek G, Smidsrod O. Alginate based new

    materials. Int J Biol Macromol 1997;21:4755.

    [79] Elcin YM, Dixit V, Gitnick G. Extensive in vivo angiogenesis

    following controlled release of human vascular endothelial cell

    growth factor: implications for tissue engineering and wound

    healing. Artif Organs 2001;25:55865.

    [80] Kikuchi A, Okano T. Pulsatile drug release control using

    hydrogels. Adv Drug Deliv Rev 2002;54:5377.

    [81] Tanihara M, Suzuki Y, Yamamoto E, Noguchi A, Mizushima

    Y. Sustained release of basic broblast growth factor and

    angiogenesis in a novel covalently crosslinked gel of heparin and

    alginate. J Biomed Mater Res 2001;56:21621.

    [82] Halberstadt C, Austin C, Rowley J, Culberson C, Loebsack A,

    Wyatt S, Coleman S, Blacksten L, Burg K, Mooney D, Holder

    Jr. W. A hydrogel material for plastic and reconstructive

    applications injected into the subcutaneous space of a sheep.

    Tissue Eng 2002;8:30919.

    [83] Marler JJ, Guha A, Rowley J, Koka R, Mooney D, Upton J,

    Vacanti JP. Soft-tissue augmentation with injectable alginate and

    syngeneic broblasts. Plast Reconstr Surg 2000;105:204958.

    [84] Watanabe E, Smith Jr. DM, Delcarpio JB, Sun J, Smart FW,

    Van Jr. MC, ClaycombWC. Cardiomyocyte transplantation in a

    porcine myocardial infarction model. Cell Transplant

    1998;7:23946.

    PRESSals 26 (2005) 18571875[85] Slevin M, Kumar S, Gaffney J. Angiogenic oligosaccharides of

    hyaluronan induce multiple signaling pathways affecting vascu-

    lar endothelial cell mitogenic and wound healing responses. J

    Biol Chem 2002;277:4104659.

    [86] Black AF, Hudon V, Damour O, Germain L, Auger FA. A

    novel approach for studying angiogenesis: a human skin

  • INateriequivalent with a capillary-like network. Cell Biol Toxicol

    1999;15:8190.

    [87] Chupa JM, Foster AM, Sumner SR, Madihally SV, Matthew

    HW. Vascular cell responses to polysaccharide materials: in vitro

    and in vivo evaluations. Biomaterials 2000;21:231522.

    [88] Elcin YM, Dixit V, Gitnick G. Controlled release of endothelial

    cell growth factor from chitosan-albumin microspheres for

    localized angiogenesis: in vitro and in vivo studies. Artif Cells

    Blood Substit Immobil Biotechnol 1996;24:25771.

    [89] Park YD, Tirelli N, Hubbell JA. Photopolymerized hyaluronic

    acid-based hydrogels and interpenetrating networks. Biomater-

    ials 2003;24:893900.

    [90] Shireman PK, Greisler HP. Mitogenicity and release of vascular

    endothelial growth factor with and without heparin from brin

    glue. J Vasc Surg 2000;31:93643.

    [91] Kuberka M, von Heimburg D, Schoof H, Heschel I, Rau G.

    Magnication of the pore size in biodegradable collagen

    sponges. Int J Artif Organs 2002;25:6773.

    [92] Weiser L, Bhargava M, Attia E, Torzilli PA. Effect of serum and

    platelet-derived growth factor on chondrocytes grown in

    collagen gels. Tissue Eng 1999;5:53344.

    [93] Wissink MJ, Beernink R, Poot AA, Engbers GH, Beugeling T,

    van Aken WG, Feijen J. Improved endothelialization of vascular

    grafts by local release of growth factor from heparinized collagen

    matrices. J Control Release 2000;64:10314.

    [94] Montanez E, Casaroli-Marano RP, Vilaro S, Pagan R.

    Comparative study of tube assembly in three-dimensional

    collagen matrix and on Matrigel coats. Angiogenesis

    2002;5:16772.

    [95] Zisch AH, Lutolf MP, Hubbell JA. Biopolymeric delivery

    matrices for angiogenic growth factors. Cardiovasc Pathol

    2003;12:295310.

    [96] Hodde JP, Record RD, Tullius RS, Badylak SF. Retention of

    endothelial cell adherence to porcine-derived extracellular matrix

    after disinfection and sterilization. Tissue Eng 2002;8:22534.

    [97] Hodde J. Naturally occurring scaffolds for soft tissue repair and

    regeneration. Tissue Eng 2002;8:295308.

    [98] Kikuchi A, Okuhara M, Karikusa F, Sakurai Y, Okano T. Two-

    dimensional manipulation of conuently cultured vascular

    endothelial cells using temperature-responsive poly(N-isopropy-

    lacrylamide)-grafted surfaces. J Biomater Sci Polym Ed

    1998;9:133148.

    [99] Chu CC. The in-vitro degradation of poly(glycolic acid)

    sutureseffect of pH. J Biomed Mater Res 1981;15:795804.

    [100] Vert M, Mauduit J, Li S. Biodegradation of PLA/GA polymers:

    increasing complexity. Biomaterials 1994;15:120913.

    [101] Lenza RF, Jones JR, Vasconcelos WL, Hench LL. In vitro

    release kinetics of proteins from bioactive foams. J Biomed

    Mater Res 2003;67A:1219.

    [102] Linn T, Erb D, Schneider D, Kidszun A, Elcin AE, Bretzel RG,

    Elcin YM. Polymers for induction of revascularization in the rat

    fascial ap: application of vascular endothelial growth factor

    and pancreatic islet cells. Cell Transplant 2003;12:76978.

    [103] Pego AP, Siebum B, Van Luyn MJ, Van Seijen XJ, Poot AA,

    Grijpma DW, Feijen J. Preparation of degradable porous

    structures based on 1,3-trimethylene carbonate and D,L-lactide

    (co)polymers for heart tissue engineering. Tissue Eng

    2003;9:98194.

    [104] Pego AP, Van Luyn MJ, Brouwer LA, Van Wachem PB,

    Poot AA, Grijpma DW, Feijen J. In vivo behavior of

    ARTICLER.Y. Kannan et al. / Biompoly(1,3-trimethylene carbonate) and copolymers of

    1,3-trimethylene carbonate with D,L-lactide or epsilon-caprolac-

    tone: Degradation and tissue response. J Biomed Mater Res

    2003;67A:104454.

    [105] Porjazoska A, Kayaman-Apohan N, Karal-Yilmaz O, Cvet-

    kovska M, Baysal K, Baysal BM. Synthesis and characterizationof glycolide, L-lactide, and PDMS-based terpolymers as a

    support for cell cultures. J Biomater Sci Polym Ed

    2002;13:111934.

    [106] Middleton JC, Tipton AJ. Synthetic biodegradable polymers as

    orthopedic devices. Biomaterials 2000;21:233546.

    [107] Pego AP, Poot AA, Grijpma DW, Feijen J. Biodegradable

    elastomeric scaffolds for soft tissue engineering. J Control

    Release 2003;87:6979.

    [108] Peter SJ, Miller ST, Zhu G, Yasko AW, Mikos AG. In vivo

    degradation of a poly(propylene fumarate)/beta-tricalcium

    phosphate injectable composite scaffold. J Biomed Mater Res

    1998;41:17.

    [109] Peter SJ, Yaszemski MJ, Suggs LJ, Payne RG, Langer R, Hayes

    WC, Unroe MR, Alemany LB, Engel PS, Mikos AG.

    Characterization of partially saturated poly(propylene fumarate)

    for orthopaedic application. J Biomater Sci Polym Ed

    1997;8:893904.

    [110] Attawia MA, Uhrich KE, Botchwey E, Fan M, Langer R,

    Laurencin CT. Cytotoxicity testing of poly(anhydride-co-imides)

    for orthopedic applications. J Biomed Mater Res

    1995;29:123340.

    [111] Erdmann L, Macedo B, Uhrich KE. Degradable poly(anhydride

    ester) implants: effects of localized salicylic acid release on bone.

    Biomaterials 2000;21:250712.

    [112] Laurencin CT, Pierre-Jacques HM, Langer R. Toxicology and

    biocompatibility considerations in the evaluation of polymeric

    materials for biomedical applications. Clin Lab Med

    1990;10:54970.

    [113] Gunatillake PA, Adhikari R. Biodegradable synthetic polymers

    for tissue engineering. Eur Cell Mater 2003;5:116.

    [114] Urry DW. Elastic molecular machines in metabolism and soft-

    tissue restoration. Trends Biotechnol 1999;17:24957.

    [115] Aamer KA, Sardinha H, Bhatia SR, Tew GN. Rheological

    studies of PLLAPEOPLLA triblock copolymer hydrogels.

    Biomaterials 2004;25:108793.

    [116] Lutolf MP, Lauer-Fields JL, Schmoekel HG, Metters AT,

    Weber FE, Fields GB, Hubbell JA. Synthetic matrix metallo-

    proteinase-sensitive hydrogels for the conduction of tissue

    regeneration: engineering cell-invasion characteristics. Proc Natl

    Acad Sci USA 2003;100:54138.

    [117] Shin H, Quinten RP, Mikos AG, Jansen JA. In vivo bone and

    soft tissue response to injectable, biodegradable oligo(poly(ethy-

    lene glycol) fumarate) hydrogels. Biomaterials 2003;24:320111.

    [118] Nguyen KT, West JL. Photopolymerizable hydrogels for tissue

    engineering applications. Biomaterials 2002;23:430714.

    [119] Schmedlen RH, Masters KS, West JL. Photocrosslinkable

    polyvinyl alcohol hydrogels that can be modied with cell

    adhesion peptides for use in tissue engineering. Biomaterials

    2002;23:432532.

    [120] Cao X, Shoichet MS. Photoimmobilization of biomolecules

    within a 3-dimensional hydrogel matrix. J Biomater Sci Polym

    Ed 2002;13:62336.

    [121] De Rosa M, Carteni M, Petillo O, Calarco A, Margarucci S,

    Rosso F, De Rosa A, Farina E, Grippo P, Peluso G. Cationic

    polyelectrolyte hydrogel fosters broblast spreading, prolifera-

    tion, and extracellular matrix production: implications for tissue

    engineering. J Cell Physiol 2004;198:13343.

    [122] Ma T, Li Y, Yang ST, Kniss DA. Tissue engineering human

    PRESSals 26 (2005) 18571875 1873placenta trophoblast cells in 3-D brous matrix: spatial effects

    on cell proliferation and function. Biotechnol Prog

    1999;15:71524.

    [123] Li Y, Ma T, Yang ST, Kniss DA. Thermal compression and

    characterization of three-dimensional nonwoven PET matrices

    as tissue engineering scaffolds. Biomaterials 2001;22:60918.

  • INateri[124] Wintermantel E, Mayer J, Blum J, Eckert KL, Luscher P,

    Mathey M. Tissue engineering scaffolds using superstructures.

    Biomaterials 1996;17:8391.

    [125] Vaissiere G, Chevallay B, Herbage D, Damour O. Comparative

    analysis of different collagen-based biomaterials as scaffolds for

    long-term culture of human broblasts. Med Biol Eng Comput

    2000;38:20510.

    [126] Shu XZ, Ghosh K, Liu Y, Palumbo FS, Luo Y, Clark RA,

    Prestwich GD. Attachment and spreading of broblasts on an

    RGD peptide-modied injectable hyaluronan hydrogel. J

    Biomed Mater Res 2004;68A:36575.

    [127] Patrick Jr. CW, Chauvin PB, Hobley J, Reece GP. Preadipocyte

    seeded PLGA scaffolds for adipose tissue engineering. Tissue

    Eng 1999;5:13951.

    [128] Halbleib M, Skurk T, deLuca C, von Heimburg D, Hauner H.

    Tissue engineering of white adipose tissue using hyaluronic acid-

    based scaffolds. I: in vitro differentiation of human adipocyte

    precursor cells on scaffolds. Biomaterials 2003;24:312532.

    [129] Garfein ES, Orgill DP, Pribaz JJ. Clinical applications of tissue

    engineered constructs. Clin Plast Surg 2003;30:48598.

    [130] De Ugarte DA, Ashjian PH, Elbarbary A, Hedrick MH. Future

    of fat as raw material for tissue regeneration. Ann Plast Surg

    2003;50:2159.

    [131] Soker S, Machado M, Atala A. Systems for therapeutic

    angiogenesis in tissue engineering. World J Urol 2000;18:108.

    [132] Sims CD, Butler PE, Cao YL, Casanova R, Randolph MA,

    Black A, Vacanti CA, Yaremchuk MJ. Tissue engineered

    neocartilage using plasma derived polymer substrates and

    chondrocytes. Plast Reconstr Surg 1998;101:15805.

    [133] Brown AN, Kim BS, Alsberg E, Mooney DJ. Combining

    chondrocytes and smooth muscle cells to engineer hybrid soft

    tissue constructs. Tissue Eng 2000;6:297305.

    [134] Masuda H, Asahara T. Post-natal endothelial progenitor cells

    for neovascularization in tissue regeneration. Cardiovasc Res

    2003;58:3908.

    [135] Kalka C, Masuda H, Takahashi T, Kalka-Moll WM, Silver M,

    Kearney M, Li T, Isner JM, Asahara T. Transplantation of ex

    vivo expanded endothelial progenitor cells for therapeutic

    neovascularization. Proc Natl Acad Sci USA 2000;97:

    34227.

    [136] Murohara T, Ikeda H, Duan J, Shintani S, Sasaki K, Eguchi H,

    Onitsuka I, Matsui K, Imaizumi T. Transplanted cord blood-

    derived endothelial precursor cells augment postnatal neovascu-

    larization. J Clin Invest 2000;105:152736.

    [137] von Degenfeld G, Ban A, Springer ML, Blau HM. Myoblast-

    mediated gene transfer for therapeutic angiogenesis and arter-

    iogenesis. Br J Pharmacol 2003;140:6206.

    [138] Tiwari A, Salacinski HJ, Hamilton G, Seifalian AM. Tissue

    engineering of vascular bypass grafts: role of endothelial cell

    extraction. Eur J Vasc Endovasc Surg 2001;21:193201.

    [139] Grenier G, Remy-Zolghadri M, Guignard R, Bergeron F, Labbe

    R, Auger FA, Germain L. Isolation and culture of the three

    vascular cell types from a small vein biopsy sample. In Vitro Cell

    Dev Biol Anim 2003;39:1319.

    [140] Hershey JC, Baskin EP, Glass JD, Hartman HA, Gilberto DB,

    Rogers IT, Cook JJ. Revascularization in the rabbit hindlimb:

    dissociation between capillary sprouting and arteriogenesis.

    Cardiovasc Res 2001;49:61825.

    [141] Hudon V, Berthod F, Black AF, Damour O, Germain L, Auger

    FA. A tissue-engineered endothelialized dermis to study the

    ARTICLER.Y. Kannan et al. / Biom1874modulation of angiogenic and angiostatic molecules on capil-

    lary-like tube formation in vitro. Br J Dermatol 2003;

    148:1094104.

    [142] Black AF, Berthod F, LHeureux N, Germain L, Auger FA. In

    vitro reconstruction of a human capillary-like network in a

    tissue-engineered skin equivalent. FASEB J 1998;12:133140.[143] Moldovan NI, Goldschmidt-Clermont PJ, Parker-Thornburg J,

    Shapiro SD, Kolattukudy PE. Contribution of monocytes/

    macrophages to compensatory neovascularization: the drilling

    of metalloelastase-positive tunnels in ischemic myocardium. Circ

    Res 2000;87:37884.

    [144]