Experimental Approaches to Constructs

Embed Size (px)

Citation preview

  • 8/17/2019 Experimental Approaches to Constructs

    1/53

    Full Terms & Conditions of access and use can be found athttp://www.tandfonline.com/action/journalInformation?journalCode=tbsp20

    Download by: [98.179.203.196] Date: 10 March 2016, At: 23:07

     Journal of Biomaterials Science, Polymer Edition

    ISSN: 0920-5063 (Print) 1568-5624 (Online) Journal homepage: http://www.tandfonline.com/loi/tbsp20

    Experimental approaches to vascularisation withintissue engineering constructs

    Md. Sarker, X.B. Chen & D.J. Schreyer

    To cite this article: Md. Sarker, X.B. Chen & D.J. Schreyer (2015) Experimental approaches to

    vascularisation within tissue engineering constructs, Journal of Biomaterials Science, PolymerEdition, 26:12, 683-734, DOI: 10.1080/09205063.2015.1059018

    To link to this article: http://dx.doi.org/10.1080/09205063.2015.1059018

    Accepted author version posted online: 08 Jun 2015.

    Submit your article to this journal

    Article views: 232

    View related articles

    View Crossmark data

    http://crossmark.crossref.org/dialog/?doi=10.1080/09205063.2015.1059018&domain=pdf&date_stamp=2015-06-08http://crossmark.crossref.org/dialog/?doi=10.1080/09205063.2015.1059018&domain=pdf&date_stamp=2015-06-08http://www.tandfonline.com/doi/mlt/10.1080/09205063.2015.1059018http://www.tandfonline.com/doi/mlt/10.1080/09205063.2015.1059018http://www.tandfonline.com/action/authorSubmission?journalCode=tbsp20&page=instructionshttp://www.tandfonline.com/action/authorSubmission?journalCode=tbsp20&page=instructionshttp://dx.doi.org/10.1080/09205063.2015.1059018http://www.tandfonline.com/action/showCitFormats?doi=10.1080/09205063.2015.1059018http://www.tandfonline.com/loi/tbsp20http://www.tandfonline.com/action/journalInformation?journalCode=tbsp20

  • 8/17/2019 Experimental Approaches to Constructs

    2/53

    REVIEW ARTICLE

    Experimental approaches to vascularisation within tissue engineeringconstructs

    Md. Sarker a *, X.B. Chena ,c and D.J. Schreyer a , b

    a Division of Biomedical Engineering, College of Engineering, University of Saskatchewan, 1A26,57 Campus Drive, Saskatoon, SK, S7N 5A9, Canada;   b Department of Anatomy and Cell Biology,College of Medicine, University of Saskatchewan, 5800 Saskatoon City Hospital, Saskatoon, SK,S7K 0M7, Canada;   c Department of Mechanical Engineering, College of Engineering, University

    of Saskatchewan, 3B47, 57 Campus Drive, Saskatoon, SK, S7N 5A9, Canada

    ( Received 26 March 2015; accepted 2 June 2015)

    Tissue engineering opens up a new area to restore the function of damaged tissue or replace a defective organ. Common strategies in tissue engineering to repair andform new tissue containing a functional vascular network include the use of cells,growth factors, extracellular matrix proteins, and biophysical stimuli. Yet, formationof well-distributed, interconnected, and stable vascular network still remainschallenging. In addition, anastomoses with host vasculature upon implantation andlong-time survival of the new blood vessel   in vivo   are other critical issues to beaddressed. This paper presents a brief review of recent advances in vascularizationin vitro   as well as   in vivo   for tissue engineering, along with suggestions for futureresearch.

    Keywords:   vascularization; angiogenesis; vasculogenesis; biomaterial; growthfactors; scaffold

    1. Introduction

    A major challenge in the creation of new tissues by tissue engineering is the limitation

    of diffusional mass transfer into new tissue having a thickness of more than

    100 – 200 μm,[1] causing ischemia and cell death. Vascularization overcomes the limit 

    of diffusional mass transfer for delivery of various types of nutrients, oxygen, growth

    factors, biochemical signaling factors, and removal of carbon dioxide, and metabolic

    waste.[2] The 100 – 200 μm limit regulates the critical distance necessary between two

    successive capillaries to prevent an ischemic condition.[3] This issue must beconsidered in any experimental or therapeutic effort to regenerate functional tissue of 

    appreciable size using a tissue engineering approach.

    Two mechanisms are responsible for the generation of a vascular network   in vivo:

    vasculogenesis and angiogenesis. Vasculogenesis generally takes place at an embryonic

    stage, while angiogenesis occurs mostly in postnatal life.[4,5] The term vasculogenesis

    denotes the assembly of endothelial progenitor cells (EPCs) that eventually form capil-

    laries and a vascular network [6] in response to various growth factors, then undergoes

    remodeling to form functional vasculature. Angiogenesis refers to the sprouting of new

     blood vessels from existing ones due to specic angiogenic signals.[4] Engineered

    *Corresponding author. Email: [email protected]

    © 2015 Taylor & Francis

     Journal of Biomaterials Science, Polymer Edition, 2015

    Vol. 26, No. 12, 683 – 734, http://dx.doi.org/10.1080/09205063.2015.1059018

    mailto:[email protected]://dx.doi.org/10.1080/09205063.2015.1059018http://dx.doi.org/10.1080/09205063.2015.1059018mailto:[email protected]

  • 8/17/2019 Experimental Approaches to Constructs

    3/53

    tissue can be vascularized by either process, but must ultimately result in anastomosis

    with host vasculature.

    2. Different types of blood vessels and their functions

    The vascular system is a geometrically complex network consisting of arteries that 

    supply blood to tissues, and veins that remove it (Figure   1). Major arteries branch out 

    into successively smaller arterioles, metarterioles, and arterial capillaries. Capillaries

    combine downstream to form venules, and then major veins.

    Arterioles typically have diameters in the range of 10 – 300 μm and are composed of 

    concentric layers of smooth muscle cells (SMC), basement membrane and endothelial

    cells (ECs).[7] A metarteriole is a short vessel which branches out from an arteriole

    with a smaller diameter. Generally, a metarteriole consists of only two layers; SMCs

    surrounding ECs. At the point where capillaries branch out from metarterioles, there

    exists a loop of SMCs called the precapillary sphincter. The function of precapillary

    sphincter is to control the blood   ow to the capillary bed and to ensure enough time tofacilitate diffusion and other mass transfer mechanism between capillaries and nearby

    cells. To keep the blood  ow continuous, a bypass vessel called the thoroughfare chan-

    nel connects the metarteriole with the venule. The capillaries consist only of ECs hav-

    ing an outer layer of protein-based basement membrane. However, pericyte cells, which

    are responsible for paracrine signaling and capillary blood   ow, can be found within

    the capillary basement membrane. The capillaries are also known as microvessels, and

    have a diameter of 4 – 10 μm. The transition from capillary to venule takes place gradu-

    ally and the approximate diameter of post-capillary venule is 10 – 50 μm. Post-capillary

    venules then fuse with a connecting venule whose diameter is 50 – 300 μm.[8]

    Capillaries are the principle site of mass transfer. The difference between the pressure of blood within the capillaries and the interstitial   uid pressure causes

    the movement of  uid into or out of capillaries. Generally, three types of capillaries are

    Figure 1. Blood circulatory system inside the body (Reproduced from [288]).

    684   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    4/53

    seen in the body: continuous, fenestrated and sinusoidal (Figure   2). Most capillarieswithin the body are the continuous type. These have tight junctions as well as inter-

    cellular clefts between the ECs, and are covered with basement membrane. Water, gas

    molecules, ions, and other water soluble molecules can pass in or out of continuous

    capillaries through diffusion, vesicles, and intercellular clefts. However, high molecular 

    weight molecules are transported across ECs by pinocytosis. Fenestrated capillaries

    have 60 – 80 nm pores in their ECs and are covered with basement membrane.

    Fenestrated capillaries carry out mass transfer through diffusion, vesicles, intercellular 

    clefts, and pores. This type of capillary is found within the endocrine glands, intestines,

     pancreas, and kidneys. Sinusoidal capillaries are leaky in nature and have 30 – 40 μm

     pores, much larger than those seen in fenestrated capillaries. Sinusoidal capillaries have

    incomplete basement membrane covering and more intercellular clefts compare to other 

    capillaries. White and red blood cell and serum protein can pass through the walls of 

    sinusoidal capillaries. Generally, sinusoidal capillaries are found in bone marrow, liver,

    spleen, and adrenal gland.[9]

    3. Mechanisms of blood vessel formation  in vivo

    3.1. Vasculogenesis

    EPCs from bone marrow or blood are mainly responsible for pre- and post-natal vascu-

    logenesis through a complex series of steps (Figure  3). At the initial stage, EPCs move

    from their source and enter unvascularized tissue in response to chemo-attractant fac-

    tors, such as granulocyte – monocyte colony stimulating factor, granulocyte colony

    stimulating factor, vascular endothelial growth factor (VEGF), basic    broblast growth

    factor (bFGF), placental growth factor, erythropoietin and stromal cell-derived factor-1

    (SDF-1).[10] Moreover, factors secreted from ischemic tissue cause nitric oxide (NO)

     production that activates extracellular proteases, especially matrix metalloproteases-9

    (MMP-9), and facilitates EPC migration.[11]

    Homing of EPCs into ischemic tissue site requires chemotaxis, adhesion, and trans-

    endothelial migration. After moving out from bone marrow traveling through blood

    vessels, EPCs undergo chemotaxis toward a particular ischemic region in response to

    gradients of chemokines, such as SDF-1, interleukin-8 (IL-8), growth-regulated onco-gene-α, and C – C chemokine.[12,13] The chemokines also stimulate EPCs to promote

    adhesion with the inner layer of blood vessels. EPCs pass through the endothelial lining

    Figure 2. Structure of different types of capillaries (Reproduced from [288]).

     Journal of Biomaterials Science, Polymer Edition   685

  • 8/17/2019 Experimental Approaches to Constructs

    5/53

    of blood vessels by a specic transendothelial migration mechanism. Once at the base-

    ment membrane, EPCs initiate their invasion mechanism by rupturing it and remodeling

     basement membrane proteins, as well as the extracellular matrix (ECM) of the intersti-

    tial space, using extracellular proteases, such as MMP-9, cathepsin L, urokinase-type

     plasminogen activator, and tissue-type plasminogen activator, to reach the ischemic tis-

    sue site.[14,15] Then EPCs accumulate at the ischemic site; form a vascular pattern bydifferentiating into ECs and interacting with existing ECs and ECM. EPCs attached to

    the ECM proliferate under the inuence of VEGF, immunoglobulin, and epidermal

    growth factor (EGF),[16,17] and differentiate due to monocyte chemoattractant protein-

    1, insulin-like growth factor-1 (IGF-1), SDF-1, VEGF, and platelet-derived growth

    factor (PDGF).[18,19]

    3.2. Angiogenesis

    During angiogenesis, new capillaries sprout from existing blood vessels in response to

    various biochemical signals and expand into ischemic tissue following tracks, gradients,or attractive or repulsive signals. Any unnecessary vascular plexus caused by random

    sprouting undergoes remodeling according to the requirements of specic tissue.

    Figure 3. Blood vessel formation by vasculogenesis (Reproduced from [289]).

    686   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    6/53

    Functional non-uniformity of ECs along the blood vessel wall caused by VEGF

    gradients plays a critical role in angiogenic sprouting. Mechanistically, specic ECs

    respond to a chemo attractant by extending as tip cell, then propagating in different 

    directions by forming stalk-like structure, which eventually acquire a lumen.[20,21] To

    communicate with the surroundings, tip cells extend multiple  lopoida which are sensi-

    tive to various guidance molecules and specic growth factors, such as VEGF-A, FGF,

    angiopoietin-1 (Ang-1), EGF, and IL-8. The spatial as well as temporal distributions of 

    these factors are very crucial for guiding the growth of new capillaries within the

    ischemic tissue.[22] For example, a VEGF-A gradient in tissue plays an important role

    for  lopodia extension from the tip cell, and the corresponding receptor for VEGF-A is

    VEGFR-2, which is expressed by the tip cell.[23] Angiogenesis in ischemic or newly

    formed tissue is a complicated process which can be demonstrated through some steps

    (Figure 4).

    At the beginning, ischemic tissue secretes VEGF which dilates the existing blood

    vessel through the redistribution of intercellular adhesion molecules as well as mod-

    ication of the cell membrane structure. It also increases vascular permeability with thehelp of NO [24,25] and assists the relocation of plasma protein to form temporary

    structures to facilitate EC migration. Besides VEGF, Ang-2 contributes to angiogenic

    sprouting through the degradation of ECM protein and the removal of SMCs from the

    outer surfaces of capillaries.[25,26] Degradation of basement membrane and ECM are

    vital for EC migration, while microvascular ECs themselves secrete some matrix met-

    alloproteases, such as MMP-2, MMP-3, MMP-9, along with protease inhibitor mainly

    tissue inhibitor, of metalloproteinase-2, which coordinately regulate basement mem-

     brane and ECM degradation [27] by causing various spatial and temporal factors to

    Figure 4. Blood vessel formation by angiogenesis in brain cancer (Reproduced from [290]).

     Journal of Biomaterials Science, Polymer Edition   687

  • 8/17/2019 Experimental Approaches to Constructs

    7/53

    generate non-uniformity along the wall of blood vessel.[28,29] In addition, another 

    important feature of extracellular proteolytic degradation is the release of some growth

    factors, such as IGF-1, VEGF, and FGF-2, which also facilitate angiogenic

    sprouting.[30]

    The progression step is associated with the migration and proliferation of ECs due

    to the inuence of VEGF, angiopoietins (Angs), FGFs, and PDGF. Among them, Ang-

    1 regulates the interaction between ECs and periendothelial cells during migration,

    [25,31] FGF facilitates EC growth via the recruitment of mesenchymal cells,[ 32] and

    PDGF enhances EC growth through the recruitment of pericytes and SMCs.[33] In

    addition, other factors involved in EC proliferation and migration include neuropep-

    tides, IGF-1, erythropoietin, hepatocyte, and interleukins.[34,35]

    After migration and proliferation of EC into ECM, ECs eventually differentiate to

    form cord-like structure and a lumen starts developing due to the inuence of VEGF

    and Ang. Interestingly, Ang-1, VEGF121,   and VEGF165   help to increase lumen

    diameter, while VEGF189   decreases lumen diameter.[36]

    4. Tissue engineering approaches to blood vessel formation

    Well-distributed and interconnected vascular network is required to maintain the viabil-

    ity of large cell population in engineered tissue. While tissue engineering is eventually

    emerging as a potential approach to regenerate tissue and organ, formation of 3D

    vascular network within the neotissue still remains challenging. Current strategies to

    form complex vascular channel within engineering construct or growing tissue can be

    categorized as scaffold-based and scaffold-free approaches (Figure   5). Strategies

    (Table 1) associated with scaffold-based approaches are, construction of 3D matrix with

    appropriate biomaterial by biofabrication technique or stacking multiple micropat-terened thin planar surfaces or micro tissue modules, engineering new tissue with

    decellularized matrix, functionalization of scaffolds with cells, angiogenic factors (AFs)

    or proteins, transfection of seeded cells by gene therapy to obtain sustained release of 

    AFs and prevascularisation to maintain viability of the seeded cells. In contrast, in scaf-

    fold-free approach cell sheets are assembled to promote vascularized tissue.

    4.1. Scaffold-based aproaches

    4.1.1. Scaffold fabrication biomaterial 

    The choice of matrix biomaterial has profound impact on scaffold vascularization in tis-

    sue engineering. Like natural ECM, the biomaterial should facilitate vascular cell – 

     biomaterial interactions, which would activate numerous signaling pathways to promote

    vascular cell survival, proliferation, differentiation, and migration. A tissue scaffold

    experiences different types of force upon implantation   in vivo, such as compression,

    shear, torsion, and tensile force. Therefore, the biomaterial should have suf cient 

    mechanical strength to sustain those forces to keep the scaffold architecture intact.

    Moreover, controlled biodegradability is desired so that over time, the scaffold could

    maintain its mechanical strength while the biomaterial is replaced with a vascular net-

    work and newly formed tissue. Furthermore, cytotoxicity has to be considered for its

    impact on implanted or host cell survival   in vivo   or during preparation   in vitro.

    Generally, polymers are used as biomaterials for the fabrication of scaffolds.[37]Biopolymers can be classied into three categories in terms of preparation or source:

    synthetic, natural, and hybrid.

    688   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    8/53

    4.1.1.1. Synthetic polymer.   Synthetic polymers used for tissue engineering offer 

    scaffold fabrication   exibility while maintaining desired mechanical properties of the

    engineering construct. However, lack of cell adhesion peptides in the molecular struc-

    ture of synthetic materials limits their application for vascular tissue engineering. A

    number of biodegradable, biocompatible, and non-toxic synthetic polymers have shown

     potential for tissue engineering applications. Among them, polyglycolic acid (PGA),

     polylactic acid (PLA), polylactic-co-glycolic acid (PLGA), poly-L-lactic acid (PLLA),

     poly-ε-caprolactone (PCL), polyethylene glycol (PEG), and polyhydroxyalkanoates

    (PHAs) have frequently been explored in different studies of tissue-engineered graft 

    vascularization.[38]

    When exposed to pulsatile conditions, PGA mesh tube scaffolds seeded with

     porcine carotid SMCs were eventually replaced by blood vessel-like structures while

    maintaining uniform SMCs distribution and collagen content close to that of native ves-

    sels.[39]   In vivo   experiments have also shown PGA scaffolds as promoters of vascular-

    ization. PGA scaffolds seeded with bovine artery cells degraded completely after 

    11 weeks of implantation in the pulmonary artery of sheep and were replaced by new

    tissue having 73.9% collagen content with respect to native tissue.[40] Similarly, in an

    in vivo   study, PEG diacrylate hydrogel was implantated subfascially into Sprague-

    Dawley rats and kept   in vivo   for three weeks. Interestingly, even in the absence of 

    incorporated growth factors, vascularized tissue ingrowth was reported throughout the

    entire PEG gels having pore size 50 – 150 μm.[41]Derivatives of PLA (polymer of lactide) have also shown their angiogenic potential

    in different studies. For example , in vivo   implantation of tubular scaffold fabricated

    Figure 5. Different approaches for vascular network formation.

     Journal of Biomaterials Science, Polymer Edition   689

  • 8/17/2019 Experimental Approaches to Constructs

    9/53

    with porous   lms of poly-D,   L-lactic-co-glycolic acid facilitated the ingrowth of 

     brovascular tissue, which eventually formed a vascularized, tubular tissue.[42] To

    investigate the combined effect of PLLA and collagen gel on vascularization, porous

    PLLA scaffold was exposed to   in vitro   condition, where the pores were   lled with the

    mixture of human aortic SMCs and collagen gel, resulting in the rapid formation of a

    smooth inner layer around the vascular graft.[43] Another composite polymer of lactidePLGA was explored to study the release prole of VEGF. VEGF was directly incorpo-

    rated into PLGA scaffolds and pre-encapsulated in PLGA microspheres. It was reported

    that direct incorporation of VEGF resulted in 40 – 60% release within   ve days   in vitro,

    while encapsulation of VEGF within microspheres resulted in prolonged release. When

    implanted into subcutaneous pockets of SCID mice, VEGF released from PLGA

    scaffolds signicantly enhanced local angiogenesis.[44]

    Like PLA, composites and derivatives of PCL have been identied as a vasculature

    enhancer in several studies. In a comprehensive review, Woodruff et al. presented the

    attractive features of PCL (e.g. inexpensive, FDA-approved, and superior rheological

    and viscoelastic properties) to demonstrate its applicability for the fabrication of longer 

    term degradable implants suitable for a specic anatomical site.[45]   In vivo   implanta-

    tion of PCL/PLA tubular scaffolds seeded with mixed cells obtained from femoral

    Table 1. Tissue engineering approaches to blood vessel formation.

    Approaches Description

    A. Scaffold based approachA.1. Scaffold fabrication

     biomaterial

    Synthetic polymers (e.g. PGA, PLA, PLGA, PLLA, PCL, PEG,

    and PHAs); natural polymers (e.g. collagen,    bronectin,   brin,elastin, silk   broin, HA, alginate, chitosan); decellularized matrix(e.g. allogenic or xenogenic tissue or organ graft)

    A.2. 3D printingapproaches

    3D Fabrication (e.g. 3D plotting, inkjet printing, laser-based biofabrication and stereolithography) technique to fabricatecomplex vascular network; microfabrication (e.g. microuidics,micromolding, lithographic, direct laser write) technique to indent micro pattern on polymer or cell seeded hydrogel to allowcomplex network formation at micro level; modular assembly of 

     patterned micro modules generated from self-assembledaggregation, microfabrication of cell seeded hydrogels, or direct 

     printing method to form macro tissue assembled by random

     packing, stacking of layers or directed assemblyA.3. Addition of cells Autologus mature ECs (e.g. human umbilical vein, human dermalmicrovascular, human vascular), stem cells (e.g. embryonic stemcell, MSC), EPCs (e.g. peripheral blood derived, umbilical cord

     blood derived), vascular wall cells (e.g. pericytes, vascular SMCs, pulmonary artery SMCs)

    A.4. Addition of growthfactors

    VEGF, FGFs, PDGF, Angs, TGF-β

    A.5. Gene therapy Viral vectors (e.g. retrovirus, lentivirus, adeno-virus and adeno-associated virus) and nonviral vectors (e.g. plasmid DNA)

    A.6. Prevascularisation   In vitro  or   in vivo  culture prior to implant to form vasculature andmaintain cell viability

    B. Scaffold free approachB.1. Cell sheet technique Temperature controlled cell attachment, culture and detachment 

    followed by stacking multiple layers

    690   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    10/53

    veins of a mongrel dog showed no evidence of stenosis, thrombus, occlusion, or 

    aneurysmal formation within the tubular graft. Moreover, the vascular graft was

    replaced by native tissue containing a large amount of ECM, where the ECs were

    found to organize themselves in a linear way on the luminal surface of scaffold.[46]

    Besides animal model study, in May 2000 a clinical trial was done on a four-year-old

    girl to implant a pulmonary bypass graft made of PCL/PLA copolymer seeded with

    autologous vein cells. This implantation was a milestone in pediatric cardiovascular sur-

    gery as the patient was doing well even after seven months of implantation and no

     postoperative complications were reported.[47] Another derivative of PCL called poly

    (lactide-co-caprolactone) (PLCL) was studied for tissue vascularization due to its bio-

    compatibility, elasticity, and slow degradation property. In a study, PLCL vascular 

    grafts seeded with rabbit aortic SMCs were exposed to pulsatile   ow in a perfusion

     bioreactor for eight weeks. It was reported that the PLCL graft signicantly regulated

    the proliferation of SMCs, increased collagen production, and promoted cell alignment 

    similar to that of native vascular smooth muscle tissues.[48]

    The angiogenic potential of PHA derivatives and its composites were demonstratedin different investigations. In a study, PHA – PGA tubular scaffolds seeded with ovine

    carotid artery cells were implanted into abdominal aortic segments of sheep for   ve

    months. No aneurysms were observed in the tubular graft; however, the percentage of 

    collagen, DNA content, as well as the mechanical strength of grown tissue, reached

    close to that of native vessels.[49] Another research group maintained static/pulsatile

    condition for PGA/PHA derivative-fabricated vascular grafts seeded with ovine vascular 

    myo broblasts and ECs. After 28 days of culture, it was reported that all samples

    gained viable, dense, and conuent smooth tissue with high collagen content.[50]

    4.1.1.2. Natural polymer.   Biopolymer or naturally derived polymers can be classiedinto two categories, such as; protein-based polymers and polysaccharidic polymers. In

    tissue engineering, natural polymers are more advantageous over synthetic polymers in

    many aspects, however, are not free from shortcomings.Some advantages and

    drawbacks of natural polymers have been presented in Table  2.

    Protein-based polymers, particularly collagen,    brin,    bronectin, elastin, and silk 

     broin, have frequently been explored in different studies for vascularization applica-

    tion. For example, it has been reported that freeze-dried collagen scaffold seeded with

    dermal    broblasts and bone marrow-derived stem cells enhance the proliferation of 

    EC.[51] Further, immobilized VEGF on collagen scaffolds promotes EC invasion and

     proliferation.[52] Another group incorporated modied VEGF into collagen scaffolds

    implanted subcutaneously into mice, resulting in an improved vascular network within

    the implanted scaffold.[53] Besides, Fibrin tissue engineering scaffolds have also pre-

    sented promising results for promoting blood vessel formation. Human microvascular 

    ECs were seeded into a 90%    brin, 10% collagen matrix with combinations of added

    growth factors; FGF-2 or VEGF, with TNF-α.[54] This investigation suggests that    b-

    rin matrix facilitates the formation of a vascular capillary network through matrix    bri-

    nolysis by ECs, which occurs from the activities of plasmin and metalloproteinases. In

    addition to providing specic cell adhesion sequences, a    brin scaffold can store and

    release angiogenic growth factors, such as VEGF-A165   and FGF-2, as well as plasmid

    DNA to improve angiogenesis.[55] Another ECM protein    bronectin has been consid-

    ered as a vasculature promoter due to its enormous contribution in vascularization. In astudy,    bronectin-coated collagen modules increased human vascular EC survival and

    vessel formation when implanted in immunodecient mice compared to collagen

     Journal of Biomaterials Science, Polymer Edition   691

  • 8/17/2019 Experimental Approaches to Constructs

    11/53

    Table 2. Natural biomaterials for scaffold fabrication.

    Biomaterial Advantages Disadvanages

    Collagen (1) Biocompatible, biodegradable, ease

    availability and modiability,nonantigenic and controlled

     protein release ability.[291](2) Hydrophilic and enhance cell

    interactions.[292]

    (1) Poor mechanical strength,[293] deformability,

    exibility and tensilestrength.[294]

    (2) High cost of purication,handling complexity,[295]rapid degradation, littlecompression forceresistivity.[296]

    (3) Sterilization causes alterationof protein structure.[291]

    Hyaluronicacid

    (1) Easily modiable,hydrophilic, nonadhesive,

     biodegradable,[297]nonantigenic and non-inammatory.[298]

    (2) Regulate cellular migration,interaction, anddifferentiation.[299]

    (1) Requires purication to avoiddisease transmission.[300]

    (2) Rapid degradation rate, poor mechanical strength,[301]and forms scar tissuein vivo.[294]

    Fibrin   (1) Non toxic, non inammatory,[302] non allergenic,[303]non immunogenic[304],economical,[305]

     biodegradable [306] and

    easily processable.[307](2) Facilitates synthesis of collagen [308] and promotesangiogenesis.[309]

    (3) Enhances attachment,migration and proliferation of smooth muscle [310] andECs.[311]

    (1) Inconsistency in polymerization, fragile andstress squeezable.

    (2) Structural weakness.[312](3) Upon implantation, solubility

    increases and structuralintegrity decreases over time.[313]

    Fibronectin   (1) Critical role for angiogenesis[314] and scaffoldvascularisation.[315]

    (2) Facilitates cell adhesion,[316]migration,[317] proliferationand alignment.[318]

    (3) Can be used to coat articial biomaterials to support celladhesion, spreading [319] and

     proliferation.[320]

    (1) High cost.[321](2) Excessive mechanical stress

    interrupts formation of 

    vasculature.[322]

    Alginate   (1) Non inammatory,[321] biocompatible, non toxic,cheap and potential for cellencapsulation and drugdelivery.[323,324]

    (1) Uncontrollable degradationkinetics due to loss of divalent ion and burst releaseof protein drugs at higher 

     pH.[325]

    (Continued )

    692   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    12/53

    modules without     bronectin coating.[56] Similarly, implantation of a    bronectin/ 

    collagen I gel seeded with EC and mesenchymal cell facilitated the formation of well-

     perfused and long-lasting vascular network after implantation into mice.[57]

    The role of structural protein elastin in vascularization is very signicant as geneti-cally modied mice lacking elastin died within four days of birth from arterial obstruc-

    tion resulting from subendothelial cell proliferation and reorganization of smooth

    muscle into the lumen of artery.[58] Cross-linked soluble alpha-elastin discs were found

    to facilitate vascular SMC adhesion,[59] whereas tropoelastin-coated surface was

    reported as a promoter of EC attachment and proliferation.[60] Elastin composites have

    also been investigated to vascularize tissue engineering grafts. For instance, in a study,

    human coronary artery SMCs were cultured for seven days on aligned nano brous

     polyurethane scaffolds blended with the mixture of elastin and collagen. It was con-

    cluded that the growth of SMCs on elastin/collagen-blended scaffold increased by

    224% compared to that of polyurethane.[61] Besides animal body-derived protein, silk-

    worm-secreted protein silk    broin showed promising results in several studies in tissue

    vascularization. In a study, co-culture of human dermal microvascular endothelial cells

    (HDMECs) and primary human osteoblast cells (HOCs) in 3D silk    broin nets caused

    ECs to form intertwined microcapillary-like structures.[62] In another study, 3D silk 

     broin scaffolds pre-cultured with HDMEC and HOC were implanted into

    immunodecient mice. After 14 days, it was found that the microcapillary structures

     pre-formed   in vitro, perfused well with the host vasculature.[63]

    Apart from protein-based polymers, polysaccharide polymers, mostly hyaluronic

    acid (HA), alginate, and chitosan have been studied for tissue vascularization. For 

    example, subcutaneously injected HA combined with recombinant gelatin facilitates the

    formation of vascular network as well as the deposition of ECM components inrats.[64] Another group achieved controlled release of two angiogenic growth factors

    (VEGF165   and PDGF-BB) from a hybrid mesh consisting of poly (3-caprolactone)-

    collagen blend and HA hydrogel. They reported that the growth factor-loaded hybrid

    mesh enhances cellular attachment and formation of vascular capillary network within

    the tissue engineering construct during the culture of ECs and    broblasts in a 3D

    model.[65] However, HA has limitations in tissue engineering applications as it exhibits

     poor cell attachment and mechanical properties.

    Seaweed-derived polysaccharide alginate does not have EC adhesion properties, but 

    can be introduced through adsorption of proteins,[66] or by covalent incorporation of 

    different functional groups into the sugar backbone.[67] To facilitate vascularizationwithin alginate scaffolds, internal pores should be well interconnected and large enough

    to allow ingrowth of blood vessels upon implantation   in vivo.[68] For example, an

    Table 2. (Continued ).

    Biomaterial Advantages Disadvanages

    (2) pH sensitive behavior facilitates drug delivery.[325]

    (2) Poor mechanicalstrength.[326]

    (3) Hydrophilic character  prevents it from adsorptionof cell adhesive protein.[327]

    (4) Lack of cell adhesion.[323]

     Journal of Biomaterials Science, Polymer Edition   693

  • 8/17/2019 Experimental Approaches to Constructs

    13/53

    alginate scaffold with 90% porosity and pore sizes ranging from 50 to 200 mm

    facilitates human embryonic stem cell aggregation and the formation of void and tube-

    like structures   in vivo.[69] Furthermore, growth factors can be incorporated in alginate

    scaffold to enhance vascularization, for instance incorporation of VEGF in alginate

    hydrogels enhance neovascularization into the matrix.[70,71]

    Due to remarkable wound healing capability, chitosan and its composites were

    explored for tissue engineering graft vascularization in different studies.[72] In a study,

     porous chitosan, glycosaminoglycans (GAGs)-chitosan and dextran sulfate (DS)-chi-

    tosan scaffolds were implanted in dorsal subcutaneous pockets of male Sprague-Dawley

    rats. The rats were sacriced after two weeks and it was noticed that chitosan, heparin – 

    chitosan, and DS-chitosan scaffolds promoted cell proliferation, tissue ingrowth, and

    formation of vascularized granulation tissue. Chitosan composites can also be used for 

    the controlled release of growth factors. To study the release prole of an AF from chi-

    tosan complexes, FGF-2 incorporated chitosan/heparin hydrogel was subcutaneously

    injected into the back of male Sprague-Dawley rats. Controlled release of FGF-2 mole-

    cules for 20 days caused micro blood vessel and    brous tissue formation around theinjected site.[73]

    4.1.1.3. Decellularized matrix.   Decellularization of tissue or whole organ causes the

    formation of ECM protein-enriched three-dimensional bioscaffold. The acellular matrix

    contains unique, tissue-specic, structural and functional molecules that regulate cellu-

    lar phenotype and function, mechano-transduction, signaling, cell – matrix interactions as

    well as tissue homeostasis.[74,75] Therefore, matrix decellularization followed by

    recellularization with autologous cell could be a promising approach in tissue engineer-

    ing. The technical dif culty of removing all cellular remnants limits the application of 

    decellularized matrix due to the risk of a host immune response after transplantation.Moreover, preserving the native properties of ECM, for example, three-dimensional

    ultrastructure, surface topography, composition, bioactivity, and density of ligand dis-

    tribution as well as internal network, such as nervous, vascular, and lymphatic net-

    works, presents another technical challenge.[76] One of the important features of using

    decellularized matrix is that it helps to rebuild the intricate vascular network within the

    organ or tissue when the intact vascular spaces within the matrix are repopulated with

    ECs.

    Tissue decellularization can be accomplished by chemical, biologic, or physical

    agents. Selection of an agent largely depends on tissue size, thickness, density, cellular-

    ity, and lipid content. Chemical agents, for example, acids and bases, hypotonic and

    hypertonic solutions, ionic, non-ionic, and zwitterionic detergents, or solvents, can be

    used to decellularize tissue based on the mechanism of solubilizing cytoplasmic compo-

    nents, denaturing proteins, dehydrating cells, disrupting nucleic acids, lipids, and pro-

    teins. However, chemical agents may cause some disruption of the ultrastructure of 

    ECM as well as damage of collagen, GAGs, and growth factors.[77,78] Biologic

    agents, such as nucleases, trypsin, or dispase, are basically enzymes that work by cat-

    alyzing the hydrolysis of RNA and DNA chains or cleaving peptide bonds. The use of 

     biologic agents can also result in complications, for example, it can cause an immune

    response, can disrupt ECM ultrastructure, or can remove collagen, laminin,    bronectin,

    elastin, and GAGs after prolonged exposure.[77,79] Physical techniques based on tem-

     perature, pressure, direct force, electroporation, perfusion, agitation, pressure gradient,or super critical   uid can also be used to remove cellular material from tissue. Physical

    694   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    14/53

    agents work by facilitating chemical exposure, bursting cells, or disrupting cell

    membranes, but can cause disruption or damage of ECM due to physical force.[77]

    Repopulation of a decullarized matrix with appropriate cells is a major engineering

    challenge to be solved. The parenchymal cells, for example, hepatocytes, cardiomy-

    ocytes, epithelium, etc., are responsible for the specic functions of the organ, while

    nonparenchymal cells, such as    broblasts and ECs, help to maintain the functional

     phenotype of the parenchymal cells and the cellular architecture of the tissue.[80,81]

    Moreover, ECs eliminate the thrombotic barrier within decellularized matrix and protect 

    the parenchymal cells from the shear stress by maintaining blood   ow within the

    vascular network.[82,83] Therefore, the success of decellularized matrix as a scaffold

    mostly depends on re-endothelialization of the intact vascular space. Autologus, allo-

    genic, progenitor, or multipotent stem cells are frequently investigated for their ability

    to repopulate decullarized tissues or organs. Non-immunogenic autologous cells are the

     best choice because no immunosuppressive drug is required. However, harvesting com-

     plexity and inadequate proliferation capability limit the use of autologous cells to

    repopulate acellular matrix. By some criteria, for example, required cell population,urgency of implantation, simplicity of cell harvesting and expansion, and capability of 

    differentiation into desired cell types, allogenic cells could be more useful [75] if 

    immunogenic reaction and the use of immunosuppressive drugs can be tolerated.

    Multipotent stem cells are a third option to repopulate decellularized matrix, although

    challenges exist here in controlling or directing differentiation along organ-specic or 

    tissue-specic cell lineages.[76] Bioreactors with appropriate biophysical stimuli might 

     be used to assist in differentiation of stem cells.

    A number of studies suggest a promising future for organ or tissue vascularization

    using decellularized matrix. For example, rat hearts decellularized by coronary

     perfusion and reseeded with cardiac or ECs were kept in a bioreactor, where a simu-lated cardiac physiology was maintained for 28 days. In this study, EC formed single

    layers in both larger and smaller coronary vessels after day seven, and recellularized

    heart exhibited pump function by day eight under physiological load with electrical

    stimulation.[84] Similarly, lungs decellularized by detergent perfusion, reseeded with

    epithelial or EC, and then perfused with blood and ventilated using physiologic pres-

    sures resulted in gas exchange similar to that of isolated native lungs by day   ve.

    Moreover, when these reseeded lung constructs were reimplanted into an orthotopic

     position, they generated gas exchange for up to 6 h after extubation.[85]

    4.1.2. 3D printing approaches to create vascular networks

    Vascular cell survival, growth, signaling, gene expression, and phenotype during matrix

    tissue culture is signicantly affected by the pore architecture, pore interconnectivity

    and mechanical stiffness of the scaffold.[86 – 88] The porous architecture of a scaffold

    includes pore size, shape, porosity, and surface topography of the pores. Proper poros-

    ity allows cell migration, proliferation, and interaction that facilitate the formation of a

    vascular network. In addition, porosity assists to transport nutrients and oxygen gas to

    cells and remove metabolic waste from cell surroundings. However, excessive porous

    structure might result in poor mechanical strength.

    In bone tissue engineering, higher porosity and larger pore size of the scaffold

    enhance vascularization and tissue growth. For example, a six-day  in vitro  investigationon the proliferation of goat bone marrow stromal cells suggests that scaffolds having

     Journal of Biomaterials Science, Polymer Edition   695

  • 8/17/2019 Experimental Approaches to Constructs

    15/53

    70% porosity and 800 μm average pore size enhance stromal cell proliferation

    compared to scaffolds possessing 60% porosity and 700 μm average pore size.[89]

    Like porosity, the average pore size of a scaffold has a profound effect on tissue

    growth and vascularization. A number of studies suggest that variation of pore size

    affects cell functionality. For example, it was reported that implanted polytetrauo-

    roethylene membrane having pore size 5 μm under rat skin signicantly facilitates the

    capillary blood vessel formation across the membrane – tissue interface.[90] When    bro-

    nectin-coated porous silicon nitride substrates seeded with 3T3    broblasts and bovine

    aortic ECs were kept in   ex vivo   condition for   ve days, EC covered the scaffold pores

    having diameter below 80  μm. Interestingly, pore size under 30  μm didn’t display any

    effect on EC coverage. Besides,    broblast could cover pores of 50 μm and less.[91]

    Further, disc-shaped porous PLA scaffolds with pore size in the range of 63 – 150 μm

    facilitated vascular SMC proliferation and matrix deposition during a four-weeks cul-

    ture period.[92] Therefore, a potential strategy would be to fabricate scaffolds with tar-

    geted pore size that would promote the desired cellular activity, for example, tissue

    ingrowth as well as blood vessel formation. The surface area of a scaffold is also animportant parameter to be considered as there exists a relationship between pore size

    and available surface area for cell attachment where an RGD binding sequence motif 

     prevails. In general, small pore size increases specic surface area, which ensures mini-

    mal ligand density for the attachment of a critical number of cells.[ 93,94] On the other 

    hand, large pore size facilitates cell migration but reduces cell density.

    Finally, pore interconnectivity within the scaffold can have a profound inuence on

    neovascularization and tissue growth. The diffusional mass transfer of nutrient and oxy-

    gen gas and removal of waste metabolites can be interrupted due to improper pore

    interconnectivity.[95] A   β-tricalcium phosphate scaffold fabricated by the casting tech-

    nique was used in a rabbit model to investigate the effect of pore parameters on vascu-larized bone tissue formation. An increase in pore size increases the size of blood

    vessels, but the increase in pore interconnectivity results in an increase of size as well

    as the number of blood vessels.[96] As well, pore sizes greater than 400  μm do not 

    support vascularization at all.[96] Scaffolds fabricated with PLGA biomaterial having a

    small inter-pore distance along with minute pore size in the range of 5 – 20 μm enhances

    EC growth signicantly.[97]

    Physical stiffness of a scaffold can enhance formation of focal adhesions and

    cytoskeletal reorganization in ECs which regulate cell migration, proliferation, and dif-

    ferentiation, as well as cell – cell and cell – scaffold adhesion.[98,99] A number of studies

    suggest that the spreading area of EC during focal adhesion increases with the increase

    of biomaterial’s stiffness,[100,101] however, in a stiffer biomaterial, ECs like to make

    cell –  biomaterial interactions, rather than cell – cell interaction that cause failure to form

    vascular network. On the other hand, ECs take an elongated morphology in compliant 

    material whose Young’s modulus is around 1 kPa, augment cell – cell interactions

    instead of cell –  biomaterial interaction [102], which result in the formation of 

    self-assembled vascular network.[100]

    4.1.2.1. Rapid protyping technique and miscellaneous.   To promote vascularization

    within tissue engineering construct, two major factors are to be considered during scaf-

    fold fabrication, one is well-interconnected microchannels or micropores, and the other 

    is controlled positioning of vascular cells. Some studies suggest that incorporation of channel or grooves into scaffold can promote the formation of vascular network as ECs

    require aligned organization to change their function and phenotype to form

    696   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    16/53

    capillaries.[103,104] More specically, the depth and topography of microgrooves has

     profound effect on cell alignment and functionality. Uttayarat et al. reported that maxi-

    mum alignment of bovine aortic ECs can be obtained on    bronectin-coated 1 μm deep

    microgrooved surface, while Hu and colleagues concluded that microgrooves having

    wavy surface display better cell attachment, alignment, and survival compared to those

    on the rectangularshaped microgrooved surface.[104,105]

    To construct functional complex tissue with vasculature, different types of cells

    should be seeded in an organized way as per their need and function, rather than in a

    random fashion. Scaffolds fabricated with conventional techniques, such as gas foam-

    ing, electrospinning, salt-leaching, porogen melting, molding,    ber deposition, and

    freeze-drying, display uneven, uncontrolled, and undesired pore size, shape, wall thick-

    ness, pore interconnectivity, and morphology.[106] To fabricate the complex vascular 

    network layer by layer in a controlled and precise manner, CAD-based rapid prototyp-

    ing (RP) techniques, such as 3D bioplotting, inkjet printing, laser-based biofabrication,

    and stereolithography, can be used. However, in many cases, acellular structures were

    seen to be fabricated by RP technique to keep cells free from the effect of unfavorablefabrication conditions (e.g. heat, pressure, light, adhesives, cytotoxic solvents, or mold-

    ing). One major   aw of acellular approach is the inef cient and inhomogeneous post-

    fabrication cell incorporation. Yet, a number of studies have been accomplished to

    fabricate intricate microvascular pattern with RP technique. Of several approaches, one

     promising approach was to incorporate sacricial  laments into scaffold by 3D bioplotter 

    to get complex and well-interconnected vascular pattern (Figure   6). Lewis group

    demonstrated the effectiveness of fugitive ink to create 3D microvascular network by

    direct write assembly method.[107] In another study, they created vascular pattern by

    direct write method using fugitive ink and encapsulated the whole 3D network into

    gelatin methacrylate gel (GelMA). Then HUVECs were injected after evacuation of fugitive ink. HUVECs were found to attach, proliferate, and differentiate to form vascu-

    lar network.[108] To avoid cytotoxic organic solvents, and sacricial template-removal

    associated extreme temperature or pressure, Wang et al. encapsulated sodium alginate-

     based sacricial vascular networks fabricated with 3D bioplotter into gelatin hydrogels

    and then dissolved the sacricial pattern with EDTA solution. HUVECs were injected

    into evacuated channel and found to form vascular lumens and network.[109] Another 

    research group, Chen and colleagues fabricated a patterned 3D network composed of 

    sacricial carbohydrate glass   lament by bioplotter, dispensed ECM (e.g.    brin gels,

    matrigel) encapsulated cells and mixture of poly (ethylene glycol) diacrylate and

    Figure 6. 3D approaches for vascular channel formation.

     Journal of Biomaterials Science, Polymer Edition   697

  • 8/17/2019 Experimental Approaches to Constructs

    17/53

    acrylate-PEG-RGDS into a rectangular mold containing the lattice   lament,

     photo-crosslinked the PEG hydrogel to encapsulate the 3D network as well as ECM

    embedded cells, and then dissolved the lattice with cell culture media to obtain the

    interconnected empty vascular lumen. HUVECs were then seeded in the vascular 

    lumen, and perfused with blood under high-pressure pulsatile   ow. Incorporated vascu-

    lar pattern was able to sustain the metabolic function of primary rat hepatocytes in the

    core of a thick, densely populated tissue constructs.[110] In another study, to access the

    effect of complex vascular geometry on diffusion gradient, they prepared vascular chan-

    nel structure in gelatin gel by photolithography, encapsulated the microuidic network 

    and therapeutic cells within collagen gel,   ushed gelatin gel by external   ow, and then

    injected HUVECs into the vascular lumen. Injected HUVECs formed conuent mono-

    layers within the open microuidic channels after attachment, alignment, and prolifera-

    tion. In addition, the relationship between the geometry of vasculature and spatial

    diffusive gradients which is responsible for angiogenic sprouting was also demon-

    strated.[111] To avoid the necrosis of large cell population in a thick tissue, vascular 

     pattern design parameter (e.g. vessel alignment, branching frequency and angles, andtortuosity) is required to be optimized to ensure ef cient convective diffusional mass

    transfer (e.g. nutrients, metabolic wastes, gases, AFs, etc.) between cells and culture

    medium or blood during   in vitro   or   in vivo  culture.[112] Therefore, several mathemati-

    cal modeling and computational studies have been accomplished to optimize the archi-

    tecture of vasculature considering the diffusional mass transfer, microuidic behavior,

    and physiological data of human microvasculature.[113]

    Controlled positioning of vascular cell into scaffold can be achieved through the

    deposition of cell-laden hydrogels. Polymer hydrogels offer structural support and 3D

    hydrated environment similar to   in vivo  condition for EC attachment, proliferation, and

    differentiation as well as keep ECs free from fabrication-induced high shear forces.[114] To maintain EC viability and function in culture, hydrogel crosslinking

    and scaffold fabrication process should be biocompatible. Several studies frequently

    applied stereolithography,[115] inkjet printing [116,117], and 3D bioplotting [118,119]

    techniques to prepare cell (e.g. ECs, SMCs) laden hydrogel to fabricate vascular pat-

    tern. Besides, precise cell positioning, microchannel or interconnected porous structure

    is required to avoid necrosis. To incorporate of EC-laden gel into microchannel, in a

    study, liquid collagen containing EC suspension was distributed above micropatterned

     poly (dimethylsiloxane) PDMS templates, the templates were then centrifuged to move

    ECs into the channels, and then collagen was crosslinked to form gel. It was reported

    that EC formed capillary tubes after culture with VEGF and bFGF over 24 – 48 h.[120]

    While microfabrication offers many promising features to pattern microcapillary net-

    work, time delay, use of nondegradable hydrogel, and handling complexity limits its

    application. Therefore,an alternative way, such as mechanically removable spacer (e.g.

    aligned array of wires), was investigated to create microchannel into hyrogel matrix.

    Wray et al. used mechanical spacer to generate unbranched microchannel ranging from

    152 to 787 μm in diameter into silk scaffolds, and then they seeded human arterial

    endothelial cells (hAECs) into the hollow channels along with bioactive agents. The

    formation of a contiguous layer of hAECs around the channel wall was reported

    seven days after cell seeding.[121] In addition, to ensure uniform cell seeding into

    incorporated microchannels, spacers can also be used to transfer self-assembled cell

    layer into hydrogel matrix. In a study, casted GelMA over EC-coated micrometric goldrods positioned in a culture chamber was photo-crosslinked, an electrical potential was

    applied to transfer the self-assembled layer, and then rods were removed to leave

    698   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    18/53

     behind the EC-assembled layer into the matrix. The same procedure was followed to

     prepare and transfer double-layered cell and gel mixture to combine microvessel stabi-

    lizing 3T3    broblast cells with HUVECs.[122] Beyond non-sacricial   laments,

    microuidic/micromolding techniques were also investigated to fabricate sacricial pat-

    tern in a microscale. In a study, sacricial gelatin meshes were prepared by micromold-

    ing in patterned PDMS stamp. Hydrogels (e.g.    brinogen, Type I collagen, matrigel)

    were used to encapsulate the sacricial pattern, and then successive melting and   ush-

    ing were done to empty the interconnected channels. The viability of injected HDMECs

    into microchannel was excellent as HDMECs were seen to attach, spread, and prolifer-

    ate.[123] Regardless of tremendous success with sacricial templates, dissolution-

    associated cytotoxic reaction limits their applications.[124] In an effort to fabricate

    sacricial templates-free scaffold, Bertassoni and colleagues bioprinted agarose    bers,

    casted PEGDA hydrogels over the bioprinted templates, and then photo-polymerized

    them. Removal of bioprinted templates was easy, as agarose didn’t adhere with casted

    hydrogels. With this approach, a perfusable network could be incorporated into hydro-

    gel matrix without producing any dissolution-related cytotoxicity.[125] Aside fromsacricial or removable   laments, laser scanning lithography (LSL) was investigated to

    create extremely precise micropattern on photo-sensitive hydrogel surface. In a study,

    West group covalently incorporated RGDs and VEGF on LSL-generated micropatterned

     poly (ethylene glycol) hydrogel surface. Tubules like formation in the micro-patterned

    area were observed within two days, whereas no tubules formation was recognized by

    day two for cells cultured on wide patterned lines.[126] As blood perfusion is very sig-

    nicant to regulate vascular pattern, West and colleagues combined microfabrication

    and self-assembly technique to study macro   –   micro-scale transport to achieve biomi-

    metic perfusion   in vitro. They prepared PDMS housing with   uid access ports by soft 

    lithographic techniques, injected PEG hydrogel containing the HUVEC-10T1/2 cellmixture into the PDMS housing, photolithographically crosslinked the cell-laden hydro-

    gel, and then initiated channel perfusion with normal physiologic  ow rates. The perfu-

    sion between the fabricated microchannel and self-assembled vascular network 

    signicantly facilitated the convective diffusional mass transport.[127] Moreover,

    signicant success for   in vivo   vascularization was reported while VEGF-mimetic pep-

    tide-bound PEGDA matrices were implanted into mice.[128] Besides microfabrication,

    Garicia and colleagues studied functionalized hydrogel to promote vascularization

    in vivo. They engineered photopolymerized PEG diacrylate hydrogel matrices contain-

    ing protease-degradable sites, cell-adhesion motifs (e.g. arginine – glycine – aspartic acid),

    and growth factors (e.g. VEGF) and implanted subcutaneously in male Lewis rats.

    They reported that a signicant number of vessels formed into the matrices at two

    weeks and the network became more densed at four weeks due to cell-demanded

    sustained release of VEGF over two weeks.[129] As hydrogel density regulates the

    mechanical strength, Putnam and colleagues investigated the effect of elevated    brin

    gel density on vasculature formation   in vitro   and   in vivo. Their   ndings suggest that 

    inhibition of vasculature formation   in vivo   due to elevated    brin matrix density can be

     partially lessened by co-delivery of mesenchymal stem cells (MSCs) with human

    umbilical vein endothelial cells (HUVECs) into hydrogel.[130]

    4.1.2.2. Microfabrication technique. Implanted, biodegradable tissue engineering scaf-

    folds containing cells and growth factors successfully vascularize thin tissue (1 – 2 mm).However, vascularization of thick tissue constructs (>2 mm) is still challenging as it 

    takes time for blood vessels to form and anastomose with the host blood supply. To

     Journal of Biomaterials Science, Polymer Edition   699

  • 8/17/2019 Experimental Approaches to Constructs

    19/53

    solve this issue, a microvascular network can be established by microfabrication within

    the implantable scaffold   in vitro.[131] Microfabrication and bio-microelectromechanical

    techniques, are attracting attention due to their spatial resolutions of less than 10 μm,

    [132] a substantial improvement over conventional scaffold fabrication techniques such

    as solvent casting and porogen leaching,[133] gas foaming [134], and three-dimensional

     printing.[135] However, some parameters, such as selection of fabrication material,

    oxygen concentrations in the microenvironment [136], and   uid shear stresses within

    the microuidic scaffold [137,138], signicantly affect the success and applicability of 

    microfabrication techniques.

    Microfabrication generally involves preparation of lithographic mask to pattern the

     blood vessel network according to a model,[113] formation of a master mold by thick 

     photoresist or plasma etching, casting the desired biopolymer into the master mold,

    incorporation of a sacricial layer to facilitate   lm removal from the master mold, and

    curing the   lms under vacuum (Figure   7). Single-layer microuidic networks are then

    stacked together to form 3D scaffolds with complex vascular microchannels before

    seeding with ECs in a bioreactor.[132] Real vascular networks consist of blood vesselshaving various diameters. The direct-write laser technique can facilitate the precise

    fabrication of multi-depth channels mimicking the   in vivo  structure.[139]

    Synthetic nondegradable materials, such as poly dimethyl siloxane or silicon, are

    often employed for microfabrication.[132] However, biodegradation and biocompatibil-

    ity are important issues related to the integration of the vascular construct into the host 

    tissue as well as the potential for toxicity or inammatory responses. The biodegradable

     polymers PLGA,[140] poly glycerol sebacate (PGS) [141], and silk    broin [142] were

    investigated for their suitability in constructing microuidic networks by microfabrica-

    tion. PLGA exhibits rigid mechanical properties,[134] undesirable bulk degradation

    kinetics,[143] limited biocompatibility [144], and cytotoxic degradation byproducts,[145] all of which limit the applicability of PLGA to microuidic scaffolds. PGS offers

    superior mechanical properties as well as better cellular response and morphology com-

     pared to PLGA.[143,144] PGS is inexpensive and attractive in terms of synthesizing

    and processing simplicity to form layers up to 100 μm.[146] A third possibility,

    Figure 7. Microfabrication technique for vascular network formation.

    700   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    20/53

    chemically functionalized silk    broin, is a promising biopolymer for microfabrication

    of vascular networks [142] because it offers improved cellular adhesion, controllable

    degradation, and enhanced mechanical property compared to PGS  lms.

    Alternative to multiple-layer stacking techniques, lithographic processes and sacri-

    cial gel encapsulation methods can be applied to generate 3D microuidic networks

    into hydrogels (alginate, collagen,    brin, etc.) that can later be seeded with ECs for 

    vascular tissue engineering. A lithographic process has been used to form microuidic

    channels within an alginate scaffold seeded with chondrocytes, and these channels

    appear to support long-term cell survival and convective diffusion of soluble fac-

    tors.[147] Examples of sacricial gel techniques include a study where Type I collagen

    was used to encapsulate patterned Matrigel and dened shaped microcavities were

    achieved within collagen after the digestion of matrigel by enzymes.[148] Similarly,

    microuidic channels as narrow as 6  μm were formed in collagen and    brin gels using

    embedded sacricial gelatin meshes. The channels generated in this way facilitate the

    transport of macromolecules into the channels and from channels into the gel

    matrix.[123]

    4.1.2.3. Modular assembly.   This sophisticated tissue engineering approach achieves

    formation of macro tissue constructs through the assembly of smaller modules. Micro

    modules can be prepared by self-assembled aggregation,[149] microfabrication of cell-

    seeded hydrogels, [150]creation of cell sheets [151], or direct printing.[152] To promote

    vascularization, micro modules containing target tissue cells have been coated with ECs

    and perivascular cells prior to assembly.[153] The small size of the micro modules

    facilitates the transport of oxygen and metabolites even if they are seeded with high

    cell density.[154] The surface of the modules can also be modied by coating with

    ECM proteins. For example,    bronectin coating of collagen modules implanted inimmunodecient mice increases human vascular EC survival as well as blood vessel

    formation.[56]

    To form macro tissue, a number of methods can be adapted to assemble the micro-

    modules, such as random packing,[155] stacking of layers,[156] or directed assem-

     bly.[157] In the random packing approach, EC-coated modules are packed together into

    a larger chamber and perfused with blood or culture medium which facilitates the

    formation of interconnected channels among the interstitial spaces between modules

    (Figure   8). Antithrombogenic ECs assemble to form vascularized tissue, and promote

    functional perfusion by delaying clotting time and inhibiting loss of platelets.[153]

    Incorporation of stem and progenitor cells along with ECs can signicantly affect the

    stabilization of blood vessels that develop when using the modular approach. For exam-

     ple, implanted collagen gel modules coated with rat aortic ECs into Sprague-Dawley

    rats caused the formation of blood vessels within the   rst seven days. The nascent 

     blood vessels eventually became mature and then lasted at least for 60 days. In these

    simple constructs, the new blood vessels connected with the host vasculature, yet some

    of them were leaky.[158] Interestingly, addition of bone marrow-derived stem cells to

    the EC coated modules lead to formation of blood vessels having similar density, but 

    less leakiness.[159] However, the random-packed modular approach causes lack of 

    mechanical integrity in the resultant macro tissue and limits the use of secondary cells

    other than ECs.

    Sequential assembly is a more directed method used to construct large tissue frommodular tissues with a specic microarchitecture.[160] In this approach, microgels con-

    taining specic architectural designs are assembled in a controlled fashion to connect 

     Journal of Biomaterials Science, Polymer Edition   701

  • 8/17/2019 Experimental Approaches to Constructs

    21/53

    the micro channels of each microgel, resulting in the formation of a bifurcating and

    interconnected network.[112] Thus, sequential assembly provides better control over 

    the relative spatial arrangement of the building blocks. For example, concentric PEG

    microgel building blocks embedded with network of microchannels were assembled

    sequentially by photo-crosslinking to achieve a natural blood vessel-like structure. The

    inner layer was seeded with HUVECs, and the outer layer was coated with SMCs to

    achieve a tube-like structure using a mineral oil immersion technique.[1] Formation of 

    vascular network by 3D fabrication technique has been summarized in Table  3.

    4.1.2.4. Comparison among different fabrication techniques.   It is well established that 

    oriented pores or microchannels are required to maintain cell viability in tissue engi-

    neering construct. In previous section, it was already stated that 3D fabrication tech-

    niques are associated with unfavorable conditions which led researchers to fabricate

    acellular construct. However, post-fabrication cell seeding into scaffold demonstrates

    many complexities in terms of ef cient and homogeneous cell positioning. In contrast,

    fabrication of cellular scaffold facilitates the controlled and homogeneous cell distribu-

    tion. Compared to other fabrication material, hydrogels are more suitable to fabricate

    cellular scaffold as it can ensure cell viability by providing   in vivo   like environment.

    However, lack of mechanical strength limits the applications of hydrogel scaffold. In

    addition, free radical photo polymerization or chemical crosslinking to polymerize

    hydrogels can negatively affect the viability of encapsulated cells. Channel, which isthe key parameter to promote mass transport and directional growth of EC, can be

    introduced into the 3D scaffold by using sacricial and non-sacricial   laments.

    Figure 8. Modular assembly technique for vascular network formation.

    702   Md. Sarker  et al.

  • 8/17/2019 Experimental Approaches to Constructs

    22/53

        T   a    b    l   e    3 .

        S   c   a    f    f   o    l    d    f   a    b   r    i   c   a    t    i   o   n    t   e   c    h   n    i   q   u   e   s    f   o   r   v   a   s   c   u    l   a   r    i   s   a    t    i   o   n .

        F   a    b   r    i   c   a    t    i   o   n

        t   e   c    h   n    i   q   u   e

        C    h   a   n   n   e    l    /   p   o   r   e    /   p   a    t    t   e   r   n

        /

       s   p   a   c   e   r

        G   e    l   a    t    i   o   n

       m   e    t    h   o    d

        S   c   a    f    f   o    l    d   m   a    t   e   r    i   a    l

        A    d    d    i    t    i   v   e   s    /   a    d    d    i    t    i   o   n

        t   e   c    h   n    i   q   u   e

        M   o    d   e    l    /    t   a   r   g   e    t

       s    i    t   e

        C    h   a   n   n   e    l   c   o   n          u   e   n   c   e    /    l   u   m   e   n

        f   o   r   m   a    t    i   o   n

        R   e    f

        3    D    B    i   o   p    l   o    t    t   e   r

        S   a   c   r    i          c    i   a    l

               l   a   m   e   n    t   s    /   p    l   u   r   o   n    i   c

        F    1    2    7

        P    h   o    t   o

        G   e    l    M    A

        H    U    V    E    C   s

         I    n    v     i    t    r    o

        F   o   r   m   e    d   c   o   n          u   e   n    t    l   a   y   e   r ,   v

        i   a    b    i    l    i    t   y

        >    9    5    %   a    f    t   e   r    4    8    h

        [    1    0    8    ]

        B   u    l    k   p   o    l   y   m   e   r    i   z   a    t    i   o   n

        M   e   c    h   a   n    i   c   a    l   s   p   a   c   e   r

        P    h   o    t   o

        G   e    l    M    A

        H    U    V    E    C   s

         I    n    v     i    t    r    o

        C   o   n          u   e   n    t    l   u   m   e   n   o    f    1    0   m   m    l   o   n   g

       a   n    d    6    1    8    ±    1    5     μ   m    d    i   a   m   e    t   e   r ,

       s    t   a    b    l   e   g   e   o   m   e    t   r   y    f   o   r    1    5    d   a   y   s

        [    1    2    2    ]

        3    D    B    i   o   p    l   o    t    t   e   r

        A   g   a   r   o   s   e   s   p   a   c   e   r

        T    h   e   r   m   a    l

        M   u    l    t    i   c   e    l    l   u    l   a   r

       s   p    h   e   r   o    i    d   s ,

        d    i   a   m   e    t   e   r    3    0    0   –

        5    0    0     μ   m

        C    H    O ,

        H    U    V    S    M    C   s ,

        H    S    F   s

         I    n    v     i    t    r    o

        S   p    h   e   r   o    i    d   s    f   u   s   e    d   w    i    t    h    i   n    5   –    7    d   a   y   s

        t   o    f   o   r   m   v   a   s   c   u    l   a   r    t   u    b   e   s   o    f

        0 .    9   –

        2 .    5   m   m    d    i   a   m   e    t   e   r

        [    3    2    8    ]

        B   u    l    k   p   o    l   y   m   e   r    i   z   a    t    i   o   n

        S   a   c   r    i          c    i   a    l

               l   a   m   e   n    t   s    /   c   a   r    b   o    h   y    d   r   a

        t   e

       g    l   a   s   s

        I   o   n    i   c ,

       p    h   o    t   o ,

        t    h   e   r   m   a    l ,

       e   n   z   y   m   a    t    i   c

        A    l   g    i   n   a    t   e ,           b   r    i   n ,

       p   e   g    d   a ,   m   a    t   r    i   g   e    l

        H    U    V    E    C   s

         I    n    v     i    t    r    o

        C   o   n          u   e   n    t    l   a   y   e   r   w    i    t    h    i   n   o   n   e    d   a   y

        [    1    1    0    ]

        B   u    l    k   p   o    l   y   m   e   r    i   z   a    t    i   o   n

        S   a   c   r    i          c    i   a    l    l   a    t    t    i   c   e

        /   s   o    d    i   u   m   a    l   g    i   n   a    t   e

        T    h   e   r   m   a    l ,

        i   o   n    i   c

        G   e    l   a    t    i   n ,   a   g   a   r   o

       s   e

       a   n    d   c   o    l    l   a   g   e   n

        H    U    V    E    C   s

         I    n    v     i    t    r    o

        C   o   n          u   e   n    t    l   a   y   e   r   o   n   g   e    l   a    t    i   n

       a   n    d

       c   o    l    l   a   g   e   n ,   s   p    h   e   r   o    i    d   s   o   n   a   g   a   r   o   s   e

       a    f    t   e   r    3   –    4    d   a   y   s

        [    1    0    9    ]

        3    D    M    i   c   r   o   m   o    l    d    i   n   g

        N   o   n   s   a   c   r    i          c    i   a    l   a   g   a   r   o

       s   e

               l   a   m   e   n    t   s

        P    h   o    t   o ,

        t    h   e   r   m   a    l

        1    0    %    G   e    l    M    A

        H    U    V    E    C   s

         I    n    v     i    t    r    o

        C   o   n          u   e   n    t    l   a   y   e   r    b   y    d   a   y   s   e

       v   e   n

        [    1    2    5    ]

        S    t   e   r   e   o    l    i    t    h

       o   g   r   a   p    h   y

        H   e   x   a   g   o   n   a    l    /   w   o   o    d   p    i    l   e

       m    i   c   r   o   p   a    t    t   e   r   n

        P    h   o    t   o

        1    0   o   r    1    5    %

        G   e    l    M    A

        H    U    V    E    C   s    /

        i   m   m   e   r   s    i   o   n   a   n    d

       a   g    i    t   a    t    i   o   n

         I    n    v     i    t    r    o

        C   o   n          u   e   n    t    l   a   y   e   r    b   y    d   a   y    f   o

       u   r

        [    3    2    9    ]

        3    D    R   o    b   o

        t    i   c

        d    i   s   p   e   n   s    i   n   g

        5    %   g   e    l   a    t    i   n           l   a   m   e   n    t ,

       c    h   a   n   n   e    l    4    0    0   –    7    0    0     μ   m

        C    h   e   m    i   c   a    l

        3   –    9   m   g    /   m    L

       c   o    l    l   a   g   e   n

        H    U    V    E    C   s    /   m    i   x    i   n   g

         I    n    v     i    t    r    o

        C   o   n          u   e   n    t    l   a   y   e   r   o   n    3   m   g    /   m    l

       c   o    l    l   a   g   e   n   a    f    t   e   r    t    h   r   e   e    d   a   y   s

        [    3    3    0    ]

        L   a   s   e   r  -   g   u    i    d   e    d    d    i   r   e   c    t

       w   r    i    t    i   n   g

        M    i   c   r   o   p   a    t    t   e   r   n   e    d   c   e    l    l   s

        T    h   e   r   m   a    l

        M   a    t   r    i   g   e    l

        H    U    V    E    C   s ,    V    E    G    F

         I    n    v     i    t    r    o

        E    l   o   n   g   a    t   e    d   s    t   r   u   c    t   u   r   e   s   a    f    t   e   r

        2    4    h

        [    3    3    1    ]

        P    h   o    t   o    l    i    t    h

       o   g   r   a   p    h   y

       a   n    d   a   n

        d

       a   s   s   e   m    b    l   y   o    f

       s    t   r   u   c    t   u

       r   e   s

        M   u    l    t    i  -    l   e   v   e    l

        i   n    t   e   r   c   o   n   n   e   c    t   e    d    l   u   m   e   n   s

        P    h   o    t   o

        P    E    G    D    A

        H    U    V    E    C   s ,    S    M    C   s    /

       e   n   c   a   p   s   u    l   a    t    i   o   n

         I    n    v     i    t    r    o

        9    0    %   c   e    l    l   v    i   a    b    i    l    i    t   y   a    f    t   e   r    t   w

       o    d   a   y   s ,

         i    n    v     i    v    o    l    i    k   e   v   e   s   s   e    l    f   o   r   m   a    t    i   o   n

        [    1    ]

        (     C    o    n    t     i    n    u    e     d    )

     Journal of Biomaterials Science, Polymer Edition   703

  • 8/17/2019 Experimental Approaches to Constructs

    23/53

        T   a    b    l   e    3 .

        (     C    o    n    t     i    n    u    e     d    ) .

        F   a    b   r    i   c   a    t    i   o   n

        t   e   c    h   n    i   q   u   e

        C    h   a   n   n   e    l    /   p   o   r   e    /   p   a    t    t   e   r   n

        /

       s   p   a   c   e   r

        G   e    l   a    t    i   o   n

       m   e    t    h   o    d

        S   c   a    f    f   o    l    d   m   a    t   e   r    i   a    l

        A    d    d    i    t    i   v   e   s    /   a    d    d    i    t    i   o   n

        t   e   c    h   n    i   q   u   e

        M   o    d   e    l    /    t   a   r   g   e    t

       s    i    t   e

        C    h   a   n   n   e    l   c   o   n          u   e   n   c   e    /    l   u   m   e   n

        f   o   r   m   a    t    i   o   n

        R   e    f

        P    h   o    t   o    l    i    t    h

       o   g   r   a   p    h   y

        (   m    i   c   r   o

        f   a    b   r    i   c   a    t    i   o   n    )

        M    i   c   r   o   p   a    t    t   e   r   n   e    d

        P    h   o    t   o ,

        t    h   e   r   m   a    l

        P    D    M    S    /    2 .    4   m   g

        /

       m    L   c   o    l    l   a   g   e   n   g

       e    l

        H    U    V    E    C   s ,    b    F    G    F ,

        V    E    G    F    /    i   m   m   e   r   s   e

       a   n    d   c   e   n    t   r    i    f   u   g   e

         I    n    v     i    t    r    o

        C   o   n          u   e   n    t    l   a   y   e   r    b   y    2    4   –    4    8

        h

        [    1    2    0    ]

        L    S    L

        M    i   c   r   o   p   a    t    t   e   r   n   e    d

        P    h   o    t   o

        P    E    G ,    P    E    G    D    A

        H    U    V    E    C   s ,    R    G    D    S

       a   n    d    V    E    G    F

         I    n    v     i    t    r    o

        l   u   m   e   n   s    f   o   r   m    i   n    t   w   o    d   a   y   s

        f   o   r

        <    3    5     μ   m   g   r   o   o   v   e

        [    1    2    6    ]

        S   o    f    t    l    i    t    h   o

       g   r   a   p    h   y

       a   n    d   p    h   o    t   o    l    i    t    h   o   g   r   a   p    h   y

        M    i   c   r   o   c    h   a   n   n   e    l   s    5    0   –

        1    0    0    0   a   n    d    1    5   –    5    0     μ   m

        P    h   o    t   o

        P    D    M    S ,    P    E    G

        R    G    D    S ,    H    U    V    E    C

       a   n    d    1    0    T    1    /    2 ,

        i   n    j   e   c    t    i   o   n

         I    n    v     i    t    r    o

        M   a    t   u   r   e   v   e   s   s   e    l    b   y    4    8   –    9    6    h

        [    1    2    7    ]

        C    O

           2

        l   a   s   e

       r

       e   n   g   r   a   v

        i   n   g   s   y   s    t   e   m

        2    0    0     μ   m    d    i   a   m   e    t   e   r

       c    h   a   n   n   e    l   s

        I   o   n    i   c

        A    l   g    i   n   a    t   e ,

       a    l   g    i   n   a    t   e  -   s   u    l    f   a    t   e

        H    U    V    E    C   s ,    R    G    D    /

        H    B    P

        M    i   c   e

        H    i