advances in tendon and ligament tissue engineering ...tendon and ligament tissue prosthesis material...

18
Review Article Advances in Tendon and Ligament Tissue Engineering: Materials Perspective Feras Alshomer , 1,2 Camilo Chaves, 1,3 and Deepak M. Kalaskar 1,4 1 UCL Centre for Nanotechnology and Regenerative Medicine, Division of Surgery & Interventional Science, University College London, London NW3 2PF, UK 2 King Khalid University Hospital Surgery Department 37, Plastic and Reconstructive Surgery Section, Riyadh 11472, Saudi Arabia 3 Universit´ e Paris Sud, 63 Rue Gabriel P´ eri, 94270 Le Kremlin-Bicˆ etre, France 4 Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery and Interventional Science, University College London, Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, Middlesex HA7 4LP, UK Correspondence should be addressed to Feras Alshomer; [email protected] Received 6 May 2018; Revised 6 July 2018; Accepted 26 July 2018; Published 7 August 2018 Academic Editor: Amit Bandyopadhyay Copyright © 2018 Feras Alshomer et al. is is an open access article distributed under the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited. Introduction. Tendons are specialised, heterogeneous connective tissues, which represent a significant healthcare challenge aſter injury. Primary surgical repair is the gold standard modality of care; however, it is highly dependent on the extent of injuries. Tissue engineering represents an alternative solution for good tissue integration and regeneration. In this review, we look at the advanced biomaterial composites employed to improve cellular growth while providing appropriate mechanical properties for tendon and ligament repair. Methodology. Comprehensive literature searches focused on advanced composite biomaterials for tendon and ligament tissue engineering. Studies were categorised depending on the application. Results. In the literature, a range of natural and/or synthetic materials have been combined to produce composite scaffolds tendon and ligament tissue engineering. In vitro and in vivo assessment demonstrate promising cellular integration with sufficient mechanical strength. e biological properties were improved with the addition of growth factors within the composite materials. Most in vivo studies were completed in small- scale animal models. Conclusions. Advanced composite materials represent a promising solution to the challenges associated with tendon and ligament tissue engineering. Nevertheless, these approaches still demonstrate limitations, including the necessity of larger-scale animal models to ease future clinical translation and comprehensive assessment of tissue response aſter implantation. 1. Introduction Traumatic tendon and ligamentous injuries represent sig- nificant healthcare and economic challenges for the future. Notably, these injuries are estimated to affect 110 million people in the United States [1], and incomplete repair is associated with variable disabilities and chronic sequelae [2, 3]. Tendon represents a specialised connective tissue in which collagen type I accounts for 80% of the net dry weight. In combination with proteoglycans and elastin, col- lagen permits high mechanical strength in tendons [4, 5]. Furthermore, tendons display a unique structural hierarchy where collagen molecules produce collagen fibrils, which group together to form collagen fibres. e multicomposite tendon units are composed of several collagen fibres, known as tropocollagen (Figure 1) [3, 4, 6]. Ligaments are another form of viscoelastic connective tissue, with a highly organised composition where collagens (types I, III, and V) constitute the bulk. Proteoglycans and chondroitin sulfate are also expressed, allowing the ligament tissue to swell in aqueous environments [7]. In the body, the attachment of tendons and ligaments to bone involves a transition zone with unmineralised and mineralised fibrocar- tilage [3, 8]. Tendons can further attach to muscles through fascia [4]. Defining the structural organisation of tendons and ligaments has improved understanding of the way these heterogeneous tissues function in synergy [9]. Surgical repair of tendons and ligaments via primary suture or autologous transfer techniques are considered Hindawi Journal of Materials Volume 2018, Article ID 9868151, 17 pages https://doi.org/10.1155/2018/9868151

Upload: others

Post on 27-Jan-2021

8 views

Category:

Documents


0 download

TRANSCRIPT

  • Review ArticleAdvances in Tendon and Ligament Tissue Engineering:Materials Perspective

    Feras Alshomer ,1,2 Camilo Chaves,1,3 and DeepakM. Kalaskar1,4

    1UCLCentre for Nanotechnology andRegenerativeMedicine, Division of Surgery & Interventional Science, University College London,London NW3 2PF, UK

    2King Khalid University Hospital Surgery Department 37, Plastic and Reconstructive Surgery Section, Riyadh 11472, Saudi Arabia3Université Paris Sud, 63 Rue Gabriel Péri, 94270 Le Kremlin-Bicêtre, France4Institute of Orthopaedics and Musculoskeletal Science, Division of Surgery and Interventional Science, University College London,Royal National Orthopaedic Hospital, Brockley Hill, Stanmore, Middlesex HA7 4LP, UK

    Correspondence should be addressed to Feras Alshomer; [email protected]

    Received 6 May 2018; Revised 6 July 2018; Accepted 26 July 2018; Published 7 August 2018

    Academic Editor: Amit Bandyopadhyay

    Copyright © 2018 FerasAlshomer et al.This is an open access article distributed under the Creative Commons Attribution License,which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

    Introduction. Tendons are specialised, heterogeneous connective tissues, which represent a significant healthcare challenge afterinjury. Primary surgical repair is the gold standard modality of care; however, it is highly dependent on the extent of injuries. Tissueengineering represents an alternative solution for good tissue integration and regeneration. In this review, we look at the advancedbiomaterial composites employed to improve cellular growth while providing appropriate mechanical properties for tendon andligament repair. Methodology. Comprehensive literature searches focused on advanced composite biomaterials for tendon andligament tissue engineering. Studies were categorised depending on the application. Results. In the literature, a range of naturaland/or synthetic materials have been combined to produce composite scaffolds tendon and ligament tissue engineering. In vitroand in vivo assessment demonstrate promising cellular integration with sufficient mechanical strength. The biological propertieswere improved with the addition of growth factors within the composite materials. Most in vivo studies were completed in small-scale animal models. Conclusions. Advanced composite materials represent a promising solution to the challenges associated withtendon and ligament tissue engineering. Nevertheless, these approaches still demonstrate limitations, including the necessity oflarger-scale animal models to ease future clinical translation and comprehensive assessment of tissue response after implantation.

    1. Introduction

    Traumatic tendon and ligamentous injuries represent sig-nificant healthcare and economic challenges for the future.Notably, these injuries are estimated to affect 110 millionpeople in the United States [1], and incomplete repair isassociated with variable disabilities and chronic sequelae [2,3].

    Tendon represents a specialised connective tissue inwhich collagen type I accounts for ∼80% of the net dryweight. In combination with proteoglycans and elastin, col-lagen permits high mechanical strength in tendons [4, 5].Furthermore, tendons display a unique structural hierarchywhere collagen molecules produce collagen fibrils, whichgroup together to form collagen fibres. The multicomposite

    tendon units are composed of several collagen fibres, knownas tropocollagen (Figure 1) [3, 4, 6].

    Ligaments are another form of viscoelastic connectivetissue, with a highly organised composition where collagens(types I, III, and V) constitute the bulk. Proteoglycans andchondroitin sulfate are also expressed, allowing the ligamenttissue to swell in aqueous environments [7]. In the body,the attachment of tendons and ligaments to bone involves atransition zone with unmineralised andmineralised fibrocar-tilage [3, 8]. Tendons can further attach to muscles throughfascia [4]. Defining the structural organisation of tendonsand ligaments has improved understanding of the way theseheterogeneous tissues function in synergy [9].

    Surgical repair of tendons and ligaments via primarysuture or autologous transfer techniques are considered

    HindawiJournal of MaterialsVolume 2018, Article ID 9868151, 17 pageshttps://doi.org/10.1155/2018/9868151

    http://orcid.org/0000-0001-9282-3845https://doi.org/10.1155/2018/9868151

  • 2 Journal of Materials

    Collagen

    FiberFibril

    Fascicle

    Tendon

    Figure 1: Complex structural hierarchy of tendon. Unique struc-tural hierarchy in which collagen molecules represent the simplestforming structure of tendon with complex arrangement up totendon fascicles producing the final tendon tissue.

    the gold standard modality of care. However, these solu-tions are often associated with challenges including reducedmechanical strength due to scar tissue formation, infections,donor site morbidity, and limited availability of autografts[10]. Furthermore, specialised physiotherapy protocols arerequired to improve the repair-site properties and limit scartissue formation [11].

    To overcome this clinical issue, additional therapeuticoptions involving the use of synthetic prosthesis or tissueengineered biomaterial constructs have been investigated inthe literature [12]. Ranges of synthetic prosthetic deviceshave been described as tendon and ligament tissue substi-tutes. Nonabsorbable and biocompatible polyester polyethy-lene terephthalate (PET) was investigated as a potentialtendon and ligament tissue prosthesis material with suit-able mechanical and tissue integrative properties [13, 14].Other materials like polytetrafluoroethylene (PTFE) werealso investigated as ligament tissue substitutes or in ten-don augmentation grafts, for their biologically inert andstrong mechanical characteristics [15, 16]. However, limi-tations such as graft failure, poor durability, poor tissueintegration, and foreign body synovitis were observed withthese materials [17]. Tissue engineering represents an alter-native option with potential for proper tissue integration ofimplants.

    Different synthetic or naturally derived materials havebeen investigated as scaffolds, which permit cell integrationand subsequent matrix deposition. Among the naturallyderived materials, collagen and chitosan have been exten-sively investigated for scaffold development due to theiroptimum biocompatibility and tissue integration potential[18]. Nevertheless, these materials exhibit limitations suchas low mechanical strength, batch variability, and difficultprocessing with latent immunogenicity [19].

    Synthetic materials, including polylactic acid (PLA), pol-yglycolic acid (PGA), and polylactic-co-glycolic acid (PLGA)have been extensively investigated for tendon and ligamentrepair [20]. Synthetics exhibit several advantages linkedto large-scale manufacturing, limited disease transmission,controlled degradation, and better host tissue integration.There are also limitations associated with synthetic biomate-rials; cell integration is challenging without further materialtreatment, degradation-related products can be cytotoxic,and the materials are mechanically weaker than healthymusculoskeletal tissues [18, 21].

    Novel tissue engineering approaches using compositematerials have also been described to mimic the complex,nonhomogenous environments in tendons and ligaments. Inthese composite materials, specialised cells have been com-bined with biomaterials to produce complex, heterogeneousscaffolds with controlled mechanical properties [1, 9]. Thisreview will present an overview of the different approachesdescribed to address the use of composite scaffolds for tendonand ligament tissue engineering. An in-depth critique ofthe mechanical properties, cell/ tissue integration, successcriteria, and limitations is discussed in detail.

    2. Materials and Methodology

    Comprehensive literature searches were conducted on thePubMed, Medline, Web of Science, and Google Scholardatabases. Various combinations of keywords were used inthe search process, including “tendon”, “ligament”, “tissue”,“engineering”, “hybrid”, “composite”, “scaffold”, “material”,“biomaterial”, “graft”, and “polymer”. Only publications inEnglish were included and there were no restrictions onyear of publication since no previous reviews were foundcovering the use of composite materials in tendon andligament tissue engineering simultaneously. All relevantmanuscripts were accessed and articles that combined tissueengineering approaches for both tendons and ligaments wereincluded. Articles that discussed different tissue engineeringapproaches of the tendon/ligament to bone interface werealso excluded due to the broadness of the topic and theimportant number of review articles on the subject [8, 22–24].

    3. Results and Discussion

    3.1. Composite Scaffolds for Tendon and Ligament TissueEngineering. Tendon injuries present significant healthcarechallenges due to slow healing rates, loss of function, andscar tissue formation around trauma sites [12]. In severe cases,tissue engineered scaffolds have been fabricated to replace thelost tissues. These artificial grafts can be composed of naturalor synthetic biomaterials [49]. The ideal scaffold in tendontissue engineering should exhibit specific properties such ascontrolled degradation rates, appropriate mechanical proper-ties, nonimmunogenicity, and suturability. Additionally, easymass processing and fabrication, while mimicking the nativetissue environment, are desirable [12].

    3.2. Synthetic CompositeMaterials. One example of materialsused for tendon tissue engineering involved the fabricationof composite, heterogeneous scaffolds from polyglycolic acid(PGA), and polylactic acid (PLA) [44]. The outer surfacewas composed of a knitted scaffold made from a 4:2 mixtureof PGA and PLA fibres. Internally, the construct containedlongitudinally arranged, unwoven PGA fibres. The wholeconstruct was folded and secured with sutures at each end toproduce a cord structure and was tested both in vitro and invivo. Results showed that the scaffold seeded with adipose-derived stem cells (ADSCs) promoted matrix depositionand the formation of mature collagen fibrils. These scaffolds

  • Journal of Materials 3

    presented subphysiological mechanical properties. Althoughthe study showed promising results, assessment of tenogenicdifferentiation of ADSCs was not elaborated.

    Another manufacturing technique was proposed byBaker B. M. et al., who investigated the use of coelectrospin-ning technique of different polymers to yield a compositescaffold with variable degradation techniques [28]. Theyevaluated the use of PCL fibres as slow absorbing elements,while PLGA (50:50 polylactic/ glycolic acid) or PCL/ PLGAfibres were applied for intermediate degradation rates in asingle scaffold. A water-soluble polyethylene oxide (PEO)was as a sacrificial fibre element with fast absorption ratesand aimed to increase the scaffold porosity with minimalinfluence on scaffold integrity. Mechanical assessment ofPCL/ PLGA/ PEO constructs showed a maximum stressyield of nearly 3.5MPa at 0.08% strain. The modulus wasaround 100MPa and the yield strain was 0.026. PCL/ PLGA-PCL/ PEO scaffolds showed a maximum stress yield of2MPa at 0.12% strain. Initial modulus of the material was25MPa and dropped to 12.5MPa after 63-day incubationin culture media. Strain increased from 0.065% to 0.12%after incubation. A 22% mass decrease of the compositescaffold after hydration was caused by the dissolution of thePEO component. However, themechanical properties changeafter hydration and dissolution of PEO are worth furtherexplanation.

    It has been suggested that woven scaffolds are superiorfor tissue integration due to their interconnected porousstructures.Nevertheless, they require challenging cell seedingtechniques and complex cell delivery systems [4, 5]. Sahoo S.et al. investigated the use of woven scaffoldsmade fromPLGAor PLLA [29]. Both f scaffolds were coated with PCL, PLGAnanofiber, or collagen type I to yield composite scaffolds andwere seeded with porcine bonemarrow-derived MSCs. Somecollagen-coated scaffolds were seeded with human dermalfibroblasts to test cell seeding and integration efficiency.Results showed that PLLA-based woven scaffolds performedinferiorly in terms of cell attachment. This was linked to thehydrophobic nature of the PLLAmaterial. Furthermore, PCLcoating of both types of knitted scaffolds was associated withhighermechanical strength but reduced cell attachment. Thiswas also linked to greater hydrophobicity in PCL than in theother polymers.

    As mechanical strength is particularly important, anapproach using PLAwith graphene nanoplatelets (GNP) andPLA with carboxyl functionalized carbon nanotubes (CNT-COOH) has been proposed by Pinto et al. [25, 26]. Invitro assessment of the composite was nontoxic to humanfibroblasts. In vivo assessment using mouse model did notshow any toxicity or local or systemic inflammatory response.The addition of nanofillers mentioned enhanced themechan-ical properties of PLA polymer films reaching Young’smodulus of 4.86±0.47 GPa for CNT-COOH scaffolds and4.92±0.15GPa for GNP scaffold groups. The tensile strengthhowever was mostly enhanced in the PLA/ CNT-COOHscaffolds reaching up to 72.22±1.52MPa. The authors did notinvestigate, however, the effect of in vivo implantation onthe associated mechanical properties mimicking real clinicalsettings.

    3.3. Biological Composite Scaffolds. Collagen type I compositescaffold was also investigated to incorporate resilin-like pro-tein. Resilin is an arthropods protein with elastic and highlystretchable structure. Sanami, M., et al. [27] investigated thefabrication of such composite.The scaffoldwasmade throughextrusion process of composite solution containing Collagenand Resilin at different concentration into polyethyleneglycol buffer. Fibres were subsequently cross-linked using 4-arm poly(ethylene glycol) ether tetrasuccinimidyl glutaratesolution. Mechanical assessment showed that resilin in non-cross-linked collagen scaffold significantly reduced stressat break and Young’s modulus values, while significantlyincreasing break strain. Cross-linked collagen/resilin scaffoldshowed significant increase in stress and strain values and asignificantly decreased Young’s modulus values. This showsan interesting effect of resilin exhibiting its natural properties.In vitro, the scaffold produced supported 100% fibroblastproliferation and alignment compared to 80% in collagen-fibre control.

    Scaffolds made from collagen and glycosaminoglycan(GAG) were recognised for their role in supporting cellu-lar proliferation and differentiation. However, this type ofscaffold lacks the mechanical characteristics required fortendon tissue engineering applications. Caliari et al., 2011,have proposed the concept of developing composite mate-rials in a core-shell fashion with the necessary mechanicalproperties [30]. The group fabricated scaffolds composedof highly porous, aligned isotropic GAG cores surroundedby strong, high density isotropic GAG membranes. Thecore-shell design was proposed to increase the mechanicalstrength of the scaffolds. The material was fabricated usingan evaporation technique to create membranes and freeze-drying to incorporate the core within the membrane shell.Dehydrothermal (DTH) cross-linking was used to increasematerial integration, as well as the mechanical properties. Invitro assessment using horse-derived tenocytes demonstratedgood cell attachment, proliferation, and cell viability up to14 days after seeding. Scaffolds exhibited high porosity andappropriate mechanical properties depending on the mem-brane thickness. Several factors however that need furtherinvestigation like cellular functional assessment (e.g., proteinexpression), nature and content of collagen after cell seeding,and the mechanical properties of the scaffold at differentstates (e.g., wet versus dry).

    Chitosan is a naturally derived polysaccharide with excel-lent potential for tissue engineering applications. Due tothe biocompatible and cell adhesive properties of chitosan,the polysaccharide has been investigated for tendon tissueregeneration [9–11]. In one example, composite scaffoldsmade from chitosan and alginates were produced through aspinning/coagulation technique to yield an alginate-0.1% chi-tosan scaffold. Alginate is an anionic polysaccharide with cal-cium chain in which the integration of chitosan improves itsbiocompatibility and cell adhesive potential and decreases itsdegradation rate. In vitro assessment using rabbit patellar ten-don fibroblasts showed that alginate-0.1% chitosan scaffoldshad significantly higher cell adhesion and matrix depositioncompared to alginate-only and polyglactin 910 controls. Thealginate-0.1% chitosan material was evaluated mechanically

  • 4 Journal of Materials

    and exhibited lower tensile strength and strain at failure thanthe polyglactin group [33]. Chitosan was also fabricated withhyaluronic acid followed by wet spinning and hybridisationto increase the mechanical properties of the construct.Different hyaluronic acid concentrations were investigatedand showed that the combination of chitosan with 0.1%hyaluronic acid showed the best cell attachment. The finalcomposite constructs were sterilised using ethylene oxide gasprior to in vitro evaluation with rabbit patellar fibroblasts.Mechanical properties of the materials were reduced withinthe first 2 hours after seeding but the consequent moduluswas maintained for 28 days of culture. Cell proliferationquantificationusingDNAcontent analysis showed significantimprovement in chitosan-0.1% hyaluronic acid compositescompared to other chitosan-based scaffolds. Todetermine theclinical potential of the composite scaffolds, authors assessedthe chitosan-0.1% hyaluronic acid composites in vivo bytreating rabbit rotator cuff injuries with cell-seeded scaffolds[31]. The scaffolds were cultured with rabbit patellar tendonfibroblasts for 4 weeks prior to implantation. Additionally,authors tested the potential for ligament tissue engineeringusing a rabbit medial collateral ligament injury model. Theyused scaffolds seeded with fibroblasts adapted from rabbitsAchilles tendon for 2 weeks prior to implantation. Forthe tendon model, results indicated collagen deposition incell-seeded scaffolds with significant improvement in themechanical properties from 4 to 12 weeks after implantation.For the ligament-engineering model, authors showed a lackof tissue integration with the bony tunnel attachment with60% recovery in failure load compared to healthy ligament.Additional research on the chitosan-hyaluronic acid scaffoldshas aimed to understand the effects ofmechanical stimulationon fibroblasts response [35]. It was found that the applicationof 90-degree rotations and 5% stretch at 0.5Hzwas associatedwith increased expression of fibromodulin, and collagens Iand III. No further assessment of the overall mechanicalinfluence of the cultured scaffolds under dynamic conditionswas made.

    Additionally, composite scaffolds made from extruded,cross-linked bovine type I collagen and chondroitin-6-sulfatewere fabricated [36]. The collagen-based constructs under-went a series of cross-linking steps using carbodiimide, 1-ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDC), andN-hydroxysuccinimide (NHS). Cross-linking was combinedwith freeze-drying to incorporate a porous chondroitin-6-sulfate. The final constructs had open and interconnectedporosity with an average pore size of 100𝜇m and axi-ally aligned collagen fibres. Mechanical assessment showedthat the cross-linked composite scaffolds could withstand61.94±15.54 N, compared to 11.75±2.62 N without cross-linking. The maximum strain was shown to be 30.17±7.17%with cross-linked fibres, while it was 15.40±3.22% with-out. Composites with cross-linked fibres presented a tensilestrength of 1.55±0.30MPa compared to 0.24±0.08MPa fornon-cross-linked fibres. It is important to mention thatscaffold porosity is inversely related to mechanical strengthand further studies are required to evaluate the cell responseof such construct for application in tendon and ligamenttissue engineering.

    Gelatin was also utilised to fabricate a composite scaffoldfor tendon tissue engineering applications. Coelectrospin-ning of aligned poly-𝜀-caprolactone (PCL) and methacry-lated gelatin (mGLT) followed by photo-cross-linking wasused in the fabrication process [34]. Scaffold films were firstproduced and then were seeded with ADSC, after whichphoto-cross-linking of 5 layers using UV radiation was madeto produce a multilayered scaffold mimicking the naturaltendon structure. In vitro assessment showed biocompatibil-ity of produced composite in which ADSCs were orientedalong the longitudinal access of aligned fibre construct andexpressed tendon-related markers (scleraxis and tenascin-C)after being stimulated with TGF𝛽-3. Mechanical assessmentof cell-seeded cross-linked scaffolds however was far inferiorfor potential clinical application (details in Table 1).

    Other groups have combined collagen type I with silkto improve scaffold properties for tendon applications [37].Sericin protein was extracted from silk fibres producedby Bombyx mori, and mixed with collagen solution to fillthe gaps in the silk fibre net. Furthermore, adherence andmechanical strength were increased though DTH cross-linking.Mesenchymal stem cells derived fromhuman embry-onic stem cells (hEC-MSCs) were used in in vitro and invivomodels and showed that themechanically loaded knittedsilk/ collagen microsponge scaffolds can induce tenogenicdifferentiation in mesenchymal stem cells and support ten-don regeneration up to 360 days after implantation (Table 1).

    In an additional study, the same composite scaffoldswere tested with the supplementation of recombinant humanstromal cell-derived factor-1 alpha (rhSDF-1 alpha), to thecollagen type I sponges [38]. SDF-1 is a chemokine thatpromotes cell recruitment and enhances tissue regeneration.In a murine Achilles tendon model, the authors showed thatthis approach permits higher expression of collagen one weekafter implantation, indicating an accelerated onset of tendonhealing. Further, the mechanical properties were marginallyhigher than in scaffolds without rhSDF-1 alpha treatment.

    In order to improve the mechanical properties andtendon regeneration, Chen X. et al. utilised the same com-posite scaffolds with hEC-MSCs [39]. Cells were geneticallyengineered to overexpress the Scleraxis (SCX) gene. SCX is atranscription factor identified for its role as a tenocytemarkerand upstream regulator of tendon-related genes. In vitro andin vivo results showed enhanced tendon regeneration andbetter quality neotendon formation, compared to the previ-ous studies with the same scaffolds. Importantly, there wasless osteogenic, chondrogenic, and adipogenic differentiationof the SCX-hEC-MSCs compared to nongenetically modifiedMSCs. Mechanical properties in a murine Achilles tendoneight weeks after implantation were inferior to native tendontissue. This research highlights that scaffold properties areimportant for tendon tissue engineering, but factors like cell-types used can influence mechanical and histological results.

    3.4. Synthetic and Biological Composite Scaffolds. Othershave also focused on incorporating PLGA with silk derivedmaterials for tendon tissue engineering applications [40]. Acomposite scaffold comprised degummed silk microfiberscoated electrospun PLGA.The authors degummed silk fibres

  • Journal of Materials 5

    Table1:Summaryo

    fthe

    literaturerelated

    tothea

    pplications

    ofhybrid

    biom

    aterialintend

    ontissuee

    ngineerin

    g.∗

    indi

    cate

    stha

    tapp

    roximaten

    umberswe

    reextractedfro

    msupp

    liedfig

    ures

    since

    noexactreference

    values

    arem

    entio

    nedin

    thetext.PD

    S:po

    ly-dioxano

    ne;SCX

    ,scleraxis;

    TGF𝛽

    ,transform

    inggrow

    thfactor

    beta;hEC

    S-MSC

    s,mesenchym

    alste

    mcells

    deriv

    edfro

    mhu

    man

    embryonics

    tem

    cells;rhSDF1,recom

    binant

    human

    strom

    alcell-deriv

    edfactor

    1;PG

    A,poly-(la

    ctide-co-glycolid

    e);P

    CL,poly-caprolactone;bFG

    F,basic

    fibroblastgrowth

    factor.

    CO

    MPO

    SITE

    /HYB

    RID

    MO

    DEL

    SCA

    FFO

    LDPR

    OPE

    RTIE

    SC

    ELL/

    TISS

    UE

    INTE

    GRA

    TIO

    N

    PGA

    /PLA

    com

    posi

    te.[

    24]

    Invitro(A

    DSC

    s)In

    vivo

    (rab

    bit)

    Afte

    rimplantatio

    nin

    Achillestendo

    n,Te

    nsile

    stre

    ngth

    :4.88±

    8.07

    MPa

    No

    repo

    rtof

    othe

    rmec

    hani

    calp

    rope

    rtie

    s.

    (i)Grossly,

    theimplantedcell-seeded

    scaffoldwas

    integrated

    with

    the

    nativ

    etissue

    interfe

    rence,with

    asmoo

    thsurfa

    cecord-like

    shapew

    ithlessno

    ticeabler

    emaining

    materialafte

    r45we

    eks.

    (ii)T

    issue

    adhesio

    ns,described

    grossly

    tobe

    lesscomparedto

    control.

    (iii)Paralleland

    morem

    aturec

    ollagenfib

    ersa

    ndlong

    itudinally

    alignedcells

    arep

    resentsthancontrol.

    P(L

    LA-C

    L)/C

    olla

    gen

    I.[2

    5]In

    vitro(Tenocytes)

    Youn

    g’sm

    odul

    us∗

    Non

    seeded

    abou

    t2.2MPa.

    Cellseededabou

    t3MPa.

    Tens

    ilest

    reng

    th∗

    Non

    seeded

    abou

    t3MPa.

    Cellseededabou

    t4.5MPa.

    (i)Sign

    ificantlyhigh

    ercellproliferatio

    nin

    then

    anoyarnscaffold

    comparedto

    otherscaffo

    ldsa

    ndcontrol.

    (ii)S

    EMshow

    edspindle-shaped

    cellin

    both

    nano

    yarn

    andaligned

    nano

    fiber

    scaffold,whilepo

    lygonaland

    rand

    ompatte

    rncells

    are

    foun

    din

    ther

    ando

    moriented

    fibers.

    (iii)Ex

    pressio

    nof

    tend

    onspecificE

    CM(ty

    peIc

    ollagen,

    type

    IIIcollagen,

    decorin

    ,tenascin-C,

    andbiglycan)w

    assig

    nificantly

    high

    erat14

    days

    inthen

    anoyarngrou

    p.

    P(L

    LA-C

    L)/C

    olla

    gen

    Ina

    noya

    rn.[

    26]

    Invitro(TDSC

    s)In

    vivo

    (mou

    se)

    Mec

    hani

    cals

    tres

    s:Dynam

    icgroupwas

    59.58±7.8

    1MPa

    Staticgroupwas

    43.18±6.58

    MPa

    Controlgroup

    was

    32.43±

    5.27%MPa

    Youn

    g’sm

    odul

    us:

    Dynam

    icgroupwas

    51.99±

    7.16MPa

    Staticgroupwas

    34.76±4.75

    MPa.

    Controlgroup

    was

    23.30±3.83

    MPa

    TDSC

    swereu

    sedin

    scaffoldseedingwith

    both

    dynamicandsta

    ticcultu

    recond

    ition

    s.In

    vitro:

    (i)TD

    SCssho

    wedelon

    gatedfib

    roblast-likem

    orph

    olog

    ywith

    asig

    nificantincreaseincellcoun

    tindynamicgrou

    pat14

    days.

    (ii)M

    orec

    ellinfi

    ltrationanddensem

    atrix

    indynamicgrou

    p.(iii)PC

    Rconfi

    rmed

    Tend

    onrelatedmRN

    Aexpressio

    nmorein

    dynamicgrou

    p.(iv

    )Western

    blottin

    gshow

    edsig

    nificantincreaseinthep

    rotein

    expressio

    nlevelsof

    Collagens

    Iand

    III,and

    tenascin-C

    inthe

    dynamicgrou

    p.In-vivo:

    (i)Sign

    ificantlylowe

    rnum

    bero

    fcellsat12

    weeksw

    ithgreaterm

    atrix

    depo

    sitionandlong

    itudinalspind

    le-shapedcells

    indynamicgrou

    pcomparedto

    others.

    (ii)C

    ollagencontentw

    ashigh

    estindynamicgrou

    preaching

    77.76±

    6.82%of

    norm

    alrabb

    itpatellartendo

    n(174.31±13.89𝜇g/mg).

    (iii)CollagenIexpressionwas

    Sign

    ificantlyhigh

    erin

    dynamicgrou

    p.

  • 6 Journal of Materials

    Table1:Con

    tinued.

    CO

    MPO

    SITE

    /HYB

    RID

    MO

    DEL

    SCA

    FFO

    LDPR

    OPE

    RTIE

    SC

    ELL/

    TISS

    UE

    INTE

    GRA

    TIO

    N

    PLA

    /Col

    lage

    n-Ie

    lect

    rosp

    unbu

    ndle

    s[27

    ]In

    Vitro(Tenocytes)

    Com

    posites

    caffo

    ldsw

    erem

    adefrom

    blends

    containing

    PLA/C

    oll-7

    5/25

    (w/w

    ).Yo

    ung’s

    mod

    ulus

    was

    98.6±12.4MPa

    Max

    imum

    stre

    sswas

    14.2±0.7MPa

    (i)Seeded

    Teno

    cytessho

    wnto

    exhibitgoo

    dcelladhesio

    nprofi

    leand

    moree

    long

    ated

    morph

    olog

    ythatwas

    bette

    roverP

    LA/C

    oll-5

    0/50

    blends

    than

    other.

    PLA

    /gra

    phen

    enan

    opla

    tele

    ts(G

    NP)

    and

    PLA

    /car

    boxy

    l-fu

    nctio

    naliz

    edca

    rbon

    nano

    tube

    s(C

    NT-

    CO

    OH

    )[2

    8,29

    ]

    Invitro(fi

    broblast)

    Invivo

    (mou

    se)

    Youn

    g’sm

    odul

    usPL

    Acontrol:3.99±0.42

    GPa

    PLA/C

    NT-COOH:4.86±0.47

    GPa

    PLA/G

    NP:

    4.92±0.15

    GPa

    Tens

    ilest

    reng

    thPL

    Acontrol:59.90±4.93

    MPa

    PLA/C

    NT-COOH:72.22±1.5

    2MPa

    PLA/G

    NP:

    58.56±3.99

    MPa

    (i)Bo

    thprod

    uced

    scaffolds

    supp

    ortedfib

    roblastsmetabolicactiv

    ityandproliferatio

    ntillfi

    nalassessm

    entp

    oint

    (72hrs).

    (ii)Invivo

    assessmentsho

    wedlack

    ofanylocalorsystemic

    inflammatoryr

    espo

    nseu

    singN-acetylglucosaminidase(NAG

    )and

    nitricoxide(

    NO)serum

    levels.

    (iii)Noassociated

    hepatotoxicityin

    histo

    logica

    ssessm

    ent.

    (iv)H

    istologicassessmento

    fexplanted

    scaffoldshow

    edform

    ationof

    thin

    capsulea

    roun

    dtheimplantw

    ithho

    mogenou

    sgranu

    latio

    ntissue.

    PCL/

    colla

    gen-

    PLLA

    /col

    lage

    n[3

    0]In

    vitro(m

    yoblasts,

    fibroblast)

    Tens

    ilest

    reng

    thwas

    0.5058±0.2130

    MPa

    Max

    imum

    stra

    inwas

    18.49%±8.210

    Youn

    g’sm

    odul

    uswas

    7.339±2.131M

    Pa

    (i)Statisticallyhigh

    erviability

    ofbo

    thmyoblastand

    fibroblastsin

    all

    region

    softhe

    scaffold.

    (ii)S

    caffo

    ldcouldsupp

    ortthe

    form

    ationof

    myotubesthatise

    ssentia

    lforn

    ormalmuscle-tend

    onjunctio

    nform

    ation.

    Alig

    ned

    PLLA

    nano

    fiber

    /Lay

    ered

    chito

    san-

    colla

    gen

    hydr

    ogel

    /Alg

    inat

    eout

    erco

    atin

    g.[3

    1]

    Invitro(Tenocytes)

    Max

    imum

    Forc

    eto

    brea

    kFo

    runcoated2and3layersscaffold:

    7.89±

    1.5Nand7.4

    5±0.3N

    ,Fo

    rgelcoated

    2and3layersscaffolds:4.76±

    0.23Nand6.49±0.09N.

    No

    repo

    rtof

    othe

    rmec

    hani

    calp

    rope

    rtie

    s.

    (i)Alginatec

    oatin

    gwas

    associated

    with

    significantly

    lessattached

    proteins

    than

    control.

    (ii)B

    othcoated

    andun

    coated

    scaffoldmaintain50%of

    their

    substancea

    fterincub

    ationwith

    PBScontaining

    104un

    its/m

    llysozym

    esolutio

    n.(iii)Alamar

    blue

    andDNAconcentrationassessmentsho

    wedhigh

    cellu

    larv

    iability,metabolicactiv

    ity,and

    proliferatio

    nup

    to7days

    after

    seeding.

    (iv)S

    eededscaffolds

    were

    show

    nto

    supp

    ortcellulara

    lignm

    ent.

  • Journal of Materials 7

    Table1:Con

    tinued.

    CO

    MPO

    SITE

    /HYB

    RID

    MO

    DEL

    SCA

    FFO

    LDPR

    OPE

    RTIE

    SC

    ELL/

    TISS

    UE

    INTE

    GRA

    TIO

    N

    Elec

    tros

    pun

    colla

    gen

    Ina

    nofib

    er/c

    olla

    gen

    mic

    rofib

    er[3

    2]

    Invitro(fi

    broblast)

    Invivo

    (rab

    bit)

    Before

    implantatio

    n:M

    axim

    umlo

    adwas

    28.33±2.19

    N.

    Max

    imum

    stre

    sswas

    2.69±0.47

    MPa

    Max

    imum

    stra

    inwas

    61.34±4.71

    %Yo

    ung’s

    mod

    ulus

    was

    43.81±

    4.19

    KPa.

    60days

    after

    implantatio

    nto

    Achillestendo

    n:M

    axim

    umlo

    adwas

    abou

    t63.72

    N.

    Max

    imum

    stre

    sswas

    abou

    t9.84N/m

    m.

    Max

    imum

    stra

    inwas

    abou

    t16.35%

    Youn

    g’sm

    odul

    uswas

    abou

    t0.62N/m

    m.

    Invitro:

    (i)Sign

    ificantlyhigh

    ercellviability

    inthea

    lignedhybrid

    scaffold

    comparedto

    others.

    (ii)S

    EMandim

    mun

    ofluo

    rescence

    proven

    superio

    ralignm

    ento

    ffib

    roblaststogether

    with

    cellproliferatio

    nwith

    closec

    ell-to-cell

    contactinthea

    lignedhybrid

    scaffoldcomparedto

    others.

    Invivo:

    (i)Sign

    ificant

    increase

    inthen

    umber,density,and

    alignm

    ento

    fthe

    collagendepo

    sitionwith

    maturee

    lasticfi

    bers.

    (ii)S

    ignificantincreaseinthen

    umbera

    ndmaturity

    ofteno

    blastsand

    teno

    cytes.

    (iii)Sign

    ificantlylowe

    rperitend

    inou

    sadh

    esions,m

    usclefi

    brosis,

    and

    atroph

    yandinflammatoryc

    ellsfoun

    din

    thetreated

    tend

    ons

    comparedto

    controls.

    Elec

    tros

    pun

    colla

    gen

    Ina

    nofib

    er/c

    olla

    gen

    mic

    rofib

    er/

    PDS

    shee

    ts.[33]

    Invivo

    (rab

    bit)

    Afte

    r60days

    ofim

    plantatio

    nto

    Achilles

    tend

    on:

    Max

    imum

    load∗was

    abou

    t74.02

    NM

    axim

    umst

    ress∗was

    abou

    t11.3

    7MPa

    Youn

    g’sm

    odul

    us∗was

    abou

    t0.754

    MPa

    (i)Sign

    ificantlyhigh

    erfib

    rillogenesis

    after

    PDStre

    atment.

    (ii)S

    ignificantly

    high

    ercollagenfib

    rilsw

    erefou

    ndin

    theP

    DStre

    ated

    scaffolds

    comparedto

    ther

    egular

    one.

    (iii)Sign

    ificant

    increase

    intheloadto

    failu

    reandload

    toyieldpo

    int

    with

    PDStre

    atment.

    (iv)S

    ignificantly

    improved

    peritendino

    usadhesio

    ns,m

    usclefi

    brosis,

    andatroph

    yscores

    thatarec

    omparableintheP

    DStre

    ated

    and

    nontreated

    collagenscaffolds.

    Cor

    e-sh

    ellC

    olla

    gen

    type

    I/G

    lyco

    sam

    inog

    lyca

    n.[34]

    Invitro(Tenocytes)

    Tens

    ilem

    odul

    usfo

    rdry

    mem

    bran

    eswas

    abou

    t636±47

    MPa

    to693±20

    MPa

    Tens

    ilem

    odul

    usfo

    rhyd

    rate

    dm

    embr

    anes

    was

    30MPa.

    Tens

    ilem

    odul

    usfo

    rdry

    alig

    ned

    core

    scaff

    oldranges

    from

    833±236to

    829±165

    KPa

    (i)Sign

    ificant

    increase

    inthen

    umbero

    fcellsatday1afte

    rseeding

    inthec

    ore-shellcom

    positec

    omparedto

    core

    alon

    escaffo

    lds.

    (ii)H

    igherm

    etabolicactiv

    ityob

    served

    atday7in

    thec

    orea

    lone

    scaffoldthatissta

    tistic

    allysig

    nificant.

    (iii)Nosig

    nificantd

    ifference

    inthem

    etabolicactiv

    ityandcell

    numberb

    etwe

    enthetwo

    grou

    psat14-day

    perio

    d.

    Col

    lage

    n-I/

    nano

    carb

    onfib

    ers

    [35]

    -Mechanicalassessm

    entinwe

    tcon

    ditio

    nYo

    ung’s

    mod

    ulus

    was

    840±140MPa

    Tens

    ilest

    reng

    thwas

    70±8MPa

    (i)Nocellwo

    rkwas

    presented.

  • 8 Journal of Materials

    Table1:Con

    tinued.

    CO

    MPO

    SITE

    /HYB

    RID

    MO

    DEL

    SCA

    FFO

    LDPR

    OPE

    RTIE

    SC

    ELL/

    TISS

    UE

    INTE

    GRA

    TIO

    N

    Col

    lage

    nty

    peI/

    Resi

    lin[3

    6]In

    vitro(Fibroblasts)

    Max

    imum

    stre

    sswas

    34.63±9.7

    5MPa

    Max

    imum

    stra

    inwas

    0.21±0.04

    MPa

    Youn

    g’sm

    odul

    uswas

    49.48±10.71M

    Pa

    (i)Hum

    anadultfi

    broblasts

    were

    used

    inthea

    ssessm

    entp

    rocess.

    (ii)Th

    eautho

    rsshow

    edthatadditio

    nof

    resilin

    andtheu

    seof

    poly(ethyleneg

    lycol)either

    tetrasuccinimidylglutarated

    idcomprom

    isethec

    ellulara

    ctivity.

    (iii)Th

    escaffo

    ldprod

    uced

    managed

    tosupp

    ortcellularp

    roliferation

    and100%

    cellu

    lara

    lignm

    entafte

    r7days

    ofseedingcomparedto

    80%

    incollagencontrol.

    poly

    -𝜀-c

    apro

    lact

    one(

    PCL)

    and

    met

    hacr

    ylat

    edge

    latin

    (mG

    LT)

    [37]

    Invitro(A

    DSC

    s)M

    axim

    umlo

    adwas

    abou

    t0.25N

    .N

    ore

    port

    ofot

    herm

    echa

    nica

    lpro

    pert

    ies.

    (i)Teno

    genicd

    ifferentia

    tionwas

    indu

    cedthroug

    hdifferentiatio

    nmedium

    having

    DMEM

    ,2%FB

    S,P/S,and10

    ng/m

    lTGF-𝛽3for7

    days

    after

    seeding.

    (ii)S

    eededconstructswe

    reshow

    nto

    exhibitelong

    ated

    morph

    olog

    yas

    wellas

    expressio

    nof

    teno

    genicm

    arkers(scleraxisandTenascin-C

    ).(iii)Stackedscaffoldwas

    show

    nto

    have

    adequatepo

    rosityforc

    ell

    diffu

    sionanddifferentiatio

    n.

    Seri

    cin

    extr

    acte

    dkn

    itted

    silk

    /cro

    ss-li

    nked

    colla

    gen

    type

    Im

    icro

    spon

    ges[38]

    Invitro

    (hES

    C-MSC

    s)In

    vivo

    (mou

    se)

    4we

    eksa

    fterimplantatio

    nto

    Achillestendo

    nof

    cell-seeded

    scaffolds:

    Max

    imum

    load

    was

    65.24±9.5

    8N

    Max

    imum

    stre

    sswas

    6.72±0.90

    MPa

    Stiff

    nesswas

    28.26±2.95

    N/m

    mYo

    ung’s

    mod

    ulus

    was

    34.91±

    5.08

    MPa

    Invitro:

    (i)Goo

    dcellattachment,proliferatio

    n,andspreadingat14

    days'

    perio

    d.(ii)S

    cx.,collagenI,andcollagenIII

    expressio

    nwe

    resig

    nificantly

    high

    erin

    dynamicgrou

    pthan

    thec

    ontro

    l.In

    vivo

    assessmenta

    fter4

    weeks

    ofim

    plan

    tatio

    n:(i)

    Grossly,

    well-integrated

    constructw

    ithnativ

    etendo

    ns.

    (ii)V

    iablec

    ellsarep

    resentssho

    wingspindle-shaped

    morph

    olog

    y.(iii)Sign

    ificantlyhigh

    ercollagenI,III,V𝛼1,V𝛼2,andTG

    F𝛽1in

    cell-seeded

    scaffoldcomparedto

    cellfre

    escaffo

    ld.

    (iv)N

    eocollagenwas

    replacingexogenou

    sone

    with

    morec

    ontent,

    andmaturem

    orph

    olog

    yin

    cell-seeded

    scaffoldcomparedto

    cellfre

    escaffold.

  • Journal of Materials 9

    Table1:Con

    tinued.

    CO

    MPO

    SITE

    /HYB

    RID

    MO

    DEL

    SCA

    FFO

    LDPR

    OPE

    RTIE

    SC

    ELL/

    TISS

    UE

    INTE

    GRA

    TIO

    N

    Seri

    cin

    extr

    acte

    dkn

    itted

    silk

    /cro

    ss-li

    nked

    colla

    gen

    type

    Im

    icro

    spon

    ges/

    rhSD

    F-1a

    lpha

    [39]

    Invitro(fi

    broblast)

    Invivo

    (rat)

    4we

    eksa

    fterimplantatio

    nin

    Achillestendo

    n:M

    axim

    umlo

    adwas

    68.5±18

    NM

    axim

    umst

    resswas

    7.02±1.7

    MPa

    Stiff

    nesswas

    39.0±6.6N/m

    mYo

    ung’s

    mod

    ulus

    was

    45.3±10.4MPa

    Invivo

    assessmenta

    fter4

    weeks

    ofim

    plan

    tatio

    n:(i)

    Morefi

    brob

    lasts

    -like

    cells

    andlessinflammatorycells

    arefou

    ndearly

    after

    implantatio

    n(4

    days,1

    week).

    (ii)M

    oreo

    rganized

    continuo

    uscollagenfib

    ersa

    refoun

    dwith

    ahigh

    erconcentrationof

    collagentype

    I.(iii)Goo

    dvascular

    compo

    nentsw

    ithlessinflammatorycells

    are

    present.

    (iv)S

    ignificantly

    high

    erexpressio

    nof

    Collagens

    Iand

    III,decorin

    intheS

    DF-1scaffo

    ldsa

    tallassessmentp

    eriods.

    (v)L

    arge

    fibrilso

    fAchilles

    tend

    onsformed

    intheS

    DF-1scaffo

    lds

    (41.6±5.5nm

    )com

    paredto

    control(37.1±2.9nm

    ).

    Seri

    cin

    extr

    acte

    dkn

    itted

    silk

    /cro

    ss-li

    nked

    colla

    gen

    type

    Im

    icro

    spon

    ges/

    SCX

    engi

    neer

    edce

    lls[40]

    Invitro

    (hES

    C-MSC

    s)In

    vivo

    (mou

    se)

    4we

    eksa

    fterimplantatio

    n:Yo

    ung’s

    mod

    ulus

    was

    60.63±17.6MPa

    Max

    imum

    stre

    sswas

    8.73±2.15

    MPa

    8we

    eksa

    fterimplantatio

    n:Yo

    ung’s

    mod

    ulus

    was

    71.3±12.4MPa

    Max

    imum

    stre

    sswas

    10.1±2.2MPa

    Invitro:

    (i)SC

    Xtre

    atmentincreases

    collagenIexpressionwith

    enhanced

    cell-sheetformation.

    (ii)D

    ecreased

    capacityof

    adipogenic,cho

    ndrogenic,andosteogenic

    differentiatio

    nof

    thec

    ellswith

    moretenogenicdifferentiatio

    n.In

    vivo:

    (i)Goo

    dproliferatio

    nandearly

    matrix

    depo

    sitionin

    theS

    CXcells

    comparedto

    control.

    (ii)Increased

    CollagenIa1,Ia2,andtend

    onrelatedtranscrip

    tion

    factor

    Eya2

    inSC

    Xcells

    comparedto

    control.

    (iii)Morefi

    brob

    last-

    likespind

    le-shapedcells

    andfewe

    rimmun

    ogenic

    cellinfiltrates

    intheS

    CXcells

    grou

    p.(iv

    )Highere

    xpressionof

    biglycan

    intheS

    CXcells

    grou

    p,indicatin

    gmorem

    aturee

    ndogenou

    scollagen.

    Kni

    tted

    silk

    coat

    edw

    ithel

    ectr

    ospu

    nco

    llage

    n-I/

    poly

    uret

    hane

    (PU

    )nan

    ofibe

    rs[4

    1]

    Invitro(Fibroblast)

    Max

    imum

    stre

    sswas

    13.5±1.5

    MPa

    Youn

    g’sm

    odul

    uswas

    21.7±4MPa

    (i)Hum

    anfib

    roblastswe

    reseeded

    oncompo

    sites

    caffo

    ldto

    testfor

    viability.

    (ii)A

    ssessm

    entw

    asmadethrou

    ghAlamar

    Blue

    assayandshow

    edthatsamples

    with

    high

    ercollagencontenth

    adhigh

    ercellu

    larv

    iability

    profi

    le(C

    OL75/PU

    25)thanotherg

    roup

    s.

  • 10 Journal of Materials

    Table1:Con

    tinued.

    CO

    MPO

    SITE

    /HYB

    RID

    MO

    DEL

    SCA

    FFO

    LDPR

    OPE

    RTIE

    SC

    ELL/

    TISS

    UE

    INTE

    GRA

    TIO

    NSi

    lkco

    ated

    with

    Poly

    capr

    olac

    tone

    (PC

    L)or

    Poly

    (3-h

    ydro

    xybu

    tyra

    te)

    (P3H

    B)na

    nofib

    ers[

    42]

    Invitro(Fibroblast)

    Max

    imum

    load

    97.6±11.4Nforsilk

    fibroin/P3H

    B110

    .5±6.6Nforsilk

    fibroin/PCL

    No

    repo

    rtof

    othe

    rmec

    hani

    calp

    rope

    rtie

    s.

    (i)Hum

    anfib

    roblastswe

    reseeded

    oncompo

    sites

    caffo

    ldandshow

    edgood

    cellu

    larv

    iabilityandno

    toxicity(assessm

    entfor

    3days).

    Deg

    umm

    edSi

    lk-fi

    broi

    nm

    eshe

    s/al

    igne

    dSi

    lk-fi

    broi

    n[43]

    Invitro(M

    SCs)

    Max

    imum

    load

    ford

    ynam

    iccu

    lture

    cond

    ition

    Alignedscaffold

    144.44±5.03

    N(7

    days)

    172.08±6.28

    N(14

    days)

    Rand

    omScaffold

    122.35±3.67

    N(7

    days)

    138.67±9.2

    2N(14

    days)

    Stiff

    ness

    Alignedscaffold

    24.33±1.4

    0N/m

    m(7

    days)

    26.93±2.40

    N/m

    m(14

    days)

    Rand

    omScaffold

    17.48±0.93

    N/m

    m(7

    days)

    23.07±

    2.54

    N/m

    m(14

    days)

    No

    repo

    rtof

    othe

    rmec

    hani

    calp

    rope

    rtie

    s.

    (i)Highlysig

    nificantcellviabilityin

    thea

    lignedscaffoldcomparedto

    ther

    ando

    mon

    eatb

    othdynamicandsta

    ticcond

    ition

    s.(ii)C

    onsistent

    cellu

    larp

    roliferationin

    both

    aligned(dyn

    amicand

    static

    )and

    dynamicrand

    omscaffold.

    (iii)Higherc

    ollagendepo

    sitionin

    thed

    ynam

    icalignedscaffold

    comparedto

    thes

    tatic

    onea

    ndther

    ando

    mscaffoldgrou

    ps.

    (iv)D

    ynam

    iccultu

    reim

    proved

    theh

    istologicalassessmento

    fboth

    alignedandrand

    omscaffoldwith

    morec

    ellelong

    ationandmatrix

    depo

    sition.

    (v)H

    ighere

    xpressionlevelofcollagenI,tenascin-C

    ,and

    teno

    mod

    ulin

    inthea

    ligneddynamicscaffolds

    comparedto

    others.

    Kni

    tted

    PLG

    A-P

    LLA

    /coa

    ting

    with

    (1)P

    CL.

    (2)P

    LGA

    nano

    fiber

    .Ty

    peIc

    olla

    gen[44]

    Invitro(BMSC

    s)

    Max

    imum

    load

    Uncoated68.4±5.37

    N.

    PCLcoated

    63.1±4.52

    N.

    PLGAcoated

    56.3±6.61

    N.

    Collagencoated

    59.5±8.3N.

    Stiffness

    Uncoated9.1±2.38

    N/m

    m.

    PCLcoated

    4.3±0.91

    N/m

    m.

    PLGAcoated

    5.8±0.70

    N/m

    m.

    Collagencoated

    5.7±0.48

    N/m

    m.

    No

    repo

    rtof

    othe

    rmec

    hani

    calp

    rope

    rtie

    s.

    (i)Effi

    cientcellseeding

    onPL

    GAnano

    fiber

    coated

    and

    collagen-coated

    scaffolds.(80-89%

    and61-69%

    ,respectively

    ).(ii)S

    ignificantly

    high

    ercellproliferatio

    nbetween2n

    dand7t

    hcultu

    redays

    ofPL

    GAnano

    fiber

    coated

    knitted

    PLGAscaffolds.

  • Journal of Materials 11

    Table1:Con

    tinued.

    CO

    MPO

    SITE

    /HYB

    RID

    MO

    DEL

    SCA

    FFO

    LDPR

    OPE

    RTIE

    SC

    ELL/

    TISS

    UE

    INTE

    GRA

    TIO

    N

    PLG

    Ana

    nofib

    er/S

    ilkm

    icro

    fiber

    [45]

    Invitro(BMSC

    s)

    Max

    imum

    load

    ofun

    seeded

    scaff

    olds

    75.3±4.79

    Non

    day0,

    and61.5±3.43

    Non

    day21

    Max

    imum

    load

    ofrolledseeded

    scaff

    olds

    68.2±6.72

    Non

    day21

    Stiffnessofun

    seeded

    scaff

    olds

    4.8±0.52

    N/m

    mon

    day0,

    and5.9±0.54

    N/m

    mon

    day21

    Stiffnessofrolledseeded

    scaff

    olds

    5.5±0.30

    N/m

    mon

    day21

    No

    repo

    rtof

    othe

    rmec

    hani

    calp

    rope

    rtie

    s.

    (i)Dualsurface

    seeded

    scaffolds

    show

    edsig

    nificantly

    high

    ercell

    proliferatio

    nratesc

    omparedto

    singles

    urface

    seeding.

    (ii)Increased

    cellproliferatio

    nbetween14

    days

    and21

    days

    after

    cultu

    re.

    (iii)Ro

    lled-up

    scaffolds

    hadno

    nsignificantly

    lowe

    rcellproliferation

    rates.

    PLG

    Ana

    nofib

    er/b

    FGF

    /Silk

    mic

    rofib

    er[46]

    Invitro

    (mesenchym

    alprogenito

    rcell)

    3we

    eksa

    fterc

    ulture

    ofrolledscaffolds:

    Max

    imum

    load

    Unseeded

    scaff

    olds∗we

    reabou

    t61.5

    NSeeded

    bFGF-fre

    escaffolds∗

    were

    abou

    t68.2

    N Seeded

    bFGFscaff

    olds∗we

    reabou

    t82.7N

    Stiffness

    Unseeded

    scaff

    olds∗we

    reabou

    t5.92N/m

    mSeeded

    bFGF-fre

    escaffolds∗

    were

    abou

    t5.53

    N/m

    mSeeded

    bFGFscaff

    olds∗we

    reabou

    t6.97

    N/m

    m

    (i)Sign

    ificantlyhigh

    ercellviability

    intheb

    FGFscaffolds

    compared

    tobF

    GF-fre

    escaffo

    lds.

    (ii)S

    ignificantly

    high

    ercollagens

    Iand

    III,fi

    bron

    ectin

    ,and

    biglycan

    14days

    after

    cultu

    rein

    theb

    FGFscaffolds

    comparedto

    bFGFfre

    escaffolds.

    (iii)Sign

    ificantlyhigh

    ercollagencontentintheb

    FGFscaffolds

    comparedto

    bFGFfre

    escaffo

    ldsb

    y3r

    dwe

    ekaft

    ercultu

    re.

  • 12 Journal of Materials

    Table1:Con

    tinued.

    CO

    MPO

    SITE

    /HYB

    RID

    MO

    DEL

    SCA

    FFO

    LDPR

    OPE

    RTIE

    SC

    ELL/

    TISS

    UE

    INTE

    GRA

    TIO

    N

    Alg

    inat

    e/0.

    1%ch

    itosa

    n[47]

    Invitro(fi

    broblast)

    Tensile

    strengthwas

    235.2±

    8.5MPa

    Max

    imum

    strainwas

    12.3±0.3%

    (i)Sign

    ificantlylowe

    rnum

    bero

    funatta

    ched

    cells

    inalginate–chitosangrou

    pcomparedto

    polyglactin

    andalginatealon

    e.(ii)F

    ibroblastswe

    respread

    onthep

    olym

    erfib

    ers.

    (iii)Prom

    inentcollagentype

    Iprodu

    ction14

    days

    after

    cultu

    rewas

    moreo

    nthes

    caffo

    ldsurfa

    ceby

    immun

    estainingwith

    noclear

    visualizationof

    both

    typesIIa

    ndIII

    collagen.

    Chi

    tosa

    n/0

    .1%hy

    alur

    onic

    acid

    [48]

    Invitro(fi

    broblast)

    Invivo

    (rat)

    (i)In

    vitr

    o:Tensile

    strength:

    Before

    seedingwas

    213.3±10

    MPa

    2hrsa

    fterseeding

    was

    60±6.7MPa

    14days

    after

    seedingwas

    66.7±6.8MPa

    28days

    after

    seedingwas

    65.1±6.6MPa

    Max

    imum

    strain:

    Before

    seedingwas

    3.2±0.6%

    (ii)I

    nvi

    vote

    ndon

    mod

    el:

    Tangentm

    odulus

    forc

    ell-seededscaff

    old:

    4we

    eksa

    fterimplantatio

    n:abou

    t58±MPa

    12we

    eksa

    fterimplantatio

    n:abou

    t85±MPa

    Invi

    volig

    amen

    tmod

    el:

    Max

    imum

    load

    forc

    ell-seededscaff

    old:

    12we

    eksa

    fterimplantatio

    n:abou

    t110±10

    N

    Invitroup

    to28

    days

    after

    cellseeding:

    (i)Grossob

    servationof

    ECM

    prod

    uctio

    nby

    light

    microscop

    y.(ii)P

    rominentcollagentype

    Iprodu

    ction14

    days

    after

    cultu

    remore

    onthes

    caffo

    ldsurfa

    ce.

    Invivo

    tend

    onmod

    el:

    (i)Ty

    peIc

    ollagenisseen

    onlyin

    cell-seeded

    scaffold.

    Invivo

    ligam

    entm

    odel:

    Non

    e.

  • Journal of Materials 13

    to achieve a more efficient sericin removal. This resulted insmoother fibre surfaces and preserved the generalmechanicalproperties of the silk. In vitro studies using rabbit bonemarrow-derivedMSCs revealed good cell viability dependingon the seeding technique applied (single or dual surfaceseeding) and whether scaffolds had a flat or rolled/ cylindricalmorphology. Rolled structures had lower cell proliferationcompared to the other constructs but, overall, the electrospunPLGA polymer provided a large surface area for cell prolifer-ation. To increase tenocyte differentiation of bone marrow-derivedMSCs, authors reported amodified protocol inwhichthe scaffold has a 1-week release of basic fibroblast growthfactor (bFGF) [45]. bFGFwas blendedwith PLGAand bovineserum albumin prior to electrospinning with knitted silkfibres. Results showed that incorporating bFGF was associ-ated with upregulation of tenogenic markers during MSCdifferentiation. Collagen expression was also increased, andthis contributed to improved mechanical properties of thescaffold. The combined effect of growth factor incorporationtogether with dynamic culturing conditions would be ofinterest for its effect over MSC differentiation and overallconstruct incorporation.

    Sharifi-AghdamM. et al. [46] have investigated a compos-ite scaffold consisting of knitted silk coated with electrospuncollagen-I/polyurethane nanofibres. The main aim of thisapproach was to incorporate a polyurethane polymer layerto increase the attachment of collagen to silk fibres. Invitro testing using seeded human fibroblast showed thatcomposite scaffold maintained adequate cellular metabolicactivity. Assessment of mechanical properties showed anultimate stress profile reaching 13.5±1.5MPa and Young’smodulus of 21.7±4MPa. However, further assessment ofseeded constructs for their biocompatibility and cell functionwas not investigated.

    Investigative work showed the incorporation of silk withnanofibres derived either from Polycaprolactone (PCL) orPoly(3-hydroxybutyrate) (P3HB) [41].The composite scaffoldwas fabricated through electrospinning process of differentpolymer material over twisted silk fibroin fibres. The mainaim of this approach was to incorporate nanofibrous struc-tures onto the composite scaffold to increase the surfaceto volume ratio and therefore increase cellular attachment.Authors showed a composite scaffold with no toxic effect ofseeded fibroblasts with good cellular viability up to day 3after seeding. Mechanical assessment of fabricated constructshowed amaximum load of 97.6±11.4 N for silk fibroin/P3HBand 110.5±6.6 N for silk fibroin/PCL with no statisticaldifference in between. Authors, however, did not furtherinvestigate the effect of cellular functions or matrix produc-tion or the effect of cell seeding on associated mechanicalproperties.

    Others have focused on the modification of silk toproduce scaffolds for tendon regeneration [42]. Degummedsilk fibroin meshes were integrated with electrospun alignedsilk fibroin cores [42]. In vitro assessment with rabbit MSCsinvestigated the effects of aligned fabrication techniques,compared to random fabrication, under static and dynamicculture conditions. Results showed that the effects ofmechan-ical stimulation on MSCs was intensified during culture

    on aligned silk fibroin scaffolds. The dynamic effect wasapplied in both translational and rotational movement tomimic in vivo environment. This enhanced cellular pro-liferation and remodelling, with an overall improvementin mechanical properties. Apart from the material used,presence of the aligned scaffold core was proven to beessential for these positive effects when compared to thesame scaffolds without alignment. Several authors haveshowed a similar effect when topography was introducedto the surface of a polymeric scaffold with improvement incellular alignment and collagen content as well as the expres-sion of different tendon-related extracellular matrix proteins[43, 50].

    Elsewhere, collagen has been combined with variouspolymers through a range of manufacturing techniques tosimulate the heterogeneous nature of tendon tissues. Col-lagen type I was electrospun with synthetic poly(L-lactide-co-caprolactone) at a 10:90 ratio, respectively [47]. Fibreswere twisted into nanoyarn to produce 150 𝜇mthick scaffolds.Scaffolds aligned randomly or in nanofibers were tested forporosity, surface morphology, and adhesion of tenocytes.Results showed improved cell proliferation in nanoyarnscaffolds, but with poor mechanical properties not useful forfuture clinical applicability.

    In a follow-up study, tendon-derived stem cells (TDSCs)were harvested from rabbit patellar tendons and seeded ontothe fibrous scaffolds [48]. In vitro and in vivo assessmentunder static and dynamic conditions revealed that the com-posite scaffolds seeded with TDSCs were a promising meansfor neotendon formation. Furthermore, mechanical dynamicstimulation of the cell-seeded constructs could significantlypromote tendon regeneration compared to static cultureconditions.

    Sensini A. et al. have investigated an electrospun bun-dled scaffold containing PLA and collagen type I [51]. Thisscaffold had sufficient mechanical properties with blendscontaining PLA/collagen 75:25 reaching aYoung’smodulus of98.6±12.4MPa as spun bundles and 205.1±73.0MPa after 14days of immersion inPBS.Maximumstress was 14.2±0.7 MPaas spun bundles and 6.8±0.6MPa after 14 days in PBSimmersion. Tenocytes were metabolically active with goodcellular alignment more on blends containing PLA/collagen50:50 ratio.

    Collagen type I has been integrated also with other com-posite synthetic polymers blends. Polycaprolactone (PCL)/collagen and poly L-lactide (PLLA)/collagen were evalu-ated as potential composite materials for tendon-musclejunction tissue engineering [52]. Triphasic scaffolds werefabricated with regions primarily composed of PCL/collagenin one part, PLLA/collagen on the other part, and a mix-ture of both composites in the middle. All constructs hadgood biodegradability and biocompatibility. PCL supportedmyoblast growth due to its low stiffness profile and thehigher stiffness of PLLA encouraged fibroblast prolifera-tion. Scaffolds were manufactured using electrospinningtechnique combined with glutaraldehyde cross-linking andcollagen to increase mechanical strength and cell attach-ment, respectively. In vitro, scaffolds presented good cellintegration, viability, and formation of myotubes as those

  • 14 Journal of Materials

    found in tendon-muscle junctions in vivo. Aligned col-lagen scaffolds were produced with surface cover havingpolydioxanone (PDS) nanoplates [32]. Assessment in rabbitAchilles tendon demonstrated the same biocompatibility asthe previously tested electrospun collagen scaffolds. Furtherdetailed assessment of the composite with and without PDSshowed significant improvement in water uptake and release.Histological evaluation showed good tenocyte alignment,neotendon formation, and an initial increase in the inflam-matory cell response. The addition of PDS sheets resultedin decreased peritendinous adhesions with higher numbersof mature tenocytes and increased collagen fibril alignment.Improved mechanical properties of the construct were alsoobserved compared to scaffolds made of collagen fibres only[32, 50].

    PLLA was also utilised to fabricate a composite scaffoldin which electrospinning process was used to form analigned nanofibres that was later on cross-linked to chitosan-collagen hydrogel mimicking the extracellular matrix ofnative tendons [53]. The scaffold was rolled and coatedon the outer surface with alginate gel aiming to producean antiadhesion layer around the construct. The scaffoldsupported cellular alignment and proliferation of tenocyteswith no toxic effect. Additionally, adsorption tests showedsignificantly less attached proteins on the coated surfacecompared to the noncoated one. Mechanical assessmentof produced scaffold showed no effect of coating processon tensile strength of produced scaffold with an effect onthe layer number having a value around 2MPa for 2-layercoated and uncoated scaffolds whereas it was around 6MPafor uncoated and 4MPa for coated 3-layer scaffolds. Thedegradation profile of the scaffold was also investigated andshowed that scaffolds maintained 50% of their substance at21 days after incubation with PBS containing 104 units/mllysozyme solution.

    E.C. Green et al. [54] investigated the fabrication ofcollagen-I/nanocarbon fibres composite scaffold for potentialtendon tissue engineering application. Fibres were madethrough gel-spinning process with the use of a filling load of0.5 and 5wt%.Thiswas followed by fibre elongation at a strainrate of 0.02mm/s and subsequent glutaraldehyde (GA) cross-linking. Material characterization showed a yielded fibreconstruct similar to native tendon collagen with enhancedmechanical properties.

    3.5. Similarities and Dissimilarities between Tendons andLigaments. Tendons and ligaments have similar structureswith different fundamental properties and functions. Ten-dons are fibrous inelastic structures that connect muscles tobones within joints while ligaments are fibrous but flexiblestructures important for supporting bone and cartilage. On astructural level, both connective tissues are dense with vari-able cellular and proteomic elements. Mechanical analysis ofhuman tendons and ligaments showed that the maximumtensile strength ranges from 4.4 to 660MPa depending ondifferent locations [55, 56]. The maximum strain of theseconnective tissues was shown to range between 18 and 30%.Young’smoduluswas shown to range between 0.2 and 1.5 GPa[57, 58]. Structurally, tendons are predominantly composed

    of a collagen type I matrix containing tenocytes andtenoblasts. In contrast, ligaments contain glycosaminoglycanand lower levels of collagen compared to tendon tissue,with fibroblasts being the main cellular element (Figure 2)[59, 60]. Kharaz Y. et al. compared the extracellular matrixcomposition of both, natural and tissue engineered, tendonsand ligaments [61]. Results showed that, although tissueengineered constructs share the same composition with thenative tissue in variable proportions, fundamental differencesexist. Specific proteins, such as asporin and tenomodulin,were limited to tendons, while versican, proteoglycan 4,and SOD3 were ligament-specific (Figure 1). Identifyingdifferences in structural protein expression indicates thattissue engineering approaches should aim to replicate thesedistinctions at the proteomic level. To date, this has not alwaysbeen the case, and the terms tendon and ligament are oftenused interchangeably in the literature. This is predominantlybecause the two connective tissues exhibit similar functionaland mechanical properties [12]. An important concept toremember is that cell source is fundamental in dictatingthe type of the matrix produced [61]. This highlights theintrinsic cell memory that is different between tendon andligament. In order to tissue engineer these specific tissues,it is important to consider their functional differences sothat biomimicry can be achieved. This will eventually helpin enhancing integration and function to be restored whenused to replace injured tendon or ligaments. From this, it isclear that although the two tissues share several features, adifference exist.

    4. Conclusion and Future Directions

    This review summarises the current strategies based oncomposite biomaterials for tendon and ligaments tissueengineering. This approach represents a way to address theheterogeneous nature shared between tendon and ligaments.Although both structures share similar mechanical prop-erties, their constituting cell-types and extracellular matrixcompositions are different. Currently, for tendon tissue engi-neering, several challenges need to be addressed. First, thenecessity for a single standard evaluation identifying themechanical requirements for successful tendon regenerationis lacking. Crucially, this should incorporate various forms ofmechanical assessment (including strain and rotation) withdifferent tissue engineering approaches to mimic the naturalenvironment. This is difficult to achieve and implementbecause of the heterogeneous nature of tendons. The secondchallenge is to address and control the response of thesurrounding tissue to the implanted scaffold. To date, this hasbeen limited to observation of the intrinsic healing responseduring in vivo applications. Additionally, most of the studiesinvestigating the use of composite materials for tendon andligament tissue engineering utilise small animals like rabbitsrather than larger models like sheep or horses [62]. Suchlimitations are based on significant weight and mechanical-related difference between in vivo models and native humantissues. Furthermore, the ability to fix the produced constructin situ (with suture, anchors) and shelf life availability shouldbe considered whenever a model is being tested. In fact

  • Journal of Materials 15

    TendonLigament

    Ligaments act to connect bones together around joints while tendons act to connect muscles to bones

    Cell type Tenocytes (Fibroblast like cells).Fibroblasts.

    Native ECM

    EngineeredconstructsECM

    MechanicalProperties

    -Less collagen content. -Glycosaminoglycan. -More water content.

    -Asporin.-Tenomodulin.

    -Abundant collagen content. -Proteoglycan.-Elastin.

    - Versican.- Proteoglycan 4. - SOD3.

    -Maximum tensile strength ranges from 4.4 up to 660 MPa for both tissue types. -Maximum strain of both structures ranges between 18 and 30%.-Estimated Young’s modulus ranges between 0.2 and 1.5 GPa for both.

    Figure 2: Comparison between natural and tissue engineered tendon and ligament constructs. Unique cellular and extracellular matrixcomposition that is different between the two types of tissues is important for future clinical translation of tendon and ligament research.SOD3, superoxide dismutase;MPa, mega-Pascal; GPa, giga-Pascal.

    the interface scaffold-tendon or scaffold-bone is the placeat risk of rupture after implantation rather the scaffoldsitself.

    The described challenges constitute a potential issue forfuture clinical application of tissue engineered tendon andligament solutions. Further research is required on the rolesof composite materials in order to mimic the appropriatestructural, mechanical, and functional characteristics of ten-don and ligaments. While synthetic materials are being usedcurrently in clinical practice for tendon and ligament repairs,it will be essential to mechanically match their propertiesto native tissue. To mimic the mechanical properties oftendon is particularly important as it would allow fasterphysiotherapy and therefor reduce scar tissue formationwhich can evolve to articular stiffness. Moreover, this willenhance integration and healing at injury site. The use ofcertain growth factors such as basic fibroblast growth factor(bFGF) [45] can also be employed during surgery to enhancetissue integration. However this requires a strict control andregulated environment. Since advanced composite materialscan be tailored to match different mechanical properties ofnative tissue, they can be incorporated with growth factorsand employed with and without cells, providing large numberof possibilities for their clinical use based on site and extentof injuries.

    Conflicts of Interest

    None of the authors have any commercial associations orfinancial relationships that would create a conflicts of interestwith the work presented in this article.

    Acknowledgments

    This study was funded by Royal Free Charity (Award no.167275).

    References

    [1] A. P. Breidenbach, S. D. Gilday, A. L. Lalley et al., “Functionaltissue engineering of tendon: Establishing biological successcriteria for improving tendon repair,” Journal of Biomechanics,vol. 47, no. 9, pp. 1941–1948, 2014.

    [2] U. G. Longo, G. Garau, V. Denaro, and N. Maffulli, “Surgicalmanagement of tendinopathy of biceps femoris tendon inathletes,” Disability and Rehabilitation, vol. 30, no. 20-22, pp.1602–1607, 2008.

    [3] C.-F. Liu, L. Aschbacher-Smith, N. J. Barthelery, N. Dyment,D. Butler, and C. Wylie, “What we should know before usingtissue engineering techniques to repair injured tendons: adevelopmental biology perspective,” Tissue Engineering Part B:Reviews, vol. 17, no. 3, pp. 165–176, 2011.

  • 16 Journal of Materials

    [4] J. H.-C. Wang, Q. Guo, and B. Li, “Tendon biomechanics andmechanobiology—a minireview of basic concepts and recentadvancements,” Journal of HandTherapy, vol. 25, no. 2, pp. 133–141, 2012.

    [5] T. W. Lin, L. Cardenas, and L. J. Soslowsky, “Biomechanics oftendon injury and repair,” Journal of Biomechanics, vol. 37, no.6, pp. 865–877, 2004.

    [6] J. Kastelic, A. Galeski, and E. Baer, “The multicompositestructure of tendon,” Connective Tissue Research, vol. 6, no. 1,pp. 11–23, 1978.

    [7] J.W. Freeman,M. D.Woods, D. A. Cromer et al., “Evaluation ofa hydrogel-fiber composite for ACL tissue engineering,” Journalof Biomechanics, vol. 44, no. 4, pp. 694–699, 2011.

    [8] H. H. Lu and S. Thomopoulos, “Functional attachment of softtissues to bone: Development, healing, and tissue engineering,”Annual Review of Biomedical Engineering, vol. 15, pp. 201–226,2013.

    [9] K. Atesok, M. N. Doral, J. Karlsson et al., “Multilayer scaffoldsin orthopaedic tissue engineering,” Knee Surgery, Sports Trau-matology, Arthroscopy, pp. 1–9, 2014.

    [10] Z. A. Glass, N. R. Schiele, and C. K. Kuo, “Informing tendontissue engineering with embryonic development,” Journal ofBiomechanics, vol. 47, no. 9, pp. 1964–1968, 2014.

    [11] V. A. Tsiampa, I. Ignatiadis, A. Papalois, P. Givissis, A.Christodoulou, and J. Fridén, “Structural and mechanicalintegrity of tendon-to-tendon attachments used in upper limbtendon transfer surgery,” Journal of Plastic Surgery and HandSurgery, vol. 46, no. 3-4, pp. 262–266, 2012.

    [12] M. T. Rodrigues, R. L. Reis, and M. E. Gomes, “Engineeringtendon and ligament tissues: Present developments towardssuccessful clinical products,” Journal of Tissue Engineering andRegenerative Medicine, vol. 7, no. 9, pp. 673–686, 2013.

    [13] A. Melvin, A. Litsky, J. Mayerson, D. Witte, D. Melvin, andN. Juncosa-Melvin, “An artificial tendon with durable muscleinterface,” Journal of Orthopaedic Research, vol. 28, no. 2, pp.218–224, 2010.

    [14] K. Gao, S. Chen, L. Wang et al., “Anterior cruciate ligamentreconstruction with LARS artificial ligament: a multicenterstudy with 3-to 5-year follow-up,” Arthroscopy: The Journal ofArthroscopic & Related Surgery, vol. 26, no. 4, pp. 515–523, 2010(Chinese).

    [15] L. E. Paulos, T. D. Rosenberg, S. R. Grewe, D. S. Tearse, andC. L.Beck, “TheGORE-TEX anterior cruciate ligament prosthesis: Along-term followup,” The American Journal of Sports Medicine,vol. 20, no. 3, pp. 246–252, 1992.

    [16] U. G. Longo, A. Lamberti, N. Maffulli, and V. Denaro, “Tendonaugmentation grafts: a systematic review,” British Medical Bul-letin, vol. 94, no. 1, pp. 165–188, 2010.

    [17] C. M. Glezos, A. Waller, H. E. Bourke, L. J. Salmon, and L. A.Pinczewski, “Disabling synovitis associated with lars artificialligament use in anterior cruciate ligament reconstruction: Acase report,” The American Journal of Sports Medicine, vol. 40,no. 5, pp. 1167–1171, 2012.

    [18] Y. Liu, H. S. Ramanath, and D.-A. Wang, “Tendon tissueengineering using scaffold enhancing strategies,” Trends inBiotechnology, vol. 26, no. 4, pp. 201–209, 2008.

    [19] A. K. Lynn, I. V. Yannas, and W. Bonfield, “Antigenicity andimmunogenicity of collagen,” Journal of Biomedical MaterialsResearch Part B: Applied Biomaterials, vol. 71, no. 2, pp. 343–354,2004.

    [20] H. W. Ouyang, J. C. H. Goh, A. Thambyah, S. H. Teoh, and E.H. Lee, “Knitted poly-lactide-co-glycolide scaffold loaded withbone marrow stromal cells in repair and regeneration of rabbitachilles tendon,”Tissue Engineering Part A, vol. 9, no. 3, pp. 431–439, 2003.

    [21] M. G. Galvez, C. Crowe, S. Farnebo, and J. Chang, “Tissueengineering in flexor tendon surgery: Current state and futureadvances,” Journal of Hand Surgery (European Volume), vol. 39,no. 1, pp. 71–78, 2014.

    [22] P. Y. Mengsteab, L. S. Nair, and C. T. Laurencin, “The past,present and future of ligament regenerative engineering,” Jour-nal of Regenerative Medicine, vol. 11, no. 8, pp. 871–881, 2016.

    [23] L. M. Cross, A. Thakur, N. A. Jalili, M. Detamore, and A. K.Gaharwar, “Nanoengineered biomaterials for repair and regen-eration of orthopedic tissue interfaces,” Acta Biomaterialia, vol.42, pp. 2–17, 2016.

    [24] S. Font Tellado, E. R. Balmayor, and M. Van Griensven,“Strategies to engineer tendon/ligament-to-bone interface: Bio-materials, cells and growth factors,” Advanced Drug DeliveryReviews, vol. 94, pp. 126–140, 2015.

    [25] V. Correia Pinto, R. Costa-Almeida, I. Rodrigues, L. Guardão,R. Soares, and R. Miranda Guedes, “Exploring the in vitroand in vivo compatibility of PLA, PLA/GNP and PLA/CNT-COOH biodegradable nanocomposites: Prospects for tendonand ligament applications,” Journal of Biomedical MaterialsResearch Part A, vol. 105, no. 8, pp. 2182–2190, 2017.

    [26] V. C. Pinto, T. Ramos, A. S. F. Alves et al., “Dispersionand failure analysis of PLA, PLA/GNP and PLA/CNT-COOHbiodegradable nanocomposites by SEM and DIC inspection,”Engineering Failure Analysis, vol. 71, pp. 63–71, 2017.

    [27] M. Sanami, Z. Shtein, I. Sweeney et al., “Biophysical and biolog-ical characterisation of collagen/resilin-like protein compositefibres,” Biomedical Materials, vol. 10, no. 6, 2015.

    [28] B. M. Baker, N. L. Nerurkar, J. A. Burdick, D. M. Elliott, and R.L.Mauck, “Fabrication andmodeling of dynamic multipolymernanofibrous scaffolds,” Journal of Biomechanical Engineering,vol. 131, no. 10, Article ID 101012-1, 2009.

    [29] S. Sahoo, J. G. Cho-Hong, and T. Siew-Lok, “Development ofhybrid polymer scaffolds for potential applications in ligamentand tendon tissue engineering,” Biomedical Materials, vol. 2, no.3, pp. 169–173, 2007.

    [30] S. R. Caliari, M. A. Ramirez, and B. A. C. Harley, “Thedevelopment of collagen-GAG scaffold-membrane compositesfor tendon tissue engineering,” Biomaterials, vol. 32, no. 34, pp.8990–8998, 2011.

    [31] T.Majima, T. Irie, N. Sawaguchi et al., “Chitosan-based hyaluro-nan hybrid polymer fibre scaffold for ligament and tendontissue engineering,” Proceedings of the Institution of MechanicalEngineers, Part H: Journal of Engineering in Medicine, vol. 221,no. 5, pp. 537–546, 2007.

    [32] A. Oryan, A. Moshiri, A. M. Parizi, and N. Maffulli, “Implan-tation of a novel biologic and hybridized tissue engineeredbioimplant in large tendon defect: An in vivo investigation,”Tissue Engineering Part: A, vol. 20, no. 3-4, pp. 447–465, 2014.

    [33] T.Majima, T. Funakosi, N. Iwasaki et al., “Alginate and chitosanpolyion complex hybrid fibers for scaffolds in ligament andtendon tissue engineering,” Journal of Orthopaedic Science, vol.10, no. 3, pp. 302–307, 2005.

    [34] G. Yang, H. Lin, B. B. Rothrauff, S. Yu, and R. S. Tuan, “Mul-tilayered polycaprolactone/gelatin fiber-hydrogel composite fortendon tissue engineering,” Acta Biomaterialia, vol. 35, pp. 68–76, 2016.

  • Journal of Materials 17

    [35] N. Sawaguchi, T. Majima, T. Funakoshi et al., “Effect of cyclicthree-dimensional strain on cell proliferation and collagen syn-thesis of fibroblast-seeded chitosan-hyaluronan hybrid polymerfiber,” Journal of Orthopaedic Science, vol. 15, no. 4, pp. 569–577,2010.

    [36] J. H. Shepherd, S. Ghose, S. J. Kew, A. Moavenian, S. M. Best,and R. E. Cameron, “Effect of fiber crosslinking on collagen-fiber reinforced collagen-chondroitin-6-sulfate materials forregenerating load-bearing soft tissues,” Journal of BiomedicalMaterials Research Part A, vol. 101, no. 1, pp. 176–184, 2013.

    [37] J. L. Chen, Z. Yin, W. L. Shen et al., “Efficacy of hESC-MSCs inknitted silk-collagen scaffold for tendon tissue engineering andtheir roles,” Biomaterials, vol. 31, no. 36, pp. 9438–9451, 2010.

    [38] W. Shen, X. Chen, J. Chen et al., “The effect of incorporationof exogenous stromal cell-derived factor-1 alpha within aknitted silk-collagen sponge scaffold on tendon regeneration,”Biomaterials, vol. 31, no. 28, pp. 7239–7249, 2010.

    [39] X. Chen, Z. Yin, and J.-L. Chen, “Scleraxis-overexpressedhuman embryonic stem cell–derived mesenchymal stem cellsfor tendon tissue engineering with knitted silk-collagen scaf-fold,” Tissue Engineering Part A, vol. 20, no. 11-12, pp. 1583–1592,2014.

    [40] S. Sahoo, S. Lok Toh, and J. C. Hong Goh, “PLGA nanofiber-coated silk microfibrous scaffold for connective tissue engineer-ing,” Journal of Biomedical Materials Research Part B: AppliedBiomaterials, vol. 95, no. 1, pp. 19–28, 2010.

    [41] E. Naghashzargar, S. Farè, V. Catto et al., “Nano/micro hybridscaffold of PCL or P3Hb nanofibers combined with silk fibroinfor tendon and ligament tissue engineering,” Journal of AppliedBiomaterials and Functional Materials, vol. 13, no. 2, pp. e156–e168, 2015.

    [42] T. K. H. Teh, S.-L. Toh, and J. C. H. Goh, “Aligned fibrousscaffolds for enhanced mechanoresponse and tenogenesis ofmesenchymal stem cells,” Tissue Engineering Part A, vol. 19, no.11-12, pp. 1360–1372, 2013.

    [43] F. Alshomer, C. Chaves, T. Serra, I. Ahmed, and D. M. Kalaskar,“Micropatterning of nanocomposite polymer scaffolds usingsacrificial phosphate glass fibers for tendon tissue engineer-ing applications,” Nanomedicine: Nanotechnology, Biology andMedicine, vol. 13, no. 3, pp. 1267–1277, 2017.

    [44] D. Deng, W. Wang, B. Wang et al., “Repair of Achilles tendondefect with autologous ASCs engineered tendon in a rabbitmodel,” Biomaterials, vol. 35, no. 31, pp. 8801–8809, 2014.

    [45] S. Sahoo, S. L. Toh, and J. C. H. Goh, “A bFGF-releasingsilk/PLGA-based biohybrid scaffold for ligament/tendon tissueengineering using mesenchymal progenitor cells,” Biomaterials,vol. 31, no. 11, pp. 2990–2998, 2010.

    [46] M. Sharifi-Aghdam, R. Faridi-Majidi, M. A. Derakhshan, A.Chegeni, and M. Azami, “Preparation of collagen/polyure-thane/knitted silk as a composite scaffold for tendon tissue engi-neering,” Proceedings of the Institution of Mechanical Engineers,Part H: Journal of Engineering in Medicine, vol. 231, no. 7, pp.652–662, 2017.

    [47] Y. Xu, J. Wu, H. Wang et al., “Fabrication of electrospun poly(L-lactide-co-𝜀-caprolactone)/collagen nanoyarn network as anovel, three-dimensional, macroporous, aligned scaffold fortendon tissue engineering,” Tissue Engineering Part C: Methods,vol. 19, no. 12, pp. 925–936, 2013.

    [48] Y. Xu, S. Dong, Q. Zhou et al., “The effect of mechanicalstimulation on the maturation of TDSCs-poly(L-lactide-co-e-caprolactone)/collagen scaffold constructs for tendon tissueengineering,” Biomaterials, vol. 35, no. 9, pp. 2760–2772, 2014.

    [49] J. T. Shearn, K. R. C. Kinneberg, N. A. Dyment et al., “Tendontissue engineering: Progress, challenges, and translation to theclinic,” Journal ofMusculoskeletal andNeuronal Interactions, vol.11, no. 2, pp. 163–173, 2011.

    [50] V. Kishore, W. Bullock, X. Sun, W. S. Van Dyke, and O.Akkus, “Tenogenic differentiation of human MSCs induced bythe topography of electrochemically aligned collagen threads,”Biomaterials, vol. 33, no. 7, pp. 2137–2144, 2012.

    [51] A. Sensini, C. Gualandi, L. Cristofolini et al., “Biofabricationof bundles of poly(lactic acid)-collagen blends mimicking thefascicles of the humanAchille tendon,”Biofabrication, vol. 9, no.1, Article ID 015025, 2017.

    [52] M. R. Ladd, S. J. Lee, J. D. Stitzel, A. Atala, and J. J. Yoo, “Co-electrospun dual scaffolding system with potential for muscle-tendon junction tissue engineering,” Biomaterials, vol. 32, no. 6,pp. 1549–1559, 2011.

    [53] S. Deepthi, M. Nivedhitha Sundaram, J. Deepti Kadavan, and R.Jayakumar, “Layered chitosan-collagen hydrogel/aligned PLLAnanofiber construct for flexor tendon regeneration,” Carbohy-drate Polymers, vol. 153, pp. 492–500, 2016.

    [54] E. C. Green, Y. Zhang, H. Li, and M. L. Minus, “Gel-spinningof mimetic collagen and collagen/nano-carbon fibers: Under-standingmulti-scale influences onmolecular ordering andfibrilalignment,” Journal of the Mechanical Behavior of BiomedicalMaterials, vol. 65, pp. 552–564, 2017.

    [55] R. De Santis, F. Sarracino, F. Mollica, P. A. Netti, L. Ambrosio,and L. Nicolais, “Continuous fibre reinforced polymers as con-nective tissue replacement,”Composites Science and Technology,vol. 64, no. 6, pp. 861–871, 2004.

    [56] G. A. Johnson, D. M. Tramaglini, R. E. Levine, K. Ohno, N.-Y.Choi, and S. L.-Y. Woo, “Tensile and viscoelastic properties ofhuman patellar tendon,” Journal of Orthopaedic Research, vol.12, no. 6, pp. 796–803, 1994.

    [57] J. Peltonen, N. J. Cronin, J. Avela, and T. Finni, “In vivomechanical response of human Achilles tendon to a single boutof hopping exercise,” Journal of Experimental Biology, vol. 213,no. 8, pp. 1259–1265, 2010.

    [58] R. B. Svensson, P. Hansen, T. Hassenkam et al., “Mechanicalproperties of human patellar tendon at the hierarchical levelsof tendon and fibril,” Journal of Applied Physiology, vol. 112, no.3, pp. 419–426, 2012.

    [59] K. Gelse, E. Pöschl, and T. Aigner, “Collagens—structure,function, and biosynthesis,” Advanced Drug Delivery Reviews,vol. 55, no. 12, pp. 1531–1546, 2003.

    [60] G. Y. F. Ng, “Ligament injury and repair: Current concepts,”Hong Kong Physiotherapy Journal, vol. 20, pp. 22–29, 2002.

    [61] Y. A. Kharaz, S. Tew, E. Canty-Laird, and E. Comerford, “Acomparison of the extracellular matrix composition of ten-don/ligament tissue and tissue engineered tendon and ligamentconstruct,”Osteoarthritis and Cartilage, vol. 22, p. S313, 2014.

    [62] J. E. Carpenter, S. Thomopoulos, and L. J. Soslowsky, “Animalmodels of tendon and ligament injuries for tissue engineeringapplications,” Clinical Orthopaedics and Related Research, no.367, pp. S296–S311, 1999.

  • CorrosionInternational Journal of

    Hindawiwww.hindawi.com Volume 2018

    Advances in

    Materials Science and EngineeringHindawiwww.hindawi.com Volume 2018

    Hindawiwww.hindawi.com Volume 2018

    Journal of

    Chemistry

    Analytical ChemistryInternational Journal of

    Hindawiwww.hindawi.com Volume 2018

    Scienti�caHindawiwww.hindawi.com Volume 2018

    Polymer ScienceInternational Journal of

    Hindawiwww.hindawi.com Volume 2018

    Hindawiwww.hindawi.com Volume 2018

    Advances in Condensed Matter Physics

    Hindawiwww.hindawi.com Volume 2018

    International Journal of

    BiomaterialsHindawiwww.hindawi.com

    Journal ofEngineeringVolume 2018

    Applied ChemistryJournal of

    Hindawiwww.hindawi.com Volume 2018

    NanotechnologyHindawiwww.hindawi.com Volume 2018

    Journal of

    Hindawiwww.hindawi.com Volume 2018

    High Energy PhysicsAdvances in

    Hindawi Publishing Corporation http://www.hindawi.com Volume 2013Hindawiwww.hindawi.com

    The Scientific World Journal

    Volume 2018

    TribologyAdvances in

    Hindawiwww.hindawi.com Volume 2018

    Hindawiwww.hindawi.com Volume 2018

    ChemistryAdvances in

    Hindawiwww.hindawi.com Volume 2018

    Advances inPhysical Chemistry

    Hindawiwww.hindawi.com Volume 2018

    BioMed Research InternationalMaterials

    Journal of

    Hindawiwww.hindawi.com Volume 2018

    Na

    nom

    ate

    ria

    ls

    Hindawiwww.hindawi.com Volume 2018

    Journal ofNanomaterials

    Submit your manuscripts atwww.hindawi.com

    https://www.hindawi.com/journals/ijc/https://www.hindawi.com/journals/amse/https://www.hindawi.com/journals/jchem/https://www.hindawi.com/journals/ijac/https://www.hindawi.com/journals/scientifica/https://www.hindawi.com/journals/ijps/https://www.hindawi.com/journals/acmp/https://www.hindawi.com/journals/ijbm/https://www.hindawi.com/journals/je/https://www.hindawi.com/journals/jac/https://www.hindawi.com/journals/jnt/https://www.hindawi.com/journals/ahep/https://www.hindawi.com/journals/tswj/https://www.hindawi.com/journals/at/https://www.hindawi.com/journals/ac/https://www.hindawi.com/journals/apc/https://www.hindawi.com/journals/bmri/https://www.hindawi.com/journals/jma/https://www.hindawi.com/journals/jnm/https://www.hindawi.com/https://www.hindawi.com/