You are on page 1of 9

Review Article

Tissue Engineering Solutions for


Tendon Repair

Abstract
MaCalus V. Hogan, MD Tendon injuries range from acute traumatic ruptures and
Namory Bagayoko, MD lacerations to chronic overuse injuries, such as tendinosis. Even
with improved nonsurgical, surgical, and rehabilitation techniques,
Roshan James, MS
outcomes following tendon repair are inconsistent. Primary repair
Trevor Starnes, MD, PhD remains the standard of care. However, repaired tendon tissue
Adam Katz, MD rarely achieves functionality equal to that of the preinjured state.
A. Bobby Chhabra, MD Poor results have been linked to alterations in cellular organization
within the tendon that occur at the time of injury and throughout the
early stages of healing. Enhanced understanding of the biology of
tendon healing is needed to improve management and outcomes.
The use of growth factors and mesenchymal stem cells and the
development of biocompatible scaffolds could result in enhanced
tendon healing and regeneration. Recent advances in tendon
bioengineering may lead to improved management following
tendon injury.

L ong-term outcomes are variable


following surgical repair of ten-
don lacerations and ruptures. Pri-
turning point in the quest to success-
fully manage tendon injury. Current
research efforts are focused on the
mary repair remains the standard of creation of alternative repair meth-
care. However, despite a multitude of ods using tissue-engineered substi-
suture techniques and therapy proto- tutes to enhance tendon healing, re-
cols, reproducible satisfactory results pair, and regeneration.
are difficult to achieve. Existing tech-
niques are insufficient to return ten-
don to its normal preinjury condi- Normal Tendon
tion. Repair and the healing process Maintenance and Healing
often lead to permanent alteration of
preinjury characteristics such as Extracellular Matrix
From the Department of strength, tissue structure, organiza- Extracellular matrix (ECM) and its
Orthopaedic Surgery (Dr. Hogan, tion, and composition. composition are critical to the devel-
Dr. Bagayoko, Dr. Starnes, and
Dr. Chhabra), the Department of
Tissue engineering has been de- opment and maintenance of healthy
Biomedical Engineering (Mr. James), fined as “the application of biologi- tendon.2 Tenocytes are the predomi-
and the Department of Plastic and cal, chemical and engineering princi- nant cell type in tendon, and they
Maxillofacial Surgery (Dr. Katz), ples toward the repair, restoration, produce collagen and maintain the
University of Virginia Health System,
Charlottesville, VA. or regeneration of living tissues using ECM microenvironment.3 The inter-
biomaterials, cells, and factors, alone action of these fibroblast-like cells
J Am Acad Orthop Surg 2011;19:
134-142
or in combination.”1 The ability to with the ECM can affect cell prolifer-
regenerate tendon with tissue pos- ation, migration, and development.4
Copyright 2011 by the American
sessing properties equal to those of The ECM is critical in tendon re-
Academy of Orthopaedic Surgeons.
preinjured tendon could serve as a sponse to mechanical loads and in-

134 Journal of the American Academy of Orthopaedic Surgeons


MaCalus V. Hogan, MD, et al

Table 1 cal delivery of a universal MMP in-


hibitor led to decreased collagen deg-
Major Components in Tendon Extracellular Matrix
radation and decreased histologic
Component Function changes at the tendon-bone interface
Type I collagen2 Predominant collagen type in native tendon tissue (highly following rotator cuff repair in
organized) rats.10
Type III collagen5 Present during early healing and in smaller amounts in Several MMPs and tissue inhibitors
native tendon (unorganized)
of metalloproteinases have been
Decorin3,6 Proteoglycan involved in collagen fibril organization; found
identified. Although their precise
in the tensile segments of tendon
Aggrecan6 Proteoglycan involved in collagen fibril organization; found
role is not known, it is well accepted
in compressed segments of tendon that this relationship plays a major
role in the turnover process in both
normal and damaged tendon.13
jury.5 Engineered tendon substitute Recent research has focused on the
must closely mimic the structure of role of matrix-remodeling genes in Tendon Development
ECM. tendon (Table 2). Matrix metallopro- Genes implicated in tendon cell de-
The major components of normal teinases (MMPs) are involved in the velopment and maturation are also
tendon ECM are essential to its over- degradation of tendon ECM compo- important to tissue engineering (Ta-
all function (Table 1). Type I colla- nents during development and reas- ble 3). Scleraxis is a transcription
gen predominates within the ECM. sembly following injury.9 MMPs are factor and highly specific marker of
This is the primary dry constituent of essential to tendon homeostasis and tenocyte precursor populations that
normal tendon. Type I collagen is repair through upregulation and regulates ECM gene expression in
well organized into parallel fibrils downregulation, and they counter- tendon fibroblasts.14 Tenomodulin is
that contribute to the tensile strength balance tissue inhibitors of metallo- a late marker of tenocyte differentia-
of tendon. Type III collagen is a less proteinases.11 Loiselle et al12 recently tion and proliferation, and it is up-
organized form of collagen within developed a murine model of pri- regulated by scleraxis.15 Together,
the ECM that is abundant during the mary tendon repair based on MMP scleraxis and tenomodulin expres-
earlier stages of healing and remodel- and neotendon gene expression in an sion serve as strong indicators of
ing.6 Several proteoglycans, including effort to further understand the role neotendon formation. Tenascin-C is
decorin and aggrecan, are present of MMP in the healing process. They an ECM glycoprotein that has been
within tendon ECM, and they play reported a biphasic response with implicated in tendon development at
an important role in collagen fibril MMPs. Early expression of MMP-9 both the embryonic and differenti-
organization and matrix assembly. was associated with inflammation ated cell levels.16 Smad8 is a marker
They also contribute to tendon vis- and damaged collagen at the zone of of mesenchymal stem cell (MSC) dif-
coelastic response to compressive injury. Late expression of MMP-2 ferentiation into tendon progenitor
forces.7 Water represents slightly and -14 was seen during tendon re- populations. Hoffmann et al17 dem-
more than half of tendon weight, modeling. Markers associated with onstrated that with specific manipu-
mostly within the ECM; water facili- neotendon development were ex- lations of the Smad8 signaling path-
tates gliding of collagen fibril by re- pressed later during the transition to way, MSCs could differentiate into
ducing friction in response to load- normal tendon, at 21 and 28 days.12 tenocytes. The role of Smad8 in ten-
ing.8 A recent study demonstrated that lo- don development was further sup-

Dr. Hogan or an immediate family member has received research or institutional support from the Orthopaedic Research and
Education Foundation and serves as a board member, owner, officer, or committee member of the American Academy of Orthopaedic
Surgeons Candidate, Resident, and Fellow Subcommittee and of the J. Robert Gladden Orthopaedic Society. Dr. Katz or an
immediate family member serves as a board member, owner, officer, or committee member of the International Federation of Adipose
Therapeutics and Science and serves as a paid consultant to or is an employee of MicroAire Surgical Instruments and LifeNet
Health. Dr. Chhabra or an immediate family member has received research or institutional support from the National Institutes of
Health (NIH/NIAMS) grant No. AR052891 and the Orthopaedic Research and Education Foundation, and serves as a board member,
owner, officer, or committee member of the American Society for Surgery of the Hand, the Virginia Orthopaedic Society, and Miller
Review Course. None of the following authors or any immediate family member has received anything of value from or owns stock in
a commercial company or institution related directly or indirectly to the subject of this article: Dr. Bagayoko, Mr. James, and
Dr. Starnes.

March 2011, Vol 19, No 3 135


Tissue Engineering Solutions for Tendon Repair

Table 2 ported by a recent murine model of


tendon repair that found elevated
Tendon Extracellular Matrix-remodeling Genes
Smad8 expression at 4 weeks after
Gene Function repair.12 This was associated with the
MMP (eg, -1, -2, -3, -9, -11, -13)8,9 ECM degradation during development and repair formation of neotendon and a reduc-
TIMP (ie, -1, -2, -3, -4)9,10 Reversible inhibition of MMPs tion in adhesions represented by im-
proved metatarsophalangeal joint
ECM = extracellular matrix, MMP = matrix metalloproteinases, TIMP = tissue inhibitors of range of motion.
metalloproteinases

Tendon Healing
Table 3 An understanding of the natural
Genes Specific to Tendon Development tendon-healing process is vital to the
Gene Function development of tools for modulating
tendon healing. Following tendon in-
13
Scleraxis Expressed in progenitor population and cells of tendon jury, the body begins a healing and scar
tissue (early marker)
formation response with three overlap-
Tenomodulin14 Late marker of tenocyte differentiation and proliferation;
upregulated by scleraxis
ping phases distinguished by their cel-
lular and biochemical events8 (Figure
Tenascin-C15 ECM glycoprotein present in embryonic and differentiated
tendon 1). This healing process reestablishes
Smad816 Intracellular signaling molecule in MSCs; upregulation the tendon fibers and the gliding
represents tendon morphology mechanism needed for tendon excur-
sion. Tensile strength improves with
ECM = extracellular matrix, MSCs = mesenchymal stem cells
time but never reaches the level of
uninjured tendon. This process oc-
Figure 1 curs over a course of 1 year.8

Inflammatory Stage
Hematoma formation occurs following
damage to blood vessels in the zone of
injury. Chemotactic factors such as
proinflammatory molecules and vaso-
dilators are released in response to the
hematoma and attract inflammatory
cells from surrounding tissues. Mono-
cytes, macrophages, and neutrophils
migrate to the injury site, where they
break down the clot and necrotic tissue
via phagocytosis. Macrophages also
aid in the recruitment of new fibro-
blasts and the release of proangiogenic
factors, resulting in the formation of a
new vascular network within the
wound.18 This phase is marked by an
increase in type III collagen, DNA, fi-
bronectin, glycosaminoglycan, and wa-
ter, which collectively stabilize the
ECM.8
Stages of tendon healing following injury. ECM = extracellular matrix,
GAG = glycosaminoglycans. (Adapted with permission from James R, Proliferative Stage
Kesturu G, Balian G, Chhabra AB: Tendon: Biology, biomechanics, repair, Continued recruitment and rapid
growth factors, and evolving treatment options. J Hand Surg Am
2008;33[1]:102-112.) proliferation of fibroblasts is the
hallmark of the proliferative stage.

136 Journal of the American Academy of Orthopaedic Surgeons


MaCalus V. Hogan, MD, et al

Type III collagen and DNA concen- dependent on many factors, includ- ing. Basic fibroblast growth factor is
trations peak, and the wound devel- ing the type and extent of trauma, expressed by fibroblasts and neutro-
ops a disorganized, scar-like appear- presence of a synovial sheath, tendon phils at the site of injury and func-
ance. The repair tissue is cellular, and location, and the amount of postop- tions in angiogenesis and stimulation
at the end of this stage, the tissue erative stress placed on the repair of cell-matrix interactions. Basic fi-
contains abundant water and ECM during motion. broblast growth factor is also mito-
components.19 genic in that it induces proliferation
Growth Factors of local fibroblasts and bone mar-
Remodeling Stage Many growth factors are involved in row–derived MSCs, leading to in-
Tissue begins to remodel approxi- the tendon healing process. The keys
creased ECM production.24
mately 6 weeks after the initial in- in optimizing tendon repair are de-
Bone morphogenetic proteins, in-
jury. A decrease in type III collagen termining which growth factors are
cluding growth differentiation factor
and matrix synthesis is seen, as well necessary and the ideal timing of ad-
(GDF)-5, -6, and -7, have been
as an overall decrease in cellularity. ministration. Localization and me-
shown to be key factors in tendon re-
Concomitantly, type I collagen syn- chanical stimulation at the site of in-
pair.25 GDF treatment induces cell
thesis increases, and these fibers be- jury is required to prevent adverse
proliferation, increases collagen syn-
come organized longitudinally along effects.20 During the inflammatory
thesis, and improves organization of
the long axis of the tendon, provid- phase, chemotactic cytokines attract
the ECM. GDFs also have been
ing mechanical strength to the repair fibroblasts, macrophages, and neu-
shown to increase the transcription
tissue. As remodeling proceeds, col- trophils to the site of injury. Trans-
of multiple genes involved in tendon
lagen crosslinking further increases forming growth factor-β concentra-
repair, including scleraxis, type I and
the tensile strength of the regenerate tion is increased at the zone of
type III collagen, and tenascin-C.8
tendon; however, the regenerate injury, which leads to cell migration.
never achieves the strength of unin- This in turn leads to a cascade of
jured tendon.2 events, resulting in increased synthe- Biomechanics
sis of collagen types I and III, initial of Repaired Tendon
Intrinsic Versus scar formation, and production of
Extrinsic Healing other growth factors.21 Typically, tendons do not fail or rup-
The cells involved in tendon healing Insulin-like growth factor-1 ture under normal conditions. As in-
are believed to arise predominantly (IGF-1) production is upregulated dividual tendon fibrils begin to fail,
through intrinsic and extrinsic heal- during the inflammatory phase, in- damage accumulates, stiffness is re-
ing pathways. In the intrinsic healing ducing chemotaxis of neutrophils duced, and failure begins with micro-
pathway, fibroblasts and inflamma- and fibroblasts to the site of injury. scopic tears. The overall behavior of
tory cells from the endotenon and IGF-1 also plays a role in the remod- tendon in response to injury depends
epitenon migrate to the site of ten- eling phase, inducing collagen and on its crimp structure (ie, the wavy
don injury, where they then prolifer- ECM synthesis.22 IGF-1 has a dose- configuration of collagen fibrils that
ate and promote tendon healing. In dependent effect; overexpression of contributes to the flexibility and
the extrinsic healing pathway model, IGF-1 can lead to stiffness. compressibility of tendon tissue) and
these cells originate from the periph- Platelet-derived growth factor acts the ultimate failure of its collagen fi-
ery. Evidence exists for both pro- as a secondary messenger, inducing brils. The complex interactions be-
cesses, and in most cases, both path- the expression of other cytokines tween the multiple components of
ways contribute to tendon healing. during the repair process. Platelet- the ECM result in viscoelasticity.
The extrinsic mechanism is activated derived growth factor also works Thus, the rate of elongation to which
earlier, but it contributes to the for- during the proliferative phase to in- tendon is subjected modulates the
mation of adhesions. The intrinsic crease production of collagen and amount of load transferred.8
pathway results in improved biome- ECM components. Timing of admin- Derangement of collagen fibril ori-
chanics and fewer complications be- istration and duration of action af- entation and ECM composition fol-
cause it does not contribute to adhe- fect the specific composition of scar lowing injury, in combination with
sions and because endotenon and and repaired tissue.23 the development of scar tissue at the
epitenon cells produce more collagen Vascular endothelial growth factor site of injury, leads to inferior biome-
and glycosaminoglycans.8 The rela- is essential to angiogenesis and is chanical properties.20 Tissue loss,
tive contribution pathway of each is necessary for tendon repair and heal- particularly segmental defects, fur-

March 2011, Vol 19, No 3 137


Tissue Engineering Solutions for Tendon Repair

ther complicates healing. Although plicated process, the growth factors Figure 2
tendon reapproximation and sutur- must be localized to the site of injury
ing may facilitate union following in- and must be added at the appropri-
jury, failure to restore proper tendon ate time during tendon healing. Scaf-
length following segmental tissue folds have been proposed for struc-
loss has severe implications on the tural support during the healing
overall balance of forces across the process and for local delivery of
region in which the tendon acts. growth factors and/or stem cells to
Thus, modalities are needed that can the repair site.28 The optimal con-
restore original tendon strength and struct has yet to be developed, but
length.
the ability to combine MSCs under
Tendon repair is a slow and com-
the influence of tendon-specific sup-
plicated process that requires appro-
portive cytokine growth factors with
priate and timely tension on the re- The elements of successful tendon
biocompatible scaffolds exposed to
pair. Repaired tendons are at risk of bioengineering: scaffolds,
ideal mechanical stresses is para- mesenchymal stem cells (MSCs),
rerupture under high tensile load.
mount to successful tendon bioengi- cytokine growth factors, and
Controlled physiotherapy is crucial
neering (Figure 2). mechanical stress.
to the healing process. However, in-
adequate limb movement promotes
adhesions with the surrounding tis- Mesenchymal bMSCs in a rabbit Achilles tendon
sues, leading to inhibition of the glid- Progenitor Cells primary repair model. A different
ing motion of the tendon under load. Successful isolation and selection of study indicated that coculture of
Any tendon-tissue engineering strat- MSCs or mesenchymal progenitor bMSCs with tenocytes led to in-
egy must aim to minimize adhesions cells as a source of multipotent cells creased expression of tendon- and
and restore normal tendon gliding is among the most important princi- ligament-specific genes.31 Hanke-
motion that will transfer muscular ples of tendon engineering. MSCs meier et al32 injected a mixture of hu-
forces to the bone in a manner simi- can differentiate into phenotypes un- man bMSCs and fibrin glue into rat
lar to that of native tendon.26,27 der the influence of the appropriate patellar tendon defects, resulting in
environmental cues. It is generally more mature tissue and more orga-
accepted that fibroblasts at the ten- nized cell structure compared with
Elements of Tendon don healing site arise from undiffer- controls. Drawbacks to the use of
Bioengineering entiated MSCs. The MSCs are pres- bMSCs include the painful bone
ent in adjacent connective tissue and marrow harvesting procedure re-
Improving Repair and in circulating blood and bone mar- quired to isolate them and the some-
Remodeling row. Autograft MSCs are processed times low cellular yield.
Multiple biomechanical studies sug- in vitro. A recent study showed that The use of adipose-derived MSCs
gest that the strength of repair is af- the use of human embryonic stems (aMSCs) has been investigated for
fected by the number of strands cells to repair the patellar tendon in application in regenerative medicine.
crossing the repair. Recent efforts to rats led to regeneration of tendon tis- Adipose tissue represents an abun-
understand the biology of tendon sue in vitro and in vivo.29 Compared dant source of adult stem cells with
healing, as well as the addition of with control subjects, treated rats the potential to differentiate along
growth factors and the use of tissue demonstrated increased expression multiple lineage pathways, including
engineering modalities, are focused of tendon-specific genes as well as tendon.33 Adipose tissue is abundant,
on improving tendon repair at the improved structural and mechanical and harvest-related morbidity is min-
cellular, molecular, and biomechani- properties. imal compared with that of bone
cal levels.8 Bone marrow–derived MSCs marrow sources. Recent literature
MSCs are multipotent cells that (bMSCs) have been transplanted to suggests that aMSC differentiation
can be induced to differentiate into various tissue injury sites, including and regenerative potential is equal to
tendon cells. This requires the addi- injured tendon, resulting in enhanced that of other sources.34 Kryger et al35
tion of specific growth factors in- tissue repair. Chong et al30 reported showed aMSCs to be comparable to
volved in the multiple stages of improved and accelerated healing tendon sheath fibroblast and bMSCs
tendon repair. In this extremely com- with intratendinous treatment with in their potential for use as engi-

138 Journal of the American Academy of Orthopaedic Surgeons


MaCalus V. Hogan, MD, et al

Figure 3 ber of transfer options are available


for most injuries. Allografts are more
widely available, and they mimic the
native tendon ECM architecture.
However, they carry a significant risk
of disease transmission and immune
rejection. In addition, the steriliza-
tion process alters the mechanical
properties of allograft. Freeze-dried
allografts are nonliving tissues that
are extensively available from cadav-
ers.
Polymers such as poly(a-hydroxy-
A, Tissue-engineered nanofiber tubular 65:35 poly(lactide-co-glycolide) esters) are US FDA-approved syn-
(PLAGA) polymer scaffold. B, Tubular 65:35 PLAGA scaffold with an 18-
gauge needle through the lumen. (Patent pending, University of Virginia.) thetic materials with highly customi-
zable physiochemical properties.
They can be designed for tendon
Figure 4 graft applications (Figure 3). These
polymers degrade by hydrolysis into
nontoxic products that are easily
eliminated from the body.
The ideal scaffold is biocompatible
and does not incite a host inflamma-
tory response. The selected scaffold
in its composition and fabricated
form must be capable of holding and
supporting the cells (Figure 4). In ad-
dition, the scaffold should be biode-
gradable, serving as a temporary
support for the cells and mechani-
cally augmenting the repaired tendon
while allowing for eventual replace-
ment by matrix components synthe-
Confocal microscopy live/dead assay of adipose mesenchymal stem cells
proliferating on electrospun nanofiber poly(lactide-co-glycolide) 65:35 scaffold sized by the implanted cells. The
at 14 days (magnification, ×10). A, Achilles tendon fibroblasts. B, Adipose scaffold should have high porosity
mesenchymal stem cells. Green = live, red = dead. (Patent pending, and a large surface area. It should
University of Virginia.)
mimic the native tendon ECM archi-
tecture to allow cells to be distrib-
uted throughout the scaffold and to
neered tendons through scaffold Scaffolds allow diffusion of nutrients and fac-
seeding and cell proliferation. An- Currently, tendon repair involves the tors that promote cellular prolifera-
other study demonstrated that aMSC use of either autograft or tendon tion as well as the production of
injections used to treat collagenase- transfer. Autograft supply is limited, ECM (Figure 5). Most important,
induced tendinitis in superficial digi- and donor site morbidity is a con- the scaffold must be mechanically
tal flexor tendons in horse forelimbs cern. Furthermore, only rarely can stable and support limited early limb
resulted in improved tendon organi- autograft be matched to the tensile movement, which is critical in pre-
zation.36 The plastic surgery commu- load of the repair tissue. Tendon venting adhesions and in accelerating
nity, based on their experience with transfer involves detachment of the tendon tissue remodeling. The scaf-
lipoplasty, is particularly interested tendon to be transferred and reat- fold should be reproducible, scal-
in the use of adipose tissue as a po- tachment of it to the injured tendon. able, and readily available, with
tential source of stem cells for a vari- This may restore function in the in- properties customized to the tendon
ety of soft-tissue applications.33 jured limb; however, a limited num- graft required.

March 2011, Vol 19, No 3 139


Tissue Engineering Solutions for Tendon Repair

Figure 5 that the source, species, age of donor, growth factors, can improve the biol-
and manufacturing process contrib- ogy of tendon healing.28 In future, it
uted to the unique properties of each is hoped that tissue engineering strat-
product. The biochemical composi- egies can be used not only to aug-
tion of the commercial matrices was ment repair but also to enhance heal-
found to be similar to that of native ing and regeneration. Translation of
tendon, but the commercial products tendon bioengineering to the clinical
were mechanically inferior. setting is on the horizon.
As the use of scaffolds for tendon Recently, GDF-5 (ie, bone morpho-
repair expands, so will the need for genetic protein [BMP]-14) gene ther-
apy was reported to have increased
improved design. There are several
rat Achilles tendon tensile strength
promising designs on the horizon.
without inducing bone or cartilage
Moffat et al40 designed a poly-
In vivo application of tubular formation within the healed ten-
poly(lactide-co-glycolide) polymer (lactide-co-glycolide) nanofiber-
don.42 Basile et al43 successfully
scaffold to bridge an Achilles based scaffold for rotator cuff repair.
tendon defect and foster tendon transduced freeze-dried flexor digito-
This aligned scaffold supported fi-
regeneration in a rat. (Patent rum longus tendon allografts with
broblasts in culture, with cells at-
pending, University of Virginia.) GDF-5 in a murine model. They
taching along the long axis of the
demonstrated accelerated healing
construct. Cell distribution, matrix
and decreased adhesion formation
deposition, and mechanical proper-
Current commercially available compared with controls. GDF-5 aug-
ties were each not only significantly
scaffold options for soft-tissue repair mentation in a murine Achilles ten-
improved versus controls (P < 0.05),
are often composed of small intestine don repair model led to decreased
but were also maintained after scaf-
submucosa (SIS) or collagens. Graft- expression of inflammatory genes
fold degradation in an in vitro
Jacket (Wright Medical Technology, and improved collagen organiza-
model. A different study indicated
Arlington, TN) is derived from hu- tion.44 Schnabel et al45 managed
that use of MSC-seeded collagen
man allograft dermis.37 Scaffolds collagenase-induced flexor digitorum
sponge composite in rabbit patellar
from bovine and porcine sources superficialis tendinitis lesions in
tendon defects resulted in improved
have recently been developed.38 Ex- horses with either IGF-1 gene-
histologic and biomechanical proper-
amples of such xenografts include enhanced MSCs or MSCs alone.
ties compared with normal tendon.41
CuffPatch Soft Tissue Reinforcement Both groups showed enhanced ten-
These studies represent only a sam-
(Biomet, Warsaw, IN) and Restore don healing properties at the cellular
ple of current advancements in scaf-
Orthobiologic Soft Tissue Implant and histologic levels. Another recent
fold engineering. More research is
(DePuy, Warsaw, IN), which are de- study showed that transforming
needed to aid in the development of
rived from porcine SIS. TissueMend growth factor-β1–transduced bMSCs
the optimal scaffold for tendon bio-
Soft Tissue Repair Matrix (Stryker, led to accelerated and improved
engineering.
Mahwah, NJ) is an ECM scaffold healing, as well to faster recovery
derived from fetal bovine dermis, and improved biomechanical proper-
whereas the Zimmer Collagen Re- The Future of Tendon ties, in an Achilles tendon injury
pair Patch (Zimmer, Warsaw, IN) is Repair model in rabbits.46 Seeherman et al47
made of porcine dermis.37 demonstrated that the delivery of re-
The aforementioned products are Delivery of growth factors via gene combinant hormone BMP-12 (ie,
US FDA-approved for certain indica- therapy techniques and attachment GDF-7) in collagen or hyaluronan
tions related to tendon repair and/or to scaffolds has led to improved lo- sponges led to accelerated healing
augmentation.37,38 Few published calization of the growth factor to in- following full-thickness rotator cuff
studies have compared their use, jured tendon. Alternatively, growth repair in sheep.
however. Derwin et al39 performed factors may be used to induce local Growth factors, MSCs, and scaf-
an in vitro analysis comparing the stem cells to differentiate into tendon folds are the most researched aspects
biophysical properties of commer- or fibroblast cells that support ten- of tissue engineering. However, re-
cially available ECM scaffolds for don healing. MSCs also can be deliv- cent studies have highlighted the im-
rotator cuff repair with canine in- ered to the site of injury and, in com- portance of mechanical stimulation
fraspinatus tendon. They concluded bination with delivered or local in the development of engineered

140 Journal of the American Academy of Orthopaedic Surgeons


MaCalus V. Hogan, MD, et al

constructs. Tenocyte-seeded SIS con- 2. Butler DL, Juncosa N, Dressler MR: 16. Aslan H, Kimelman-Bleich N, Pelled G,
Functional efficacy of tendon repair Gazit D: Molecular targets for tendon
structs were shown to have improved processes. Annu Rev Biomed Eng 2004; neoformation. J Clin Invest 2008;118(2):
biomechanical properties following 6:303-329. 439-444.
cyclic loading in vitro.48 Butler et al27 3. Kannus P: Structure of the tendon 17. Hoffmann A, Pelled G, Turgeman G,
combined MSCs, type I collagen connective tissue. Scand J Med Sci Sports et al: Neotendon formation induced by
2000;10(6):312-320. manipulation of the Smad8 signalling
sponges, and bioreactors for me-
pathway in mesenchymal stem cells.
chanical stimulation to create a con- 4. Xu Y, Murrell GA: The basic science of J Clin Invest 2006;116(4):940-952.
struct that was able to withstand tendinopathy. Clin Orthop Relat Res
2008;466(7):1528-1538. 18. Gelberman RH, Chu CR, Williams CS,
peak in vivo forces in a patellar ten- Seiler JG III, Amiel D: Angiogenesis in
5. Kjaer M: Role of extracellular matrix in healing autogenous flexor-tendon grafts.
don rabbit repair model. These re-
adaptation of tendon and skeletal muscle J Bone Joint Surg Am 1992;74(8):1207-
searchers previously showed that the to mechanical loading. Physiol Rev 1216.
use of stem cell–collagen sponge con- 2004;84(2):649-698.
19. Gomez M: The physiology and
structs treated with mechanical stim- 6. Woo SL, Debski RE, Zeminski J, biochemistry of soft tissue healing, in
ulation in vitro led to improved bio- Abramowitch SD, Saw SS, Fenwick JA: Griffin L, ed: Rehabilitation of the
Injury and repair of ligaments and Injured Knee, ed 2. St. Louis, MO,
mechanical and histologic properties tendons. Annu Rev Biomed Eng 2000;2: Mosby, 1995, pp 34-44.
when implanted in vivo.49 83-118.
20. Aspenberg P: Stimulation of tendon
7. Karousou E, Ronga M, Vigetti D, Passi repair: Mechanical loading, GDFs and
A, Maffulli N: Collagens, proteoglycans, platelets. A mini-review. Int Orthop
Summary MMP-2, MMP-9 and TIMPs in human 2007;31(6):783-789.
achilles tendon rupture. Clin Orthop
Relat Res 2008;466(7):1577-1582. 21. Klein MB, Yalamanchi N, Pham H,
Many knowledge gaps exist in the clin- Longaker MT, Chang J: Flexor tendon
8. James R, Kesturu G, Balian G, Chhabra healing in vitro: Effects of TGF-beta on
ical and basic science aspects of tendon AB: Tendon: Biology, biomechanics, tendon cell collagen production. J Hand
healing. Faster, more reliable healing is repair, growth factors, and evolving Surg Am 2002;27(4):615-620.
treatment options. J Hand Surg Am
needed. The ultimate goal is tendon re- 2008;33(1):102-112. 22. Abrahamsson SO, Lohmander S:
pair that leads to earlier and improved Differential effects of insulin-like growth
9. Riley GP: Gene expression and matrix factor-I on matrix and DNA synthesis in
rehabilitation, minimal complications, turnover in overused and damaged various regions and types of rabbit
and regeneration of tissue with charac- tendons. Scand J Med Sci Sports 2005; tendons. J Orthop Res 1996;14(3):370-
15(4):241-251. 376.
teristics that are as good as or better
than those of normal tendon. Growth 10. Bedi A, Kovacevic D, Hettrich C, et al: 23. Wang XT, Liu PY, Tang JB: Tendon
The effect of matrix metalloproteinase healing in vitro: Genetic modification of
factors, MSCs, and biocompatible scaf- inhibition on tendon-to-bone healing in a tenocytes with exogenous PDGF gene
folds could lead to better healing and rotator cuff repair model. J Shoulder and promotion of collagen gene
Elbow Surg 2010;19(3):384-391. expression. J Hand Surg Am 2004;29(5):
regeneration. Future research in tissue 884-890.
engineering could lead to changes in 11. Magra M, Maffulli N: Matrix
metalloproteases: A role in overuse 24. Chan BP, Fu S, Qin L, Lee K, Rolf CG,
our approach to managing tendon in- tendinopathies. Br J Sports Med 2005; Chan K: Effects of basic fibroblast
jury. 39(11):789-791. growth factor (bFGF) on early stages of
tendon healing: A rat patellar tendon
12. Loiselle AE, Bragdon GA, Jacobson JA, model. Acta Orthop Scand 2000;71(5):
et al: Remodeling of murine intra- 513-518.
References synovial tendon adhesions following
injury: MMP and neotendon gene 25. Wolfman NM, Hattersley G, Cox K,
expression. J Orthop Res 2009;27(6): et al: Ectopic induction of tendon and
Evidence-based Medicine: Levels of 833-840. ligament in rats by growth and
differentiation factors 5, 6, and 7,
evidence are described in the table of 13. Jones GC, Corps AN, Pennington CJ, members of the TGF-beta gene family.
contents. In this article, no level I, II, et al: Expression profiling of J Clin Invest 1997;100(2):321-330.
metalloproteinases and tissue inhibitors
III, or IV studies are cited. References of metalloproteinases in normal and 26. DeFranco MJ, Derwin K, Iannotti JP:
1, 2, 8, 26, 28, 34, and 38 are level degenerate human achilles tendon. New therapies in tendon reconstruction.
Arthritis Rheum 2006;54(3):832-842. J Am Acad Orthop Surg 2004;12(5):
V expert opinion. Most of the refer- 298-304.
ences are basic science studies. 14. Schweitzer R, Chyung JH, Murtaugh
LC, et al: Analysis of the tendon cell fate 27. Butler DL, Juncosa-Melvin N, Boivin
References printed in bold type are using Scleraxis, a specific marker for GP, et al: Functional tissue engineering
those published within the past 5 tendons and ligaments. Development for tendon repair: A multidisciplinary
2001;128(19):3855-3866. strategy using mesenchymal stem cells,
years. bioscaffolds, and mechanical
15. Shukunami C, Takimoto A, Oro M, stimulation. J Orthop Res 2008;26(1):
1. Laurencin CT, Ambrosio AM, Borden Hiraki Y: Scleraxis positively regulates 1-9.
MD, Cooper JA Jr: Tissue engineering: the expression of tenomodulin, a
Orthopedic applications. Annu Rev differentiation marker of tenocytes. Dev 28. Huang D, Balian G, Chhabra AB:
Biomed Eng 1999;1:19-46. Biol 2006;298(1):234-247. Tendon tissue engineering and gene

March 2011, Vol 19, No 3 141


Tissue Engineering Solutions for Tendon Repair

transfer: The future of surgical 36. Nixon AJ, Dahlgren LA, Haupt JL, engineering scaffolds for Gdf5 gene
treatment. J Hand Surg Am 2006;31(5): Yeager AE, Ward DL: Effect of adipose- delivery. Mol Ther 2008;16(3):466-473.
693-704. derived nucleated cell fractions on
tendon repair in horses with collagenase- 44. Hogan MV, Kesturu G, James R, Balian
29. Chen X, Song XH, Yin Z, et al: Stepwise induced tendinitis. Am J Vet Res 2008; G, Hurwitz S, Chhabra A: Growth and
differentiation of human embryonic stem 69(7):928-937. differentiation factor-5 regulation of
cells promotes tendon regeneration by extracellular matrix gene expression in
secreting fetal tendon matrix and 37. Chen J, Xu J, Wang A, Zheng M: murine tendon fibroblasts. J Tissue Eng
differentiation factors. Stem Cells 2009; Scaffolds for tendon and ligament repair: Regen Med 2010;Jul 23:[Epub ahead of
27(6):1276-1287. Review of the efficacy of commercial print].
products. Expert Rev Med Devices 2009;
30. Chong AK, Ang AD, Goh JC, et al: Bone 6(1):61-73. 45. Schnabel LV, Lynch ME, van der Meulen
marrow-derived mesenchymal stem cells MC, Yeager AE, Kornatowski MA,
influence early tendon-healing in a rabbit 38. Laurencin CT, El-Amin SF: Nixon AJ: Mesenchymal stem cells and
achilles tendon model. J Bone Joint Surg Xenotransplantation in orthopaedic insulin-like growth factor-I gene-
Am 2007;89(1):74-81. surgery. J Am Acad Orthop Surg 2008; enhanced mesenchymal stem cells
16(1):4-8. improve structural aspects of healing in
31. Luo Q, Song G, Song Y, Xu B, Qin J, Shi equine flexor digitorum superficialis
Y: Indirect co-culture with tenocytes 39. Derwin KA, Baker AR, Spragg RK, tendons. J Orthop Res 2009;27(10):
promotes proliferation and mRNA Leigh DR, Iannotti JP: Commercial 1392-1398.
expression of tendon/ligament related extracellular matrix scaffolds for rotator
genes in rat bone marrow mesenchymal cuff tendon repair: Biomechanical, 46. Hou Y, Mao Z, Wei X, et al: Effects of
stem cells. Cytotechnology 2009;61(1-2): biochemical, and cellular properties. transforming growth factor-beta1 and
1-10. J Bone Joint Surg Am 2006;88(12):2665- vascular endothelial growth factor 165
2672. gene transfer on Achilles tendon healing.
32. Hankemeier S, van Griensven M, Matrix Biol 2009;28(6):324-335.
Ezechieli M, et al: Tissue engineering of 40. Moffat KL, Kwei AS, Spalazzi JP, Doty
tendons and ligaments by human bone SB, Levine WN, Lu HH: Novel 47. Seeherman HJ, Archambault JM, Rodeo
marrow stromal cells in a liquid fibrin nanofiber-based scaffold for rotator cuff SA, et al: rhBMP-12 accelerates healing
matrix in immunodeficient rats: Results repair and augmentation. Tissue Eng of rotator cuff repairs in a sheep model.
of a histologic study. Arch Orthop Part A 2009;15(1):115-126. J Bone Joint Surg Am 2008;90(10):2206-
Trauma Surg 2007;127(9):815-821. 2219.
41. Juncosa-Melvin N, Boivin GP, Gooch C,
33. Gimble JM, Katz AJ, Bunnell BA: et al: The effect of autologous 48. Androjna C, Spragg RK, Derwin KA:
Adipose-derived stem cells for mesenchymal stem cells on the Mechanical conditioning of cell-seeded
regenerative medicine. Circ Res 2007; biomechanics and histology of gel- small intestine submucosa: A potential
100(9):1249-1260. collagen sponge constructs used for tissue-engineering strategy for tendon
rabbit patellar tendon repair. Tissue Eng repair. Tissue Eng 2007;13(2):233-243.
34. Parker AM, Katz AJ: Adipose-derived 2006;12(2):369-379.
stem cells for the regeneration of 49. Juncosa-Melvin N, Shearn JT, Boivin
damaged tissues. Expert Opin Biol Ther 42. Bolt P, Clerk AN, Luu HH, et al: GP, et al: Effects of mechanical
2006;6(6):567-578. BMP-14 gene therapy increases tendon stimulation on the biomechanics and
tensile strength in a rat model of Achilles histology of stem cell-collagen sponge
35. Kryger GS, Chong AK, Costa M, Pham tendon injury. J Bone Joint Surg Am constructs for rabbit patellar tendon
H, Bates SJ, Chang J: A comparison of 2007;89(6):1315-1320. repair. Tissue Eng 2006;12(8):2291-
tenocytes and mesenchymal stem cells 2300.
for use in flexor tendon tissue 43. Basile P, Dadali T, Jacobson J, et al:
engineering. J Hand Surg Am 2007; Freeze-dried tendon allografts as tissue-
32(5):597-605.

142 Journal of the American Academy of Orthopaedic Surgeons

You might also like