You are on page 1of 13

ARTICLE IN PRESS

Journal of Biomechanics 37 (2004) 865–877

Biomechanics of tendon injury and repair


Tony W. Lin, Luis Cardenas, Louis J. Soslowsky*
McKay Orthopaedic Research Laboratory, University of Pennsylvania, 424 Stemmler Hall, 36th and Hamilton Walk, Philadelphia,
PA 19104-6081, USA
Accepted 6 November 2003

Abstract

Many clinical and experimental studies have investigated how tendons repair in response to an injury. This body of work has led
to a greater understanding of tendon healing mechanisms and subsequently to an improvement in their treatment. In this review
paper, characterization of normal and healing tendons is first covered. In addition, the debate between intrinsic and extrinsic healing
is examined, and the cellular and extracellular matrix response following a tendon injury is detailed. Next, clinical and experimental
injury and repair methods utilizing animal models are discussed. Animal models have been utilized to study the effect of various
activity levels, motions, injury methods, and injury locations on tendon injury and repair. Finally, current and future treatment
modalities for improving tendon healing, such as tissue engineering, cell therapy, and gene therapy, are reviewed.
r 2003 Elsevier Ltd. All rights reserved.

Keywords: Tendon; Tendon healing; Soft tissue injury; Soft tissue repair; Animal model

1. Introduction cepts to address this problem, from creating scaffolds


and constructs out of relevant biomaterials and cells to
Tendons are soft connective tissues consisting of applying cytokines exogenously to injured tissue, or
parallel collagen fibers embedded within an extracellular using gene and cell therapy. This paper provides an
matrix. This organized structure allows tendons to overview of these studies, which demonstrate that the
withstand and transmit large forces between muscle functional outcome of injured tendons is dependent
and bone. However, as tendons are subjected to upon many factors.
repeated motion and degeneration over time, they are
prone to both acute and chronic injuries. After injury,
the healing process in tendons results in the formation of
2. Normal tendon
a fibrotic scar. The structural, organizational, and
mechanical properties of this healed tissue are inferior
Tendons connect muscle to bone and form a
to normal tendon (Frank et al., 1992, 1983). Although
musculotendinous unit whose primary function is to
these properties improve over time, they do not return
transmit tensile loads generated by muscles to move and
to normal levels, even after long periods (Frank et al.,
stabilize joints. Under normal loads, it has been shown
1997). In an attempt to better understand tendon
that tendons maintain smooth physiological mechanics
healing mechanisms and to improve these inferior
throughout range of motion. While subjected to higher
properties, researchers have investigated a broad range
loads, tendons prevent joint displacement beyond
of factors believed to affect tendon injury and repair,
anatomical barriers, thus preventing injury and main-
such as activity level, motion after injury, various injury
taining normal function (O’Brien, 1992; Dykyj and
modalities, and different injury locations. In addition,
Jules, 1991). Within the tendon there exists a structural
many researchers have applied tissue engineering con-
hierarchy where tendons can be further subdivided into
fascicles, fibrils, subfibrils, microfibrils, and tropocolla-
*Corresponding author. Tel.: +1-215-898-8653; fax: +1-215-573- gen (Fig. 1). The composition of tendon contains
2133. relatively few cells with the predominant cell type being
E-mail address: soslowsk@mail.med.upenn.edu (L.J. Soslowsky). the rod or spindle-shaped fibroblasts. Tendons contain

0021-9290/$ - see front matter r 2003 Elsevier Ltd. All rights reserved.
doi:10.1016/j.jbiomech.2003.11.005
ARTICLE IN PRESS
866 T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877

Fig. 1. Tendon hierarchical structure. Detail A represents the


macromolecular organization of the extracellular matrix. Fibril
associated proteoglycans may be oriented such that their glycosami-
noglycan side chains extend into the extracellular space, perhaps
Fig. 2. Wound healing response in tendons. (Adapted from Gomez,
linking matrix constituents. (Derwin, K., 1998. A quantitative
M., 1995. The physiology and biochemistry of soft tissue healing. In:
investigation of structure-function relationships in a tendon fascicle
Griffin, L., (Eds.), Rehabilitation of the Injured Knee, 2nd Edition.
model. Ph.D. Thesis, University of Michigan, Ann Arbor; with
Mosby Company, St. Louis, MO, pp. 34-44; with permission.)
permission.)

86% collagen, 1–5% proteoglycan, and 2% elastin as and aggressively engage in the phagocytosis of necrotic
measured by dry weights, and water is responsible for tissue and debris and break down the blood clot.
60–80% of the total wet weight of the tendon (Woo Macrophages aid in the recruitment of new fibroblasts
et al., 2000a, b; Jozsa and Kannus, 1997). Tendons glide and release of angiogenesis promoting factors to initiate
over bone and often move through canals and sheaths growth of capillary networks within the wound (Fen-
that help direct their path and limit the amount of wick et al., 2002; Gelberman et al., 1992). During this
friction they must overcome. Tendon sheaths consist of phase, there is an increase in DNA, fibronectin,
two layers, in between which contains synovial fluid that glycosaminoglycan, water, and collagen type III con-
improves lubrication. Tendons that lack this two layer tent, which collectively stabilizes the newly formed
sheath are surrounded by a loose areolar connective extracellular matrix (Woo et al., 2000a, b; Jozsa and
tissue called paratenon, which acts to provide a division Kannus, 1997; Gomez, 1995; Montgomery, 1989;
from and permit free motion of the tendon in relation to Grinnell, 1984).
the surrounding tissue (Jozsa and Kannus, 1997).
3.2. Proliferation/fibroplasia

3. Tendon healing During this phase, a disorganized matrix of granula-


tion tissue is present at the injury site. Histologically, the
When tendons are injured, the body initiates a process predominant cell types are fibroblasts along with a
of healing and scar formation that can be divided into smaller number of macrophages and mast cells. Electron
phases, which are briefly described below, are distin- microscopy studies have shown an increase in the
guishable by specific peaks in a cascade of cellular and endoplasmic reticulum of fibroblasts, which is indicative
biochemical events. These phases overlap and their of active matrix synthesis (Jozsa and Kannus, 1997), and
duration can vary greatly due to location of injury or type III collagen and DNA concentrations reach their
disease (Gomez, 1995; Montgomery, 1989). Although peak amounts during the entire reparative process.
the tensile strength of the healing tendon improves over These changes are believed to help with the optimization
time, it does not reach the levels of uninjured, normal of collagen synthesis and the gradual conversion of type
tissue (Fig. 2). III to type I collagen (Gomez, 1995; Woo and
Buckwalter, 1988).
3.1. Hemostasis/inflammation
3.3. Remodeling/maturation
This phase of tendon healing occurs almost immedi-
ately after tendon injury. First, the injury to the Changes in the healing tissue can be seen with the
surrounding vascular vessels causes the formation of a naked eye as the previous red scar now becomes pinkish
hematoma. Next, the resultant clot and hemostasis and translucent, forming a connection between the two
activates a cascade of vasodilators and platelets, as well ends. Histologically, fibroblasts have decreased in size
as the release of pro-inflammatory chemicals from mast and slowed their matrix synthesis, and collagen fibers
cells. Inflammatory cells are attracted to the injury site have begun to orient themselves longitudinally along the
ARTICLE IN PRESS
T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877 867

long axis of the tendon. As the scar enters maturation caused by local factors such as infection, disease, tissue
there is a notable return of type III to type I collagen hypoxia due to vascular deficiencies, and malnutrition,
ratio, collagen crosslinks, and glycosaminoglycan, and external factors such as rigid fixation and prolonged
water, and DNA concentrations. However, healed immobilization (Woo et al., 2000a, b). As a result, it is
tendon has been shown in many studies to take upwards often difficult to re-establish the continuity of collagen
to a year to more closely approach the functional fibers and restore the sliding surface of the tendon.
strength of uninjured tissue (Buckwalter and Hunziker, Failure to do either makes tendon healing a ‘‘double-
1996; Gomez, 1995; Woo and Buckwalter, 1988). edged sword’’—while adhesions can contribute to
healing, excessive scarring can restrict tendon gliding,
3.4. Intrinsic vs. extrinsic healing but deficient scarring may lead to premature rupture at
the injury site. Therefore, a combination of many
The biology of tendon healing is dominated by two factors must be simultaneously balanced in order to
theories, extrinsic and intrinsic healing. There has been achieve a functionally healed tendon.
much debate in the literature and much experimental
evidence to prove or disprove each healing mechanism.
In extrinsic healing, it is believed that the tendon has no 4. Tendon injury and repair
internal ability to heal on its own and thus requires the
formation of adhesions, the infiltration of inflammatory Because tendons heal poorly, tendon injury and repair
cells and fibroblasts, and an extratendinous blood mechanisms have been studied extensively in both the
supply to heal properly (Potenza, 1969, 1964, 1963, clinical and experimental setting. Clinical examples of
1962). In intrinsic healing, it is believed that the tendon different types of injury and widely used treatment
is healed by the proliferation of epitenon and endotenon methods are discussed, followed by a review of both
cells within the tendon, an intratendinous blood supply, in vitro and in vivo experimental models. As described
and the lack of adhesion formation (Mass and Tuel, below, although clinical and experimental studies have
1991; Lundborg et al., 1985; Manske et al., 1984). their own inherent advantages and disadvantages, they
Although there are credible arguments for both extrinsic both provide important information on tendon injury
and intrinsic healing, there is no clear evidence as to and repair. The correct choice of study is dependent
which theory is correct. It is plausible that tendon upon the specific hypotheses it is intended to address.
healing most likely occurs as a combination of both
processes and is dependent on tendon location, the 4.1. Clinical injury/repair
magnitude of tendon trauma, availability of synovial
fluid and a blood supply, and degree of tendon Tendon injury can be classified by types of injury and
mobilization. A better understanding of this balance are generally considered to be acute or chronic and
between processes may allow surgeons to eventually either direct or indirect (Hyman and Rodeo, 2000).
tailor their surgical procedures to best reestablish Direct acute tendon injuries occur due to either
tendon continuity and function (Jaibaji, 2000; Manske, contusion, non-penetrating blunt injury from accidents
1988; Lundborg et al., 1985; Manske et al., 1985). and sports injuries, or laceration by a sharp object.
Indirect, tendon injuries are often the result of acute
3.5. Cellular and ECM response tensile overload and repetitive microtrauma as seen in
overuse injuries. Most commonly, tensile overload and
As described earlier, the process of tendon repair is a overuse result in injuries to either the musculotendinous
complex, orchestrated series of physiological events junction (sprains, strains and rupture), or to the
involving significant synthesis, migration, and degrada- osteotendinous junction (avulsion fractures or bone
tion of extracellular matrix (ECM) components (Liu detachment). These types of injuries predominate over
et al., 1995; Leadbetter, 1992; Doillon et al., 1985). After mid-tendon ruptures because healthy tendon can with-
trauma, the ECM degradation products are essential for stand higher tensile loads than the muscle or most bone
tendon healing because they provide chemotactic signals junctions (Woo et al., 2000a, b; Hyman and Rodeo,
for fibroblasts, leukocytes, and endothelial cells, and 2000; Taylor et al., 1993; Silver et al., 1983).
serve as a reservoir for cytokines (Jaibaji, 2000). Despite There are a variety of different suture techniques used
the similarity of having cells and extracellular matrix clinically, each with their preferred suture material that
proteins becoming more active in response to an injury, can be employed at the discretion of the surgeon. Some
tendons do exhibit a differential capacity to heal. This is of these techniques include Lin locking, Savage, Kessler,
due to the intrinsic properties of the tendon and external Becker, modified Kessler, Tajima, and epitenon suture
factors such as nutrition, location and environment method. It has been shown that the repair strength is
(Woo et al., 1999). Despite this differential healing directly proportional to the number of strands crossing
capacity, there still exists a poor healing environment a repair site (Fig. 3), ruptures usually occur at the knots
ARTICLE IN PRESS
868 T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877

(Noguchi et al., 1993; Savage, 1985). In addition, many


in vitro studies have focused on the effect of applying
cyclic loads to tendons (Archambault et al., 2002;
Ditsios et al., 2002; Schechtman and Bader, 1997;
Almekinders et al., 1993). Flexor tendons cultured
in vitro not only survived, but the tendon cells were
also actively dividing, migrating, and synthesizing new
collagen when cultured in a cell-free environment (Mass
and Tuel, 1989). In vitro culture of rabbit flexor tendons
also demonstrated that these cells exhibit a differential
proliferative and healing capacity when compared to
tendon sheaths (Wiig et al., 1997). In vitro studies of
flexor tendon repair have also compared gliding func-
tion and tensile properties of injured tendons repaired
Fig. 3. Comparison of ultimate force vs. time between eight and four- using different suture methods. These parameters
strand suture techniques. The asterisks denote a significant difference provide a measure of the range of motion and strength
(po0:05). (Boyer, M.I., Gelberman, R.H., Burns, M.E., Dinopoulos, of the repair, which are both important because they
H., Hofem, R., Silva, M.J., 2001. Intrasynovial flexor tendon repair.
allow a tendon to withstand early mobilization and to
An experimental study comparing low and high levels of in vivo force
during rehabilitation in canines. Journal of Bone and Joint Surgery— avoid adhesion formation (Noguchi et al., 1993; Savage,
American 83-A, 891–899; with permission.) 1985).
In vitro cyclic testing of extensor tendons was
performed to describe its fatigue properties, which
in the suture, and epitenon sutures increase repair may serve as a stimuli for tendon remodeling. The
strength over core sutures alone. Gap strength was not effects of cyclic loading are important to characterize
significantly greater among any of the techniques because they can cause microdamage, which accumu-
examined (Woo et al., 2000a, b; Taras and Lamb, lates over time and could contribute to potential tendon
1999; Wang, 1998; Silfverskiold and Andersson, 1993; rupture (Schechtman and Bader, 1997). An example of
Woo and Buckwalter, 1988). an in vitro model studying the effects of repetitive
Timing of the repair can be categorized as primary, motion involves culturing tendon fibroblasts on flexible-
delayed primary, secondary and late secondary. A bottomed wells, where the amount and rate of
primary repair is done within 12 h of injury, while deformation can be precisely controlled. Using this
delayed primary is within 14 days. Secondary is model, the application of repetitive strain to tendon
classified as between 2 and 4 weeks with late secondary fibroblasts caused an increase in prostaglandin E2
after that period (Wright, 1999). Whenever feasible, (Almekinders et al., 1993) and the combination of both
tendon repair should be done as early as possible. cyclic strain and inflammatory cytokines results in
Clinically, ultrasound, electrical stimulation and inject- greater extracellular matrix damage in tendon than
able growth factors have been used or postulated to either factor acting alone (Archambault et al., 2002).
hasten tendon repair (Woo et al., 2000a, b). Post- Repaired canine flexor tendons undergoing a different
operatively, the surgeon faces the problem of when to cyclic loading protocol showed that repetitive loading
introduce active vs. passive motion and remobilization/ has a conditioning effect that affects repair-site rigidity
immobilization. The dilemma becomes restoring normal and strain in a dose-dependent manner (Ditsios et al.,
strength and range of motion while minimizing adhe- 2002). It is clear that the results from these in vitro
sions and protecting the healing tendon from rupture studies provide a foundation for developing hypotheses
and gapping at the tendon repair site. and designing in vivo experiments.

4.2. Experimental injury/repair—cell culture (in vitro) 4.3. Experimental injury/repair—animal model (in vivo)

Experimental in vitro studies allow researchers to The majority of experimental research on tendon
study the effect of a single isolated factor. In vitro injury and repair has been performed in animal models,
experiments are inexpensive and often serve as pre- which possess advantages such as lower cost, fewer
cursors for more complicated and expensive animal ethical issues, less variability, and greater availability of
models, which are simulated in vitro using different cell numbers when compared to clinical trials. However,
culture environments. In the case of tendon injury and animal models also have their shortcomings as well and
repair, some examples of in vitro models have cultured researchers must be cautious in applying their results to
flexor tendon cells (Wiig et al., 1997; Mass and Tuel, humans, just as care must be taken when extrapolating
1989) and tested the strength of flexor tendon repair in vitro results for in vivo settings. Some disadvantages
ARTICLE IN PRESS
T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877 869

of animal models include inherent biologic variability,


metabolic and hormonal differences from humans,
anatomical differences, and potential difficulty in work-
ing with animals, such as noncompliance. Despite these
factors, animal models have been an invaluable tool as
investigators have utilized animal models to study
specific aspects of tendon injury and repair, including
the effects of activity level and motion as well as
different types of injury modalities and locations.
Finally, animal models have been used to study how
tissue engineering therapies can be used to improve
tendon repair.

4.4. Activity level

The effects of activity levels on tendons have been


studied extensively in animal models. These activity
levels range from overuse, such as exercising, to disuse,
such as immobilization and stress shielding. As a result
of increased or decreased loads, tendon remodeling Fig. 4. Photographs and schematics depicting the rat shoulder to be an
leads to structural, biochemical, and mechanical anatomically appropriate model for a human shoulder. (Soslowsky,
changes. Investigators have used animal models to L.J., Carpenter, J.E., DeBano, C.M., Banerji, I., Moalli, M.R., 1996.
Development and use of an animal model for investigations on rotator
simulate these activity levels in a controlled manner in
cuff disease. Journal of Shoulder and Elbow Surgery 5, 383–392; with
order to study how the tendon responds and character- permission.)
ize its resulting properties.
Clinically, most overuse tendon injuries are due to
repetitive motion, which creates ‘‘microtrauma’’ that ostasis; the long-term effect of exercise on tendons
accumulates over time (Hess et al., 1989). Therefore, appears to be positive, but periods of weakness can
experimental animal models studying overuse have occur during a training period. Therefore, sufficient rest
incorporated exercise protocols that repetitively load is often required to allow the tendon to recover through
tendons to simulate the microdamage that leads to an an adaptive cellular response necessary to maintain
overuse injury. In addition to this intrinsic accumulated function. Although these exercise models allow for the
damage, there are other factors that predispose a tendon study of tendon overuse in a controlled manner, tendon
to injury. Additional intrinsic factors include muscular loads during exercise are not directly measured, so the
imbalance and muscular insufficiency while extrinsic magnitude of the overuse is unknown. If in situ loads
factors include anatomical factors, training errors, and can be monitored, then a better understanding of how
environmental conditions (Soslowsky et al., 1996; Hess tendons react and adapt to exercise can be used to
et al., 1989). Examples of animal exercise protocols have prescribe a clinically safe exercise regimen (Archambault
involved running miniature swine or rats on a motorized et al., 1995).
treadmill (Soslowsky et al., 2000; Woo et al., 1982). On the other spectrum of activity level is tendon
Short term exercise (3 months) of swine flexor tendons disuse, which is clinically relevant in patients who are
had minimal effects on tendon properties, but long-term bedridden or immobilized post-surgery or immobilized
(12 months) exercised tendons exhibited an increase in following repair. Patients are generally immobilized to
max stress and mass and a decrease in max strain (Woo allow tendons to sufficiently heal before undergoing a
et al., 1982). An anatomically appropriate animal model physical therapy regimen. Experimental animal models
for studying rotator cuff injuries has been developed by simulating immobilization have focused on different
our lab (Fig. 4) (Soslowsky et al., 1996). Using this methods to immobilize limbs and its duration. Immo-
model, an exercise regimen, which lasted for 4, 8, and 16 bilization protocols can incorporate a wide variety of
weeks, imposed on rat supraspinatus tendons resulted in techniques and methods, but they generally achieve the
significantly larger cross-sectional areas at all three time same result of either immobilizing to control range of
points and decreased modulus and maximum stress motion or also denervating to prevent loading. Im-
values (Soslowsky et al., 2000). It was also shown with mobilization methods used on tendons include both
this model that a combination of exercise with either an internal and external fixators, such as splints, casts, and
intrinsic or extrinsic factor led to a more severe injury frames (Palmes et al., 2002; Murrell et al., 1994;
than overuse alone (Carpenter et al., 1998a, b). In Enwemeka et al., 1988; Carlstedt et al., 1986). Although
general, the duration of exercise affects tissue home- immobilization of sutured tendons minimizes gap
ARTICLE IN PRESS
870 T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877

formation and suture pullout during healing (Nystrom to the tendon through varying amount of stress
and Holmlund, 1983), tendons that are immobilized shielding and to potentially remobilize the tendon after
have been shown to exhibit decreased mechanical stress alteration. Some disadvantages include the in-
properties, deter functional recovery, and promote vasive nature of the procedure, lack of denervation to
adhesion formation (Palmes et al., 2002; Murrell et al., avoid any loading, and the uncertainty as to how the
1994; Gelberman et al., 1983). The principal advantages loading pattern is changed.
of these models are also its main disadvantages.
Immobilizing a tendon simulates disuse, but there still 4.5. Motion
may be loads applied across the tendon. Denervation
removes any potential loading, but the tendon cannot be Many studies have examined the effect of motion or
reloaded if a remobilization protocol is desired. lack thereof following tendon repair. In general, it has
Another example that animal models have been used been shown that mobilization after repair has resulted in
to study tendon disuse is stress shielding. These studies improved tensile properties of the repair site, and in the
have mainly investigated how tendons remodel and case of tendons, enhanced gliding function (Gelberman
adapt in response to stress alteration. A technique et al., 1983; Woo et al., 1981; Piper and Whiteside,
developed to shield rabbit patellar tendons from stress 1980). Two types of mobilization have been studied
utilizes internal fixation to prevent gross movement of extensively, active and passive motion. Active mobiliza-
the knee joint and to completely release tension from the tion involves motion of the tendon within the sheath and
patellar tendon (Yamamoto et al., 1993). Using this application of tension to the tendon across the injury
model, investigators have found that tensile strength site through muscle contraction. Passive mobilization
and modulus up to 6 weeks post-surgery have decreased only involves motion of the repaired tendon without the
rapidly and markedly (Fig. 5), while cross-sectional area application of musculotendinous forces.
increased initially before decreasing at 6 weeks (Yama- Animal models investigating the effect of active
moto et al., 1993). In separate studies testing the motion on tendon healing have mainly studied flexor
mechanical properties of stress shielded rabbit patellar tendons. Flexor tendons allowed constrained active
tendons in either the longitudinal or transverse direc- mobilization showed gains in strength compared to
tions, there were marked decreases in both tangent tendons that were immobilized (Fig. 6) (Hitchcock et al.,
modulus and tensile strength (Yamamoto et al., 2000; 1987; Gelberman et al., 1986). Because injured tendons
Yamamoto et al., 1993). The advantages of these models are often repaired with sutures, the application of active
is their ability to control the magnitude of stress applied mobilization may be deleterious if it ruptures the suture
repair. Injured canine flexor tendons repaired by three
different repair techniques were allowed immediate
unrestricted active motion without any weight bearing
for up to 21 days. Tendon rupture occurred in two-
strand repairs, but not in the six-strand ones. The

Fig. 5. Average stress-strain curves for completely stress-shielded and Fig. 6. Comparison of tensile strengths between immobilized and
control rabbit patellar tendons. (Yamamoto, N., Ohno, K., Hayashi, actively mobilized flexor tendons after injury and repair. (Gelberman,
K., Kuriyama, H., Yasuda, K., Kaneda, K., 1993. Effects of stress R.H., Manske, P.R., Akeson, W.H., Woo, S.L., Lundborg, G., Amiel,
shielding on the mechanical properties of rabbit patellar tendon. D., 1986. Flexor tendon repair. Journal of Orthopaedic Research 4,
Journal of Biomechanical Engineering 115, 23–28; with permission.) 119–128; with permission.)
ARTICLE IN PRESS
T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877 871

strength of the repair is important because the tendon to occur clinically, injuries only to the middle half leave
must be able to withstand early active mobilization if it the marginal fibers intact, circumventing the need for
is applied (Aoki et al., 1997). Finally, it has been shown suture repair and allows for immediate postoperative
that the combined effects of both motion and tension on tendon mobilization (Chan et al., 1998; Kubota et al.,
the healing response of injured chicken flexor tendons 1996).
resulted in significantly greater tendon repair strength Complete transections have been shown to be
and cellular activity when compared to either motion or reproducible in location, pattern, and extent of damage
tension applied alone (Kubota et al., 1996). These (Walsh and Frank, 1988). As a result of a complete
animal models support the use of active mobilization as transection, there exists a gap between the two free ends.
long as the forces generated do not exceed the breaking Sutures are utilized to achieve tendon apposition in
strength of the repair technique. order to minimize scar tissue formation, hasten collage-
A number of studies have investigated the effect of nization, and avoid the presence of any non-tendinous
passive motion on tendon healing as well. Injured canine tissue between the two ends (O’Donoghue et al., 1961).
or chicken flexor tendons were repaired and subjected to Animal model studies have confirmed the clinical
either immobilization or controlled passive motion by finding that the highest suture strand technique from
manually flexing and extending the joints of the digits. each study provided the highest tensile strength (Dino-
Tendons undergoing controlled passive motion showed poulos et al., 2000; Winters et al., 1998, 1997; Noguchi
superior tensile properties and gliding function when et al., 1993). When using sutures, other factors to
compared to immobilized tendons (Feehan and Beau- consider include suture knot location, suture material,
chene, 1990; Woo et al., 1981). Similarly, injured human and suture size (Barrie and Wolfe, 2001). One dis-
flexor tendons were repaired and managed by either 312 advantage with using sutures is that they may disrupt
weeks of immobilization followed by gradual increased the intrinsic blood supply and reduce overall tensile
motion or passive motion within 5 days of repair for 412 strength of the repair site (Bishop et al., 1986).
weeks, after which active motion was allowed. A Investigators have also utilized complete transection
grading system assessing return of motion revealed that models without sutures, allowing tendon healing to
tendons undergoing passive motion had better overall commence by gap formation. The application of force
results and fewer ruptures (Strickland and Glogovac, on the tendon repair can increase gap formation
1980). Generally, enhanced tendon function following (Dinopoulos et al., 2000), which can occur immediately
passive motion may be due to a combination of factors, due to inadequate initial repair strength or can develop
such as the presence of fewer adhesions and improved slowly due to cyclic loading. Because the presence of a
tensile strength of the repair. Therefore these increased gap leads to poor healing and is possibly a major factor
levels of load and greater magnitudes of tendon repair in adhesion formation, gap formation should be
site excursion due to passive mobilization may enhance minimized to improve tensile strength. It has been
the healing response of repaired flexor tendons (Feehan shown that the tensile strength of a gap that is greater
and Beauchene, 1990; Strickland and Glogovac, 1980). than 3 mm does not improve over time (Silva et al.,
Although motion has been shown to promote cellular 2002; Gelberman et al., 1999).
activity and minimize adhesion formation, it should not Animal models have also been used to study partial
be applied at the expense of a potential rupture. transections, which involve injuring a percentage of the
Therefore, a fine balance exists between the progression tendon. Some examples include window defects, re-
of tendon healing and applying mobilization. Even- moval of the central third of the tendon, and transection
tually, this experimental data can be used to create a of the medial half of the tendon. Because partial
clinical rehabilitation protocol, which consists of a lacerations in tendons may be difficult to create
combination of immobilization and activity that allows reproducibly, caliper-based devices have been designed
for optimal tendon healing (Enwemeka et al., 1988). and tested to be accurate (Erhard et al., 2002). Although
a partial transection injury is not as clinically relevant as
4.6. Transection a complete transection injury, it however still allows the
tendon healing response to be studied. In addition,
In order to study tendon healing in an animal model, partial injuries have the advantage of not requiring
a distinct and reproducible injury must be created. The sutures to reduce gap formation since the tendon is still
two most common types of tendon transection models partially attached. It has also been shown that partially
have been complete and partial transections. Complete lacerated tendons are better off without suture repair
transections are a better simulation of what occurs (Bishop et al., 1986; Reynolds et al., 1976). In the case of
clinically, but the effect of sutures, which are necessary a window injury, a full-thickness transverse partial
to reduce gap formation, must be taken into account. transection is created (Carpenter et al., 1998a, b; Chan
Partial transections can be made in the middle half or et al., 1998; Kubota et al., 1996). The marginal tendons
along one side of the tissue. Although they are unlikely on either side of the transection provide enough support
ARTICLE IN PRESS
872 T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877

to allow for any potential early active motion (Kubota Much of the research done at the osteotendinous
et al., 1996). Mechanical testing of the healed specimens junction (OTJ) has focused primarily on the structure,
are performed after removal of marginal fibers (Car- nature and healing properties of the OTJ, which,
penter et al., 1998a, b; Chan et al., 1998). The central mechanically, is the weakest area of the early post-
third of the patellar tendon is often used as an autograft surgery. The OTJ is a complex transitional region
for ACL reconstruction. However, problems with the between bone and tendon and it has been demonstrated
remaining patellar tendon include donor site morbidity, that the short transition between tendon and bone
rupture, and a decrease in quadriceps strength (Mar- necessitates a variation in properties along the entire
umoto et al., 1996; Burks et al., 1990). Studies on these length of the insertion. This is important in order to
partially transected patellar tendons in either rabbits or functionally optimize the load transfer that must occur
dogs have shown opposite trends on the tensile proper- over this small area (Thomopoulos et al., 2003).
ties of the healing tendon. When compared to normal Histological and biomechanical analysis of extensor
contralateral controls up to 6 months post-operatively, tendon healing in a bone tunnel have indicated that
the failure strength of rabbit patellar tendons increased healing occurs by progressive ‘‘bone ingrowth and
while they decreased for canine patellar tendons mineralization’’ into the fibrovascular tissue in the early
(Beynnon et al., 1995; Burks et al., 1990). Differences stages of healing with subsequent reestablishment of the
in these results may be due to testing protocols as one collagen fiber continuity that is characteristic of the OTJ
study gripped the patella while the other removed it. (Rodeo et al., 1993).
Finally, some studies have also created a partial
transection in the medial half of patellar tendons 4.8. Tissue engineering
(Natsu-ume et al., 1997; Nakamura et al., 1996),
however, injuries that are only on one side of the tissue In addition to being used to study tendon healing
have been shown to be inconsistent with some injuries mechanisms, animal models have been important in
leading to complete rupture (Hefti et al., 1991; developing and testing tissue engineering methods to
O’Donoghue et al., 1966). improve the tendon healing process. Many of the studies
have focused on creating and using composites made
from various biomaterials and cells to facilitate healing
4.7. Junctions or to serve as potential replacement tissue. More
recently, the development of these repairs and replace-
Clinically, many indirect muscle injuries are localized ments have adhered to the approach of functional tissue
at or near the musculotendinous junction, including engineering, which proposes to measure in vivo forces in
delayed onset muscle soreness and muscle strain uninjured tissue and to use this information as a
(Noonan and Garrett, 1992). Much of the research in guideline in designing appropriate constructs that can
this area has been performed on rabbit, rat, and canine withstand in vivo loads (Butler et al., 2000). In addition,
models and has yielded interesting insights into the much research has been done on the exogenous
structure of the musculotendinous junction (MTJ) and application of cytokines to injured tendons to promote
localization of tendon tears (Noonan and Garrett, 1992; healing.
Best et al., 1989; Garrett and Tidball, 1988; Eisenberg The main factors to consider when creating a robust
and Milton, 1984; Armstrong et al., 1983; Abraham, and functional tissue engineered tendon construct
1977).The MTJ is morphologically characterized by a include a scaffold, cells, cytokines, and mechanical
continuous membrane which is extensively folded and stress. Traditionally, many studies determining the
seems to ‘‘interdigitate’’ with the muscle cell and biocompatibility of a composite have used a scaffold
extracellular connective tissue of the tendon (Andreev seeded with or without cells, which was then used as a
and Wassilev, 1986). At this junction we seem to find the replacement for native tissue. Not only should this
greatest weakness due to inherent ability of the tendon scaffold allow any seeded cells to proliferate rapidly, but
to withstand higher tensile and strain loads than muscle. the cells must initially survive after the composite is
Further studies done primarily on rabbit models have implanted in order to synthesize new tendinous tissue
shown that immediate and prolonged muscle fiber death (Butler and Awad, 1999). For example, a composite
in this region can be attributed to an ongoing calcium- constructed from polyester fibers around an elastometric
induced injury process (Reddy et al., 1991). Although it core was used as a replacement for rabbit Achilles
was originally believed that reduced extensibility of tendons (Goodship et al., 1985). Porcine small intestinal
sarcomeres resulted in rupture at the MTJ, more recent submucosa (SIS) was used to repair a segmental Achilles
studies have reported that ‘‘stiffer’’ sarcomeres do not tendon defect or to replace a completely resected
entirely account for the localization of fiber rupture and infraspinatus tendon (Dejardin et al., 2001; Badylak
that fibers do not actually tear at the MTJ but rather a et al., 1995). Although all of these replacement
short distance from the junction (LeCroy et al., 1989). biomaterials showed promising results to serve as a
ARTICLE IN PRESS
T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877 873

scaffold for cell growth and collagen deposition, they do years ago and has general clinical acceptance, its specific
not fulfill the biomechanical requirements of the original physiological mechanisms and therapeutic effects have
in vivo tendon. More recently, there has been more been a source of conflict among researchers. Some
research done on the effect of mechanical stress and earlier studies had concluded no beneficial effect of
cytokines. ultrasound treatment to tendon and ligament healing
Functional tissue engineering (FTE) is a new (Williamson et al., 1986; Roberts et al., 1982). However,
approach that places an emphasis on the importance more recent investigations have demonstrated that
of mechanical stress in determining the success of a therapeutic ultrasound may facilitate fibroblast prolif-
construct. Because one of the main goals of FTE is to eration and protein synthesis, with a significant increase
establish design parameters and safety factors for tissue- in tensile strength, rate of collagen synthesis, and energy
engineered implants, more studies that take into account absorption capacity of the tendons (Jackson et al., 1991;
the in vivo function of the replaced tissues must be done Enwemeka, 1989). The widespread use of this modality
(Butler et al., 2000). For example, the determination of by medical and surgical practitioners suggest efficacy
in vivo patellar forces in rabbits during inactivity or but a clearer understanding of the precise effects of
hopping on an inclined treadmill showed that peak ultrasound on tendon healing is needed.
forces and stress during vigorous activity never exceeded
10% of their ultimate values. Peak tendon forces and the
rates of rise and fall in these forces were shown to be 5. Future directions
positively correlated with activity level (Juncosa et al.,
2003). By taking different design criteria into considera- Over the past decade, new developments and our
tion, more functional and effective composites can be understanding of molecular biology have progressed
designed with a greater likelihood for success in vivo. and allowed investigators to apply new therapies to
A large number of studies have looked at the effects of improve tendon healing. Cell therapies involve the
cytokines on uninjured and injured tendons both in vitro delivery of mesenchymal stem cells directly to the injury
and in vivo. Although it is relatively easy to experimen- site, and gene therapies allow for the introduction of
tally add cytokines, there are many factors to consider, genetic material into the cells participating in healing.
some of which include how much to add (dosage), when Each of these methodologies has its own advantages,
to add (timing), and which ones to add (combination), and both are very promising for treating tendon injuries.
and how to add it (delivery) (Evans, 1999). It has been Mesenchymal stem cells (MSC) are progenitor cells
shown that in a healing environment, there are changes that differentiate into the body’s specialized cells during
in the expression of both cytokines and their receptors embryonic development. Because fibroblasts are be-
simultaneously, indicating a complex interaction of lieved to have their lineage traced to mesenchymal cells
influences leading to a healed structure (Sciore et al., (Caplan, 1994), there has been considerable interest in
1998; Panossian et al., 1997; Duffy et al., 1995). The utilizing MSCs as a potential therapy for tendon
aforementioned factors and the complex interaction healing. MSCs can be isolated from bone marrow and
between cells, ECM proteins, and cytokines often lead then most often seeded onto a collagen gel to create a
to the reporting of conflicting results between different tissue engineered composite, which is implanted in an
studies. For example, the addition of higher doses of injured tendon. Although MSC treated injuries only
GDF5 and GDF6 to injured Achilles tendons resulting showed slight improvements in material properties of
in an increase in failure load compared to lower doses rabbit patellar tendons compared to untreated injuries
(Aspenberg and Forslund, 1999). However, it was (Awad et al., 1999), composites that were contracted
shown that tendon healing in bone tunnels was onto a pretensioned suture prior to implantation in an
accelerated and superior with lower doses of rhBMP-2 injured rabbit Achilles tendon exhibited much improved
(Rodeo et al., 1999). These differences may also be due structural properties compared to controls (Fig. 7)
to the bell-shaped dose response curve of cytokines, so (Young et al., 1998). A study that injected the MSCs
the application of additional cytokines may not always without seeded onto a collagen gel showed no significant
be more effective (Evans, 1999). Finally, because healing disruption to tendon healing and demonstrated feasi-
occurs in a progressive, multifunctional way, optimal bility for use in an equine tendon healing model (Smith
healing may require the sequential application of several et al., 2003). The main advantage of using MSCs is their
cytokines in various combinations (Evans, 1999; Abra- pluripotent ability to differentiate into a desired cell
hamsson, 1997; Spindler et al., 1996). lineage.
The delivery of genetic material to cells can be
4.9. Ultrasound accomplished through both viral and non-viral methods.
Some examples of viruses include retrovirus, adeno-
Although the concept of using ultrasound in the virus, and adeno-associated virus, while non-viral
healing of tendon injuries was first proposed over 75 examples include liposomes and naked DNA (Mulligan,
ARTICLE IN PRESS
874 T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877

contribute towards a more thorough understanding of


tendon healing mechanisms. In addition, tissue engi-
neering concepts provide novel and exciting therapies to
improve the healing process. Although our cumulative
knowledge base is vast, it is still incomplete and more
research needs to be done in tendon injury and repair
before we can achieve the ultimate goal of directing the
healing process of an injured tendon to form tissue that
is structurally and functionally comparable to that of a
normal tendon.

References

Abraham, W.M., 1977. Factors in delayed muscle soreness. Medicine


and Science in Sports 9, 11–20.
Abrahamsson, S.O., 1997. Similar effects of recombinant human
insulin-like growth factor-I and II on cellular activities in flexor
tendons of young rabbits: experimental studies in vitro. Journal of
Fig. 7. Comparison of average stress-strain curves between MSC- Orthopaedic Research 15, 256–262.
treated and untreated rabbit Achilles tendons after injury and repair. Almekinders, L.C., Banes, A.J., Ballenger, C.A., 1993. Effects of
(Young, R.G., Butler, D.L., Weber, W., Caplan, A.I., Gordon, S.L., repetitive motion on human fibroblasts. Medicine and Science in
Fink, D.J., 1998. Use of mesenchymal stem cells in a collagen matrix Sports and Exercise 25, 603–607.
for Achilles tendon repair. Journal of Orthopaedic Research. 16, 406- Andreev, D.P., Wassilev, W.A., 1986. Specialized contacts between
413; with permission.) sarcolemma and sarcoplasmic reticulum at the ends of muscle fibers
in the diaphragm of the rat. Cell and Tissue Research 243, 415–420.
Aoki, M., Kubota, H., Pruitt, D.L., Manske, P.R., 1997. Biomecha-
nical and histologic characteristics of canine flexor tendon repair
1993). Using specific vectors, the genes of interest can be using early postoperative mobilization. Journal of Hand Surgery—
American Volume 22, 107–114.
introduced into the cells inside (in vivo) or outside (ex Archambault, J.M., Wiley, J.P., Bray, R.C., 1995. Exercise loading of
vivo) of the host. For example, in vivo gene transfer of tendons and the development of overuse injuries. A review of
PDGF-B by liposomes into cells from healing rat current literature. Sports Medicine 20, 77-89.
patellar tendons resulted in enhanced expression of Archambault, J., Tsuzaki, M., Herzog, W., Banes, A.J., 2002. Stretch
PDGF-B, promotion of angiogenesis, and enhancement and interleukin-1beta induce matrix metalloproteinases in rabbit
tendon cells in vitro. Journal of Orthopaedic Research 20, 36–39.
of matrix synthesis 4 weeks later (Nakamura et al., Armstrong, R.B., Ogilvie, R.W., Schwane, J.A., 1983. Eccentric
1998). In vivo gene transfer of pp125FAK, a protein exercise-induced injury to rat skeletal muscle. Journal of Applied
that may play a role in adhesion formation, by Physiology: Respiratory, Environmental and Exercise Physiology
adenovirus into chicken tendon cells found an over- 54, 80–93.
Aspenberg, P., Forslund, C., 1999. Enhanced tendon healing with
expression of FAK protein induces tendon adhesion
GDF 5 and 6. Acta Orthopaedica Scandinavica 70, 51–54.
formation (Lou et al., 1997). The advantages of using a Awad, H.A., Butler, D.L., Boivin, G.P., Smith, F.N., Malaviya, P.,
viral vector over a non-viral one are that the genetic Huibregtse, B., Caplan, A.I., 1999. Autologous mesenchymal stem
material can be amplified easily and increased efficiency, cell-mediated repair of tendon. Tissue Engineering 5, 267–277.
however, its disadvantages are its immunogenicity and Badylak, S.F., Tullius, R., Kokini, K., Shelbourne, K.D., Klootwyk,
toxicity (Yao and Eriksson, 2000). T., Voytik, S.L., Kraine, M.R., Simmons, C., 1995. The use of
xenogeneic small intestinal submucosa as a biomaterial for achilles
tendon repair in a dog model. Journal of Biomedical Materials
Research 29, 977–985.
6. Summary Barrie, K.A., Wolfe, S.W., 2001. The relationship of suture design to
biomechanical strength of flexor tendon repairs. Hand Surgery 6,
The clinical treatment of tendon injuries has improved 89–97.
Best, T.M., Glisson, R.R., Seaber, A.V., Garrett Jr., W.E., 1989. The
in no small part due to therapies and rehabilitation response of muscle tendon units of varying architecture to cyclic
protocols developed from both clinical and experimental passive stretching. Transactions of the Orthopaedic Research
research. Experimental animal models, whether in vitro Society, Las Vegas, NV.
or in vivo, allow us to focus on and investigate a certain Beynnon, B.D., Proffer, D., Drez Jr., D.J., Stankewich, C.J., Johnson,
R.J., 1995. Biomechanical assessment of the healing response of the
aspect or a combination of factors affecting tendon
rabbit patellar tendon after removal of its central third. American
healing. Whether the study design incorporates activity Journal of Sports Medicine 23, 452–457.
level and mobilization or looks at different types of Bishop, A.T., Cooney, W.P., Wood, M.B., 1986. Treatment of partial
injuries and injury locations, the findings of each study flexor tendon lacerations: the effect of tenorrhaphy and early
ARTICLE IN PRESS
T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877 875

protected mobilization. Journal of Trauma-Injury Infection and tendons: a new device for biomechanical studies. Journal of
Critical Care 26, 301–312. Biomechanics 35, 999–1002.
Buckwalter, J.A., Hunziker, E.B., 1996. Orthopaedics. healing of Evans, C.H., 1999. Cytokines and the role they play in the healing of
bones, cartilages, tendons, and ligaments: a new era. Lancet ligaments and tendons. Sports Medicine 28, 71–76.
348(suppl. II), 18. Feehan, L.M., Beauchene, J.G., 1990. Early tensile properties of
Burks, R.T., Haut, R.C., Lancaster, R.L., 1990. Biomechanical and healing chicken flexor tendons: early controlled passive motion
histological observations of the dog patellar tendon after removal versus postoperative immobilization. Journal of Hand Surgery—
of its central one-third. American Journal of Sports Medicine 18, American Volume 15, 63–68.
146–153. Fenwick, S.A., Hazleman, B.L., Riley, G.P., 2002. The vasculature and
Butler, D., Goldstein, S., Guilak, F., 2000. Functional tissue its role in the damaged and healing tendon. Arthritis Research 4,
engineering: the role of biomechanics. Transactions of the ASME 252–260.
122, 570–575. Frank, C., Woo, S.L., Amiel, D., Harwood, F., Gomez, M., Akeson,
Butler, D.L., Awad, H.A., 1999. Perspectives on cell and collagen W., 1983. Medial collateral ligament healing. A multidisciplinary
composites for tendon repair. Clinical Orthopaedics and Related assessment in rabbits. American Journal of Sports Medicine 11,
Research S324–332. 379–389.
Caplan, A.I., 1994. The mesengenic process. Clinics in Plastic Surgery Frank, C., McDonald, D., Bray, D., Bray, R., Rangayyan, R.,
21, 429–435. Chimich, D., Shrive, N., 1992. Collagen fibril diameters in the
Carlstedt, C.A., Madsen, K., Wredmark, T., 1986. Biomechanical and healing adult rabbit medial collateral ligament. Connective Tissue
biochemical studies of tendon healing after conservative and Research 27, 251–263.
surgical treatment. Archives of Orthopaedic and Traumatic Frank, C., McDonald, D., Shrive, N., 1997. Collagen fibril diameters
Surgery 105, 211–215. in the rabbit medial collateral ligament scar: a longer term
Carpenter, J.E., Flanagan, C.L., Thomopoulos, S., Yian, E.H., assessment. Connective Tissue Research 36, 261–269.
Soslowsky, L.J., 1998a. The effects of overuse combined with Garrett Jr., W.E., Tidball, J.G., 1988. Myotendinous junction:
intrinsic or extrinsic alterations in an animal model of rotator cuff structure, function, and failure. In: Woo, S.L., Buckwalter, J.
tendinosis. American Journal of Sports Medicine 26, 801–807. (Eds.), Injury and Repair of the Musculoskeletal Soft Tissues.
Carpenter, J.E., Thomopoulos, S., Flanagan, C.L., DeBano, C.M., American Academy of Orthopaedic Surgeons, Park Ridge, IL.
Soslowsky, L.J., 1998b. Rotator cuff defect healing: a biomecha- Gelberman, R.H., Vande Berg, J.S., Lundborg, G.N., Akeson, W.H.,
nical and histologic analysis in an animal model. Journal of 1983. Flexor tendon healing and restoration of the gliding surface.
Shoulder and Elbow Surgery 7, 599–605. An ultrastructural study in dogs. Journal of Bone and Joint
Chan, B.P., Fu, S.C., Qin, L., Rolf, C., Chan, K.M., 1998. Pyridinoline Surgery—American Volume 65, 70–80.
in relation to ultimate stress of the patellar tendon during healing: Gelberman, R.H., Manske, P.R., Akeson, W.H., Woo, S.L., Lund-
an animal study. Journal of Orthopaedic Research 16, 597–603. borg, G., Amiel, D., 1986. Flexor tendon repair. Journal of
Dejardin, L.M., Arnoczky, S.P., Ewers, B.J., Haut, R.C., Clarke, R.B., Orthopaedic Research 4, 119–128.
2001. Tissue-engineered rotator cuff tendon using porcine small Gelberman, R.H., Chu, C.R., Williams, C.S., Seiler 3rd, J.G., Amiel,
intestine submucosa. Histologic and mechanical evaluation in dogs. D., 1992. Angiogenesis in healing autogenous flexor-tendon grafts.
American Journal of Sports Medicine 29, 175–184. Journal of Bone and Joint Surgery—American Volume 74,
Dinopoulos, H.T., Boyer, M.I., Burns, M.E., Gelberman, R.H., Silva, 1207–1216.
M.J., 2000. The resistance of a four- and eight-strand suture Gelberman, R.H., Boyer, M.I., Brodt, M.D., Winters, S.C., Silva,
technique to gap formation during tensile testing: an experimental M.J., 1999. The effect of gap formation at the repair site on the
study of repaired canine flexor tendons after 10 days of in vivo strength and excursion of intrasynovial flexor tendons. An
healing. Journal of Hand Surgery—American Volume 25, 489–498. experimental study on the early stages of tendon-healing in dogs.
Ditsios, K.T., Burns, M.E., Boyer, M.I., Gelberman, R.H., Silva, M.J., Journal of Bone and Joint Surgery—American Volume 81,
2002. The rigidity of repaired flexor tendons increases following ex 975–982.
vivo cyclic loading. Journal of Biomechanics 35, 853–856. Gomez, M., 1995. The physiology and biochemistry of soft tissue
Doillon, C.J., Dunn, M.G., Bender, E., Silver, F.H., 1985. Collagen healing. In: Griffin, L. (Ed.), Rehabilitation of the Injured Knee,
fiber formation in repair tissue: development of strength and 2nd Edition. Mosby Company, St. Louis, MO, pp. 34–44.
toughness. Collagen and Related Research 5, 481–492. Goodship, A.E., Wilcock, S.A., Shah, J.S., 1985. The development of
Duffy Jr., F.J., Seiler, J.G., Gelberman, R.H., Hergrueter, C.A., 1995. tissue around various prosthetic implants used as replacements for
Growth factors and canine flexor tendon healing: initial studies in ligaments and tendons. Clinical Orthopaedics and Related
uninjured and repair models. Journal of Hand Surgery—American Research 196, 61–68.
Volume 20, 645–649. Grinnell, F., 1984. Fibronectin and wound healing. Journal of Cellular
Dykyj, D., Jules, K.T., 1991. The clinical anatomy of tendons. Journal Biochemistry 26, 107–116.
of the American Podiatric Medical Association 81, 358–365. Hefti, F.L., Kress, A., Fasel, J., Morscher, E.W., 1991. Healing of the
Eisenberg, B.R., Milton, R.L., 1984. Muscle fiber termination at the transected anterior cruciate ligament in the rabbit. Journal of Bone
tendon in the frog’s sartorius: a stereological study. American and Joint Surgery—American Volume 73, 373–383.
Journal of Anatomy 171, 273–284. Hess, G.P., Cappiello, W.L., Poole, R.M., Hunter, S.C., 1989.
Enwemeka, C.S., 1989. The effects of therapeutic ultrasound on Prevention and treatment of overuse tendon injuries. Sports
tendon healing. A biomechanical study. American Journal of Medicine 8, 371–384.
Physical Medicine and Rehabilitation 68, 283–287 (Erratum Hitchcock, T.F., Light, T.R., Bunch, W.H., Knight, G.W., Sartori,
appears in Am J Phys Med Rehabil 1990 Oct;69(5):258). M.J., Patwardhan, A.G., Hollyfield, R.L., 1987. The effect of
Enwemeka, C.S., Spielholz, N.I., Nelson, A.J., 1988. The effect of immediate constrained digital motion on the strength of flexor
early functional activities on experimentally tenotomized achilles tendon repairs in chickens. Journal of Hand Surgery—American
tendons in rats. American Journal of Physical Medicine and Volume 12, 590–595.
Rehabilitation 67, 264–269. Hyman, J., Rodeo, S.A., 2000. Injury and repair of tendons and
Erhard, L., Schultz, F.M., Zobitz, M.E., Zhao, C., Amadio, P.C., An, ligaments. Physical Medicine and Rehabilitation Clinics of North
K.N., 2002. Reproducible volar partial lacerations in flexor America 11, 267–288.
ARTICLE IN PRESS
876 T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877

Jackson, B.A., Schwane, J.A., Starcher, B.C., 1991. Effect of transfer of platelet-derived growth factor (PDGF)-B into healing
ultrasound therapy on the repair of achilles tendon injuries in patellar ligament. Gene Therapy 5, 1165–1170.
rats. Medicine and Science in Sports and Exercise 23, 171–176. Natsu-ume, T., Nakamura, N., Shino, K., Toritsuka, Y., Horibe, S.,
Jaibaji, M., 2000. Advances in the biology of zone II flexor tendon Ochi, T., 1997. Temporal and spatial expression of transforming
healing and adhesion formation. Annals of Plastic Surgery. 45, growth factor-beta in the healing patellar ligament of the rat.
83–92. Journal of Orthopaedic Research 15, 837–843.
Jozsa, L., Kannus, P., 1997. Human Tendons: Anatomy, Physiology, Noguchi 3rd, M., Seiler, J.G., Gelberman, R.H., Sofranko, R.A.,
and Pathology. Human Kinetics, Champaign, IL. Woo, S.L., 1993. In vitro biomechanical analysis of suture methods
Juncosa, N., West, J.R., Galloway, M.T., Boivin, G.P., Butler, D.L., for flexor tendon repair. Journal of Orthopaedic Research 11,
2003. In vivo forces used to develop design parameters for tissue 603–611.
engineered implants for rabbit patellar tendon repair. Journal of Noonan, T.J., Garrett Jr., W.E., 1992. Injuries at the myotendinous
Biomechanics 36, 483–488. junction. Clinics in Sports Medicine 11, 783–806.
Kubota, H., Manske, P.R., Aoki, M., Pruitt, D.L., Larson, B.J., Nystrom, B., Holmlund, D., 1983. Experimental evaluation of
1996. Effect of motion and tension on injured flexor tendons in immobilization in operative and non-operative treatment of
chickens. Journal of Hand Surgery—American Volume 21, achilles tendon rupture. A radiographic study in the rabbit. Acta
456–463. Chirurgica Scandinavica 149, 669–673.
Leadbetter, W.B., 1992. Cell-matrix response in tendon injury. Clinics O’Brien, M., 1992. Functional anatomy and physiology of tendons.
in Sports Medicine 11, 533–578. Clinics in Sports Medicine 11, 505–520.
LeCroy, C.M., Seaber, A.V., Reedy, M.K., Garrett Jr., W.E., 1989. O’Donoghue, D.H., Rockwood Jr., C.A., Zaricznyj, B., Kenyon, R.,
Limited sarcomere extensibility and strain injury in rabbit skeletal 1961. Repair of knee ligaments in dogs: the lateral collateral
muscle. Transactions of the Orthopaedic Research Society, Las ligament. Journal of Bone and Joint Surgery 43A, 1167–1178.
Vegas, NV. O’Donoghue, D.H., Rockwood Jr., C.A., Frank, G.R., Jack, S.C.,
Liu, S.H., Yang, R.S., al-Shaikh, R., Lane, J.M., 1995. Collagen in Kenyon, R., 1966. Repair of the anterior cruciate ligament in dogs.
tendon, ligament, and bone healing. A current review. Clinical Journal of Bone and Joint Surgery—American Volume 48,
Orthopaedics and Related Research 318, 265–278. 503–519.
Lou, J., Kubota, H., Hotokezaka, S., Ludwig, F.J., Manske, P.R., Palmes, D., Spiegel, H.U., Schneider, T.O., Langer, M., Stratmann,
1997. In vivo gene transfer and overexpression of focal adhesion U., Budny, T., Probst, A., 2002. Achilles tendon healing: long-term
kinase (pp125FAK) mediated by recombinant adenovirus-induced biomechanical effects of postoperative mobilization and immobi-
tendon adhesion formation and epitenon cell change. Journal of lization in a new mouse model. Journal of Orthopaedic Research
Orthopaedic Research 15, 911–918. 20, 939–946.
Lundborg, G., Rank, F., Heinau, B., 1985. Intrinsic tendon healing. A Panossian, V., Liu, S.H., Lane, J.M., Finerman, G.A., 1997.
new experimental model. Scandinavian Journal of Plastic and Fibroblast growth factor and epidermal growth factor receptors
Reconstructive Surgery 19, 113–117. in ligament healing. Clinical Orthopaedics and Related Research
Manske, P.R., 1988. Flexor tendon healing. Journal of Hand 342, 173–180.
Surgery—British Volume 13, 237–245. Piper, T.L., Whiteside, L.A., 1980. Early mobilization after knee
Manske, P.R., Gelberman, R.H., Vande Berg, J.S., Lesker, P.A., 1984. ligament repair in dogs: an experimental study. Clinical Orthopae-
Intrinsic flexor-tendon repair. A morphological study in vitro. dics and Related Research 150, 277–282.
Journal of Bone and Joint Surgery—American Volume 66, Potenza, A., 1962. Tendon healing within the flexor digital sheath in
385–396. the dog. Journal of Bone and Joint Surgery 44A, 49–64.
Manske, P.R., Gelberman, R.H., Lesker, P.A., 1985. Flexor tendon Potenza, A., 1963. Critical evalution of flexor tendon healing and
healing. Hand Clinics 1, 25–34. adhesion formation within artificial digital sheaths: an experi-
Marumoto, J.M., Mitsunaga, M.M., Richardson, A.B., Medoff, R.J., mental study. Journal of Bone and Joint Surgery 45A, 1217–1233.
Mayfield, G.W., 1996. Late patellar tendon ruptures after removal Potenza, A., 1964. Prevention of adhesions to healing digital flexor
of the central third for anterior cruciate ligament reconstruction. A tendons. The Journal of the American Medical Association 187,
report of two cases. American Journal of Sports Medicine 24, 99–103.
698–701. Potenza, A., 1969. Mechanisms of healing of digital flexor tendons.
Mass, D.P., Tuel, R., 1989. Human flexor tendon participation in the Hand 1, 40–41.
in vitro repair process. Journal of Hand Surgery—American Reddy, A.S., Reedy, M.K., Best, T.M., Lucaveche, C., Seaber, A.V.,
Volume 14, 64–71. Garrett Jr., W.E., 1991. Evaluation of strain injuries in rabbit
Mass, D.P., Tuel, R.J., 1991. Intrinsic healing of the laceration site in skeletal muscle with a single fiber model. Surgical Forum 42,
human superficialis flexor tendons in vitro. Journal of Hand 540–542.
Surgery—American Volume 16, 24–30. Reynolds, B., Wray Jr., R.C., Weeks, P.M., 1976. Should an
Montgomery, R.D., 1989. Healing of muscle, ligaments, and tendons. incompletely severed tendon be sutured? Plastic and Reconstructive
Seminars in Veterinary Medicine and Surgery (Small Animal) 4, Surgery 57, 36–38.
304–311. Roberts, M., Rutherford, J.H., Harris, D., 1982. The effect of
Mulligan, R.C., 1993. The basic science of gene therapy. Science 260, ultrasound on flexor tendon repairs in the rabbit. Hand 14, 17–20.
926–932. Rodeo, S.A., Arnoczky, S.P., Torzilli, P.A., Hidaka, C., Warren, R.F.,
Murrell, G.A., Lilly III, E.G., Goldner, R.D., Seaber, A.V., Best, 1993. Tendon-healing in a bone tunnel. A biomechanical and
T.M., 1994. Effects of immobilization on achilles tendon healing in histological study in the dog. Journal of Bone and Joint Surgery—
a rat model. Journal of Orthopaedic Research 12, 582–591. American Volume 75, 1795–1803.
Nakamura, N., Horibe, S., Matsumoto, N., Tomita, T., Natsuume, T., Rodeo, S.A., Suzuki, K., Deng, X.H., Wozney, J., Warren, R.F., 1999.
Kaneda, Y., Shino, K., Ochi, T., 1996. Transient introduction of a Use of recombinant human bone morphogenetic protein-2 to
foreign gene into healing rat patellar ligament. Journal of Clinical enhance tendon healing in a bone tunnel. American Journal of
Investigation 97, 226–231. Sports Medicine 27, 476–488.
Nakamura, N., Shino, K., Natsuume, T., Horibe, S., Matsumoto, N., Savage, R., 1985. In vitro studies of a new method of flexor tendon
Kaneda, Y., Ochi, T., 1998. Early biological effect of in vivo gene repair. Journal of Hand Surgery—British Volume 10, 135–141.
ARTICLE IN PRESS
T.W. Lin et al. / Journal of Biomechanics 37 (2004) 865–877 877

Schechtman, H., Bader, D.L., 1997. In vitro fatigue of human tendons. synovial sheaths and the effects of hyaluronan: an experimental
Journal of Biomechanics 30, 829–835. study in vivo and in vitro. Journal of Hand Surgery—American
Sciore, P., Boykiw, R., Hart, D.A., 1998. Semiquantitative reverse Volume 22, 818–825.
transcription-polymerase chain reaction analysis of mRNA for Williamson, J.B., George, T.K., Simpson, D.C., Hannah, B., Brad-
growth factors and growth factor receptors from normal and bury, E., 1986. Ultrasound in the treatment of ankle sprains. Injury
healing rabbit medial collateral ligament tissue. Journal of 17, 176–178.
Orthopaedic Research 16, 429–437. Winters, S.C., Seiler 3rd, J.G., Woo, S.L., Gelberman, R.H., 1997.
Silfverskiold, K.L., Andersson, C.H., 1993. Two new methods of Suture methods for flexor tendon repair. A biomechanical analysis
tendon repair: an in vitro evaluation of tensile strength and gap during the first six weeks following repair. Annales de Chirurgie de
formation. Journal of Hand Surgery—American Volume 18, 58–65 la Main et du Membre Superieur 16, 229–234.
(comment). Winters, S.C., Gelberman, R.H., Woo, S.L., Chan, S.S., Grewal, R.,
Silva, M.J., Boyer, M.I., Gelberman, R.H., 2002. Recent progress in Seiler 3rd, J.G., 1998. The effects of multiple-strand suture
flexor tendon healing. Journal of Orthopaedic Science 7, 508–514. methods on the strength and excursion of repaired intrasynovial
Silver, I.A., Brown, P.N., Goodship, A.E., Lanyon, L.E., McCullagh, flexor tendons: a biomechanical study in dogs. Journal of Hand
K.G., Perry, G.C., Williams, I.F., 1983. A clinical and experimental Surgery—American Volume 23, 97–104.
study of tendon injury, healing and treatment in the horse. Equine Woo, S.L., Gelberman, R.H., Cobb, N.G., Amiel, D., Lothringer, K.,
Veterinary Journal (Supplement) 1, 1–43. Akeson, W.H., 1981. The importance of controlled passive
Smith, R.K., Korda, M., Blunn, G.W., Goodship, A.E., 2003. mobilization on flexor tendon healing. A biomechanical study.
Isolation and implantation of autologous equine mesenchymal Acta Orthopaedica Scandinavica 52, 615–622.
stem cells from bone marrow into the superficial digital flexor Woo, S.L., Gomez, M.A., Woo, Y.K., Akeson, W.H., 1982.
tendon as a potential novel treatment. Equine Veterinary Journal Mechanical properties of tendons and ligaments. II. The relation-
35, 99–102. ships of immobilization and exercise on tissue remodeling.
Soslowsky, L.J., Carpenter, J.E., DeBano, C.M., Banerji, I., Moalli, Biorheology 19, 397–408.
M.R., 1996. Development and use of an animal model for Woo, S.L., Buckwalter, J.A., 1988. AAOS/NIH/ORS workshop.
investigations on rotator cuff disease. Journal of Shoulder and Injury and repair of the musculoskeletal soft tissues. Savannah,
Elbow Surgery 5, 383–392. Georgia, June 18–20, 1987. Journal of Orthopaedic Research 6,
Soslowsky, L.J., Thomopoulos, S., Tun, S., Flanagan, C.L., Keefer, 907–931.
C.C., Mastaw, J., Carpenter, J.E., 2000. Neer award 1999. Overuse Woo, S.L., Hildebrand, K., Watanabe, N., Fenwick, J.A., Papageor-
activity injures the supraspinatus tendon in an animal model: a giou, C.D., Wang, J.H., 1999. Tissue engineering of ligament and
histologic and biomechanical study. Journal of Shoulder and tendon healing. Clinical Orthopaedics and Related Research 367,
Elbow Surgery 9, 79–84. S312–S323.
Spindler, K.P., Imro, A.K., Mayes, C.E., Davidson, J.M., 1996. Woo, S.L., An, K., Frank, C., Livesay, G., Ma, A., Zeminski, J.,
Patellar tendon and anterior cruciate ligament have different Wayne, J., Myers, B., 2000a. Anatomy, biology, and bio-
mitogenic responses to platelet-derived growth factor and trans- mechanics of tendon and ligament. In: Buckwalter, J., Einhorn,
forming growth factor beta. Journal of Orthopaedic Research 14, T., Simon, S. (Eds.), Orthopaedic Basic Science: Biology and
542–546. Biomechanics of the Musculoskeletal System, 2nd Edition..
Strickland, J.W., Glogovac, S.V., 1980. Digital function following American Academy of Orthopaedic Surgeons, Rosemont, IL,
flexor tendon repair in zone II: a comparison of immobilization pp. 582–616.
and controlled passive motion techniques. Journal of Hand Woo, S.L., Debski, R.E., Zeminski, J., Abramowitch, S.D., Saw, S.S.,
Surgery—American Volume 5, 537–543. Fenwick, J.A., 2000b. Injury and repair of ligaments and tendons.
Taras, J.S., Lamb, M.J., 1999. Treatment of flexor tendon injuries: Annual Review of Biomedical Engineering 2, 83–118.
surgeons’ perspective. Journal of Hand Therapy 12, 141–148. Wright, P., 1999. Flexor and extensor tendon injuries. In: Canale, S.
Taylor, D.C., Dalton Jr., J.D., Seaber, A.V., Garrett Jr., W.E., 1993. (Ed.), Campbell’s Operative Orthopaedics. Mosby, St Louis, MO,
Experimental muscle strain injury. Early functional and structural pp. 3318–3363.
deficits and the increased risk for reinjury. American Journal of Yamamoto, E., Hayashi, K., Yamamoto, N., 2000. Effects of stress
Sports Medicine 21, 190–194. shielding on the transverse mechanical properties of rabbit patellar
Thomopoulos, S., Williams, G.R., Gimbel, J.A., Favata, M., tendons. Transactions of the ASME 122, 608–614.
Soslowsky, L.J., 2003. Variation of biomechanical, structural, Yamamoto, N., Ohno, K., Hayashi, K., Kuriyama, H., Yasuda, K.,
and compositional properties along the tendon to bone insertion Kaneda, K., 1993. Effects of stress shielding on the mechanical
site. Journal of Orthopaedic Research 21, 413–419. properties of rabbit patellar tendon. Journal of Biomechanical
Walsh, S., Frank, C., 1988. Two methods of ligament injury: a Engineering 115, 23–28.
morphological comparison in a rabbit model. Journal of Surgical Yao, F., Eriksson, E., 2000. Gene therapy in wound repair and
Research 45, 159–166. regeneration. Wound Repair and Regeneration 8, 443–451.
Wang, E.D., 1998. Tendon repair. Journal of Hand Therapy 11, Young, R.G., Butler, D.L., Weber, W., Caplan, A.I., Gordon, S.L.,
105–110. Fink, D.J., 1998. Use of mesenchymal stem cells in a collagen
Wiig, M., Abrahamsson, S.O., Lundborg, G., 1997. Tendon repair— matrix for achilles tendon repair. Journal of Orthopaedic Research
cellular activities in rabbit deep flexor tendons and surrounding 16, 406–413.

You might also like