ArticlePDF Available

Model for Muscle Regeneration around Fibrotic Lesions in Recurrent Strain Injuries

Authors:

Abstract and Figures

The purpose of this study was to establish an in vivo model for muscle regeneration after strain injury in the presence of a fibrotic discontinuity. The musculus soleus of 5-wk-old male rats was exposed, completely lacerated, and sutured together with or without a collagen scaffold in between the muscle ends. The scaffold represents a fibrotic discontinuity in the muscle. Muscle healing was evaluated after 14 d by general histology and staining for myofibroblasts, satellite cells (activated), and inflammatory cells. Around all wounds, satellite cells were activated. Inside the collagen scaffolds, satellite cells were absent, indicating that muscle regeneration was impaired. In the wounds without a collagen scaffold, the lacerated and the sutured myofibers contacted and had already started to regenerate, whereas this did not occur with an implanted scaffold. A fibrotic discontinuity, such as an implanted collagen scaffold, delays muscle regeneration in skeletal muscle. This model is suitable to study skeletal muscle regeneration in the presence of a fibrotic lesion and to evaluate new treatment modalities for muscle strain injuries.
Content may be subject to copyright.
by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.Copyright @ 2010
Methodological Advances
Model for Muscle Regeneration around
Fibrotic Lesions in Recurrent Strain Injuries
SANDER GREFTE
1
, ANNE MARIE KUIJPERS-JAGTMAN
1
, RUURD TORENSMA
2
,
and JOHANNES W. VON DEN HOFF
1
1
Department of Orthodontics and Oral Biology, and
2
Department of Tumor Immunology, Radboud University
Nijmegen Medical Centre, Nijmegen, THE NETHERLANDS
ABSTRACT
GREFTE, S., A. M. KUIJPERS-JAGTMAN, R. TORENSMA, and J. W. VON DEN HOFF. Model for Muscle Regeneration around
Fibrotic Lesions in Recurrent Strain Injuries. Med. Sci. Sports Exerc., Vol. 42, No. 4, pp. 813–819, 2010. Purpose: The purpose of this
study was to establish an in vivo model for muscle regeneration after strain injury in the presence of a fibrotic discontinuity. Methods:The
musculus soleus of 5-wk-old male rats was exposed, completely lacerated, and sutured together with or without a collagen scaffold in
between the muscle ends. The scaffold represents a fibrotic discontinuity in the muscle. Muscle healing was evaluated after 14 d by general
histology and staining for myofibroblasts, satellite cells (activated), and inflammatory cells. Results: Around all wounds, satellite cells were
activated. Inside the collagen scaffolds, satellite cells were absent, indicating that muscle regeneration was impaired. In the wounds without
a collagen scaffold, the lacerated and the sutured myofibers contacted and had already started to regenerate, whereas this did not occur
with an implanted scaffold. Conclusions: A fibrotic discontinuity, such as an implanted collagen scaffold, delays muscle regeneration in
skeletal muscle. This model is suitable to study skeletal muscle regeneration in the presence of a fibrotic lesion and to evaluate new
treatment modalities for muscle strain injuries. Key Words: MUSCLE STRAIN INJURIES, FIBROSIS, MUSCLE HEALING,
SATELLITE CELLS
Muscle strain injuries occur regularly in profes-
sional athletes as well as in the general popula-
tion (14). The hamstring is the most common
muscle group affected and is characterized by a recurrence
rate of 30% within the first year after injury. This indicates
that full recovery of a hamstring strain injury is often not
obtained (7,34,35). Magnetic resonance imaging analysis
shows that during the healing of a hamstring injury, fibrotic
tissue is formed, preventing full recovery (10,38).
In muscle strain injuries, the muscle is sheared, which
results in a total rupture of the myofibers and their plasma
membrane (21,24). At this site, necrosis of the myofibers
begins but is restricted to the injury site by contraction
bands inside the myofibers (19). After injury, satellite cells,
which are located between the sarcolemma and the basal
lamina of the muscle fibers (29,32), are released, activated,
and migrate to the site of injury. There they proliferate,
differentiate, and fuse to each other or to damaged myo-
fibers to regenerate the skeletal muscle (8,37). However,
blood vessels are also torn, and a hematoma is formed,
filling the gap between the damaged muscle ends. This
forms a primary matrix not only for inflammatory cells but
also for fibroblasts, which synthesize extracellular matrix
components (21,24). These fibroblasts firstly produce
fibronectin, followed by collagen type III, and finally
collagen type I (22). This might lead to a fibrotic tissue
that inhibits growth of muscle fibers and thus impairs
regeneration and muscle function (15,18,24,25). It has been
shown that recurrent muscle strains occur in proximity of
this fibrotic discontinuity, probably because of its different
stiffness and contractility properties (34,38). Furthermore,
recurrent injuries are also more severe and take a longer
time to heal than primary strain injuries (7,27). It is there-
fore important to prevent or to minimize the formation of
such a fibrotic discontinuity to reduce the risk of recurrence.
To reduce fibrosis and to optimize muscle regeneration,
several strategies have been evaluated. The injection of
growth factors such as insulin-like growth factor, fibroblast
growth factor 2, nerve growth factor, and granulocyte
SPECIAL COMMUNICATIONS
Address for correspondence: Johannes W. Von den Hoff, Ph.D.,
Department of Orthodontics and Oral Biology, Radboud University
Nijmegen Medical Centre, PO Box 9101, 6500 HB Nijmegen, The
Netherlands; E-mail: H.vondenHoff@dent.umcn.nl.
Submitted for publication June 2009.
Accepted for publication August 2009.
0195-9131/10/4204-0813/0
MEDICINE & SCIENCE IN SPORTS & EXERCISE
Ò
Copyright Ó2010 by the American College of Sports Medicine
DOI: 10.1249/MSS.0b013e3181beeb52
813
by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.Copyright @ 2010
colony-stimulating factor improves muscle regeneration
(25,36,39). More importantly, the administration of decorin,
an inhibitor of transforming growth factor A, reduces fi-
brosis (12,36). The direct delivery of isolated muscle cells
is another approach (9,16,31). Although the latter yields
promising results, a major problem is the poor cell survival
and limited migration of the injected cells (3,11). Alterna-
tively, several different scaffold materials have been used
for improving muscle regeneration but with varying results
(5,17,26,41). However, a model to study impaired healing
in the presence of a fibrotic lesion is not yet available.
Therefore, the aim of this study is to establish an in vivo
model for a fibrotic discontinuity in healing skeletal muscle
by implanting a collagen scaffold.
MATERIALS AND METHODS
Animals. All animal experiments were approved by the
Animal Experiments Committee of the Radboud University
Nijmegen Medical Centre in accordance to the Dutch laws
and regulations on animal experiments, which conform to
the American College of Sports Medicine animal care
standards. Twenty-four 5-wk-old male Sprague–Dawley
rats (Janvier, Le Genest, France) were used for the
experiments. The rats were housed under normal laboratory
conditions, but in the first week after the experimental
procedure, they were housed individually. All the rats were
fed normal rat chow and water ad libitum. Before the start
of the experiments, the rats had been acclimatized to the
animal facility for 1 wk.
Preparation of the collagen scaffolds. The collagen
scaffolds were prepared and chemically cross-linked as
previously described (6). Briefly, a 1% (w/v) homogenized
collagen suspension was prepared using insoluble type I
collagen from bovine Achilles tendon (Sigma Chemical Co.,
St. Louis, MO). The collagen suspension was degassed to
remove air bubbles, frozen overnight at j25-Cinaluminum
trays, and lyophilized. The dried collagen scaffolds were
cross-linked using 1-ethyl-3-(3 dimethylaminopropyl) car-
bodiimide hydrochloride and n-hydroxy-succinimide (43).
Experimental procedures. At the day of surgery, the
rats received 0.02 mgIkg
j1
body weight buprenorphine
(Temgesic; Schering Plough, Brussels, Belgium) subcuta-
neously as an analgesic and also at the next 2 d with a
12-h interval. Under 5% (induction) followed by 2%–3%
(continuation) isoflurane anesthesia (Pharmachemie BV,
Haarlem, The Netherlands), the left lower limb of the rats
was shaved. After a longitudinal incision in the skin and
underlying fascia, the musculus soleus was gently exposed
and transversally lacerated. The two ends were sutured
together using a 7-0 Polysorb suture (Tyco Healthcare UK,
Gosport, UK) with or without the collagen scaffold in
between. Before implantation, the collagen scaffolds were
sterilized by immersion in 70% ethanol for 1 h and then
washed three times with sterile phosphate-buffered saline
(PBS). The animals were divided into four groups of six
rats according to the suturing method and the presence of a
collagen scaffold: A) knot suturing without collagen
scaffold, B) knot suturing with collagen scaffold, C) con-
tinuous suturing without collagen scaffold, and D) contin-
uous suturing with collagen scaffold. The easiest method
for suturing is with one continuous suture around the
muscle. However, if this one suture breaks, the wound
opens and the scaffold might be lost. To be sure, we also
used a method with multiple sutures. However, none of the
sutures had broken, and there was no different response
between the two suturing methods. We therefore decided to
group the animals together (A + C and B + D). The fascia
and the skin were closed with 5-0 Polysorb and 5-0 Vicryl
sutures (Johnson-Johnson, Langhorne, PA), respectively.
To minimize muscle tension, the paw was splinted with an
aluminum strip at an angle approximately 45-with respect
to the tibia for 1 wk. In group B, the paws were swollen and
reddish when the aluminum strips were removed. These rats
therefore received 1 mgIkg
j1
enrofloxacin two times a day
(Bayer Healthcare, Brussels, Belgium) for 7 d. After 14 d,
the rats were sacrificed according to the standard CO
2
/O
2
protocol.
Histology and immunohistochemistry. After sacri-
fice, the left (wound) and the right (internal control) musculus
soleus of three rats of each group were fixed in freshly pre-
pared 4% paraformaldehyde in PBS for 4–6 h and processed
for paraffin embedding. The left and the right musculus
soleus of the other three rats of each group were immediately
frozen in optimal cutting temperature compound (OCT)
embedding compound (CellPath, Newtown, UK) using
isopentane precooled in liquid nitrogen. The muscles were
cut longitudinally, and 5-Hm sections were collected on
superfrost plus slides (Menzel-Gla¨ser, Braunschweig,
Germany). For general morphology, paraffin sections were
stained with hematoxylin and eosin (H&E).
Paraffin sections were also stained with the following
antibodies: mouse anti-alpha-smooth muscle actin (>-SMA;
Sigma), rabbit anti-Ki67 (Research Diagnostics Inc., Flan-
ders, NJ), mouse anti-ED1 (CD68, Serotec; DPC, Breda, The
Netherlands), and mouse anti-MyoD (DAKO; Dakopatts,
Glostrup, Denmark). Briefly, the sections were deparaffinated,
rehydrated, treated with 3% H
2
O
2
for 20 min to inactivate
endogenous peroxidase, and postfixed with 4% formalde-
hyde in PBS. For >-SMA and ED1 staining, the sections
were heated in citrate buffer (pH 6.0) for 10 min at 70-C. For
Ki67 and MyoD staining, the sections were heated to 100-C
for 10 and 40 min, respectively. After rinsing with 0.075%
glycine in PBS, the sections were preincubated with 10%
normal donkey serum (NDS; Chemicon, Temecula, CA)
followed by the antibodies against >-SMA (1:1600), ED1
(1:100), Ki67 (1:50), or MyoD (1:25) for 60 min. Subse-
quently, the biotinylated secondary antibodies goat-anti-
mouse immunoglobulin G (IgG; H + L) (1:500; Jackson
Labs, West Grove, PA) for >-SMA, ED1, and MyoD and
goat-anti-rabbit IgG (H + L) (1:500; Jackson Labs) for Ki67
were added. The bound antibodies were visualized using a
http://www.acsm-msse.org814 Official Journal of the American College of Sports Medicine
SPECIAL COMMUNICATIONS
by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.Copyright @ 2010
preformed biotinylated horse radish peroxidase and avidin
complex (Vector Laboratories, Burlingame, CA).
The frozen sections were double stained with the antibodies
rabbit anti-collagen IV (Euro-Diagnostica BV, Arnhem, The
Netherlands) and mouse anti-Pax7 (Developmental Studies
Hybridoma Bank, Iowa City, CA). Briefly, the sections were
dried in air overnight and postfixed with 1% paraformalde-
hyde in PBS for 10 min. After rinsing with 0.05% Triton X-
100 in PBS, the sections were preincubated with 10% NDS
followed by rabbit anti-collagen IV (1:100) for 60 min.
Collagen IV was then detected using the biotinylated donkey
anti-rabbit IgG (H + L) (1:500; Jackson Labs) for 60 min and
an AlexaFluor-488-labeled avidin (1:500; Molecular Probes,
Eugene, OR) for 60 min. Thereafter, the sections were again
preincubated with 10% NDS and then incubated with mouse
anti-Pax7 (1:100) overnight at 4-C. Pax7 was detected using
an AlexaFluor-594-labeled goat-anti-mouse IgG (H + L)
(1:200; Molecular Probes). All sections were photographed
with the Zeiss Imager.Z1 together with the AxioCam MRc5
camera using the AxioVision 4.6.3 software (Carl Zeiss
Microimaging GmbH, Jena, Germany).
Statistical analysis. The numbers of Pax7- and
MyoD-positive cells were counted in 1) the control muscle
(C), 2) the cutting zone without the collagen scaffold (W),
3) the cutting zone with the collagen scaffold (W + S), 4)
inside the collagen scaffold (S), and 5) in the noninjured
muscle tissue of the wounded musculus soleus (NI). To
count the Pax7-positive cells, the images were divided into
50 squares. In five random squares, the total number of
Pax7-positive cells and DAPI-stained nuclei was counted.
The total number of MyoD-positive cells and nuclei of
every group was determined in three different fields of an
overview image. The numbers of Pax7- and MyoD-positive
cells were expressed as a percentage TSD of the total
number of cells. The differences in the percentages of Pax7-
and MyoD-positive cells were tested for significance using
a Kruskal–Wallis one-way ANOVA on Ranks followed by
Dunn’s method. A value of PG0.05 was considered to be
significant.
RESULTS
Of the 24 rats, 1 rat in group A without a collagen
scaffold did not survive the surgery. After an initial growth
arrest, all rats in every group had gained about 25% body
weight at the 10th day. The groups were not significantly
different. The immobilization of the left hind leg did not
affect the growth of the rats. Macroscopically, the wounded
muscle adhered partly to the surrounding tissues. Further-
more, the collagen scaffolds were not visible anymore and
appeared to be integrated into the muscle tissue. The su-
tures did not break, and the different suturing methods
had no effect on muscle morphology and gave the same
results regarding muscle regeneration. Therefore, the ani-
mals of groups A and C and groups B and D were grouped
together.
General histology. H&E staining (Fig. 1A) revealed
properly arranged longitudinal myofibers in the controls (C)
but not in the wounded muscles. Within the wounds,
regenerating myofibers were present, indicated by centrally
located nuclei (Fig. 1A, magnification). Some myofibers in
the cutting zone had fused properly in the group without the
collagen scaffold. On the contrary, the implantation of a
collagen scaffold prevented fusion of the myofibers (W + S).
The collagen scaffolds were surrounded by giant cells and a
cell layer (an interphase).
Immunostainings. Paraffin sections were stained with
antibodies against >-SMA, ED1 (CD68), Ki67, and MyoD
to identify blood vessels and myofibroblasts, inflammatory
cells, proliferating cells, and activated satellite cells,
respectively (Fig. 1B). In the controls (C), ED1-positive
inflammatory cells and Ki67-positive proliferating cells
were present. However, the controls hardly contained any
MyoD-positive nuclei (indicated by arrows). As expected,
>-SMA–positive cells were not present in the muscle tissue
of the controls but only in blood vessels. Without a collagen
scaffold (W), there was an increase in the number of ED1-
and Ki67-positive cells of which the majority surrounded
the sutures. More importantly, many MyoD-positive nuclei
were present (a few are indicated by arrows). The number
of >-SMA–positive blood vessels was also increased, and
there were also >-SMA–positive cells in the muscle tissue.
The implantation of a collagen scaffold (W + S) caused an
infiltration of ED1-positive giant cells and other inflamma-
tory cells, which surrounded the scaffold. Even inside the
scaffolds, inflammatory cells were present. Proliferating
Ki67-positive cells were present in the muscle tissue, in the
interphase, and also inside the collagen. Again, the muscle
tissue around the scaffold (W + S) contained many MyoD-
positive nuclei (a few are indicated by arrows), but all the
cells inside the scaffolds were negative for MyoD. The
expression pattern of >-SMA was similar to the wounds
without the scaffold (W). Inside the scaffolds, >-SMA–
positive blood vessels were also found.
To identify the resident satellite cells, cryosections were
stained with the Pax7 antibody (Fig. 2). In the controls (C),
only a few satellite cells were present, but around the
cutting zone in the wounded muscle tissue with (W + S) or
without (W) a collagen scaffold, the number of satellite
cells was increased. However, no satellite cells were present
within the collagen scaffolds.
Quantifications. The percentage of MyoD- and Pax7-
positive cells were determined on the paraffin (Fig. 1B) and
cryosections (Fig. 2), respectively (Fig. 3). The controls
contained only a low number of Pax7-positive satellite cells
(2.7% T0.4%), which significantly (PG0.05) increased to
7.2% T0.6% and 6.2% T0.6% in the wounded tissue
without (W) or with (W + S) the collagen scaffold, re-
spectively. The number of MyoD-positive cells also signifi-
cantly (PG0.05) increased from 6.2% T1.1% in the
controls to 16% T4.3% and 15.9% T4.9% in the wounds
without (W) or with (W + S) the collagen scaffold,
EFFECT OF SCARRING DURING MUSCLE HEALING Medicine & Science in Sports & Exercise
d
815
SPECIAL COMMUNICATIONS
by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.Copyright @ 2010
FIGURE 1—Histology of the musculus soleus at 14 d after surgery. A. H&E staining of the control (C), wound without the collagen scaffold (W;
group C), and wound with the collagen scaffold (W + S; group D) revealed the disruption of the aligned myofibers at the cutting zone after laceration.
The implanted collagen scaffold is surrounded by an interphase and prevented myofiber fusion. B. Immunohistochemistry of the control (C), wound
without the collagen scaffold (W; group C), and wound with the collagen scaffold (W + S; group D) with antibodies directed against ED1, Ki67,
MyoD, and >-SMA. Only a few ED1-, Ki67-, and MyoD-positive cells (indicated by arrows) and >-SMA–positive blood vessels are present in the
control (C). In the wound (W), the number of these cells is higher and >-SMA–positive cells are present. The collagen scaffold (W + S) is surrounded
by an ED1-positive interphase. In the wounded muscles and around the scaffold, many Ki67-positive, MyoD-positive (a few are indicated by arrows),
and >-SMA–positive cells are present. The scaffold also contains ED1-, Ki67, and >-SMA–positive cells and blood vessels but no MyoD-positive cells.
http://www.acsm-msse.org816 Official Journal of the American College of Sports Medicine
SPECIAL COMMUNICATIONS
by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.Copyright @ 2010
respectively. Furthermore, there was a slight but nonsig-
nificant increase of Pax7-positive (3.9% T0.5%) and
MyoD-positive cells (9.1% T1.9%) in the noninjured area
(NI) of the wounded muscles compared with the controls.
However, no Pax7- and MyoD-positive cells were found
inside the collagen scaffolds.
DISCUSSION
The successful treatment of muscle strains in sports
medicine is still a problem. Fibrotic lesions are often formed
during muscle regeneration, causing incomplete functional
recovery. More importantly, recurrent muscle injuries may
occur near this fibrotic tissue (10,34,38). Because fibrotic
tissue consists mainly of collagen type I (20,23), we
developed an in vivo model for a fibrotic discontinuity by
implanting a type I collagen scaffold between the lacerated
muscle ends. Using this method, it is possible to standardize
the wounds with a collagen scaffold, but it is important to
be aware that this is an extreme version of a muscle strain.
In this model, we evaluated muscle regeneration after a
2-wk healing period. The numbers of Pax7- and MyoD-
positive (activated) satellite cells or myoblasts were in-
creased about twofold in the wounded muscle tissue and
around the collagen scaffolds compared with the control
muscle. This indicates that the muscle fibers were regener-
ating and that the scaffold did not inhibit the activation of
satellite cells in the adjacent muscle tissue. However, inside
the collagen scaffold, these cells were absent. Thus, in the
presence of a fibrotic discontinuity, the skeletal muscle
cannot regenerate properly because activated satellite cells
do not migrate into the fibrotic tissue. Similar to our results,
others have also shown that after a strain injury, inflamma-
tion occurs, followed by the production of fibrous tissue,
which could eventually develop into a fibrotic lesion (14,33).
Another study on rectus femoris strain in humans showed a
chronic inflammation and a mixture of regenerating muscle
fibers and fibrotic tissue in the wound (40). Although
muscle regeneration was only evaluated after 2 wk in this
initial study, collagen scaffolds can persist in the muscle
tissue for up to 50 d (28). Therefore, our model can be used
to evaluate treatment strategies for recurrent muscle strains.
Optimal treatment should diminish or prevent the forma-
tion of fibrotic tissue and reduce the risk of recurrence. We
and others (1,30) observed that suturing the lacerated
muscle ends directly together allows full regeneration of
the muscle. Currently, the treatment principle of muscle
strains consists of rest, ice, compression, and elevation (22).
With specific compression, which could serve as a splint, it
may also be possible to bring the muscle ends to each other
and diminish the onset of fibrosis. Surgical treatment to
suture the muscle ends together is only indicated in cases
with extensive injury to the muscle (22). If a fibrotic tissue
FIGURE 2—Fluorescent immunohistochemistry of the musculus soleus at 14 d after surgery. The control (C), wound without the collagen scaffold
(W; group A), and wound with the collagen scaffold (W + S; group B) were stained with the antibody directed against Pax7. In the control, only a few
Pax7-positive cells are present, whereas in the wound (W), the number of these cells is increased. In the wounded muscle tissue around the collagen
scaffold (W + S), the number of Pax7-positive cells is also increased. On the contrary, these cells are absent in the interphase and the collagen
scaffold.
EFFECT OF SCARRING DURING MUSCLE HEALING Medicine & Science in Sports & Exercise
d
817
SPECIAL COMMUNICATIONS
by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.Copyright @ 2010
from a previous injury is already present, additional
treatment with matrix metalloproteinase 1 (MMP-1) might
offer a solution. Previous research has shown that treatment
with injection of MMP-1 improves muscle regeneration and
that a fibrotic lesion can be partially resolved (4,23). Thus,
combining the injection of MMP-1 with specific compres-
sion therapy might diminish a preexisting fibrotic discon-
tinuity or minimize the risk of a secondary fibrosis.
In this study, the musculus soleus in rats is used as a
wound model because all the myofibers run parallel. How-
ever, the musculus soleus consists mainly of type I (slow)
fibers (42), whereas the hamstring, which is the most
common muscle group affected in muscle strains, consist of
type II (fast) fibers (13). It has been shown that type II
muscles regenerate better than type I muscles, which more
often develop fibrotic lesions (2). This indicates that the
results obtained in this study may differ from a hamstring
injury, in which the regeneration process could be more
efficient. However, it also demonstrates that the musculus
soleus is a good model to study the effects of the presence
of a fibrotic discontinuity on muscle regeneration.
In this study, we only analyzed 14 d after surgery be-
cause satellite cell activation is a relatively early event in
muscle healing (8,37). In future studies, analysis at later
time points is necessary to exclude the possibility that im-
plantation of a collagen scaffold only delays muscle regen-
eration. In addition, it is important that functional studies
are performed to further evaluate this model.
In conclusion, we generated a model for the regeneration
of skeletal muscle in the presence of a fibrotic discontinuity.
This model can be used to evaluate new treatment strategies
for recurrent muscle strains.
The Pax7 antibody developed by Atsushi Kawakami was ob-
tained from the Developmental Studies Hybridoma Bank developed
under the auspices of the National Institute of Child Health and
Human Development and maintained by The University of Iowa,
Department of Biological Sciences, Iowa City, IA 52242. This study
was supported by a grant from the Radboud University Nijmegen
Medical Centre, The Netherlands. For this research, we did not
receive any funding from the National Institutes of Health, the
Wellcome Trust, the Howard Hughes Medical Institute, or any other
organization. The results of the present study do not constitute
endorsement by the American College of Sports Medicine.
REFERENCES
1. A
¨a¨rimaa V, Ka¨a¨ria¨ inen M, Vaittinen S, et al. Restoration of
myofiber continuity after transection injury in the rat soleus.
Neuromuscul Disord. 2004;14(7):421–8.
2. Bassaglia Y, Gautron J. Fast and slow rat muscles degenerate and
regenerate differently after whole crush injury. J Muscle Res Cell
Motil. 1995;16(4):420–9.
3. Beauchamp JR, Morgan JE, Pagel CN, Partridge TA. Dynamics
of myoblast transplantation reveal a discrete minority of precur-
sors with stem cell-like properties as the myogenic source. J Cell
Biol. 1999;144(6):1113–22.
4. Bedair H, Liu TT, Kaar JL, et al. Matrix metalloproteinase-1
therapy improves muscle healing. J Appl Physiol. 2007;102(6):
2338–45.
5. Beier JP, Stern-Straeter J, Foerster VT, Kneser U, Stark GB,
Bach AD. Tissue engineering of injectable muscle: three-
dimensional myoblast-fibrin injection in the syngeneic rat animal
model. Plast Reconstr Surg. 2006;118(5):1113–21.
6. Bladergroen BA, Siebum B, Siebers-Vermeulen KG, et al. In vivo
recruitment of hematopoietic cells using stromal cell-derived
factor 1 alpha-loaded heparinized three-dimensional collagen
scaffolds. Tissue Eng Part A. 2009;15(7):1591–9.
7. Brooks JH, Fuller CW, Kemp SP, Reddin DB. Incidence, risk,
and prevention of hamstring muscle injuries in professional rugby
union. Am J Sports Med. 2006;34(8):1297–306.
8. Charge
´SB, Rudnicki MA. Cellular and molecular regulation of
muscle regeneration. Physiol Rev. 2004;84(1):209–38.
9. Collins CA, Olsen I, Zammit PS, et al. Stem cell function, self-
renewal, and behavioral heterogeneity of cells from the adult
muscle satellite cell niche. Cell. 2005;122(2):289–301.
10. Connell DA, Schneider-Kolsky ME, Hoving JL, et al. Longitudi-
nal study comparing sonographic and MRI assessments of acute
and healing hamstring injuries. AJR Am J Roentgenol. 2004;
183(4):975–84.
11. Fan Y, Maley M, Beilharz M, Grounds M. Rapid death of injected
myoblasts in myoblast transfer therapy. Muscle Nerve. 1996;
19(7):853–60.
12. Fukushima K, Badlani N, Usas A, Riano F, Fu FH, Huard J. The
use of an antifibrosis agent to improve muscle recovery after
laceration. Am J Sports Med. 2001;29(4):394–402.
13. Garrett WE Jr, Califf JC, Bassett FH 3rd. Histochemical correlates
of hamstring injuries. Am J Sports Med. 1984;12(2):98–103.
14. Garrett WE Jr. Muscle strain injuries. Am J Sports Med.
1996;24(suppl 6):S2–8.
15. Grefte S, Kuijpers-Jagtman AM, Torensma R, Von den Hoff JW.
FIGURE 3—Quantification of Pax7- and MyoD-positive (activated)
satellite cells. The number of Pax7- and MyoD-positive cells is
expressed as a mean percentage TSD of the total number of cells.
Compared with the controls (C; N= 11), the number of Pax7- and
MyoD-positive cells is significantly increased in the wounded muscle
tissue of wounds with (W + S; N= 6) or without (W; N= 5) the
collagen scaffold. In the noninjured area of the wounded muscles (NI;
N= 11), the number of Pax7- and MyoD-positive cells is also increased
but not significant. In the scaffolds (S; N= 6), no Pax7- and MyoD-
positive cells are found. *Significantly increased (PG0.05) compared
with the control.
http://www.acsm-msse.org818 Official Journal of the American College of Sports Medicine
SPECIAL COMMUNICATIONS
by the American College of Sports Medicine. Unauthorized reproduction of this article is prohibited.Copyright @ 2010
Skeletal muscle development and regeneration. Stem Cells Dev.
2007;16(5):857–68.
16. Gross JG, Morgan JE. Muscle precursor cells injected into
irradiated mdx mouse muscle persist after serial injury. Muscle
Nerve. 1999;22(2):174–85.
17. Hill E, Boontheekul T, Mooney DJ. Regulating activation of
transplanted cells controls tissue regeneration. Proc Natl Acad Sci
USA. 2006;103(8):2494–9.
18. Huard J, Li Y, Fu FH. Muscle injuries and repair: current trends in
research. J Bone Joint Surg Am. 2002;84(5):822–32.
19. Hurme T, Kalimo H, Lehto M, Ja¨rvinen M. Healing of skeletal
muscle injury: an ultrastructural and immunohistochemical study.
Med Sci Sports Exerc. 1991;23(7):801–10.
20. Hurme T, Kalimo H, Sandberg M, Lehto M, Vuorio E.
Localization of type I and III collagen and fibronectin produc-
tion in injured gastrocnemius muscle. Lab Invest. 1991;64(1):
76–84.
21. Ja¨rvinen TA, Ka¨a¨ria¨inen M, Ja¨ rvinen M, Kalimo H. Muscle strain
injuries. Curr Opin Rheumatol. 2000;12(2):155–61.
22. Ja¨rvinen TAH, Ja¨rvinen TLN, Ka¨a¨ria¨ inen M, Kalimo H, Ja¨ rvinen
M. Muscle injuries: biology and treatment. Am J Sports Med.
2005;33(5):745–64.
23. Kaar JL, Li Y, Blair HC, et al. Matrix metalloproteinase-1
treatment of muscle fibrosis. Acta Biomater. 2008;4(5):1411–20.
24. Ka¨a¨ ria¨ inen M, Ja¨ rvinen T, Ja¨ rvinen M, Rantanen J, Kalimo H.
Relation between myofibers and connective tissue during muscle
injury repair. Scand J Med Sci Sports. 2000;10(6):332–7.
25. Kasemkijwattana C, Menetrey J, Somogyl G, et al. Development
of approaches to improve the healing following muscle contusion.
Cell Transplant. 1998;7(6):585–98.
26. Kin S, Hagiwara A, Nakase Y, et al. Regeneration of skeletal
muscle using in situ tissue engineering on an acellular collagen
sponge scaffold in a rabbit model. ASAIO J. 2007;53(4):506–13.
27. Koulouris G, Connell DA, Brukner P, Schneider-Kolsky M. Mag-
netic resonance imaging parameters for assessing risk of recurrent
hamstring injuries in elite athletes. Am J Sports Med. 2007;35(9):
1500–6.
28. Kroehne V, Heschel I, Schugner F, Lasrich D, Bartsch JW,
Jokusch H. Use of a novel collagen matrix with oriented pore
structure for muscle cell differentiation in cell culture and in
grafts. J Cell Mol Med. 2008;12(5A):1640–8.
29. Mauro A. Satellite cell of skeletal muscle fibers. J Cell Biol.
1961;9(2):493–5.
30. Menetrey J, Kasemkijwattana C, Fu FH, Moreland MS, Huard J.
Suturing versus immobilization of a muscle laceration: a morpho-
logical and functional study in a mouse model. Am J Sports Med.
1999;27(2):222–9.
31. Montarras D, Morgan JE, Collins CA, et al. Direct isolation
of satellite cells for skeletal muscle regeneration. Science.
2005;309(5743):2064–7.
32. Muir AR, Kanji AHM, Allbrook D. The structure of the satellite
cells in skeletal muscle. J Anat. 1965;99( pt 3):435–44.
33. Nikolaou PK, McDonald BL, Glisson RR, Seaber AV, Garrett
WE Jr. Biomechanical and histological evaluation of muscle after
controlled strain injury. Am J Sports Med. 1987;15(1):9–14.
34. Orchard J, Best TM. The management of muscle strain injuries: an
early return versus the risk of recurrence. Clin J Sport Med.
2002;12(1):3–5.
35. Orchard J, Best TM, Verrall GM. Return to play following muscle
strains. Clin J Sport Med. 2005;15(6):436–41.
36. Sato K, Li Y, Foster W, et al. Improvement of muscle healing
through enhancement of muscle regeneration and prevention of
fibrosis. Muscle Nerve. 2003;28(3):365–72.
37. Seale P, Rudnicki MA. A new look at the origin, function, and
‘stem-cell’ status of muscle satellite cells. Dev Biol. 2000;218(2):
115–24.
38. Silder A, Heiderscheit BC, Thelen DG, Enright T, Tuite MJ. MR
observations of long-term musculotendon remodeling following
a hamstring strain injury. Skeletal Radiol. 2008;37(12):1101–9.
39. Stratos I, Rotter R, Eipel C, Mittlmeier T, Vollmar B.
Granulocyte-colony stimulating factor enhances muscle prolifer-
ation and strength following skeletal muscle injury in rats. J Appl
Physiol. 2007;103(5):1857–63.
40. Temple HT, Kuklo TR, Sweet DE, Gibbons CL, Murphey MD.
Rectus femoris muscle tear appearing as a pseudotumor. Am J
Sports Med. 1998;26(4):544–8.
41. van Wachem PB, Brouwer LA, van Luyn MJ. Absence of muscle
regeneration after implantation of a collagen matrix seeded with
myoblasts. Biomaterials. 1999;20(5):419–26.
42. Voytik SL, Przyborski M, Badylak SF, Konieczny SF. Differen-
tial expression of muscle regulatory factor genes in normal and
denervated adult rat hindlimb muscles. Dev Dyn. 1993;198(3):
214–24.
43. Wissink MJB, Beernink R, Pieper JS, et al. Immobilization of
heparin to EDC/NHS-crosslinked collagen. Characterization and
in vitro evaluation. Biomaterials. 2001;22(2):151–63.
EFFECT OF SCARRING DURING MUSCLE HEALING Medicine & Science in Sports & Exercise
d
819
SPECIAL COMMUNICATIONS
... n Immunohistochemistry Sections were deparaffinated, rehydrated, treated with 3% H 2 O 2 for 20 min to inactivate endogenous peroxidase and postfixed with 4% formaldehyde in PBS. The sections were incubated with mouse anti-a-smooth muscle actin (a-SMA, 1:1600; Sigma), mouse anti-ED1 (CD68, 1:100; Serotec, DPC, Breda, The Netherlands) and mouse anti-MyoD (1:25; DAKO, Dakopatts, Glostrup, Denmark) overnight at 4°C as described previously [36]. Paraffin sections were also incubated with mouse anti-Pax7 (1:100; Developmental Studies Hybridoma Bank, Iowa City, CA, USA), mouse anticollagen type I (1:1000; Sigma), rabbit anticollagen type III (1:1600; Chemicon International, Temecula, USA) and mouse anti-Hsp47 (1:24000). ...
... It is possible that macrophages are attracted by the collagen scaffold itself, and induce the increase of Pax7 + cells, MyoD + cells and myotubes in the regenerative zone. However, in a previous laceration wound model, we implanted an empty crosslinked collagen scaffold, which induced a similar inflammatory response and influx of macrophages [36]. This did not increase the number of Pax7 + cells ( Figure 6). ...
... This model can be used to study therapeutic modalities to improve muscle regeneration. Implantation of an SDF-1aloaded collagen scaffold into the defect increases the number of Pax7 + satellite cells, and MyoD + myoblasts and myofibers in the regenerative zone Adapted with permission from [36]. ...
Article
AIM: To develop a model for muscle fibrosis based on full-thickness muscle defects, and to evaluate the effects of implanted stromal-derived factor (SDF)-1α-loaded collagen scaffolds. METHODS: Full-thickness defects 2 mm in diameter were made in the musculus soleus of 48 rats and either left alone or filled with SDF-1α-loaded collagen scaffolds. At 3, 10, 28 and 56 days postsurgery, muscles were analyzed for collagen deposition, satellite cells, myofibroblasts and macrophages. RESULTS: A significant amount of collagen-rich fibrotic tissue was formed, which persisted over time. Increased numbers of satellite cells were present around, but not within, the wounds. Satellite cells were further upregulated in regenerating tissue when SDF-1α-loaded collagen scaffolds were implanted. The scaffolds also attracted macrophages, but collagen deposition and myofibroblast numbers were not affected. CONCLUSION: Persistent muscle fibrosis is induced by full-thickness defects 2 mm in diameter. SDF-1α-loaded collagen scaffolds accelerated muscle regeneration around the wounds, but did not reduce muscle fibrosis.
... Moreover, during regenerative processes of the muscle tissue, MuSCs are supported by other cell types 10,11 . However, despite their remarkable regenerative potential, MuSCs fail to restore tissue structure and function when faced with the extensive damage associated with VML lesions 10,12 . Importantly, recent findings showed that acellular muscle architecture is indeed required to properly guide MuSCs during muscle regeneration 13 . ...
... VML is an injury of the skeletal muscle caused by major trauma, such as battle wounds or tumour excision 1,2 . Endogenous MuSCs, despite their remarkable regenerative capacity, are unable to restore the muscle structure and function compromised by VML injuries 12,40 . Current treatments are limited to scar tissue debridement and autologous tissue transfer at the site of tissue defect 4,6 . ...
Article
Full-text available
Volumetric muscle loss (VML) is associated with loss of skeletal muscle function, and current treatments show limited efficacy. Here we show that bioconstructs suffused with genetically-labelled muscle stem cells (MuSCs) and other muscle resident cells (MRCs) are effective to treat VML injuries in mice. Imaging of bioconstructs implanted in damaged muscles indicates MuSCs survival and growth, and ex vivo analyses show force restoration of treated muscles. Histological analysis highlights myofibre formation, neovascularisation, but insufficient innervation. Both innervation and in vivo force production are enhanced when implantation of bioconstructs is followed by an exercise regimen. Significant improvements are also observed when bioconstructs are used to treat chronic VML injury models. Finally, we demonstrate that bioconstructs made with human MuSCs and MRCs can generate functional muscle tissue in our VML model. These data suggest that stem cell-based therapies aimed to engineer tissue in vivo may be effective to treat acute and chronic VML.
... The prolonged presence of myofibroblasts in the wound leads to fibrosis [29,49]. Since activated SatCs do not migrate into fibrotic tissue, this may impair muscle regeneration and functional recovery of the muscle tissue after injury [51]. ...
... In our study, Pax7-, MyoD-, and MyoG-positive cells were found at the wound edges, demonstrating the presence of activated SatCs and differentiating myofibers. In limb muscles, activated SatCs and regenerating myofibers also appear after about one week [48,51,55,56]. In limb muscles, SatCs proliferate extensively within the first 2-3 days after injury [57]. ...
... PLOS Complete muscle recovery after a serious injury is usually affected by the establishment of a fibrotic scar tissue at the site of injury. Sportsmen who demand highly efficient muscle functioning can suffer with even small fibrosis, and recurrent injuries can occur at the same site during physical exercises [4,5]. Fibrosis is part of the final step of muscle recovery and is characterized by intense synthesis of extracellular matrix (ECM) proteins by myofibroblasts, which results in the formation of connective tissue scar [6]. ...
Article
Full-text available
Regeneration of injured skeletal muscles is affected by fibrosis, which can be improved by the administration of angiotensin II (AngII) receptor (ATR) blockers in normotensive animals. However, the role of ATR in skeletal muscle fibrosis in hypertensive organisms has not been investigated yet. The tibialis anterior (TA) muscle of spontaneously hypertensive (SHR) and Wistar rats (WR) were lacerated and a lentivector encoding a microRNA targeting AngII receptor type 1 (At1) (Lv-mirAT1a) or control (Lv-mirCTL) was injected. The TA muscles were collected after 30 days to evaluate fibrosis by histology and gene expression by real-time quantitative PCR (RT-qPCR) and Western blot. SHR’s myoblasts were analyzed by RT-qPCR, 48 h after transduction. In the SHR’s TA, AT1 protein expression was 23.5-fold higher than in WR without injury, but no difference was observed in the angiotensin II receptor type 2 (AT2) protein expression. TA laceration followed by suture (LS) produced fibrosis in the SHR (23.3±8.5%) and WR (7.9±1.5%). Lv-mirAT1 treatment decreased At1 gene expression in 50% and reduced fibrosis to 7% 30 days after. RT-qPCR showed that reduction in At1 expression is due to downregulation of the At1a but not of the At1b. RT-qPCR of myoblasts from SHR transduced with Lv-mirAT1a showed downregulation of the Tgf-b1, Tgf-b2, Smad3, Col1a1, and Col3a1 genes by mirAT1a. In vivo and in vitro studies indicate that hypertension overproduces skeletal muscle fibrosis, and AngII-AT1a signaling is the main pathway of fibrosis in SHR. Moreover, muscle fibrosis can be treated specifically by in loco injection of Lv-mirAT1a without affecting other organs.
... The recovery of injured muscle tissue follows a timeline that lasts for three to four weeks and the muscle repair consists of molecular and cellular events 4 responsible for the complete functional recovery through inter-related phases such as degeneration/inflammation, repair and remodeling. Frequently, fibrosis is one of the scar end product that causes functional incapacity, pain and possible lesion recurrence 5 . ...
Article
Full-text available
Implement a laceration protocol of the rat lateral gastrocnemius (LG) and following-up its repair with ultrasound biomicroscopy (UBM), contractility tests and histology. Sixty-three male Wistar rats were distributed into two groups. One, with sub-groups GI, GII and GIII (n=12), each containing right LG lacerated (n=6), control and sham (n=3) animals. LG muscles in GI, GII and GIII were inspected by UBM (40 MHz) immediately after, 14 and 28 days post-surgery and thereafter excised with four (GI), 14 (GII) and 28 (GIII) days post-surgery for histology. Animals in second group were distributed into right LG lacerated and control sub-groups. LG muscles in lacerated sub-group were submitted to contractility tests at four (n=8), 14 (n=8) and 28 (n=8) days post-surgery, while in the control sub-group (n=5) were submitted to contractility tests along the course of the experiments. Descriptive findings agreed between the lesion model, muscle repair, UBM images and histology. Contractility results for right LG were different (p<0.05) between control and injured muscle with four and 14 days post-surgery, at tetanic stimulating frequencies (50 and 70 Hz). A laceration protocol of the lateral gastrocnemius was implemented and ultrasound biomicroscopy, contractility and histology findings agreed regarding the following-up of injured muscle repair.
... (30) In a laboratory study, the lack of activated myogenic satellite cells within the fibrotic discontinuity area (scar tissue) was suggested to be the phenomenon responsible for the delay in healing of recurrent muscle injuries. (31) Female athletes with muscle injuries in the present study took a longer time (more than six weeks) to recover compared to male athletes. While the reason for this is unclear, it could be due to the difference in the circulating sex hormones between males and females. ...
Article
Full-text available
The purpose of this study was to investigate the pattern of muscle injuries and the factors that predict the return-to-play duration among Malaysian athletes. This is a retrospective review of the case notes of athletes who attended the National Sports Institute Clinic in Malaysia. The medical records of athletes with muscle injury, diagnosed on clinical assessment and confirmed by diagnostic ultrasonography, were included for final analysis. From June 2006 to December 2009, 397 cases of muscle injury were diagnosed among 360 athletes. The median age of the athletes with muscle injuries was 20.0 years. Muscle injuries were mostly diagnosed among national-level athletes and frequently involved the lower limb, specifically the hamstring muscle group. Nearly all of the athletes (99.2%) were treated conservatively. The median return-to-play duration was 7.4 weeks. Athletes who waited more than one week before seeking medical attention, those with recurrent muscle injuries and female athletes were significantly more likely (p < 0.05) to take more than six weeks before returning to the sport. Grade 2 lower limb muscle injury was commonly diagnosed among national-level athletes in this study. The frequency of weekly physiotherapy sessions did not affect the return-to-play duration. Factors such as initial consultation at more than one week post injury, recurrent muscle injuries and female gender were significant predictors of return-to-play duration among Malaysian athletes. These predictive factors should be kept in mind during clinical assessment so as to aid in prognosticating recovery after muscle injury.
Thesis
Full-text available
Purpose: Repetitive strain injury (RSI) is accompanied by an increase in extracellular matrix (ECM), atrophy, and reduces the muscle power particularly in the elderly. Resistance training (RT) have potential positive effects on muscle function and morphology in elderly. Firstly, this study investigates changes in muscle tissues resulting from performance of a 6-weeks chronic eccentric contraction in aged rats. Using this model, the effects of eight-week therapeutic resistance training on recovery of pathological changes after chronic strain injury were examined. contraction. Methodology: in an experimental design, 48 elderly male Wistar rats were divided into six groups randomly. Three RSI groups underwent 6weeks (5 set of 10 repetitions, 5 days/week) of fast velocity submaximal eccentric contractions (5 sec trains of 0.2 msec pulses, voltage 40 V at 70 Hz), while the three control (CTL) groups were inactive. After 14 days, one of the RSI (RSI-1) and one of the control (CTL-1) were sacrificed for initial assessment of RSI-induced adaptations. Both RSI-RT and CTL-RT groups performed 8 weeks progressive resistance training (1 set of 6 repetitions using 50-100% 1RM, 3 days/week) and the other two groups (RSI-Re & CTL-2) were inactive without any modalities. Gastrocnemius muscle response was assessed by isometric force(IF) and muscle wet mass. Quantitative histopathological analysis and immunoblotting of myogenin protein were also done in all groups. Results: Raw and relative (percent to body weight) measures of isometric force and wet muscle mass of gastrocnemius in CTL-1 group are significantly greater than RSI-1 group. Masson Trichrome and Hematoxylin & eosin (H&E) stains also showed histopathologic changes were present in RSI-1 group that included increase in fibrosis and non-contractile area, and decrease of myofiber area (MA). After 10 weeks of injury protocol, fibrosis and decrease in MA and IF of gastrocnemius were remained in RSI-Re group, but muscle wet mass was recovered. RT significantly improved IF and MA in both training groups, but non-contractile area was not changed. Only in RSI-RT, the protein level of myogenin was greater than control group. Conclusion: These results suggest that in aged rat force deficit and histopathological changes of gastrocnemius muscle after chronic strain injury were reminded after 10 weeks. Therapeutic Resistance training with an emphasis on concentric phase, low velocity and adequate rest can attenuate functional and histopathological changes in muscle after chronic strain injury in elderly rats.
Article
Full-text available
Background and purpose: Repetitive strain injury (RSI) is accompanied by an increase in extracellular matrix (ECM), atrophy, and reduces the muscle power. Resistance training (RT) have potential positive effects on muscle function and morphology in elderly. This research examined the effects of eight week therapeutic resistance training on recovery of pathological changes after 6weeks chronic eccentric contraction. Materials and methods: Eighteen elderly male Wistar rats were divided into three groups (control, RSI-rest, RSI-RT). Two experimental groups underwent 6weeks (5 days/week) of fast velocity submaximal eccentric contractions while the control group was inactive. After two weeks rest, RSI-RT group performed 8 weeks resistance training and RSI-rest group was detrained for 8 weeks. Gastrocnemius muscle response was assessed by isometric force and muscle wet mass. Quantitative histopathological analysis was also done in all groups. Results: Gastrocnemius tissue of injured limbs showed increase in ECM and decrease in myofiber area and isometric force after 6 weeks RSI model in RSI-rest group, but muscle wet mass did not change. RT significantly improved isometric force, myofiber area percent but decreased non-contractile area percentage. However, pathological changes after RSI were not fully recovered by RT. Conclusion: Therapeutic Resistance training with an emphasis on concentric phase, low velocity and adequate rest can attenuate functional and histopathological changes in muscle after chronic strain injury in elderly rats.
Article
Full-text available
To report the expression of decorin and TGF-β in partial myotomy of the extraocular muscle in rats.
Article
Full-text available
Muscle satellite cells contribute to muscle regeneration. We have used a Pax3(GFP/+) mouse line to directly isolate (Pax3)(green fluorescent protein)-expressing muscle satellite cells, by flow cytometry from adult skeletal muscles, as a homogeneous population of small, nongranular, Pax7+, CD34+, CD45-, Sca1- cells. The flow cytometry parameters thus established enabled us to isolate satellite cells from wild-type muscles. Such cells, grafted into muscles of mdx nu/nu mice, contributed both to fiber repair and to the muscle satellite cell compartment. Expansion of these cells in culture before engraftment reduced their regenerative capacity.
Article
Full-text available
The whole-crush injured rat skeletal muscle was used as a model to explore the regenerating potentialities of fast and slow muscles. Laminin was chosen to follow changes in basal lamina and desmin to visualize new muscular elements; they were revealed by immunofluorescence on cryostat sections of either fast (extensor digitorum longus) or slow (soleus) regenerating muscle. Soleus myolysis was rapid, extensive and heterogeneous. Basal laminae were nearly destroyed. In contrast, extensor digitorum longus maintained its basal lamina framework during myolysis. Soleus reconstruction began early, following the pattern of remaining basal laminae as closely as possible, but regeneration stagnated from day 16 and the regenerated muscle was fibrotic. In extensor digitorum longus, reconstruction progressed slower than in soleus, but regularly from the periphery toward the centre of the muscle. The regenerated extensor digitorum longus showed a quasi-normal structure from day 16. At the end of the process, the elimination of old basal lamina was completed in extensor digitorum longus, but was not achieved in soleus. We propose that the old basal lamina should help the initiation of reconstruction. This new model also underlines the importance of the turnover of basal laminae in muscular regeneration, and will be useful to understand the background of the different regenerative response of both muscles.
Article
Full-text available
The objective of this study was to use magnetic resonance (MR) imaging to investigate long-term changes in muscle and tendon morphology following a hamstring strain injury. MR images were obtained from 14 athletes who sustained a clinically diagnosed grade I-II hamstring strain injury between 5 and 23 months prior as well as five healthy controls. Qualitative bilateral comparisons were used to assess the presence of fatty infiltration and changes in morphology that may have arisen as a result of the previous injury. Hamstring muscle and tendon-scar volumes were quantified in both limbs for the biceps femoris long head (BFLH), biceps femoris short head (BFSH), the proximal semimembranosus tendon, and the proximal conjoint biceps femoris and semitendinosus tendon. Differences in muscle and tendon volume between limbs were statistically compared between the previously injured and healthy control subjects. Increased low-intensity signal was present along the musculotendon junction adjacent to the site of presumed prior injury for 11 of the 14 subjects, suggestive of persistent scar tissue. The 13 subjects with biceps femoris injuries displayed a significant decrease in BFLH volume (p < 0.01), often accompanied by an increase in BFSH volume. Two of these subjects also presented with fatty infiltration within the previously injured BFLH. The results of this study provide evidence of long-term musculotendon remodeling following a hamstring strain injury. Additionally, many athletes are likely returning to sport with residual atrophy of the BFLH and/or hypertrophy of the BFSH. It is possible that long-term changes in musculotendon structure following injury alters contraction mechanics during functional movement, such as running and may contribute to reinjury risk.
Article
Myoblast transplantation has been proposed as a potential therapy for Duchenne muscular dystrophy (DMD). A Y-chromosome-specific probe was used to track the fate of donor male myoblasts injected into dystrophic muscles of female mdx mice (which are an animal model for DMD). In situ analysis with the Y-probe showed extremely poor survival of isolated normal male (C57B1/10Sn) donor myoblasts after injection into injured or uninjured muscles of dystrophic (mdx) and normal (C57B1/10Sn) female host mice. A decrease in the numbers of donor (male) myoblasts was seen from 2 days and was marked by 7 days after injection: few or no donor myoblasts were detected in host muscles examined at 3–12 months. There was limited movement of the injected donor myoblasts and fusion into host myofibers was rare. The results of this study strongly suggest that the failure of clinical trials of myoblast transplantation therapy in boys with DMD may have been due to rapid and massive death of the donor myoblasts soon after myoblast injection. © 1996 John Wiley & Sons, Inc.
Article
Myoblast transplantation has been proposed as a potential therapy for Duchenne muscular dystrophy (DMD). A Y-chromosome-specific probe was used to track the fate of donor male myoblasts injected into dystrophic muscles of female mdx mice (which are an animal model for DMD). In situ analysis with the Y-probe showed extremely poor survival of isolated normal male (C57B1/10Sn) donor myoblasts after injection into injured or uninjured muscles of dystrophic (mdx) and normal (C57B1/10Sn) female host mice. A decrease in the numbers of donor (male) myoblasts was seen from 2 days and was marked by 7 days after injection: few or no donor myoblasts were detected in host muscles examined at 3–12 months. There was limited movement of the injected donor myoblasts and fusion into host myofibers was rare. The results of this study strongly suggest that the failure of clinical trials of myoblast transplantation therapy in boys with DMD may have been due to rapid and massive death of the donor myoblasts soon after myoblast injection. © 1996 John Wiley & Sons, Inc.
Article
Satellite cells are situated beneath the basal lamina that surrounds each myofiber and function as myogenic precursors for muscle growth and repair. The source of satellite cell renewal is controversial and has been suggested to be a separate circulating or interstitial stem cell population. Here, we transplant single intact myofibers into radiation-ablated muscles and demonstrate that satellite cells are self-sufficient as a source of regeneration. As few as seven satellite cells associated with one transplanted myofiber can generate over 100 new myofibers containing thousands of myonuclei. Moreover, the transplanted satellite cells vigorously self-renew, expanding in number and repopulating the host muscle with new satellite cells. Following experimental injury, these cells proliferate extensively and regenerate large compact clusters of myofibers. Thus, within a normally stable tissue, the satellite cell exhibits archetypal stem cell properties and is competent to form the basal origin of adult muscle regeneration.
Article
Magnetic resonance (MR) imaging has established its usefulness in diagnosing hamstring muscle strain and identifying features correlating with the duration of rehabilitation in athletes; however, data are currently lacking that may predict which imaging parameters may be predictive of a repeat strain. This study was conducted to identify whether any MR imaging-identifiable parameters are predictive of athletes at risk of sustaining a recurrent hamstring strain in the same playing season. Cohort study; Level of evidence, 3. Forty-one players of the Australian Football League who sustained a hamstring injury underwent MR examination within 3 days of injury between February and August 2002. The imaging parameters measured were the length of injury, cross-sectional area, the specific muscle involved, and the location of the injury within the muscle-tendon unit. Players who suffered a repeat injury during the same season were reimaged, and baseline and repeat injury measurements were compared. Comparison was also made between this group and those who sustained a single strain. Forty-one players sustained hamstring strains that were positive on MR imaging, with 31 injured once and 10 suffering a second injury. The mean length of hamstring muscle injury for the isolated group was 83.4 mm, compared with 98.7 mm for the reinjury group (P = .35). In the reinjury group, the second strain was also of greater length than the original (mean, 107.5 mm; P = .07). Ninety percent of players sustaining a repeat injury demonstrated an injury length greater than 60 mm, compared with only 58% in the single strain group (P = .01). Only 7% of players (1 of 14) with a strain <60 mm suffered a repeat injury. Of the 27 players sustaining a hamstring strain >60 mm, 33% (9 of 27) suffered a repeat injury. Of all the parameters assessed, only a history of anterior cruciate ligament sprain was a statistically significant predictor for suffering a second strain during the same season of competition. A history of anterior cruciate ligament injury was the only statistically significant risk factor for a recurrent hamstring strain in our study. Of the imaging parameters, the MR length of a strain had the strongest correlation association with a repeat hamstring strain and therefore may assist in identifying which athletes are more likely to suffer further reinjury.