Вы находитесь на странице: 1из 13

Scand J Med Sci Sports 2008: 18: 3–15 Copyright & 2007 The Authors

Printed in Singapore . All rights reserved Journal compilation & 2007 Blackwell Munksgaard
DOI: 10.1111/j.1600-0838.2007.00746.x

Review

Chronic tendinopathy tissue pathology, pain mechanisms, and


etiology with a special focus on inflammation
U. Fredberg1, K. Stengaard-Pedersen2
1
Department of Medicine, Region Hospital Silkeborg, Silkeborg, Denmark, 2Department of Rheumatology, University Hospital of
Aarhus, Aarhus, Denmark
Corresponding author: U. Fredberg, Department of Medicine, Region Hospital Silkeborg, DK-8600 Silkeborg, Denmark.
E-mail: fredberg@sportnetdoc.dk
Accepted for publication 24 September 2007

Continuing progress in research in molecular biology and different growth factors, and neuropetides). Because of the
biomechanics has provided considerable new information complex interaction between the classic proinflammatory
and has given rise to new hypotheses in chronic tendino- agents and the neuropeptides, it seems impossible and
pathy. Overloading is still, however, crucial in the develop- somewhat irrelevant to distinguish sharply between chemi-
ment of tendinopathy. Most of the histologic findings in cal and neurogenic inflammation. Furthermore, glucocorti-
tendinopathy represent chronic degeneration, regeneration, coids are, at the moment, the most effective treatment in
and microtears of the tendinous tissue. The prevailing tendinopathy with regard to reduction of pain, tendon
opinion is that no histological evidence of acute inflamma- thickness, and neovascularization. This review indicates –
tion has been documented, but in newer studies using despite a great deal of uncertainty regarding the concepts –
immunohistochemistry and flow cytometry inflammatory that an inflammatory process may be related not only to the
cells have been detected. The existing data indicate that development of tendinopathy but also chronic tendinopathy.
the initiators of the tendinopathic pathway include many More attention should be directed towards the ‘‘tendinitis
proinflammatory agents (e.g. cytokines, prostaglandins, myth’’ in the future.

Chronic tendon pain in Achilles and patella tendons is reduction in pain and tendon thickening measured by
very common. In the general population, the lifetime ultrasonography (US) and an increased pain detec-
cumulative incidence of Achilles tendinopathy is 5.9% tion threshold measured by pressure algometry were
among sedentary people and 50% among elite endur- found only 1 week after administration of ultra-
ance athletes (Kujala et al., 2005), and the overall sound-guided peritendinous corticosteroid injections
prevalence of patellar tendinopathy in an athletic po- in chronic Achilles and patella tendinopathy. These
pulation has been reported to be in the range of 7–40% changes induced by corticosteroids are difficult to
(Kujala et al., 1986; Lian et al., 2005). Despite the explain if the process is degenerative. The time frame
frequency, there are still many unsolved questions and is too short to expect that the corticosteroids could
differences of opinion concerning pathology, pain me- have influenced processes normally connected with
chanisms, etiology, and even terminology. degeneration of connective tissue, such as collagen
A few years ago, the pain in chronic tendon overuse synthesis, fibroblast migration, etc., which are pro-
was believed to be due to a chronic inflammatory cesses that normally change slowly. Although the
process, but because no inflammatory cells could be effect of the injected corticosteroid might be chemical
demonstrated in ruptured tendons, the opinion chan- or mediated through vasoconstriction and thereby
ged from inflammation (‘‘tendinitis’’) to degeneration hypoxia of the accompanying nerves, the dramatic
(‘‘tendinosis’’). A large amount of scientific data have effect could obviously be explained by the anti-
so far not shown any direct evidence of inflammation inflammatory effect of the corticosteroid.
in chronic tendinopathy (Jozsa et al., 1990; Kannus
& Józsa, 1991; Astrom & Rausing, 1995; Movin et al.,
1997b; Alfredson et al., 1999, 2003a; Khan et al.,
Terminology
1999; Alfredson & Lorentzon, 2002). Today, most
authors have even abandoned the ‘‘tendinitis myth’’ The ‘‘peritendon’’ is the loose tissue surrounding the
(Khan et al., 1999, 2002; Alfredson, 2004). In a recent tendon, and it consists of the ‘‘epitenon’’ and the
study (Fredberg et al., 2004), however, a significant ‘‘paratenon’’ (Kirkendall & Garret, 1997) (see Fig. 1).

3
Fredberg & Stengaard-Pedersen

Fig. 1. Structure and model of a tendon (after Kirkendall & Garret, 1997).

In tendons without a synovial sheath, the epitenon biopsy showing degeneration or inflammation. If
is tightly bound to the tendon. Generally, ‘‘tendinitis’’ symptoms are present for more than 3 months, the
(or ‘‘tendonitis’’) is primarily used as a histopatholo- tendinopathy is categorized as ‘‘chronic,’’ for symp-
gic term describing a condition in which the primary toms present between 6 and 12 weeks as ‘‘subacute,’’
site of involvement is the tendon and with an inflam- and for symptoms present between 0 and 6 weeks as
matory response being seen within the tendon ‘‘acute.’’
(Järvinen et al., 1997; Sharma & Maffulli, 2006). The
condition is often associated with reactive ‘‘parateno-
nitis’’ or ‘‘peritendinitis,’’ which is an inflammation of Diagnosis
the paratenon (Järvinen et al., 1997). ‘‘Tendinosis’’ is
not correlated with clinical symptoms (Peers & Ly- Tendinopathy is characterized by the gradual onset
sens, 2005), but it has been widely used for patients of morning stiffness in the tendon, decreased func-
with chronic tendon pain, and with biopsy, radio- tion, localized swelling, and sometimes neovascular-
graphic, ultrasonographic, or magnetic resonance ization (Khan et al., 1999; Boesen et al., 2006). Fibrin
imaging (MRI) showing tendon abnormalities precipitated from the fibrinogen-rich fluid around
(Khan et al., 1999; Alfredson, 2003). Today, ‘‘tendi- the tendon can result in palpable crepitation (Jòzsa &
nosis’’ is primarily used to describe a histopathologic Kannus, 1997).
finding with intratendinous degeneration and no The diagnosis can be made clinically and is verifi-
sign of inflammation (Järvinen et al., 1997; Sharma & able by US or MRI. The diagnosis tendinopathy is
Maffulli, 2006). ‘‘Tendinopathy’’ is used to signify the mainly based on patient complaints (sensation of
combination of tendon pain and impaired perfor- pain) and palpation of the tendon, its surrounding
mance often associated with swelling of the tendon tissue, and its insertion, even though the clinical
and intratendinous changes (Alfredson, 2003, 2005) diagnosis of Achilles (Maffulli et al., 2003) and
evaluated by US or MRI. The diagnosis of tendino- patellar tendinopathy (Cook et al., 2001), even in
pathy can, in contrast to tendinitis and tendinosis, be experienced hands, is not straightforward, and ex-
made clinically without histopathologic examination. perienced examiners may have problems in reprodu-
No specific time criteria are used to classify tendi- cing the results of clinical examination based on
nopathy as acute or chronic. It has been suggested simple tests (Maffulli et al., 2003). Many of the cases
that tendon symptoms present for o2 weeks be were incorrectly diagnosed using only clinical exam-
described as ‘‘acute,’’ for 2–6 weeks as ‘‘subacute,’’ ination (Fredberg et al., 2004), and in some cases
and for more than 6 weeks as ‘‘chronic’’ (el Hawary even total ruptures were misdiagnosed (Ljungqvist,
et al., 1997). These somewhat arbitrary distinctions 1967; Resnick et al., 1977; Shields, 1978; Siwek &
are not based on histopathologic or clinical criteria. Rao, 1981; O’Brian, 1984; Ballas et al., 1998). Based
It is recommended that the term tendinopathy be on these facts, US (or MRI) is recommended in all
used as a clinical diagnosis for patients with pain cases of tendinopathy if shooting pain is present or if
in the tendons. Tendinosis and tendinitis require a there is no positive progress during treatment.

4
Chronic tendinopathy tissue pathology
US is more accurate than MRI in confirming tion of glycosaminoglycans (Movin et al., 1997a), (C)
clinically diagnosed patellar tendinopathy (Warden more prominent and numerous tenocytes without
et al., 2007). US has several significant advantages their normal, fine spindle shape, and with more
over MRI: tissue with few mobile protons emits little rounded nuclei (Colosimo & Bassett, 1990; Fritschy
or no signal, and, therefore, the internal architecture & Wallensten, 1993), and (D) neovascularization as
of the tendon is not well demonstrated with MRI. In seen on color and power Doppler US (Alfredson
contrast, US shows the fine internal structure of et al., 2003b).
tendons, and US therefore pictures the anatomic Histologic examination of specimens removed dur-
border of the tendon more precisely than does MRI ing surgery for tendinopathy shows hypoxic degen-
(Kamel et al., 2004). In agreement with this, the eration (Kannus & Józsa, 1991), mucoid or myxoid
‘‘standard deviation’’ and ‘‘range of the mean differ- degeneration (Jòzsa & Kannus, 1997) and fibrinoid
ence’’ from repeated measurement are less with US necrosis (Jozsa et al., 1990; Khan et al., 1996), fatty
than with MRI (Koivunen-Niemela & Parkkola, degeneration or tendolipomatosis (Kannus & Józsa,
1995). The US examination is interactive. The ex- 1991), collagen degeneration (Cetti et al., 2003),
aminer is with the patient, and any site of reported pseudocyst change (Ferretti et al., 1983), randomized
pain or tenderness can be directly correlated with its collagen with an irregular fiber structure and poor
real-time scan appearance. The ultrasonographer can fiber orientation and neovascularization and teno-
make use of the dynamic real-time character of US, cyte infiltration (Kälebo et al., 1991; Astrom &
so that tendons can be studied throughout their Rausing, 1995; Khan et al., 1999; Ohberg & Alfredson,
range of motion. Side-to-side comparison is always 2002), tenocyte necrosis (Cetti et al., 2003), micro-
available during the US examination. The spatial tears of the tendinous tissue (Cook et al., 1997),
resolution of US is much better than that of MRI chronic inflammatory cell infiltration (Mourad et al.,
(Erickson, 1997). Furthermore, US can demonstrate 1988; Raatikainen et al., 1994), acute inflammation
the neovascularization in tendinopathy. Today, US is (Cetti et al., 2003), granulation tissue (Kälebo et al.,
a well-established first-choice modality and is re- 1991), small foci with iron-positive hemosidero-
garded as the examiner’s extended hand in daily phages (Schubert et al., 2005), focal degeneration
practice, which will never be the case for MRI, and near the bone–tendon insertion (Raatikainen et al.,
MRI has only a limited place in tendinopathy 1994), hyalin degeneration and fibrocartilagionous
(Richards et al., 2001; Shalabi et al., 2007). and bony metaplasia (Myllymaki et al., 1990), calci-
In patients with chronic tendinopathy, US shows fying tendinopathy (Kannus & Józsa, 1991), angiofi-
thickening of the tendon, discontinuity of the fibers, broblastic tendinosis (Yu et al., 1995), grayish
focal hypoechoic intratendinous areas, loss of fasci- discoloration of ground substance, tendon edema,
cle organization, intratendinous focal calcification, and different combinations of these entities (Kannus
partial or complete ruptures, and thickening of the & Józsa, 1991; Jòzsa & Kannus, 1997; Paavola et al.,
hypoechoic paratenon with poorly defined borders, 2002).
bursitis, and adherences between the epitenon and Virtually every study of the pathology of Achilles
paratenon (Gibbon et al., 2000; Blankstein et al., and patellar tendinopathy has reported that there
2001; Fornage, 2003), and the contours of the were more conspicuous and more numerous cells
tendon may be deformed with a bumpy appearance than in healthy tendons and inflammatory cells
(Fornage, 1993). were absent. Most of these histologic findings above
represent
1. chronic degeneration (hypoxic degeneration,
Histologically mucoid or myxoid degeneration, fatty degenera-
In stark contrast to the glistening white normal tion, collagen degeneration, fibrinoid necrosis,
tendon, in symptomatic tendinopathy tendons ap- tenocyte necrosis, pseudocyst change, focal de-
pear gray or yellow-brown and amorphous to the generation, hyalin degeneration),
naked eye, and microscopy reveals discontinuous 2. regeneration (neovascularization or angiofibro-
and disorganized collagen fibers that lack reflectivity blastic tendinosis, tenocyte infiltration, chronic
under polarized light (Karlsson et al., 1992; Raati- and acute inflammation), and
kainen et al., 1994; Khan et al., 1999). 3. microtears of the tendinous tissue (the positive
Compared with normal tendons, the characteristic hemosiderophages).
features of tendinopathy under light microscopy are
(A) disrupted collagen, and the collagen fibers are The prevailing opinion is that no histological
thinner than normal and the characteristic hierarch- evidence of acute inflammation has been documented
ical structure is lost (Åstrøm & Rausing, 1995), (B) in ruptured tendons (Alfredson & Lorentzon, 2002;
increased ground substance with a high concentra- Khan et al., 2002) or tendinopathic tendons under-

5
Fredberg & Stengaard-Pedersen
going surgery (Benazzo et al., 1996; Cook et al., 2005) [which are upregulated by inflammatory
1997) or biopsies (Martinoli et al., 1993). cytokines and highly expressed in Achilles tendino-
In a recent study (Cetti et al., 2003), however, pathy] (Pufe et al., 2001, 2005; Petersen et al., 2002),
immunohistochemical staining confirmed acute in-  increase the expression level of cytosolic phospho-
flammation in all of 60 ruptured Achilles tendons. lipase-A2 and activity level of secretory phospholi-
The neutrophils had a morphology reminiscent of pase-A2 [which are involved in the production of
necrotic tenocytes, and their presence was confirmed PGE2 and other inflammatory mediators] (Wang
on immunohistochemical staining. Using monoclo- et al., 2004), and
nal antibodies (CD3 for detection of T lymphocytes,
 increase activation of stress-activated protein kinase
CD 20 for detection pf B lymphocytes, and CD 68
(Arnoczku et al., 2002) [which is activated from pro-
for detection of macrophages), Schubert et al. (2005)
inflammatory cytokines, indicating that this signal
demonstrated that B and T lymphocytes and macro-
pathway may contribute to the inflammatory re-
phages were increased in Achilles tendinopathy sam-
sponses] (Ip & Davis, 1998).
ples. These two studies need more confirmation.
Areas of altered collagen fiber structure and in- However, many of these investigations have used
creased interfibrillar ground substance, which has non-physiologic strain patterns or the addition of
been shown to consist of hydrophilic glucosamino- external factors to elicit these cell responses. Thus,
glycans, in Achilles tendinopathy correspond to the the clinical relevance of many of the studies must be
increased signal on MRI (Movin et al., 1998a) and called into question.
the hypoechogenic regions on US (Maffulli et al., COX-2 expression is usually low but can be
1987; Movin et al., 1998b). Areas with increased induced by numerous factors, including neurotrans-
signals on MRI (Yu et al., 1995; Khan et al., 1996) mitters, growth factors, pro-inflammatory cytokines,
and granulomas and hypoechogenic regions on US lipopolysaccharides, calcium, phorbol esters, and
(Myllymaki et al., 1990; Maffulli et al., 1992) in small peptide hormones (O’Banion, 1999) and can
patellar tendinopathy appear to correspond to mu- be reduced by glucocorticoids.
coid degeneration (Khan et al., 1996). Tissue injury is associated with inflammation and
increased prostanoid synthesis and pain hypersensi-
tivity. Prostanoids influence inflammation and im-
Biochemically mune responses, and their administration reproduces
the major signs of inflammation, including augmen-
Several cell types in and around the tendon respond
ted pain sensitivity (Tilley et al., 2001). Peripheral
to physical activity and can produce and respond to
inflammation increases prostanoid levels at the site of
many inflammatory mediators.
inflammation, and this local release contributes di-
Endothelial cells can express and respond to a
rectly to inflammation and pain. More recently,
network of inflammatory mediators, such as inter-
peripheral inflammation has also been shown to
leukins, prostaglandins (PGE1, PGE2), and nitric
increase central prostanoid levels (Dirig & Yaksh,
oxide (NO) (Scott et al., 2004).
1999; Vanegas & Schaible, 2001). Constitutive pro-
Tendon cells subjected to cyclic strain increase the
duction of prostanoids is normally low, but can be
production of:
increased within minutes by inflammatory stimuli
 COX-2 (Wang et al., 2003) [which is not expressed acting on constitutively expressed prostanoid syn-
in resting connective tissue, but is induced by inter- thetic enzymes (Funk, 2001). Pro-inflammatory sig-
leukin-1 (IL-1) and tumor necrosis factor (TNF)], nals trigger multiple transcriptional and post-
 PGE2 (Almekinders et al., 1993, 1995; Wang et al., translational changes that alter the synthetic enzyme
2003, 2004; Li et al., 2004), levels and activity, and this leads to early, massive,
and sustained increases in prostanoid levels (Samad
 IL-6 (Skutek et al., 2003),
et al., 2003).
 IL-1 b [which results in increased production of In tendons from patients with patellar tendinopa-
COX-2, matrix metalloproteinases (MMP-1, MMP- thy, both the tendon tissue itself and harvested cells
3, MMP-13, which cause matrix destruction and a express higher levels of COX-2 and PGE2 than do
loss of tendon biomechanical properties), PGE2, healthy control patellar tendons (Fu et al., 2002).
intracellular calcium [which can lead to apoptosis Human microdialysis studies have shown that
(Arnoczku et al., 2002)] and strongly downregulates peritendinous pro-inflammatory agents like PGE2
an apoptose-inhibitor gene, which could contribute (Langberg et al., 1999a, b), IL-6 (Langberg et al.,
to increased cell death] (Archambault et al., 2002; 2002), and thromboxane B2 (Langberg et al., 1999a)
Tsuzaki et al., 2003; Banes et al., 2007), are increased after exercise, indicating that the pro-
 vascular endothelial growth factor (Senger et al., duction is local (Langberg et al., 1999a, b), and
1983; Ferrara, 1999; Neufeld et al., 1999; Pufe et al., that peritendinous pro-inflammatory agents like

6
Chronic tendinopathy tissue pathology
PGE2 are increased 50% in patients with chronic In a recent study (Danielson et al., 2006b), an
tendinopathy compared with normal tendons upregulation of the cholinergic system was found
(Alfredson et al., 1999, 2001b), although the differ- concerning levels of expression of the muscarin
ences were not significant in this very small study of receptors M2 and choline acetyltransferrase in tendi-
only four patients. nopathy, and the tenocytes were suggested to be a
The nociceptive substance P (SP) and calcitonin source of acetylcholinesterase production. It is
gene-related peptide (CGRP) positive nerve fibers known that cytokines can induce upregulation of
are significantly increased in chronic tendinopathy the M2 receptors (Ebriques de Salamanca et al.,
(Forsgren et al., 2005; Schubert et al., 2005). 2005). Thus, non-neural acetylcholinesterase produc-
Using microdialysis techniques, Alfredson et al. tion may have effects on immune function, cell
(1999) found a high level of the excitatory neuro- proliferation and differentiation, and several other
transmitter glutamate in tendons from patients basic cell functions (Wessler et al., 2001).
with Achilles tendinopathy [and the occurrence of In contrast, in a study using cDNA arrays and
glutamate N-methyl-D-aspartate receptors (Alfredson real-time PCR (Alfredson et al., 2003a) on biopsies
et al., 2001a)]. from tendons with tendinopathy, Alfredson found
Neuropeptides have been found to exert trophic that the mRNA for several cytokines and cytokines
effects in different tissues in addition to their noci- receptors was not upregulated in Achilles tendino-
ceptive and pro-inflammatory actions (Strand et al., pathy. Based on the findings in these tendon biopsies,
1991; Schwartz, 1992; Hökfelt et al., 2000). SP and Alfredson concluded that there is no chemical in-
CGRP, representing the sensory system, participate flammation involved in the chronic stage of tendino-
in the regulation of fibroblast and synoviocyte pathy, but there could be a neurogenic inflammation
proliferation and of angiogenesis (Brain et al., involving neuropeptides like SP and CGRP. How-
1985; Haegerstrand et al., 1990). They have also ever, there was a mixture of all cell types in the
been implicated in the synthesis and release of biopsies, and consequently, a theoretically possible,
cytokines and growth factors (Broome & Miyan, isolated upregulation of fibroblast or endothelial
2000; Monneret et al., 2000). SP upregulates COX cells could have been missed.
2 and IL-b in the peritendon (Hart et al., 1998).
Neurogenic inflammation could initiate peritendini-
tis, with both SP and GCRP implicated in this Pain mechanism
pathway (Hart et al., 1998), and long-term periten-
dinitis can lead to degenerative changes in the tendon The pain mechanism is partly unknown. Traditional
(Sullo et al., 2001). theories state that pain arises through inflammation
In nociception, CGRP potentiates the effects of or due to separation of collagen fibers in more severe
SP (Wiesenfeld-Hallina et al., 1984). Galanin, also forms of tendinopathy. Other theories include
occurring in primary afferents, has been shown to biochemical stimulation of the nociceptors due to
mitigate nociception and inflammation (Heppelmann extravasation of glucosaminoglycans, especially
et al., 2000). chondroitin sulfates (Benazzo et al., 1996; Khan
SP and CGRP have a stimulatory role in the et al., 1996; Jòzsa & Kannus, 1997) and other
proliferation of cultured fibroblasts (Nilsson et al., biochemical irritants. In biopsies from athletes with
1985). SP and CGRP are also known to stimulate the patellar tendinopathy, Danielson et al. (2006b) re-
proliferation of endothelial cells (Nilsson et al., 1985; cently found that tenocytes produce acetylcholine and
Haegerstrand et al., 1990; Ziche et al., 1990). that nerve fibers showing immunoreactions for the
Not only is the level of SP increased significantly in acetylcholine-receptor M2 were observed in associa-
chronic tendinopathy, but SP is also increased in the tion with the small blood vessels in tendinopathy.
synovial fluid in a typical inflammatory disease like Prostaglandins, prostacyclins, and thromboxanes
rheumatoid arthritis (RA) (Westermark et al., 2001). (prostanoids) contribute to the development of pain
The synovial fibroblast in RA can produce SP (Inoue by acting both peripherally and centrally. Peripher-
et al., 2001), and neuropeptides have been shown ally, they play a major role in generating peripheral
to modulate immune function directly through ex- sensitization by increasing the sensitivity of the
pressed receptors and undergo distinct alteration in peripheral terminals of high-threshold pain fibers
RA (Sedo et al., 2005). (nociceptors). They increase excitability, reduce the
It is known that the neuroendocrine, immunologic, pain threshold, and potentiate the action of pain-
and microvascular systems interact in RA (Masi et al., producing stimuli, such as heat or irritant molecules
1999; Hernanz et al., 2003; Sedo et al., 2005) thus, it is like bradykinin (Khasar et al., 1998; Gold, 1999).
an obvious conclusion that the same could be seen in Currently, investigations are increasingly focused
tendinopathy. Some of the mechanisms in tendinopa- on the nerve supply to the tendons. Neuropeptide-
thy and inflammatory RA seem to be the same. containing nerve fibers have both afferent and effer-

7
Fredberg & Stengaard-Pedersen
ent roles with respect to bone cell regulation, and a reduction in water content. In biopsies from
they may be involved in the healing of tendons and Achilles tendinopathy tendons and normal tendons,
fractures. The nerve fibers are mainly located in the water content was the highest in the tendinopathy
periosteum, synovium, the fat pad (Witonski & tendons (de Mos et al., 2007). The effect of gluco-
Wagrowska-Danielewicz, 1997), and the loose peri- corticoids could theoretically be due to an analgesic
tendinous connective tissue. However, nerve in- effect on the neuropeptides (CGRP and SP), which,
growth is known to occur as a response to tendon as mentioned above, are increased in tendinopathy,
injury (Ackermann et al., 2002), and a number of but it seems unlikely that this could explain the
studies have demonstrated new nerve ingrowth in the dramatic reduction in tendon thickness.
tendon proper in tendinopathy (Schubert et al., 2005; Glucocorticoids regulate vascular reactivity by
Lian et al., 2006). In tendinopathy, nerve fibers acting on both endothelial and vascular smooth
accompany the blood vessels into the tendon (Da- muscle cells. Glucocorticoid receptor protein and
nielson et al., 2006a). It has been suggested that these mRNA have been identified in endothelial and vas-
nerves are a potential origin of the pain in tendino- cular smooth muscle cells. In endothelial cells, glu-
pathy (Alfredson et al., 2003b). cocorticoids suppress the production of vasodilators,
Free-sprouting SP and CGRP fibers are found such as prostacyclin and NO (Suzuki et al., 2003;
around newly formed blood vessels in ruptured Yang & Zhans, 2004). Glucocorticoids are to some
Achilles tendons, and Ackermann et al. (2003) de- extent vasoconstrictors, which may explain the
monstrated that the healing process in tendon in the change in vascularity, and secondly, the reduction
inflammatory and early proliferation phase of healing in thickness and pain, due to a reduction in the
(weeks 1–2) is associated with new nerve ingrowth supply of different noxious stimuli and pro-inflam-
and a specific temporal pattern of neuropeptide matory agents like PGE, cytokines, and neuropep-
occurrence. The rate of change in peripheral neuro- tides, whose effects will further be reduced by
peptides occurrence is related to nociceptive thres- steroids.
holds, which presumably reflect a regulatory role in Glucocorticoids are the most effective treatment in
both nociception and tissue repair. This was followed tendinopathy with regard to reduction of pain,
by nerve fiber withdrawal (weeks 6–12) from the tendon thickness (Fredberg et al., 2004), and neo-
tendon tissue. It is however, well known that the vascularization. The effects when glucocorticoids are
pain continues even during weeks 6–12, as well as injected around chronic tendinopathies and into
afterwards. The level of the excitatory neurotransmit- inflammatory joints of patients with RA, which is a
ter glutamate and the number of nociceptive SP and well-established inflammatory chronic disease, are
CGRP positive nerve fibers are also known to be nearly the same. Because glucocorticoids do not
significantly increased in chronic tendinopathy (Al- cure either tendinopathy or RA, the symptoms often
fredson et al., 2001a; Forsgren et al., 2005; Schubert relapse in both diseases. The two diseases have many
et al., 2005) in both vessels and nerve fascicles, symptoms in common (rubor, dolor, tumor, calor,
indicating that the peptides not only have an effect functio laesae), and because the clinical responses to
in relation to blood flow regulation but could also glucocorticoids in chronic tendinopathy and inflam-
have effects within the nerve fascicles. matory RA are comparable, a conclusion that
Acetylcholinesterase may have an effect on sensory immediately suggests itself is that the effect of
nerve fibers, and in this way, the acetylcholinesterase glucocorticoids in chronic tendinopathy is due to
in non-neural cells may play a role in modulating their anti-inflammatory properties as in inflamma-
peripheral nociception (Weiss et al., 2003). tory arthritis.
Why glucocorticoids have the same dramatic clin-
ical effect on pain and hyperemia in tendinopathy as
they do in RA (Terslev et al., 2003; Koenig et al., Etiology
2004) is still partly unknown, but many of the same
pro-inflammatory agents are found in both diseases. The exact pathogenesis of chronic tendinopathy
It has been postulated that the dramatic reduction in remains largely unknown but seems to be a multi-
tendon thickness (and maybe the pain) after steroid factorial process. The following are a wide range of
treatment is due to glucocorticoid reducing the water suggested intrinsic and extrinsic etiological factors
content in the tendons. However, in an experimental that are assumed to be the mechanisms of tendino-
animal study (Sullo et al., 2001), the intratendinous pathy (Williams, 1986; Murphy et al., 2003): age
water in PGE1-induced tendinopathy was close to the [with decreased arterial blood flow with local hy-
water content of the normal control tendons. This poxia, less nutrition, impaired metabolism, and free
animal study does not indicate that the considerable radicals (Archambault et al., 1995; Langberg et al.,
reduction in the tendon thickness and pain 1 week 2001; Kettunen et al., 2006)]; vascular perfusion
after steroid injection (Fredberg et al., 2004) is due to [ischemia occurs when a tendon is under maximal

8
Chronic tendinopathy tissue pathology
tensile load and microdialysis studies have demon- ditions also being seen in physically inactive indivi-
strated high intratendinous concentrations of lactate duals (Movin, 1998; Alfredson & Lorentzon, 2000).
in chronic, painful Achilles tendons (Alfredson et al., At the moment, the mechanical strain theory is the
2002) and on relaxion, reperfusion occurs, generating most accepted theory to explain the injury mecha-
oxygen free radicals (Goodship et al., 1994; Bestwick nisms of tendon overload injuries (Stanish et al., 1985;
& Maffulli, 2004)]; nutrition; exercise-induced hy- Archambault et al., 1995; Khan et al., 1999): repeated
perthermia (Arancia et al., 1989; Brich et al., 1997); heavy loading may produce initial pathological
anatomic variants: various alignments such as Q- changes in either the extracellular matrix or the cellular
angle, hyperpronation (Clement et al., 1984; Nigg, components of a tendon. When the load exceeds the
2001), limited range of motion of the ankle joint tendon’s strength (resistance), the progressive damage
(Kvist, 1991), excessive motion of the hindfoot in the (the basal ability of the tissue to repair itself after being
frontal plane (Kaufman et al., 1999), especially a overwhelmed by the repetitive microtraumatic process)
lateral heel strike with compensatory pronation, may lead to the structure of the tendon being disrupted
varus deformity of the forefoot (Clement et al., micro- and macroscopically by this repetitive strain
1984; Kvist, 1991), pes cavus, pes planus (Williams (often eccentric by nature), and collagen fibers begin to
et al., 2001), lateral ankle instability, leg-length dis- slide past one another (causing breakage of their cross-
crepancy (Kannus, 1997), impingement (Johnson linked structure) and denature (with inflammation
et al., 1996; Schmidt et al., 2002), and other biome- edema and pain), causing a focal area of intratendi-
chanical factors (Kvist, 1994); muscle weakness/im- nous degeneration, partial tears, and complete rup-
balance (Wityrouw et al., 2001; Mahieu et al., 2006); tures (Jòzsa & Kannus, 1997; Kannus, 1997). The
increased tightness of the gastrocnemius (Kaufman cumulative trauma is thought to weaken collagen
et al., 1999): physical load (sport/occupation); exces- cross-linking and the non-collagenous matrix and
sive force; repetitive loading; abnormal/unusual vascular elements of the tendon, and finally leads to
movement; poor technique; training errors: fast pro- tendinopathy.
gression and high intensity; fatigue; shoes and equip- It is highly probable that overload exercise plays a
ment; environmental conditions; temperature and decisive role in tendinopathy because the lifetime
running surface (Kvist, 1991); gender (Kannus, cumulative incidence of Achilles tendinopathy is
1997); genetic (Józsa et al., 1989; Kannus & Natri, nearly 10 times higher among elite endurance athletes
1997; Mokone et al., 2005, 2006) and genetically than among sedentary people (Kujala et al., 2005).
determined collagen abnormalities; infectious dis- Moreover, exercise has important modulatory effects
ease; neurological conditions; hyperparathyroidism on immunocyte dynamics and possibly on the im-
(Preston, 1972); hypertension (Holmes & Lin, 2006); mune function. These effects are mediated by diverse
body weight (Holmes & Lin, 2006); increased serum factors, including, among others, exercise-induced
lipid (Qzgurtas et al., 2003); glycogen storage disease release of classical stress hormones, hemodynamic
(Carvès et al., 2003); systemic disease/treatment effects involving cell distribution (Pedersen & Hoff-
[direct injection of corticosteroids (Shrier et al., mann-Goetz, 2000), and release of a soup of pro-
1996; Fredberg, 1997)]; systemic corticosteroid inflammatory mediators, as for example, cytokines
(Newham et al., 1991; Khurana et al., 2002); oral (which can be detected in plasma and urine during
contraceptives (Holmes & Lin, 2006); fluoroquino- and after exercise) (Ostrowski et al., 1999), prosta-
lones (Malaguti et al., 2001; Chhajed et al., 2002); RA glandins, and neuropeptides (Lind et al., 1996; Hasbak
(Peiro et al., 1975); psoriasis (Aydingöz & Aydingöz, et al., 2002; Karahan et al., 2002). The tendon and
2002); systemic lupus erythematosus (Pritchard & endothelial cells seem to be able to produce most of
Berney, 1989; Jakobsen et al., 2000); chronic renal these mediators.
failure (Kricun & Kricun, 1980); hyperuricemia (Hof- Animal studies support both the overload theory
mann et al., 1990); hyperthyroidism; arteriosclerosis; and the notion that cytokines and prostaglandins
and diabetes mellitus (Webb & Bannister, 1999; play a role in the etiology of tendinopathy. Backman
Holmes & Lin, 2006). et al. (1990) demonstrated that exercised rabbits
The scientific background for most of these sug- showed light microscopic degenerative changes in
gestions is lacking, and they must to be characterized tendons and increased numbers of capillaries, infil-
as non-proven theories, and, above all, their clinical trates of inflammatory cells, edema, and fibrosis in
importance is not well known. the paratenon.
The traditional view of tendinopathy is a tendon In animal studies, injections of collagenase, cyto-
injury associated with overuse (Curwin, 1994; Arch- kines, and inflammatory prostaglandins (PGE2),
ambault et al., 1995; Jòzsa & Kannus, 1997) from which, as mentioned above, are increased in exercise,
repetitive mechanical load, microtears, and acute and have been shown to cause tendinitis and tendinosis
then chronic phases of inflammatory ‘‘tendinitis’’ (Stone et al., 1999; Sullo et al., 2001; Cilli et al., 2004;
that lead to tendon degeneration, despite these con- Khan et al., 2005).

9
Fredberg & Stengaard-Pedersen
Load induced tendon abnormalities sic and intrinsic (including genetic) factors, are
predisposed.
Symptoms
It seems plausible that tendons have a baseline
mechanical strength, which depends on the loading
Pain detection threshold history of the tendon (training level). Once a rapid
increase in training load, frequency, or duration
No symptoms occurs, the tendon may not be able to adapt fast
enough to these changes. The mechanical strength of
the tendon may be exceeded, and a small injury may
occur. Under normal circumstances, this small injury
Time will heal as a normal part of tendon remodeling, but
if the training and overloading continues, these small
Fig. 2. The tendinopathic ‘‘iceberg.’’
injuries result in progressive tendon changes that,
after an asymptomatic period of several months,
slowly aggravate and finally reach the pain limit
Some people may have a genetic predisposition and become symptomatic (see Fig. 2).
toward developing tendinopathies (Mokone et al., It seems plausible that tendinopathy begins with
2005, 2006), which may explain why many patients cellular activation and inflammation and proceeds
with tendinopathies do not go in for sport. through phases of increased ground substance, col-
Most Achilles tendon ruptures occur without lagen separations, and eventually neovascularization,
warning symptoms, but in nearly all the ruptured and that the corticosteroid-sensitive mechanisms
tendons, degenerative changes can be demonstrated play a crucial role in this process.
(Kannus & Józsa, 1991), and several studies show Virtually every study of the pathology Achilles and
ultrasonographic abnormalities in patellar tendons patellar tendinopathy has reported that there are
of asymptomatic athletes playing volleyball, basket- more conspicuous and more numerous cells than in
ball, soccer, and track and field athletes (Gibbon healthy tendons and inflammatory cells are absent.
et al., 1999; Cook et al., 2000; Fredberg & Bolvig, Two newer studies, in which immunohistochemical
2002; Major & Helm, 2002; Maffulli et al., 2003). staining and monoclonal antibodies were used for
Even severe tendinopathies are asymptomatic for detection of T and B lymphocytes and macrophages,
long periods. Thus, chronic tendinopathy can be confirmed the presence of inflammation in both
compared with an iceberg, pain being the tip of the ruptured and non-ruptured chronic Achilles tendi-
iceberg (see Fig. 2). nopathies.
This ‘‘iceberg theory’’ can explain the frequent The existing data indicate that the initiators of the
relapse of symptoms when athletes resume the sport tendinopathic pathway include traumatic events, or a
activity after too short a rehabilitation period, during prolonged repetitive motion injury induces the pro-
which pain recedes to just below the detection thresh- duction of many pro-inflammatory agents (including
old while most of the intratendinous abnormalities in cytokines such as IL-1-b, prostaglandins such as
the tendon still exist. PGE2, NO, different growth factors, and neurope-
A study (Fredberg & Bolvig, 2002) showed that tides). The pro-inflammatory mediators induce apop-
US can identify these asymptomatic athletes who tosis, elaboration of pain mediators, and MMP,
have an increased risk of developing serious tendon which degrade collagens and proteoglycans. The
injuries in the future. end result is a weak tendon with an increased risk
of ruptures. The tendon cells can produce these
agents when subjected to cyclic stress, and in animal
studies these inflammatory agents can be used
Conclusion to produce experimental chronic tendinopathy.
Furthermore, many of the pro-inflammatory media-
Even in experienced hands, the diagnosis of tendino- tors and neuropeptides are also found in chronic
pathy is not straightforward, and experienced exam- tendinopathy. Because of the complex interaction
iners may have problems in reproducing the results between the pro-inflammatory agents and the neuro-
of clinical examination based on simple tests. There- peptides, it seems impossible and partly irrelevant to
fore, the diagnosis should be verified by US, which is distinguish sharply between chemical and neurogenic
a more accurate modality than MRI in confirming inflammation.
clinically diagnosed tendinopathy (Warden et al., This review indicates – without definitive proof –
2007). that an inflammatory process may be related to the
Overuse is crucial in the development of tendino- development of tendinopathy and that the inflamma-
pathy in individuals who, perhaps because of extrin- tion may also play a role in chronic tendinopathy.

10
Chronic tendinopathy tissue pathology
The major questions for the future are therefore: More attention should be directed toward the
is it advantageous to block this inflammatory tendinitis myth in the future.
cascade, and what is the most effective way to
block it with the smallest possible number of side Key words: tendonitis, tendinosis, tendinitis, tendino-
effects? pathy, Achilles tendon.

References
Ackermann PW, Ahmed M, Kreicberg A. Alfredson H, Ohberg L, Forsgren S. Is in a patient with psoriasis. Clin
Early nerve regeneration after Achilles vasculo-neural ingrowth the cause of Imaging 2002: 26: 209–211.
tendon rupture: a prerequisite for pain in chronic Achilles tendinosis? An Backman C, Boquist L, Friden J,
healing? A study in the rat. J Orthop investigation using ultrasonography Lorentzon R, Toolanen G. Chronic
Res 2002: 20: 849–856. and colour Doppler, immuno- achilles paratenonitis with tendinosis:
Ackermann PW, Li J, Lundeberg T, histochemistry, and diagnostic an experimental model in the rabbit.
Kreicberg A. Neuronal plasticity in injections. Knee Surg Sports J Orthop Res 1990: 8: 541–547.
relation to nociception and healing of Traumatol Arthrosc 2003b: 11: Ballas MT, Tytko J, Mannarino F.
rat achilles tendon. J Orthop Res 2003: 334–338. Commonly missed orthopedic problems.
21: 432–441. Alfredson H, Thorsen K, Lorentzon R. In Am Fam Physican 1998: 57: 267–274.
Alfredson H. Chronic midportion situ microdialysis in tendon tissue: high Banes AJ, Tsuzaki M, Wall M, Qi J, Yang
Achilles tendinopathy: an update on levels of glutamate, but not X, Bynum D, Karas S, Hart DA,
research and treatment. Clin Sports prostaglandin E2 in chronic Achilles Nation A, Fox AM, Almekinders LC.
Med 2003: 22: 727–741. tendon pain. Knee Surg Sports The molecular biology of
Alfredson H. Letter to editor. Scand J Traumatol Arthrosc 1999: 7: 378–381. tendinopathy: signaling and response
Med Sci Sports 2004: 14: 269. Almekinders LC, Banes AJ, Ballenger pathway in tenocytes. In: Woo SL,
Alfredson H. The chronic painful Achilles CA. Effects of repetitive motion on Renström P, Arnoczku SP, eds.
and patellar tendon: research on basic human fibroblasts. Med Sci Sports Tendinopathy in athletes. Malden,
biology and treatment. Scand J Med Exerc 1993: 25: 603–607. MA, USA: Blackwell Publisher, 2007:
Sci Sports 2005: 15: 252–259. Almekinders LC, Banes AJ, Bracey LW. 1, 29–45.
Alfredson H, Forsgren S, Thorsen K, An in vitro investigation into the effect Benazzo F, Stennardo G, Valli M.
Fahlstrom M, Johansson H, Lorentzon of repetitive motion and nonsteroidal Achilles and patellar tendinopathies in
R. Glutamate NMDAR1 receptors antiinflammatory medication on athletes: pathogenesis and surgical
localised to nerves in human Achilles human tendon fibroblasts. Am J Sports treatment. Bull Hosp Jt Dis 1996:
tendons. Implications for treatment? Med 1995: 23: 119–123. 54(4): 236–240.
Knee Surg Sports Traumatol Arthrosc Arancia G, Trovalusci CP, Mariutti G, Bestwick CS, Maffulli N. Reactive oxygen
2001a: 9: 123–126. Mondovi B. Ultrastructural changes species and tendinopathy: do they
Alfredson H, Forsgren S, Thorsen K, induced by hyperthermia in Chinese matter? Br J Sports Med 2004: 38:
Lorentzon R. In vivo microdialysis and hamster V79 fibroblasts. Int J 672–674.
immunohistochemical analyses of Hyperthermia 1989: 5: 341–350. Blankstein A, Cohen I, Diamant L, Heim
tendon tissue demonstrated high Archambault JM, Tsuzaki M, Herzog W, M, Dudkiewicz I, Israeli A, Ganel A,
amounts of free glutamate and Banes AJ. Stretch and interleukin- Chechick A. Achilles tendon pain and
glutamate NMDAR1 receptors, but no 1beta induce matrix metalloproteinases related pathologies: diagnosis by
signs of inflammation, in Jumper’s in rabbit tendon cells in vitro. J Orthop ultrasonography. Isr Med Assoc J
knee. J Orthop Res 2001b: 19: Res 2002: 20: 36–39. 2001: 3: 575–578.
881–886. Archambault JM, Wiley JP, Bray RC. Boesen MI, Koenig MJ, Torp-Pedersen S,
Alfredson H, Forsgren S, Thorsen K, Exercise loading of tendons and the Bliddal H, Langberg H. Tendinopathy
Lorentzon R. High intratendinous development of overuse injuries. A and Doppler activity: the vascular
lactate levels in painful chronic Achilles review of current literature. Sports response of the achilles tendon to
tendinosis. An investigation using Med 1995: 20: 77–89. exercise. Scand J Med Sci Sports 2006:
microdialysis technique. J Orthop Res Arnoczku SP, Tian T, Lavagnino M, 16: 463–469.
2002: 20: 934–938. Gardnet K, Schuler P, Morse P. Brain SD, Williams JR, Tippins JR,
Alfredson H, Lorentzon R. Chronic Activation of stress-activated protein Morris HR, MacIntyre I. Calcitonin
Achilles tendinosis: recommendations kinase (SAPK) in tendon cells gene-related peptide is a potent
for treatment and prevention. Sports following cyclic strain: the effects of vasodilator. Nature 1985: 313: 54–56.
Med 2000: 29: 135–146. strain frequency, strain magnitude, and Brich HL, Wilson AM, Goodship AE.
Alfredson H, Lorentzon R. Chronic cytosolic calcium. J Orthop Res 2002: The effect of exercise-induced localised
tendon pain: no signs of chemical 20: 947–952. hyperthermia on tendon cell survival. J
inflammation but high concentrations Astrom M, Rausing A. Chronic Achilles Exp Biol 1997: 200: 1703–1708.
of the neurotransmitter glutamate. tendinopathy. A survey of surgical and Broome CS, Miyan JA. Neuropeptide
Implications for treatment? Curr Drug histological findings. Clin Orthop 1995: control of bone marrow neutrophil
Targets 2002: 3: 43–54. 12: 246–252. production. A key axis for
Alfredson H, Lorentzon M, Backman S, Åstrøm M, Rausing A. Chronic Achilles neuroimmunomodulation. Ann NY
Backman C, Lerner UH. cDNA-arrays tendinopathy. A survey of surgical and Acad Sci 2000: 917: 424–434.
and real-time quantitative PCR histopathologic findings. Clin Orthop Carvès C, Duquenoy A, Toutain F,
techniques in the investigation of 1995: 316: 151–164. Trioche P, Zarnitski C, Le Roux P, Le
chronic Achilles tendinosis. J Orthop Aydingöz U, Aydingöz O. Spontaneous Luyer B. Gouty tendinitis revealing
Res 2003a: 21: 970–975. rupture of the tibialis anterior tendon glycogen storage disease Type Ia in two

11
Fredberg & Stengaard-Pedersen
adolescents. Jt Bone Spine 2003: 70: de Mos M, van El B, Degroot J, Jahr H, study. Scand J Rheumatol 2004: 33:
149–153. van Schie HT, van Arkel ER, Tol H, 94–101.
Cetti R, Junge J, Vyberg M. Spontaneous Heijboer R, van Osch GJ, Verhaar JA. Fritschy D, Wallensten R. Surgical
rupture of the Achilles tendon is Achilles tendinosis: changes in treatment of patellar tendinitis. Knee
preceded by widespread and bilateral biochemical composition and collagen Surg Sports Traumatol Arthrosc 1993:
tendon damage and ipsilateral turnover rate. Am J Sports Med 2007: 1: 131–133.
inflammation: a clinical and 35(9): 1549–1556. Fu SC, Wang W, Pau HM, Wong YP,
histopathologic study of 60 patients. Dirig DM, Yaksh TL. Spinal synthesis Chan KM, Rolf CG. Increased
Acta Orthop Scand 2003: 74: 78–84. and release of prostanoids after expression of transforming growth
Chhajed PN, Plit ML, Hopkins PM, peripheral injury and inflammation. factor-beta1 in patellar tendinosis. Clin
Malouf MA, Glanville AR. Achilles Adv Exp Med Biol 1999: 469: 401–408. Orthop Relat Res 2002: 174–183.
tendon disease in lung transplant Ebriques de Salamanca A, Siemasko KF, Funk CD. Prostaglandins and
recipients: association with Diebold Y, Calonge M, Juarez-Campo leukotrienes: advances in eicosanoid
ciprofloxacin. Eur Respir J 2002: 19: M, Stern ME. Expression of biology. Science 2001: 294: 1871–1875.
469–471. muscarinic and adrenergic receptors in Gibbon WW, Cooper JR, Radcliffe GS.
Cilli F, Khan M, Fu F, Wang JH. normal human conjunctival epithelium. Sonographic incidence of tendon
Prostaglandin E2 affects proliferation Invest Ophthalmol Vis Sci 2005: 46: microtears in athletes with chronic
and collagen synthesis by human 504–513. Achilles tendinosis. Br J Sports Med
patellar tendon fibroblasts. Clin J Sport el Hawary R, Stanish WD, Curwin S. 1999: 33: 129–130.
Med 2004: 14: 232–236. Rehabilitation of tendon injuries in Gibbon WW, Cooper JR, Radcliffe GS.
Clement DB, Taunton JE, Smart GW. sport. Sports Med 1997: 24: 347–358. Distribution of sonographically
Achilles tendinitis and peritendinitis: Erickson S. High-resolution imaging of detected tendon abnormalities in
etiology and treatment. Am J Sports the musculoskeletal system. Radiology patients with a clinical diagnosis of
Med 1984: 12: 179–184. 1997: 205: 593–618. chronic achilles tendinosis. J Clin
Colosimo AJ, Bassett FH III. Jumper’s Ferrara N. Molecular and biological Ultrasound 2000: 28: 61–66.
knee. Diagnosis and treatment. Orthop properties of vascular endothelial Gold MS. Tetrodotoxin-resistant Na1
Rev 1990: 19: 139–149. growth factor. J Mol Med 1999: 77: currents and inflammatory
Cook JL, Khan KM, Harcourt P, Grant 527–543. hyperalgesia. Proc Natl Acad Sci USA
M, Young DA, Bonar F. A cross Ferretti A, Ippolito E, Mariani P, Puddu 1999: 96: 7645–7649.
sectional study of 100 athletes with G. Jumper’s knee. Am J Sports Med Goodship AE, Birch HL, Wilson AM.
jumper’s knee managed conservatively 1983: 11: 58–62. The patholobiology and repair of
and surgically. The Victorian institute Fornage BD, Tendons. In: Cosgrove D, tendon and ligament injury. Vet Clin
of sport tendon study group. Br J Meire H, Dewbury K, eds. Clinical North Am Equine Pract 1994: 10: 329–
Sports Med 1997: 31: 332–336. ultrasound a comprehensive text. 349.
Cook JL, Khan KM, Kiss ZS, Griffiths L. Abdominal and general ultrasound. Haegerstrand A, Dalsgaard CJ, Jonzon
Patellar tendinopathy in junior New York: Churchill Livingstone, B, Larsson O, Nilsson J. Calcitonin
basketball players: a controlled clinical 1993: 816–823. gene-related peptide stimulates
and ultrasonographic study of 268 Fornage BD. Achilles tendon: US proliferation of human endothelial
patellar tendons in players aged 14–18 examination. Radiology 2003: 159: cells. Proc Natl Acad Sci USA 1990:
years. Scand J Med Sci Sports 2000: 10: 759–764. 87: 3299–3303.
216–221. Forsgren S, Danielson P, Alfredson H. Hart DA, Archambault JM, Kydd A,
Cook JL, Khan KM, Kiss ZS, Purdam Vascular NK-1 receptor occurrence in Reno C, Frank CB, Herzog W. Gender
CR, Griffiths L. Reproducibility and normal and chronic painful Achilles and neurogenic variables in tendon
clinical utility of tendon palpation to and patellar tendons: studies on biology and repetitive motion
detect patellar tendinopathy in young chemically unfixed as well as fixed disorders. Clin Orthop Relat Res 1998:
basketball players. Victorian institute specimens. Regul Pept 2005: 126: 351: 44–56.
of sport tendon study group. Br J 173–181. Hasbak P, Lundby C, Olsen NV, Schifter
Sports Med 2001: 35: 65–69. Fredberg U. Local corticosteroid S, Kanstrup IL. Calcitonin gene-
Curwin S. The aetiology and treatment of injection in sport: review of literature related peptide and adrenomedullin
tendinitis. In: Harris M, Williams C, and guidelines for treatment. Scand J release in humans: effects of exercise
Stannish WD, Michelis L, eds. Oxford Med Sci Sports 1997: 7: 131–139. and hypoxia. Regul Pept 2002: 108: 89–
textbook of sports medicine. Oxford: Fredberg U, Bolvig L. Significance of 95.
Oxford University Press, 1994: 512–528. ultrasonographically detected Heppelmann B, Just A, Pawlak P.
Danielson P, Alfredson H, Forsgren S. asymptomatic tendinosis in the patellar Galanin influences the
Distribution of general (PGP 9.5) and and achilles tendons of elite soccer mechanosensitivity of sensory endings
sensory (substance P/CGRP) players: a longitudinal study. Am J in the rat knee joint. Eur J Neurosci
innovations in the human patellar Sports Med 2002: 30: 488–491. 2000: 12: 1567–1572.
tendon. Knee Surg Sports Traumatol Fredberg U, Bolvig L, Pfeiffer-Jensen M, Hernanz A, Medina S, de Miguel E,
Arthrosc 2006a: 14: 125–132. Clemmensen D, Jakobsen BW, Martin-Mola E. Effect of calcitonin
Danielson P, Alfredson H, Forsgren S. Stengaard-Pedersen K. gene-related peptide, neuropeptide Y,
Immunohistochemical and Ultrasonography as a tool for substance P, and vasoactive intestinal
histochemical findings favoring the diagnosis, guidance of local steroid peptide on interleukin-1beta,
occurrence of autocrine/paracrine as injection and together with pressure interleukin-6 and tumor necrosis
well as nerve-related cholinergic effects algometry monitoring of the treatment factor-alpha production by peripheral
in chronic painful patellar tendon of athletes with chronic Jumper’s knee whole blood cells from rheumatoid
tendinosis. Microsc Res Technol and Achilles tendinitis: a randomised, arthritis and osteoarthritis patients.
2006b: 69: 808–819. double-blind, placebo-controlled Regul Pept 2003: 115: 19–24.

12
Chronic tendinopathy tissue pathology
Hofmann GO, Weber T, Lob G. Tendon study of 891 patients. J Bone Jt Surg Scand J Med Sci Sports 2004: 14: 100–
rupture in chronic kidney insufficiency Am 1991: 73: 1507–1525. 106.
– ‘‘uremic tendonopathy’’? A Kannus P, Natri A. Etiology and Koivunen-Niemela T, Parkkola K.
literature-supported documentation of pathophysiology of tendon ruptures in Anatomy of the Achilles tendon (tendo
3 cases. Chirurg 1990: 61: 434–437. sports. Scand J Med Sci Sports 1997: 7: calcaneus) with respect to tendon
Hökfelt T, Broberger C, Xu Z-QD, 107–112. thickness measurements. Surg Radiol
Sergeyev V, Ubink R, Diez M. Karahan S, Kincaid SA, Baird AN, Anat 1995: 17: 263–268.
Neuropeptides – an overview. Kammermann JR. Distribution of Kricun R, Kricun ME. Patellar tendon
Neuropharmacology 2000: 39: beta-endorphin and substance P in the rupture with underlying systemic
1337–1356. shoulder joint of the dog before and disease. Am J Radiol 1980: 135:
Holmes GB, Lin J. Etiologic factors after a low impact exercise programme. 803–807.
associated with symptomatic achilles Anat Histol Embryol 2002: 31: 72–77. Kujala UM, Kvist M, Osterman K. Knee
tendinopathy. Foot Ankle Int 2006: 27: Karlsson J, Kalebo P, Goksor LA, injuries in athletes. Review of exertion
952–959. Thomee R, Sward L. Partial rupture of injuries and retrospective study of
Inoue H, Shimayama Y, Hirabayashi K, the patellar ligament. Am J Sports Med outpatient sports clinic material. Sports
Kajigaya H, Yamamooto S, Oda H, 1992: 20: 390–395. Med 1986: 3: 447–460.
Koshihara Y. Production of Kaufman KR, Brodine SK, Shaffer RA, Kujala UM, Sarna S, Kaprio J.
neuropeptide substance P by synovial Johnson CW, Cullison TR. The effect Cumulative incidence of achilles
fibroblasts from patients with of foot structure and range of motion tendon rupture and tendinopathy in
rheumatoid arthritis and osteoarthritis. on musculoskeletal overuse injuries. male former elite athletes. Clin J Sport
Neurosci Lett 2001: 303: 149–152. Am J Sports Med 1999: 27: 585–593. Med 2005: 15: 133–135.
Ip YT, Davis RJ. Signal transduction by Kettunen JA, Kujala UM, Kaprio J, Kvist M. Achilles tendon injuries in
the c-Jun N-terminal kinase (JNK) – Sarna S. Health of master track and athletes. Ann Chir Gynaecol 1991: 80:
from inflammation to development. field athletes: a 16-year follow-up 188–201.
Curr Opin Cell Biol 1998: 10: 205–219. study. Clin J Sport Med 2006: 16: 142– Kvist M. Achilles tendon injuries in
Jakobsen LP, Knudsen TB, Bloch T. 148. athletes. Sports Med 1994: 18:
Spontaneous infrapatellar tendon Khan KM, Bonar F, Desmond PM, 173–201.
rupture in a patient with systemic lupus Cook JL, Young DA, Visentini PJ, Langberg H, Olesen JL, Gemmer C,
erythematosus. Ugeskr Laeger 2000: Fehrmann MW, Kiss ZS, O’Brian PA, Kjaer M. Substantial elevation of
162: 5088–5089. Harcourt P, Dowling RJ, O’Sullivan interleukin-6 concentration in
Järvinen M, Jòzsa L, Kannus P, Järvinen RM, Crichton KJ, Tress BM, Wark peritendinous tissue, in contrast to
TJN, Kvist M, Leadbetter WB. JD. Patellar tendinosis (jumper’s knee): muscle, following prolonged exercise in
Histopathological findings in chronic findings at histopathologic humans. J Physiol 2002: 542: 985–990.
tendon disorders. Scand J Med Sci examination, US, and MR imaging. Langberg H, Olesen JL, Skovgaard D,
Sports 1997: 7: 85–95. Radiology 1996: 200: 821–827. Kjaer M. Age related blood flow
Johnson DP, Wakeley CJ, Watt I. Khan KM, Cook JL, Bonar F, Harcourt around the Achilles tendon during
Magnetic resonance imaging of patellar PAM. Histopathology of common exercise in humans. Eur J Appl Physiol
tendonitis. J Bone Jt Surg Br 1996: 78: tendinopathies. Update and 2001: 84: 246–248.
452–457. implications for clinical management. Langberg H, Skovgaard D, Karamouzis
Józsa L, Balint JB, Kannus P, Reffy A, Sports Med 1999: 27: 393–408. M, Bulow J, Kjaer M. Metabolism and
Barzo M. Distribution of blood groups Khan KM, Cook JL, Kannus P, Maffulli inflammatory mediators in the
in patients with tendon rupture. An N, Bondesteam S. Time to abandon the peritendinous space measured by
analysis of 832 cases. J Bone Jt Surg Br ‘‘tendinitis’’ myth (editorial). BMJ microdialysis during intermittent
1989: 71: 272–274. 2002: 324: 626–627. isometric exercise in humans. J Physiol
Jòzsa L, Kannus P. Human tendons. Khan MH, Li Z, Wang JHC. Repeated 1999a: 15: 919–927.
Anatomy, physiology and pathology. exposure of tendon to prostaglandin- Langberg H, Skovgaard D, Peterson L,
Canada: Human Kinetics, 1997. E2 leads to localized tendon Bulow J, Kjaer M. Type I collagen
Jozsa L, Reffy A, Kannus P, Demel S, degeneration. Clin J Sport Med 2005: synthesis and degradation in periten-
Elek E. Pathological alterations in 15: 27–33. dinous tissue after exercise determined
human tendons. Arch Orthop Trauma Khasar SG, Gold MS, Levine JD. A by microdialysis in humans. J Physiol
Surg 1990: 110: 15–21. tetrodotoxin-resistant sodium current 1999b: 521: 299–306.
Kälebo P, Sward L, Karlsson J, Peterson mediates inflammatory pain in the rat. Li Z, Khan M, Stone D, Woo SL, Wang
L. Ultrasonography in the detection of Neurosci Lett 1998: 256: 17–20. JH. Inflammatory response of human
partial patellar ligament ruptures Khurana R, Torzillo PJ, Horsley M, tendon fibroblasts to cyclic mechanical
(jumper’s knee). Skeletal Radiol 1991: Mahoney J. Spontaneous bilateral stretching. Am J Sports Med 2004: 32:
20: 285–289. rupture of the Achilles tendon in a 435–440.
Kamel M, Eid H, Mansour R. patient with chronic obstructive Lian O, Dahl A, Ackermann PW,
Ultrasound detection of knee patellar pulmonary disease. Respirology 2002: Frihagen F, Engebretsen L, Bahr R.
enthesitis: a comparison with magnetic 7: 161–163. Pronociceptive and antinociceptive
resonance imaging. Ann Rheum Dis Kirkendall DT, Garret WE. Function neuromediatory in patellar
2004: 63: 213–214. and biomechanics of tendons. Scand J tendinopathy. Am J Sports Med 2006:
Kannus P. Etiology and pathophysiolohy Med Sci Sports 1997: 7: 62–66. 34(11): 1801–1808.
of chronic tendon disorders in sports. Koenig MJ, Torp-Pedersen S, Qvistgaard Lian O, Engebretsen L, Bahr R.
Scand J Med Sci Sports 1997: 7: 78–85. E, Terslev L, Bliddal H. Preliminary Prevalence of jumper’s knee among
Kannus P, Józsa L. Histopathological results of colour Doppler-guided elite athletes from different sports: a
changes preceding spontaneous intratendinous glucocorticoid injection cross-sectional study. Am J Sports Med
ruptures of a tendon. A controlled for Achilles tendonitis in five patients. 2005: 33: 561–567.

13
Fredberg & Stengaard-Pedersen
Lind H, Brudin L, Lindholm L, Edvisson imaging of jumper’s knee-patellar and strenuous exercise: TNF-alpha, IL-
L. Different levels of sensory tendinitis. Clin Radiol 1988: 39: 162– 2beta, IL-6, IL-1ra, sTNF-r1, sTNF-
neuropeptides (calcitonin gene-related 165. r2, and IL-10. J Physiol (London) 1999:
peptide and substance P) during and Movin T. Aspects of aetiology, 515: 287–291.
after exercise in man. Clin Physiol pathoanatomy and diagnostic methods Paavola M, Kannus P, Jarvinen TA,
1996: 16: 73–82. in chronic midportion Achilledynia. Khan KM, Jozsa L, Jarvinen M.
Ljungqvist R. Subcutaneous partial Stockholm, Sweden: Karolinska Achilles tendinopathy. J Bone Jt Surg
rupture of the Achilles tendon. Acta Institute, 1998. Am 2002: 84-A: 2062–2076.
Orthop Scand 1967: 113(Suppl.):11. Movin T, Gad A, Reinholt FP, Rolf C. Pedersen BK, Hoffmann-Goetz L.
Maffulli N, Kenward MG, Testa V, Tendon pathology in long-standing Exercise and the immune system:
Capasso G, Regine R, King JB. achillodynia. Biopsy findings in 40 regulation, integration, and
Clinical diagnosis of Achilles patients. Acta Orthop Scand 1997a: 68: adaptation. Physiol Rev 2000: 80:
tendinopathy with tendinosis. Clin J 170–175. 1055–1079.
Sport Med 2003: 13: 11–15. Movin T, Gunter P, Gad A, Rolf C. Peers KH, Lysens RJJ. Patellar
Maffulli N, Regine R, Angelillo M, Ultrasonography-guided percutaneous tendinopathy in athletes: current
Capasso G, Filice S. Ultrasound core biopsy in Achilles tendon disorder. diagnostic and therapeutic
diagnosis of Achilles tendon pathology Scand J Med Sci Sports 1997b: 7: 244– recommendations. Sports Med 2005:
in runners. Br J Sports Med 1987: 21: 248. 35: 71–78.
158–162. Movin T, Kristoffersen-Wiberg M, Rolf Peiro A, Ferrandis R, Garcia L, Alcazar
Maffulli N, Regine R, Carrillo F, Minelli C, Aspelin P. MR imaging in chronic E. Simultaneous and spontaneous
S, Beaconsfield T. Ultrasonographic Achilles tendon disorders. Acta Orthop bilateral rupture of the patellar tendon
scan in knee pain in athletes. Br J Scand 1998a: 68: 126–132. in rheumatoid arthritis. A case report.
Sports Med 1992: 26: 93–96. Movin T, Kristoffersen-Wiberg M, Acta Orthop Scand 1975: 46:
Mahieu NN, Wityrouw E, Stevns V, Van Shalabi A, Gad A, Aspelin P, Rolf C. 700–703.
Tiggelen D, Roget P. Intrinsic risk Intratendinous alterations as imaged Petersen W, Pufe T, Kurz B, Mentlein R,
factors for the development of Achilles by ultrasound and contrast medium- Tillmann B. Angiogenesis in fetal
tendon overuse injury: a prospective enhanced magnetic resonance in tendon development: spatial and
study. Am J Sports Med 2006: 34(2): chronic achillodynia. Foot Ankle Int temporal expression of the angiogenic
226–235. 1998b: 19: 311–317. peptide vascular endothelial cell
Major NM, Helm CA. MR imaging of Murphy DF, Connell DAJ, Beynnon BD. growth factor. Anat Embryol (Berlin)
the knee: findings in asymptomatic Risk factors for lower extremity injury: 2002: 205: 263–270.
collegiate basketball players. Am J a review of the literature. Br J Sports Preston ET. Avulsion of both quadriceps
Roentgenol 2002: 179: 641–644. Med 2003: 37: 13–29. tendons in hyperparathyroidism.
Malaguti M, Triolo L, Biagina M. Myllymaki T, Bondesteam S, Suramo I, JAMA 1972: 221: 406–407.
Ciprofloxacin-associated Achilles Cederberg A, Peltokallio P. Pritchard CH, Berney S. Patellar tendon
tendon rupture in a hemodialysis Ultrasonography of jumper’s knee. rupture in systemic lupus
patient. J Nephrol 2001: 14: 431–432. Acta Radiol 1990: 31: 147–149. erythematosus. J Rheumatol 1989: 16:
Martinoli C, Derchi LE, Pastorino C, Neufeld G, Cohen T, Gengrinovitch S, 786–788.
Bertolotto M, Silvestri E. Analysis of Poltorak Z. Vascular endothelial Pufe T, Petersen WJ, Mentlein BN,
echotexture of tendons with US. growth factor (VEGF) and its Tillmann BN. The role of vasculature
Radiology 1993: 186: 839–843. receptors. FASEB J 1999: 13: and angiogenesis for the pathogenesis
Masi AT, Bijsma JW, Chikanza IC, 9–22. of degenerative tendons disease. Scand
Pitzalis C, Cutolo M. Neuroendocrine, Newham DM, Douglas JG, Leggs JS, J Med Sci Sports 2005: 15: 211–222.
immunologic, and microvascular Friend JA. Achilles tendon rupture: an Pufe T, Petersen W, Tillmann B, Mentlein
systems interactions in rheumatoid underrated complication of R. The angiogenic peptide vascular
arthritis: physiopathogenetic and corticosteroid treatment. Thorax 1991: endothelial growth factor is expressed
therapeutic perspectives. Semin 46: 853–854. in foetal and ruptured tendons.
Arthritis Rheum 1999: 29: 65–81. Nigg BM. The role of impact forces and Virchows Arch 2001: 439: 579–585.
Mokone GG, Gajjar M, September AV, foot pronation: a new paradigm. Clin J Qzgurtas T, Yildiz C, Serdar M, Atesalp
Schwellnus MP, Greenberg J, Noakes Sport Med 2001: 11: 2–9. S, Kutluay T. Is high concentration of
TD, Collins M. The guanine–thymine Nilsson J, von Euler AM, Dalsgaard CJ. serum lipids a risk factor for Achilles
dinucleotide repeat polymorphism Stimulation of connective tissue cell tendon rupture? Clin Chim Acta 2003:
within the tenascin-c gene is associated growth by substance P and substance 331: 25–28.
with Achilles tendon injuries. Am K. Nature 1985: 315: 61–63. Raatikainen T, Karpakka J, Puranen J,
Orthop Soc Sports Med 2005: 33: O’Banion MK. Cyclooxygenase-2: Orava S. Operative treatment of partial
1016–1021. molecular biology, pharmacology, and rupture of the patellar ligament. A
Mokone GG, Schwellnus MP, Noakes neurobiology. Crit Rev Neurobiol study of 138 cases. Int J Sports Med
TD, Collins M. The COL5A1 gene and 1999: 13: 45–82. 1994: 15: 46–49.
Achilles tendon pathology. Scand J O’Brian T. The needle test for complete Resnick D, Feingold ML, Curd J,
Med Sci Sports 2006: 16: 19–26. rupture of the Achilles tendon. J Bone Niwayama G, Goergen TG. Calcaneal
Monneret G, Pachot A, Laroche B, Jt Surg Am 1984: 66: 1099–1101. abnormalities in articular disorders.
Picollet J, Bienvenu J. Procalcitonin Ohberg L, Alfredson H. Ultrasound Rheumatoid arthritis, ankylosing
and calcitonin gene-related peptide guided sclerosis of neovessels in painful spondylitis, psoriatic arthritis, and
decrease LPS-induced tnf production chronic Achilles tendinosis: pilot study Reiter syndrome. Radiology 1977: 125:
by human circulating blood cells. of a new treatment. Br J Sports Med 355–366.
Cytokine 2000: 12: 762–764. 2002: 36: 173–175. Richards PJ, Dheer AK, McCall IM.
Mourad K, King JB, Guggiana P. Ostrowski K, Rhode T, Asp S, Schjerling Achilles tendon (TA) size and power
Computed tomography and ultrasound P, Pedersen BK. The cytokine balance Doppler ultrasound (PD) changes

14
Chronic tendinopathy tissue pathology
compared to MRI: a preliminary stretching enhances secretion of imaging and ultrasonography in
observational study. Clin Radiol 2001: interleukin 6 in human tendon confirming clinically diagnosed patellar
56: 843–850. fibroblasts. Knee Surg Sports tendinopathy. Am J Sports Med 2007:
Samad TA, Sapirstein A, Woolf JC. Traumatol Arthrosc 2003: 9: 322–326. 35: 427–436.
Prostanoids and pain: unraveling Stanish WD, Curwin S, Rubinovich M. Webb JM, Bannister GC. Percutaneous
mechanisms and revealing therapeutic Tendinitis: the analysis and treatment repair of the ruptured tendo Achillis. J
targets. Trends Mol Med 2003: 8: 390– for running. Clin Sports Med 1985: 4: Bone Jt Surg Br 1999: 81: 877–880.
396. 593–609. Weiss J, Duttaroy A, Gomeza J, Zhans
Schmidt MR, Hodler J, Cathrein P, Stone D, Green C, Rao U, Aizawa H, W, Yamada M, Felder CC, Bernardini
Duewell S, Jacob HA, Romeo J. Is Yamaji T, Niyibizi C, Carlson CS. N, Reeh PW. Muscarinic receptor
impingement the cause of jumper’s Cytokine-induced tendinitis: a subtypes mediating central and
knee? Dynamic and static magnetic preliminary study in rabbits. J Orthop peripheral antinociception studied with
resonance imaging of patellar tendinitis Res 1999: 17: 168–177. muscarinic receptor knockout mice: a
in an open-configuration system. Am J Strand FL, Rose LZLA, Kume J, Alves review. Life Sci 2003: 72: 2047–2054.
Sports Med 2002: 30: 388–395. SE, Anatonawich FJ, Garrett LY. Wessler I, Kilbinger H, Bittinger F,
Schubert TE, Weidler C, Lerch K, Neuropeptide hormones as Kirkpatrick CJ. The biological role of
Hofstadter F, Straub RH. Achilles neurotrophic factors. Physiol Rev non-neuronal acetylcholine in plants
tendinosis is associated with sprouting 1991: 71: 1017–1046. and humans. Jpn J Pharmacol 2001: 85:
of substance P positive nerve fibres. Sullo A, Maffulli N, Capasso G, Testa V. 2–10.
Ann Rheum Dis 2005: 64: 1083–1086. The effects of prolonged peritendinous Westermark T, Rantapaa-Dalqvist S,
Schwartz JP. Neurotransmitters as administration of PGE1 to the rat Wallberg-Jonsson S, Kjorell U,
neurotrophic factors: a new set of Achilles tendon: a possible animal Forsgren S. Increased content of
functions. Int Rev Neurobiol 1992: 34: model of chronic Achilles tendinopathy. bombesin/GRP in human synovial
1–23. J Orthop Sci 2001: 6(4): 349–357. fluid in early arthritis: different pattern
Scott A, Khan KM, Roberst CR, Suzuki T, Nakamura Y, Moriya T, compared with substance P. Clin Exp
Duronio V. What do we mean by the Sasano H. Effects of steroid hormones Rheumatol 2001: 19: 715–720.
term ‘‘inflammation’’? A contemporary on vascular functions. Microsc Res Wiesenfeld-Hallina Z, Hökfelt T,
basic science update for sports Technol 2003: 60: 76–84. Lundberg JM, Forssmannd WG,
medicine. Br J Sports Med 2004: 38: Terslev L, Qvistgaard E, Danneskiold- Reineckee M, Tschoppf FA, Fischerg
372–380. Samsøe B, Bliddal H. Estimation of JA. Immunoreactive calcitonin gene-
Sedo A, Duke-Cohan JS, Balaziova E, inflammation by Doppler ultrasound: related peptide and substance P coexist
Sedova LR. Dipeptidyl peptidase IV quantitative changes after intra- in sensory neurons to the spinal cord
activity and/or structure homologs: articular treatment in rheumatoid and interact in spinal behavioral
contributing factors in the pathogenesis arthritis. Ann Rheum Dis 2003: 62: responses of the rat. Neurosci Lett
of rheumatoid arthritis? Arthritis Res 1049–1053. 1984: 52: 199–204.
Ther 2005: 7: 253–269. Tilley SL, Coffman TM, Konnttinen Y. Williams DS III, McClay IS, Hamill J. Arch
Senger DR, Gali SJ, Dvorak AM, Peruzzi Mixed messages: modulation of structure and injury patterns in runners.
CA, Harvey VS, Dvorak HF. Tumor inflammation and immune responses Clin Biomech 2001: 16: 341–347.
cells secrete a vascular permeability by prostaglandins and thromboxanes. J Williams JGP. Achilles tendon lesions in
factor that promotes accumulation of Clin Invest 2001: 108: 15–23. sport. Sports Med 1986: 3: 114–135.
ascites fluid. Science 1983: 219: 983– Tsuzaki M, Guyton G, Garret W, Witonski D, Wagrowska-Danielewicz M.
985. Archambault JM, Herzog W, Distribution of substance-P nerve
Shalabi A, Kristoffersen-Wiberg M, Almekinders LC, Bynum D, Yang X, fibres in the knee joint in patients with
Svensson L, Aspelin P, Movin T. Banes AJ. IL-1 beta induces COX2, anterior knee pain syndrome. A
Eccentric training of the MMP-1, -3 and -13, ADAMTS-4, IL-1 preliminary report. Knee Surg Sports
gastrocnemius-soleus complex in beta and IL-6 in human tendon cells. J Traumatol Arthrosc 1997: 7: 177–183.
chronic Achilles tendinopathy results in Orthop Res 2003: 21: 256–264. Wityrouw E, Bellemans J, Lysens R,
decreased tendon volume and Vanegas H, Schaible HG. Prostaglandins Danneels L, Cambier D. Intrinsic risk
intratendinous signal as evaluated by and cyclooxygenases [correction of factors for the development of patellar
MRI. Am J Sports Med 2007: 32: cycloxygenases] in the spinal cord. Prog tendonitis in an athletic population: a
1286–1296. Neurobiol 2001: 64: 327–363. two-years prospective study. Am J
Sharma P, Maffulli N. Biology of tendon Wang JH, Jia F, Yang G, Yang S, Sports Med 2001: 29: 190–195.
injury: healing, modeling and Cambell BH, Stone D, Woo SL. Cyclic Yang S, Zhans L. Glucocorticoids and
remodeling. J Musculoskelet Neuronal mechanical stretching of human tendon vascular reactivity. Curr Vasc
Interact 2006: 6: 181–190. fibroblasts increases the production of Pharmacol 2004: 2: 1–12.
Shields CL. The Cybex II evaluation on prostaglandin E2 and levels of Yu JS, Popp JE, Kaeding CC, Lucas J.
surgical repaired Achilles tendon rup- cyclooxygenase expression: a novel in Correlation of MR imaging and
tures. Am J Sports Med 1978: 7: 15–17. vitro model study. Connect Tissue Res pathologic findings in athletes
Shrier I, Matheson GO, Kohl HW III. 2003: 3–4: 128–133. undergoing surgery for chronic patellar
Achilles tendonitis: are corticosteroid Wang JH, Li Z, Yang G, Khan M. tendinitis. Am J Roentgenol 1995: 165:
injections useful or harmful? Clin J Repetitively stretched tendon 115–118.
Sport Med 1996: 6: 245–250. fibroblasts produce inflammatory Ziche M, Morbidelli L, Pacinni M,
Siwek CW, Rao JP. Ruptures of the mediators. Clin Orthop Relat Res Geppetti P, Alessandri G, Maggi CA.
extensor mechanism of the knee joint. J 2004: 422: 243–250. Substance P stimulates
Bone Jt Surg Am 1981: 63: 932–937. Warden SJ, Kiss ZS, Malara FA, Aoi AB, neovascularization in vivo and
Skutek M, van Griensven M, Zeichen J, Cook J, Crossley KM. Comparative proliferation of cultured endothelial
Brauer N, Bosch U. Cyclic mechanical accuracy of magnetic resonance cells. Microvasc Res 1990: 40: 264–278.

15

Вам также может понравиться