header advert
Bone & Joint Research Logo

Receive monthly Table of Contents alerts from Bone & Joint Research

Comprehensive article alerts can be set up and managed through your account settings

View my account settings

Visit Bone & Joint Research at:

Loading...

Loading...

Open Access

Systematic Review

Cytokines in tendon disease

A Systematic Review



Download PDF

Abstract

Objectives

Emerging evidence indicates that tendon disease is an active process with inflammation that is critical to disease onset and progression. However, the key cytokines responsible for driving and sustaining inflammation have not been identified.

Methods

We performed a systematic review of the literature using MEDLINE (U.S. National Library of Medicine, Bethesda, Maryland) in March 2017. Studies reporting the expression of interleukins (ILs), tumour necrosis factor alpha (TNF-α) and interferon gamma in diseased human tendon tissues, and animal models of tendon injury or exercise in comparison with healthy control tissues were included.

Results

IL-1β, IL-6, IL-10, and TNF-α are the cytokines that have been most frequently investigated. In clinical samples of tendinopathy and tendon tears, the expression of TNF-α tended not to change but IL-6 increased in tears. Healthy human tendons showed increased IL-6 expression after exercise; however, IL-10 remained unchanged. Animal tendon injury models showed that IL-1β, IL-6, and TNF-α tend to increase from the early phase of tendon healing. In animal exercise studies, IL-1β expression showed a tendency to increase at the early stage after exercise, but IL-10 expression remained unchanged with exercise.

Conclusions

This review highlights the roles of IL-1β, IL-6, IL-10, and TNF-α in the development of tendon disease, during tendon healing, and in response to exercise. However, there is evidence accumulating that suggests that other cytokines are also contributing to tendon inflammatory processes. Further work with hypothesis-free methods is warranted in order to identify the key cytokines, with subsequent mechanistic and interaction studies to elucidate their roles in tendon disease development.

Cite this article: W. Morita, S. G. Dakin, S. J. B. Snelling, A. J. Carr. Cytokines in tendon disease: A Systematic Review. Bone Joint Res 2017;6:656–664. DOI: 10.1302/2046-3758.612.BJR-2017-0112.R1.

Article focus

  • To investigate gene and protein expression of cytokines in the development and progression of tendon disease and during tendon healing compared with healthy tendon tissues in both humans and animals.

  • To investigate how exercise affects the gene and protein expression of cytokines in tendon tissues.

  • To determine how cytokines affect the expression of tendon extracellular matrix (ECM) genes and proteins.

Key messages

  • The most frequently investigated cytokines in the development and progression of tendon disease, during tendon healing or in response to exercise were interleukin (IL)-1β, IL-6, IL-10, and tumour necrosis factor alpha (TNF-α), with a paucity of research on others that may also contribute.

  • IL-6 was the only cytokine involved in human tendon disease and was increased in tendon tears, whereas IL-1β, IL-6, and TNF-α tended to be increased in animal models of tendon injury.

  • The effects of cytokines on the expression of tendon ECM genes and proteins could not be determined due to the lack of studies which, in turn, warrants further investigation.

Strengths and limitations

  • This review encompasses current literature by a systematic review and organises the evidence in human and animal studies separately.

  • The key cytokines and their dominant role in the development of tendon disease could not be determined due to the small number and heterogeneity of samples and models.

Introduction

Tendon disease is increasingly common and comprises a third of all musculoskeletal complaints.1 The tendon tissue of early-stage disease, commonly referred to as tendinopathy, is characterised by the development of fibrosis: disoriented collagen fibres; altered composition of extracellular matrix (ECM) proteins; formation of new vessels; and rounding of tendon cells.2 The accumulation of fibrotic tissue predisposes to injury and tendon tear.3,4 The aetiology of tendon disease is acknowledged to be multifactorial, with overuse, trauma, ageing, and genetic predisposition as notable factors;5 however, the involvement of inflammation has long been debated.6 Today, emerging evidence indicates a strong inflammatory component to the pathogenesis of tendon disease, with inflammatory cells and cytokines as important regulators of the tendon ECM.7,8 Cytokines such as interleukins (IL), tumour necrosis factor alpha (TNF-α) and interferon gamma (IFN-γ), alongside growth factors such as transforming growth factor beta (TGF-β) and platelet-derived growth factor, are released from tendon stromal and immunoregulatory cells in response to tissue injury, mechanical stress, and malfunction.3,9,10 They alter the cellular phenotype of the local cells and the persistence of the cellular change by chronic inflammation results in the production of excessive and inappropriate matrix proteins and fibrosis.10 Similar responses have been widely studied in fibrotic diseases in organs such as the liver, kidney, and lung.11

The aim of this study was to systematically review the key cytokines that are involved in the development of tendon disease with a focus on fibrosis. The first objective was to investigate the gene and protein expression of cytokines in diseased tendons along the development of tendinopathy to tear, in comparison with that of healthy tendon. The second objective was to investigate how exercise affects the expression of these cytokines in tendon tissues. We also reviewed how tendon cells respond to these cytokines in the expression of ECM genes and proteins. We hypothesised that the cytokines expressed would vary during the process of tendon disease development or injury healing, and that the tendon cells from normal tissue, early disease, late disease, and healing would have differential responses to these cytokines.

Materials and Methods

This systematic review was designed, undertaken, and reported based on the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) statement. Scientific literature was obtained using the MEDLINE (U.S. National Library of Medicine, Bethesda, Maryland) electronic database using the term “tendon AND cytokine” in March 2017.

The studies were included if they reported the expression of ILs, TNF-α, and IFN-γ as these were found to be the cytokines that were consistently considered through the preliminary screening. The cytokines of the TGF-β superfamily were excluded as we have reviewed these previously.12 Studies that compared the expression of the cytokines in diseased human tendon tissues and animal models of tendon injury or exercise with that of healthy control tissue were included. The tendon tissues included in the search criteria were from the mid-substance of tendon or tendon-to-bone enthesis. Studies on muscle-tendon junctions and ligament reconstruction using tendon grafts and other soft tissues (muscles, ligaments, cartilage, fat, bursa, and synovial tissues), as well as fetal, knockout animal models, animal studies of endocrine disorders (hyperglycaemia, menopause), and ex vivo experimental studies were excluded. The in vitro studies that investigated the effects of the cytokines on the expression of tendon ECM genes and proteins (collagens, elastin, proteoglycans, metalloproteinases (MMPs), tissue inhibitor of metalloproteinases (TIMPs), and a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS)) by healthy, diseased, or injured tendon cells were also included. Review articles, protocols, commentaries, case reports, and studies that were not reported in English were excluded. There was no limitation as to the year of publication.

Our search yielded 784 results. One article was identified by searching through references of listed articles. Following title screening, 594 abstracts were screened to determine eligibility and 57 papers met the inclusion criteria (Fig. 1). The papers that met the criteria are summarised in the additional files (supplementary tables i to iii).

Fig. 1 
          Flowchart of the systematic review protocol.

Fig. 1

Flowchart of the systematic review protocol.

Study characteristics

The studies that compared the expression of cytokines in diseased human tendons obtained tissue samples from healthy, tendinopathic, torn, and healing tendons after surgical repair of the rotator cuff (RC), Achilles, patella, posterior tibialis, digital flexors, and extensor carpal radialis brevis. The effect of exercise was studied in the Achilles and patellar tendons. Five out of 17 studies on human tendon tissues used a gene micro or cytometric bead array to determine the cytokines of interest in diseased human tendon tissues (supplementary table i).

The animal tendon injury models were wide-ranging, using the Achilles, RC, or flexor digitorum (FD) tendons in rats, dogs, and rabbits, with injuries created by collagenase, crush injury, partial transection, and full transection. Depending on the study, the transected tendons were repaired surgically or left to heal spontaneously. The animal tendon exercise models used the patellar, Achilles, RC, or FD tendons of rats or rabbits. Studies on horse tendon tissues obtained clinical samples of FD tendinopathy and compared the expression of cytokines with that of healthy tendon tissues. There were three out of 30 animal studies that used an array to determine the cytokines involved (supplementary table ii).

The in vitro studies investigated the effects of cytokines on the expression of tendon ECM genes and proteins in healthy, disease, or injured human and animal tendons. Human tendon cells were obtained from normal or tendinopathic tendons. The anatomy and species differed in all of the studies using animal tendon cells, which included healthy horse FD, injured mouse Achilles, and both healthy and injured rat patellar tendons (supplementary table iii).

Study methodology and assessing the risk of bias

The quality of study methodology was assessed in all papers by referring to the modified scoring system by Dean et al8 and Morita et al12 in order to highlight the studies with a high risk of potential bias. The median score was 8 (interquartile range, 7 to 9) out of 10 (supplementary table iv). Eight human tissue studies, six animal model tissue studies, and five in vitro experiment studies did not fully describe the age and gender of the included subjects. All of the included studies clearly described the control group. One study obtained the control tissue from the unaffected region of the tendinopathic tendon under confirmation by ultrasonography. There were three studies that sampled diseased tissues based on gross inspection, which may be a risk of bias. All except four studies clearly described the experimental procedures of tissue sampling and analysis, and 32 documented the validity or reliability of the methods used. Seven studies did not use quantitative measures or statistical analysis for comparison. Of the 51 studies that used quantitative analysis with statistical comparison, 45 stated the statistical level of significance, but only eight checked the data for normal distribution. Study limitations were not addressed in 21 studies. A meta-analysis was not carried out due to the heterogeneity of the data from clinical samples and animal models.

Results

A total of 20 cytokines were implicated in the development of tendon disease, healing or in response to exercise: IL-1α, IL-1β, IL-2, IL-3, IL-4, IL-6, IL-8, IL-10, IL-11, IL-12, IL-13, IL-14, IL-15, IL-17, IL-18, IL-27, TNF-α, and IFN-γ were identified by array-based studies (seven studies); and IL-21 and IL-33 were guided by literature. IL-1β, IL-6, IL-10, and TNF-α had been investigated in numerous (more than ten) hypothesis-free and literature-guided studies (Fig. 2). Hence, the results on these four cytokines were summarised (Table I). Results of other cytokines are summarised in the additional files (supplementary table v). Effects of cytokines on the expression of tendon ECM genes and proteins in tendon cells have been presented for IL-1β, IL-6, IL-10, and TNF-α, similarly (Table II). Results of other cytokines are shown in the additional files (supplementary table vi).

Fig. 2 
          Number of studies of human tendon tissues and animal tendon injury or exercise models for each cytokine.

Fig. 2

Number of studies of human tendon tissues and animal tendon injury or exercise models for each cytokine.

Table I.

Expression of interleukins IL-1β, IL-6, IL-10, and tumour necrosis factor alpha (TNF-α) in tissues of diseased human tendon, animal models of tendon injury or exercise versus healthy control tendon. Arrows indicate increased (↑), unchanged (→), or decreased (↓) expression of cytokines in tissues of diseased human tendon, animal models of tendon injury or exercise versus healthy control tendon. If two arrows are given, this indicates that more than one change in expression has been reported (for example, →/↑ indicates that both unchanged and increased expression have been reported)

Cytokine Animal Disease model Increased, unchanged, decreased in diseased vs control
Gene Protein
IL-1β Human Rotator cuff, tear 13 ↑ (descriptive)15
Achilles, tear, post-operative (2 wks) *16
Achilles, tendinopathy *17
Achilles, tendinopathy + exercise (1 hr run) 14
Rat Rotator cuff, tendinopathy model, transection + repair ↑ (1 wk)60
Achilles, partial transection ↑ (1 day) → (4 days)20
Achilles, transection + repair ↑ (3 days; 1, 2, 4 wks)18
Achilles, crush ↑ (1 day)21,22 → (3 days)22 ↑ (5 days)51 ↑ (1 wk)22
Achilles, collagenase ↑ (1, 2 wks)19
Achilles, exercise → (7 wks)27
Achilles, transection + exercise (post-operative day 5) ↑ (1, 3 hrs) → (12 hrs)28
↑ (1, 3 hrs) → (12 hrs)(IL-1RA)28
Achilles/patella, stress deprivation ↑ (2, 6 wks)31
Patella, cyclic exercise by surgery ↑ (high strain) ↓ (low strain)29 ↑ (high strain) ↓ (low strain)29
Flexor digitorum, exercise ↑ (training)26 →/↑ (3, 6, 8 wks)32-36 → (9 wks)36 →/↑ (12 wks)33,36-38 → (18, 24 wks)26
Rabbit Rotator cuff, partial transection (defect) ↑ (1, 3 days; 1 wk) → (3 wks)(descriptive)23 ↑ (1, 3 days; 1 wk) → (3 wks)23
Flexor digitorum superficialis, transection + repair ↑ (3, 6 days) → (12, 24 days)24
Flexor, electrical stimulation → (14 wks)30
Dog Flexor, transection + repair ↑ (1, 3, 9 days)25
Horse Flexor digitorum superficialis, tendinopathy ↑ (descriptive)61
IL-6 Human Rotator cuff, tear 4041 40
Rotator cuff/Achilles/patella/biceps/ECRB/flexor, tear ↑ (OSM)62 ↓ (IL-6R)62
Rotator cuff/posterior tibialis, tendinopathy 41,43 → (IL-6, OSM, LIF, IL-6R)42
Achilles, tear ↑ (IL-6, OSM, LIF)42 ↓ (IL-6R)42
Achilles, tear, post-operative (2 wks) *16
Achilles, tendinopathy 4243 ↓ (IL-6R)42 → (OSM, LIF)42 *17
Achilles, exercise (1 hr/36 km run) * (post-exercise 2-6 hrs; 1, 2 days)44,45
Patella, exercise (knee strenuous extension) → (post-exercise 1, 3 days)*63
Achilles, tendinopathy + exercise (1 hr run) 14
Rat Rotator cuff, exercise ↑ (4 wks)40
Achilles, transection + repair ↑ (3 days; 1, 2, 4 wks)18
Achilles, crush ↑(1 day)21
Achilles, collagenase ↑ (2 hrs)46 ↑ (1, 2 wks)19
Achilles, exercise → (7 wks)27
Flexor, exercise ↑ (training)26 → (3, 6 wks)36 ↑ (9 wks)36 →/↑(12 wks)36,38 → (18 wks)26 ↑ (24 wks)26
IL-10 Human Rotator cuff, tear 1341
Rotator cuff/Achilles, tendinopathy 41 ↑ (descriptive)*17
Achilles, tear, post-operative (2 wks) *16
Achilles, tendinopathy + exercise (1 hr run) 14
Rat Achilles, partial transection → (1, 4 days)20
Achilles, transection + repair → (3 days; 1, 2 wks) ↑ (4 wks)18
Achilles, crush → (1 day)21
Achilles, collagenase ↑ (2 hrs)46 → (1, 2 wks)48,49
Flexor digitorum, exercise → (training, 3, 6, 8, 9, 12, 18, 24 wks)26,32,34,36,38
TNFα Human Rotator cuff, tear 4041 ↓ (TNFR1)13
Rotator cuff/Achilles tendinopathy 41 →(TNFα, TNFR2) ↑ (TNFR1)50
Achilles, tear, post-operative (2 wks) *16
Achilles, tendinopathy + exercise (1 hr run) 14
Rat Rotator cuff, tendinopathy model, transection + repair → (1 wk)60
Rotator cuff, exercise ↑ (4 wks)40
Achilles, partial transection → (1 day) ↑ (4 days)20
Achilles, crush → (1 day)21 ↑ (5 days)51
Achilles, collagenase ↑ (2 hrs)46,64 ↑ (1 wk) → (2 wks)19
Patella, stress deprivation ↑ (2, 6 wks)31
Flexor, exercise ↑ (training)26,34 → (3, 6 wks)32,34,36 ↑ (8 wks)34,35 → (9 wks)36 ↓/→/↑ (12 wks)34,36-38 → (18 wks)26 ↑ (24 wks)26,34
Dog Flexor, transection + repair ↑ (1, 3, 9 days)25
Horse Superficial flexor digitorum, tendinopathy ↑ (acute) ↓ (chronic) (descriptive)65 ↑ (acute) ↓ (chronic) (descriptive) (TNFα, R1, TRAF2)65 ↑ (descriptive)61
  1. *

    studies that obtained clinical samples of tendon by microdialysis

  1. studies that sampled control tissues from the healthy region of the same tendon

  1. IL-1RA, interleukin-1 receptor antagonist; OSM, oncostatin M; IL-6R, interleukin 6 receptor; LIF, leukemia inhibitory factor; TNFR, tumour necrosis factor receptor; TRAF, TNF receptor-associated factor; ECRB, extensor carpi radialis brevis

Table II.

Cellular responses to treatment by cytokines in tendon cells. Arrows indicate increased (↑), unchanged (→), or decreased (↓) expression of cytokines in tissues of diseased human tendon, animal models of tendon injury or exercise versus healthy control tendon

Treatment Cells Increased, unchanged, decreased in response to treatment versus control
Gene Protein
IL-1β Human, various, normal Collagen I ↓ MMP1 ↑ MMP2 → MMP3 ↑ MMP13 ↑ TIMP1 → TIMP2→ ADAMTS-4 →66,67 MMP1 ↑ MMP3 ↑67
Human, not described, tendinopathy MMP1 ↑ MMP2 → MMP3 ↑68 MMP1 ↑ MMP2 ↑ MMP3 ↑68
Rat, patella, normal MMP13 ↑*39
Rat, patella, injured MMP13 ↑*39
Mouse, Achilles, injured Collagen I ↓ Collagen III ↓ Biglycan ↓ Decorin ↑ Fibromodulin ↓ Lumican → Aggrecan ↓ MMP13 ↑69
IL-6, IL-10 Human, various, normal Elastin → MMP1 →47 Collagen I →47
TNFα Human, various, normal Elastin ↑ MMP1 ↑47 Collagen I ↓47
Horse, FDS, normal Collagen I ↑ MMP9 → MMP13 ↓52
  1. *

    significant difference between tendon cells from normal and injured patellar tendons (p < 0.05)

  1. IL, interleukin; MMP, matrix metalloproteinase; TIMP, tissue inhibitor of metalloproteinase; ADAMTS, a disintegrin and metalloproteinase with thrombospondin motifs; TNF, tumour necrosis factor; FDS, flexor digitorum superficialis

IL-1β

In clinical samples, gene expression was decreased and protein expression increased in torn RC, but it remained unchanged in torn Achilles tendon tissues and after repair, exercise, or tendinopathy.13-17 The role of IL-1β in human tendon disease or after exercise could not be concluded. In animal injury models, gene and protein expression of IL-1β tended to increase at the early stages of tendon injury or healing until two weeks after the intervention.18-25 Similarly, exercise tended to increase the gene and protein expression of IL-1β in the early stages.26-38 Treatment of tendon cells with IL-1β showed catabolic effects such as increased expression of matrix metalloproteinase (MMPs), with a significant difference between cells from healthy and injured patellar tendons.39

IL-6

Gene and protein expression of IL-6 tended to be increased in RC and Achilles tendon tear patient samples,40-42 which continued until two weeks post-surgical repair,16 but not in patients with Achilles, RC, or posterior tibialis tendinopathy.17,41-43 Increased IL-6 protein expression was noted after prolonged running in healthy tendons,44,45 but not in tendinopathic Achilles tendons.14 In animal injury models, gene, and protein expression of IL-6 was increased from two hours to four weeks after the intervention.18,19,21,26,46 The effect of exercise was inconsistent in animal models.26,27,36,38,40 IL-6 treatment on tendon cells did not have any effect on the expression of tendon ECM genes and proteins.47

IL-10

The expression of IL-10 was inconsistent in clinical samples13,14,16,17,41 and animal injury models.18,20,21,46,48,49 No effects of exercise on the expression of IL-10 were noted in either humans or animals.26,32,34,36,38 IL-10 treatment on tendon cells did not have any effect on the expression of tendon ECM genes and proteins.47

TNF-α

Gene and protein expression of TNF-α tended not to change in clinical samples of RC and Achilles tendinopathy and tear patients.16,40,41,50 The effect of exercise on the expression of TNF-α in healthy human tendons has not been reported. In animal injury models, gene expression of TNF-α was elevated from two hours to nine days and declined at two weeks after the intervention,19,20,21,25,51 whereas the protein expression increased after four days. The effect of exercise on the gene and protein expression of TNF-α was inconclusive in animal models.26,31,32,34-38 The effects of TNF-α on tendon cells could not be concluded as genes of interest differed between studies.47,52

Discussion

This systematic review shows that IL-1β, IL-6, IL-10, and TNF-α are the most frequently investigated cytokines in the development and progression of tendon disease, during tendon healing and in response to exercise. Most studies focused on inflammatory cytokines based on previous literature, and only a few studies used hypothesis-free approaches to define the implicated cytokines.

Expression of IL-1β, IL-6, IL-10, and TNF-α differed depending on the stage of the tendon disease development, injury healing, and in response to exercise: IL-1β tended to increase in the early stage of tendon injury or exercise in animal models; IL-6 was suggested to increase at tendon tear, after prolonged exercise in healthy human tendons, and in the early stage of tendon injury in animal models; IL-10 remained unchanged in response to exercise in both humans and animals; and TNF-α tended to increase in the early stage of animal tendon injury models. Their functional mechanism in tendon disease development or healing could not be determined due to the small number of studies. The cellular response to IL-1β treatment significantly differed between injured and healthy tendon cells, but the involvement of other cytokines in tendon disease, healing, or after exercise could not be determined due to the paucity of studies or the inconsistency of the results thus far.

We were not able to identify the key cytokines through array studies alone due to the small number of studies, and due to the heterogeneity of clinical samples and animal models. An arbitrary number of more than ten studies was set to focus on the cytokines that had been investigated frequently in the literature. However, there was a wide variety of the anatomical locations of the diseased and control tendons, diagnostic criteria of tendinopathy, intervention, and the methods of gene or protein expression analysis in both human and animal studies. Tendons respond differently despite similar intervention based on anatomical location, function,53 and the content of exercises.37 Variances of the study limited the performing of a meta-analysis.

Schulze-Tanzil et al3 and Millar, Murrell, and McInnes54 have indicated through narrative reviews that multiple cytokines such as IL-1β, IL-4, IL-6, IL-13, IL-15, IL-17, IL-18, IL-21, IL-33, and TNF family members alongside TGF-β contribute to the development of tendon disease. By carrying out a systematic review, we captured all current literature and organised the evidence in human and animal studies separately. This largely supported the data presented in previous narrative reviews. Animal studies have been helpful in understanding tendon healing and the effect of exercise, but only represent limited features of the pathophysiology and clinical diseases.55 Our data on the four most investigated cytokines suggest that the expression of cytokines in diseased human tendons and animal models does not act in the same manner.

Numerous cytokines have been proposed to contribute to the development of tendon pathology, but there is a clear shortage of analyses on human-derived tendon tissue and cells. There is a risk of noting IL-1β, IL-6, IL-10, and TNFα as the key cytokines in tendon disease just for their frequent investigation. More work is warranted through individual interrogation to specify the cytokines that actually play prominent roles. Dakin et al41 indicated that advanced-stage disease tendon tissues from large to massive tears have a tissue inflammation signature characterised by the activation of signal transducer and activator of transcription 6, which is predominantly activated by cytokines such as IL-4. Millar et al,56,57 through mechanistic studies, have proposed IL-33 as an alarmin that triggers inflammation and IL-17A as an inflammatory modulator regulating cytokines, which contribute to the development and progress of tendon disease. There is clearly a strong inflammatory component in the development of tendon disease, with cytokines, which thus far have not been investigated frequently, potentially playing a substantial role. In the case of rheumatoid arthritis, TNF-α unexpectedly turned out to be the master regulator of the pro-inflammatory cytokines contributing to the disease.58 Moreover, it may also be important to consider interplay of numerous inflammatory processes with regard to different cell types, receptors, and biological and physical environments, and not just to focus on a limited number of molecules.

The cellular response of tendon cells to cytokines in the expression of tendon ECM genes differed depending on cell phenotype. A study by Tohyama et al39 suggested that infiltrating fibroblasts show significantly decreased expression of an ECM gene (MMP13) in response to IL-1β treatment when compared with the response of healthy tendon cells, and this comes in line with the studies by Dakin et al41,59 reporting that diseased tendon stromal cells may be primed for inflammation. Although the directions of the stimulatory or inhibitory effect of IL-1β in the gene and protein expression of collagens and MMPs were similar, it should be noted that the magnitude of the change may differ in cells isolated from diseased or healing tissues in comparison with healthy cells.59 Currently, only a few studies have focused on the mechanisms of action of the cytokines in cells derived from tissues of different phenotypes.

Determining both the temporal expression of cytokines during tendon disease progression and the mechanism of interaction of tendon cells with focus on cell phenotype is essential in order to improve our understanding of tendon disease pathophysiology and tendon healing. A systematic approach using well-defined clinical specimens to identify the cytokines that play a prominent role in the development of tendon disease is warranted. Further mechanistic studies on cytokine biology based on context of expression and surrounding inflammatory milieu should conduce identification of fundamental therapies to improve disease management through enhancing the quality of tissue repair or slowing disease progression.


A.J. Carr; email:
Author Contribution

W. Morita: Study concept and design, Data collection, Analysis, Interpretation of data, Drafting the manuscript.

S. G. Dakin: Analysis, Interpretation of data, Critical revision of manuscript.

S. J.B Snelling: Analysis, Interpretation of data, Critical revision of manuscript.

A. J. Carr: Supervision of study, Interpretation of data, Critical revision of manuscript.


  • Supplementary material

    Tables and figures showing the main characteristics of the papers included, results of assessment of study methodology quality, and summarised data of the expression of other cytokines in tendon tissues are available alongside the online version of this article at www.bjr.boneandjoint.org

  • Funding Statement

    The research was funded by the National Institute for Health Research (NIHR) Oxford Biomedical Research Centre (BRC). Dr Morita was funded by 28th term scholarship No. 274 of the INOAC International Education and Scholarship Foundation. Dr Dakin is a recipient of an Oxford UCB Prize Fellowship in Biomedical Research.

    We would like to thank Miss J. Mimpen and Dr M. Baldwin for critical feedback of the manuscript.

  • Conflict of Interest Statement

    None declared

  • References

    1 Forde MS , Punnett L , Wegman DH . Prevalence of musculoskeletal disorders in union ironworkers. J Occup Environ Hyg2005;2:203-212.CrossrefPubMed Google Scholar

    2 Dean BJ , Franklin SL , Carr AJ . A systematic review of the histological and molecular changes in rotator cuff disease. Bone Joint Res2012;1:158-166.CrossrefPubMed Google Scholar

    3 Schulze-Tanzil G , Al-Sadi O , Wiegand E , et al. The role of pro-inflammatory and immunoregulatory cytokines in tendon healing and rupture: new insights. Scand J Med Sci Sports2011;21:337-351.CrossrefPubMed Google Scholar

    4 Neer CS , Welsh RP . Shoulder in Sports. Orthop Clin North Am1977;8:583-591. Google Scholar

    5 Wang JH , Iosifidis MI , Fu FH . Biomechanical basis for tendinopathy. Clin Orthop Relat Res2006;443:320-332.CrossrefPubMed Google Scholar

    6 Rees JD , Stride M , Scott A . Tendons–time to revisit inflammation. Br J Sports Med2014;48:1553-1557. Google Scholar

    7 Millar NL , Dean BJ , Dakin SG . Inflammation and the continuum model: time to acknowledge the molecular era of tendinopathy. Br J Sports Med2016 (Epub ahead of print) PMID: 27259752.CrossrefPubMed Google Scholar

    8 Dean BJ , Gettings P , Dakin SG , Carr AJ . Are inflammatory cells increased in painful human tendinopathy? A systematic review. Br J Sports Med2016;50:216-220.CrossrefPubMed Google Scholar

    9 Sziksz E , Pap D , Lippai R , et al. Fibrosis related inflammatory mediators: role of the IL-10 cytokine family. Mediators Inflamm2015;2015:764641.CrossrefPubMed Google Scholar

    10 Medzhitov R . Origin and physiological roles of inflammation. Nature2008;454:428-435.CrossrefPubMed Google Scholar

    11 Wynn TA . Cellular and molecular mechanisms of fibrosis. J Pathol2008;214:199-210.CrossrefPubMed Google Scholar

    12 Morita W , Snelling SJ , Dakin SG , Carr AJ . Profibrotic mediators in tendon disease: a systematic review. Arthritis Res Ther2016;18:269.CrossrefPubMed Google Scholar

    13 Chaudhury S , Xia Z , Thakkar D , Hakimi O , Carr AJ . Gene expression profiles of changes underlying different-sized human rotator cuff tendon tears. J Shoulder Elbow Surg2016;25:1561-1570.CrossrefPubMed Google Scholar

    14 Pingel J , Fredberg U , Mikkelsen LR , et al. No inflammatory gene-expression response to acute exercise in human Achilles tendinopathy. Eur J Appl Physiol2013;113:2101-2109.CrossrefPubMed Google Scholar

    15 Gotoh M , Hamada K , Yamakawa H , et al. Significance of granulation tissue in torn supraspinatus insertions: an immunohistochemical study with antibodies against interleukin-1 beta, cathepsin D, and matrix metalloprotease-1. J Orthop Res1997;15:33-39.CrossrefPubMed Google Scholar

    16 Ackermann PW , Domeij-Arverud E , Leclerc P , Amoudrouz P , Nader GA . Anti-inflammatory cytokine profile in early human tendon repair. Knee Surg Sports Traumatol Arthrosc2013;21:1801-1806.CrossrefPubMed Google Scholar

    17 Waugh CM , Morrissey D , Jones E , et al. In vivo biological response to extracorporeal shockwave therapy in human tendinopathy. Eur Cell Mater2015;29:268-280.CrossrefPubMed Google Scholar

    18 Sugg KB , Lubardic J , Gumucio JP , Mendias CL . Changes in macrophage phenotype and induction of epithelial-to-mesenchymal transition genes following acute Achilles tenotomy and repair. J Orthop Res2014;32:944-951.CrossrefPubMed Google Scholar

    19 Pires D , Xavier M , Araujo T , et al. Low-level laser therapy (LLLT; 780 nm) acts differently on mRNA expression of anti- and pro-inflammatory mediators in an experimental model of collagenase-induced tendinitis in rat. Lasers Med Sci2011;26:85-94. Google Scholar

    20 Da Ré Guerra F , Vieira CP , Oliveira LP , et al. Low-level laser therapy modulates pro-inflammatory cytokines after partial tenotomy. Lasers Med Sci2016;31:759-766.CrossrefPubMed Google Scholar

    21 Haslerud S , Lopes-Martins RA , Frigo L , et al. Low-level laser therapy and cryotherapy as mono- and adjunctive therapies for Achilles tendinopathy in rats. Photomed Laser Surg2017;35:32-42.CrossrefPubMed Google Scholar

    22 de Jesus JF , Spadacci-Morena DD , dos Anjos , Rabelo ND , et al. Low-level laser therapy in IL-1β, COX-2, and PGE2 modulation in partially injured Achilles tendon. Lasers Med Sci2015;30:153-158.CrossrefPubMed Google Scholar

    23 Koshima H , Kondo S , Mishima S , et al. Expression of interleukin-1beta, cyclooxygenase-2, and prostaglandin E2 in a rotator cuff tear in rabbits. J Orthop Res2007;25:92-97.CrossrefPubMed Google Scholar

    24 Berglund M , Hart DA , Wiig M . The inflammatory response and hyaluronan synthases in the rabbit flexor tendon and tendon sheath following injury. J Hand Surg Eur Vol2007;32:581-587.CrossrefPubMed Google Scholar

    25 Manning CN , Havlioglu N , Knutsen E , et al. The early inflammatory response after flexor tendon healing: a gene expression and histological analysis. J Orthop Res2014;32:645-652.CrossrefPubMed Google Scholar

    26 Gao HG , Fisher PW , Lambi AG , et al. Increased serum and musculotendinous fibrogenic proteins following persistent low-grade inflammation in a rat model of long-term upper extremity overuse. PLoS One2013;8:e71875.CrossrefPubMed Google Scholar

    27 Pingel J , Wienecke J , Kongsgaard M , et al. Increased mast cell numbers in a calcaneal tendon overuse model. Scand J Med Sci Sports2013;23:e353-e360.CrossrefPubMed Google Scholar

    28 Eliasson P , Andersson T , Aspenberg P . Influence of a single loading episode on gene expression in healing rat Achilles tendons. J Appl Physiol (1985)2012;112:279-288.CrossrefPubMed Google Scholar

    29 Sun HB , Li Y , Fung DT , et al. Coordinate regulation of IL-1beta and MMP-13 in rat tendons following subrupture fatigue damage. Clin Orthop Relat Res2008;466:1555-1561.CrossrefPubMed Google Scholar

    30 Asundi KR , King KB , Rempel DM . Evaluation of gene expression through qRT-PCR in cyclically loaded tendons: an in vivo model. Eur J Appl Physiol2008;102:265-270.CrossrefPubMed Google Scholar

    31 Uchida H , Tohyama H , Nagashima K , et al. Stress deprivation simultaneously induces over-expression of interleukin-1beta, tumor necrosis factor-alpha, and transforming growth factor-beta in fibroblasts and mechanical deterioration of the tissue in the patellar tendon. J Biomech2005;38:791-798.CrossrefPubMed Google Scholar

    32 Frara N , Abdelmagid SM , Tytell M , et al. Growth and repair factors, osteoactivin, matrix metalloproteinase and heat shock protein 72, increase with resolution of inflammation in musculotendinous tissues in a rat model of repetitive grasping. BMC Musculoskelet Disord2016;17:34.CrossrefPubMed Google Scholar

    33 Fedorczyk JM , Barr AE , Rani S , et al. Exposure-dependent increases in IL-1beta, substance P, CTGF, and tendinosis in flexor digitorum tendons with upper extremity repetitive strain injury. J Orthop Res2010;28:298-307.CrossrefPubMed Google Scholar

    34 Barbe MF , Elliott MB , Abdelmagid SM , et al. Serum and tissue cytokines and chemokines increase with repetitive upper extremity tasks. J Orthop Res2008;26:1320-1326.CrossrefPubMed Google Scholar

    35 Coq JO , Barr AE , Strata F , et al. Peripheral and central changes combine to induce motor behavioral deficits in a moderate repetition task. Exp Neurol2009;220:234-245.CrossrefPubMed Google Scholar

    36 Kietrys DM , Barr-Gillespie AE , Amin M , et al. Aging contributes to inflammation in upper extremity tendons and declines in forelimb agility in a rat model of upper extremity overuse. PLoS One2012;7:e46954.CrossrefPubMed Google Scholar

    37 Barbe MF , Gallagher S , Massicotte VS , et al. The interaction of force and repetition on musculoskeletal and neural tissue responses and sensorimotor behavior in a rat model of work-related musculoskeletal disorders. BMC Musculoskelet Disord2013;14:303.CrossrefPubMed Google Scholar

    38 Elliott MB , Barr AE , Clark BD , et al. High force reaching task induces widespread inflammation, increased spinal cord neurochemicals and neuropathic pain. Neuroscience2009;158:922-931.CrossrefPubMed Google Scholar

    39 Tohyama H , Yasuda K , Uchida H , Nishihira J . The responses of extrinsic fibroblasts infiltrating the devitalised patellar tendon to IL-1beta are different from those of normal tendon fibroblasts. J Bone Joint Surg [Br]2007;89-B:1261-1267.CrossrefPubMed Google Scholar

    40 Millar NL , Wei AQ , Molloy TJ , Bonar F , Murrell GA . Cytokines and apoptosis in supraspinatus tendinopathy. J Bone Joint Surg [Br]2009;91-B:417-424.CrossrefPubMed Google Scholar

    41 Dakin SG , Martinez FO , Yapp C , et al. Inflammation activation and resolution in human tendon disease. Sci Transl Med2015;7:311ra173.CrossrefPubMed Google Scholar

    42 Legerlotz K , Jones ER , Screen HR , Riley GP . Increased expression of IL-6 family members in tendon pathology. Rheumatology (Oxford)2012;51:1161-1165.CrossrefPubMed Google Scholar

    43 Courneya JP , Luzina IG , Zeller CB , et al. Interleukins 4 and 13 modulate gene expression and promote proliferation of primary human tenocytes. Fibrogenesis Tissue Repair2010;3:9.CrossrefPubMed Google Scholar

    44 Gump BS , McMullan DR , Cauthon DJ , et al. Short-term acetaminophen consumption enhances the exercise-induced increase in Achilles peritendinous IL-6 in humans. J Appl Physiol (1985)2013;115:929-936.CrossrefPubMed Google Scholar

    45 Langberg H , Olesen JL , Gemmer C , Kjaer M . Substantial elevation of interleukin-6 concentration in peritendinous tissue, in contrast to muscle, following prolonged exercise in humans. J Physiol2002;542:985-990.CrossrefPubMed Google Scholar

    46 Torres-Silva R , Lopes-Martins RA , Bjordal JM , et al. The low level laser therapy (LLLT) operating in 660 nm reduce gene expression of inflammatory mediators in the experimental model of collagenase-induced rat tendinitis. Lasers Med Sci2015;30:1985-1990. Google Scholar

    47 John T , Lodka D , Kohl B , et al. Effect of pro-inflammatory and immunoregulatory cytokines on human tenocytes. J Orthop Res2010;28:1071-1077.CrossrefPubMed Google Scholar

    48 Xavier M , de Souza RA , Pires VA , et al. Low-level light-emitting diode therapy increases mRNA expressions of IL-10 and type I and III collagens on Achilles tendinitis in rats. Lasers Med Sci2014;29:85-90.CrossrefPubMed Google Scholar

    49 Casalechi HL , Leal-Junior EC , Xavier M , et al. Low-level laser therapy in experimental model of collagenase-induced tendinitis in rats: effects in acute and chronic inflammatory phases. Lasers Med Sci2013;28:989-995.CrossrefPubMed Google Scholar

    50 Gaida JE , Bagge J , Purdam C , et al. Evidence of the TNF-alpha system in the human Achilles tendon: expression of TNF-alpha and TNF receptor at both protein and mRNA levels in the tenocytes. Cells Tissues Organs2012;196:339-352. Google Scholar

    51 Dohnert MB , Venâncio M , Possato JC , et al. Gold nanoparticles and diclofenac diethylammonium administered by iontophoresis reduce inflammatory cytokines expression in Achilles tendinitis. Int J Nanomedicine2012;7:1651-1657.CrossrefPubMed Google Scholar

    52 Hosaka YZ , Uratsuji T , Ueda H , Uehara M , Takehana K . Comparative study of the properties of tendinocytes derived from three different sites in the equine superficial digital flexor tendon. Biomed Res2010;31:35-44.CrossrefPubMed Google Scholar

    53 Davies MR , Ravishankar B , Laron D , et al. Rat rotator cuff muscle responds differently from hindlimb muscle to a combined tendon-nerve injury. J Orthop Res2015;33:1046-1053.CrossrefPubMed Google Scholar

    54 Millar NL , Murrell GA , McInnes IB . Inflammatory mechanisms in tendinopathy - towards translation. Nat Rev Rheumatol2017;13:110-122.CrossrefPubMed Google Scholar

    55 Dirks RC , Warden SJ . Models for the study of tendinopathy. J Musculoskelet Neuronal Interact2011;11:141-149.PubMed Google Scholar

    56 Millar NL , Akbar M , Campbell AL , et al. IL-17A mediates inflammatory and tissue remodelling events in early human tendinopathy. Sci Rep2016;6:27149.CrossrefPubMed Google Scholar

    57 Millar NL , Gilchrist DS , Akbar M , et al. MicroRNA29a regulates IL-33-mediated tissue remodelling in tendon disease. Nat Commun2015;6:6774.CrossrefPubMed Google Scholar

    58 Feldmann M , Maini RN . Lasker Clinical Medical Research Award. TNF defined as a therapeutic target for rheumatoid arthritis and other autoimmune diseases. Nat Med2003;9:1245-1250.CrossrefPubMed Google Scholar

    59 Dakin SG , Buckley CD , Al-Mossawi MH , et al. Persistent stromal fibroblast activation is present in chronic tendinopathy. Arthritis Res Ther2017;19:16.CrossrefPubMed Google Scholar

    60 Tucker JJ , Riggin CN , Connizzo BK , et al. Effect of overuse-induced tendinopathy on tendon healing in a rat supraspinatus repair model. J Orthop Res2016;34:161-166.CrossrefPubMed Google Scholar

    61 Hosaka Y , Kirisawa R , Yamamoto E , et al. Localization of cytokines in tendinocytes of the superficial digital flexor tendon in the horse. J Vet Med Sci2002;64:945-947.CrossrefPubMed Google Scholar

    62 Jelinsky SA , Rodeo SA , Li J , et al. Regulation of gene expression in human tendinopathy. BMC Musculoskelet Disord2011;12:86.CrossrefPubMed Google Scholar

    63 Pingel J , Langberg H , Skovgard D , et al. Effects of transdermal estrogen on collagen turnover at rest and in response to exercise in postmenopausal women. J Appl Physiol (1985)2012;113:1040-1047.CrossrefPubMed Google Scholar

    64 Marcos RL , Leal-Junior EC , Arnold G , et al. Low-level laser therapy in collagenase-induced Achilles tendinitis in rats: analyses of biochemical and biomechanical aspects. J Orthop Res2012;30:1945-1951.CrossrefPubMed Google Scholar

    65 Hosaka Y , Kirisawa R , Ueda H , Yamaguchi M , Takehana K . Differences in tumor necrosis factor (TNF)alpha and TNF receptor-1-mediated intracellular signaling factors in normal, inflamed and scar-formed horse tendons. J Vet Med Sci2005;67:985-991.CrossrefPubMed Google Scholar

    66 Thampatty BP , Li H , Im HJ , Wang JH . EP4 receptor regulates collagen type-I, MMP-1, and MMP-3 gene expression in human tendon fibroblasts in response to IL-1 beta treatment. Gene2007;386:154-161.CrossrefPubMed Google Scholar

    67 Tsuzaki M , Guyton G , Garrett W , et al. IL-1 beta induces COX2, MMP-1, -3 and -13, ADAMTS-4, IL-1 beta and IL-6 in human tendon cells. J Orthop Res2003;21:256-264.CrossrefPubMed Google Scholar

    68 Corps AN , Harrall RL , Curry VA , et al. Ciprofloxacin enhances the stimulation of matrix metalloproteinase 3 expression by interleukin-1beta in human tendon-derived cells. A potential mechanism of fluoroquinolone-induced tendinopathy. Arthritis Rheum2002;46:3034-3040.CrossrefPubMed Google Scholar

    69 Zhang K , Asai S , Yu B , Enomoto-Iwamoto M . IL-1β irreversibly inhibits tenogenic differentiation and alters metabolism in injured tendon-derived progenitor cells in vitro. Biochem Biophys Res Commun2015;463:667-672. Google Scholar

    70 Campbell AL , Smith NC , Reilly JH , et al. IL-21 receptor expression in human tendinopathy. Mediators Inflamm2014;2014:481206.CrossrefPubMed Google Scholar

    71 Attia M , Huet E , Gossard C , et al. Early events of overused supraspinatus tendons involve matrix metalloproteinases and EMMPRIN/CD147 in the absence of inflammation. Am J Sports Med2013;41:908-917.CrossrefPubMed Google Scholar

    72 Leumann A , Longino D , Fortuna R , et al. Altered cell metabolism in tissues of the knee joint in a rabbit model of Botulinum toxin A-induced quadriceps muscle weakness. Scand J Med Sci Sports2012;22:776-782.CrossrefPubMed Google Scholar

    73 Wang W , Tang X , Zhang J , Yan X , Ma Y . Complete stress shielding of the Achilles tendon: ultrastructure and level of interleukin-1 and TGF-beta. Orthopedics2010;33:810. Google Scholar