Next Article in Journal
Learning from the Community to Predict Nutrition Status of Children Aged 6–24 Months in Gulu District, Northern Uganda: A Case Control Study
Previous Article in Journal
Primary and Secondary Progressive Aphasia in Posterior Cortical Atrophy
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Tendon and Ligament Genetics: How Do They Contribute to Disease and Injury? A Narrative Review

by
William J. Ribbans
1,2,*,
Alison V. September
3,4,5 and
Malcolm Collins
3,4,5
1
School of Health, The University of Northampton, Northampton NN1 5PH, UK
2
The County Clinic, Northampton NN1 5DB, UK
3
Division of Physiological Sciences, Department of Human Biology, Health Sciences Faculty, University of Cape Town, Cape Town 7700, South Africa
4
Health Through Physical Activity, Lifestyle and Sport Research Centre (HPALS), Department of Human Biology, Health Sciences Faculty, University of Cape Town, Cape Town 7700, South Africa
5
International Federation of Sports Medicine (FIMS), Collaborative Centre of Sports Medicine, Department of Human Biology, University of Cape Town, Cape Town 7700, South Africa
*
Author to whom correspondence should be addressed.
Life 2022, 12(5), 663; https://doi.org/10.3390/life12050663
Submission received: 27 March 2022 / Revised: 24 April 2022 / Accepted: 26 April 2022 / Published: 29 April 2022
(This article belongs to the Section Genetics and Genomics)

Abstract

:
A significant proportion of patients requiring musculoskeletal management present with tendon and ligament pathology. Our understanding of the intrinsic and extrinsic mechanisms that lead to such disabilities is increasing. However, the complexity underpinning these interactive multifactorial elements is still not fully characterised. Evidence highlighting the genetic components, either reducing or increasing susceptibility to injury, is increasing. This review examines the present understanding of the role genetic variations contribute to tendon and ligament injury risk. It examines the different elements of tendon and ligament structure and considers our knowledge of genetic influence on form, function, ability to withstand load, and undertake repair or regeneration. The role of epigenetic factors in modifying gene expression in these structures is also explored. It considers the challenges to interpreting present knowledge, the requirements, and likely pathways for future research, and whether such information has reached the point of clinical utility.

1. Introduction

It has been estimated that tendon and ligament injuries account for 30–50% of all sporting injuries [1]. The impact on people’s overall well-being, ability to work, and participate in exercise is significant. The health economic burden of managing such conditions is huge [2,3]. Is the prevalence of such injuries simply related to increased societal involvement in sport increasing exposure to extrinsic risk factors, such as running or other high impact activities? Alternatively, has the more sedentary lifestyle of many, combined with increasing levels of obesity, simply led to deconditioning and increased loading causing failure of these critical structures? How much does our inherited genotype influence our risk profile? This review considers the impact of genetic factors on the development of tendon and ligament injury and disease. What is our present state of knowledge of tendon and ligament structure and function? What do we understand about the influence of genetic factors and how might this knowledge be used in the future to reduce risk?

2. The Injury Causation Model and Jar Model

Tendon and ligament pathologies usually follow the injury causation model described by Meeuwisse in 1994 [4] and refined by Bittencourt in 2016 [5]. A complex interaction of intrinsic (including genetic factors) and extrinsic factors influences an individual’s specific profile along the ‘reduced to increased risk (predisposed) spectrum’. Following an inciting episode, such as an injurious event, the problem becomes symptomatic to the patient (Figure 1). An injury may manifest itself as an acute episode or chronic condition. For the latter, there is often initial damage that is asymptomatic. Further subsequent inciting episodes, which can be identified by the patient, causes the damage to become symptomatic [6].
People inherit genetic factors which can result in an increased or reduced risk for many potential tendon and ligament injuries. However, the possession of such factors does not necessarily lead to the clinical development of such multifactorial conditions. Austin’s Jar Model for genetic risk was developed for use in psychiatric counselling [7] and applied to other areas of medicine, including cancers. It can be used in discussing risk for developing musculoskeletal damage (Figure 2).
Extrinsic factors can be overwhelming, leading to injury such as in a tackle-playing sport causing ligament injury. However, for many instances of damage (usually sub-acute or chronic), both genetic and non-genetic intrinsic elements combine with extrinsic or environmental factors. Once the jar has ‘overflowed’, the damage becomes apparent. Careful review of modifiable risk factors (such as flexibility and body composition variables) combined with attempts to reduce their influence and the adoption of protective factors (such as improved nutrition and rest intervals) can improve tendon and ligament health. Medications such as steroids and certain fluoroquinolone antibiotics (examples include ciprofloxacin), can initiate tendon damage that can be ameliorated following their cessation [8,9].

3. Genetic Involvement in Musculoskeletal Condition

Most intrinsic risk factors for common tendon and ligament injuries have a genetic contribution [10]. For instance, flexibility has a heritability component estimated to between 64 and 70% [11,12]. Familial studies have reported a 40% heritability between twins for lateral epicondylitis [13], a five times increased risk for rotator cuff issues in siblings [14], and twin studies describing a knee anterior cruciate ligament (ACL) tear risk heritability of 69% [15].
Some rare orthopaedic conditions, such as pseudoachondroplasia and osteogenesis imperfecta, are caused by gene mutations [16,17]. However, most sports injuries are caused by extrinsic factors (such as training load) interacting with an individual’s genetic background and other intrinsic factors [18]. Additionally, some medical conditions with a genetic background increase the risk of tendon pathology, including various seropositive and seronegative rheumatological conditions (such as gout and ankylosing spondylitis), Ehlers–Danlos syndrome, and other endocrine and metabolic disorders [19].
We note a spectrum of genetic-related connective tissue disorders. At one extreme are conditions associated with a classical Mendelian inheritance pattern—such as osteogenesis imperfecta [20], Ehlers–Danlos syndrome [21] and Marfan’s syndrome [22]. Establishing the genetic pattern of these conditions usually involves techniques such as linkage analysis or direct gene sequencing [23]. At the other end of the spectrum are conditions with a multifactorial aetiology often involving multiple and complex interactions between various genes with non-genetic intrinsic and extrinsic factors. These include gene–gene and gene with non-genetic factor interactions, making the investigation of the genetic contribution to such conditions more complicated.

4. Types of Genetic Studies

Depending on the prevalence of the phenotype and, therefore, the sample size, several study designs can be applied to investigate genetic influences on the development of multifactorial phenotypes such as tendon and ligament injuries.

4.1. Family Studies

The most basic of these formats are family studies. Disease inheritance patterns between family members indicate the degree of hereditability of such conditions. Examples of autosomal dominant (e.g., Huntington’s disease), autosomal recessive (e.g., phenylketonuria), and sex-linked related (e.g., Vitamin D resistant rickets with hypophosphatemia) are readily identified. However, the interactions of many factors (including genetic and non-genetic factors), such as a shared environmental load, influence the susceptibility to complex multifactorial clinical conditions such as tendon and ligament injuries. Therefore, identifying several members of a family with an identical injury is not common and, for this reason, a classical inheritance pattern is usually not identifiable. Twin studies represent a unique opportunity to identify the shared heritability component between twins together with a potential shared environmental exposure.

4.2. Case-Control Studies

Case-control genetic association studies allow interrogation and comparison of large data sets both within a population and between populations of different geographical locations and ancestry. This has been a popular method of identifying genetic risk factors of common multifactorial phenotypes including tendon and ligament pathologies [24]. However, it requires rigour in the phenotyping of cases and controls. Cases should be well defined, and diagnoses confirmed using preferably ‘gold standard’ methodology, such as imaging or surgical confirmation. Additionally, the multifactorial nature of the susceptibility, a comprehensive medical history, sporting history (including training regimes), medicines’ use, and familial injury history should be recorded for both cases and controls.
Controls’ selection is equally important, and individuals should be matched for sex, age, body mass index (BMI), and sports participation exposure and level, and any other potential confounders. Both the cases and controls may harbour genetic and non-genetic elements, which may confer an increased or decreased risk to sustaining a potential tendon or ligament injury. It is the balance of these multifactorial risk factors which will determine if the injury presents.

4.3. Hypothesis-Free Approach

Almost all past and current research has focused on the candidate gene approach where knowledge of a given gene and the injury is assumed (Section 4.2 above). There has been progress towards a hypothesis-free approach using the application of next generation sequencing technologies, such as genome wide association studies (GWAS). Most GWAS studies have used canine models [25,26,27] although one human study highlighted three independent DNA sequence variants associated with ACL rupture—albeit of borderline significance [28]. A whole exome sequencing (WES) approach was also recently undertaken in a twin family study, where 11 novel variants were highlighted for further exploration in ACL injury susceptibility [29]. Recently, Gibbons used a hybrid approach of WES on targeted participants and applied a tiered filtering strategy to identify potentially biologically relevant new candidate-variants within previously implicated genes. This allowed further prioritisation in larger independent cohorts [30]. Currently, there are no whole genome sequencing datasets specific to exercise-related injury phenotypes, such as tendon and ligament injuries. In the future, genetic susceptibility would gain from research characterising the genome in relation to tendon and ligament injury.

5. Tendon: Structure, Function and Genetic Research

Tendons transfer the forces generated within muscles to their bone insertions. Tendons are composed of a heterogeneous population of tendon cells embedded within an extracellular matrix (ECM) consisting of collagen fibres, elastin fibres, proteoglycans, glycosaminoglycans, and glycoproteins.

5.1. Tendon Cells

Ninety to 95% of the cellular population within the mature tendon, which synthesise and regulate the components of the ECM, consist predominately of tenocytes and immature tenoblasts [31]. Tenocytes are predominately found within the fascicles between the collagen fibres and have an intricate network of connections using processes producing intercellular links via gap junctions [32]. The more rounded and metabolically active tenoblasts are primarily situated between fascicle units within the inter-fascicular matrix (IFM) [33,34]. Tendon cell activity alters with exposure to normal stresses, injury, and ageing. One to 4% of the cells within tendons are tendon stem/progenitor cells (TSPCs) which have similar characteristics to mesenchymal stem cells [35].

5.2. Collagen

Within the ECM, collagen is the major structural protein and constitutes 60–85% of the dry tendon weight. The collagen is arranged in a hierarchical manner [36] (Figure 3).

5.2.1. Type I Collagen

Type I collagen accounts for up to 90% of total collagen content. Type I collagen fibrils are the tendon’s primary structural elements. They provide tensile strength enhanced by cross-linking. Fibrils aggregate to fibres and, once again, fibres combine to form fascicles. Each fascicle is surrounded by an endotenon or IFM. Fascicles combine to form the tendon entity, which is bound together by a surrounding epitenon. A paratenon surrounds many tendons, such as the Achilles (Table 1).
Two studies, a South African cohort of Achilles tendon disease and a Turkish cohort of lateral epicondylitis of the elbow, investigated the COL1A1 rs1800012 functional DNA sequence variant with tendon disease and neither found an association [47,48]. However, later sub-analysis of acute Achilles tendon rupture in the South African cohort found that possession of a relatively rare TT genotype protects against injury [49]. More recently Gibbon [50] explored the COL1A1 rs1007946-rs1800012 haplotype and implicated G-T to be associated with reduced risk of Achilles tendinopathy and rupture (Table 2). Other collagen types, although in a minority in terms of mass, play an important part in determining collagen configuration, strength, and optimal function.

5.2.2. Type III Collagen

Type III collagen, encoded by COL3A1, is the second most common type comprising up to 10% of total collagen (Table 1). To date, no studies have investigated the association of COL3A1 variants with tendon injuries (although they have been undertaken in ligament injuries—see Section 6.1.3).

5.2.3. Type V Collagen

Type V collagen performs a similar role to Type III collagen. COL5A1 encodes for its α1 chain and is found close to the ABO gene on chromosome 9q34 (Table 1). A South African cohort study described four variants within the 3’-untranslated region (3’-UTR), which were independently associated with the development of chronic Achilles’ tendinopathy (AT). Additionally, a protective genotype for a single variant was identified [51]. The variants located within the 3’-UTR appear to influence the stability of the mRNA. This region appears to play an important role in post-transcriptional regulation [52,53]. The authors hypothesise that the C and T alleles of rs12722 influences COL5A1 mRNA stability and possibly through alternate mRNA structural forms, which may affect type V collagen expression. They further hypothesise that the mRNA stability is enhanced with the functional form associated with increased chronic AT, leading to increased type V collagen [54]. Collins further postulated on how increased type V collagen content was associated with altered mechanical properties of tendons and increased injury susceptibility [55]. A later study reported other genotypes, also within the functional COL5A1 3’-UTR, in South African and Australian populations that showed increased risk for chronic AT [56]. More recently, RNA sequencing analyses in torn rotator cuff tissue vs. control tissue, showed that COL5A1 gene expression was markedly (3.01×) increased in tears, demonstrating its role in healing and/or remodelling [57].
The effect of altered genotypes in the MIR608 gene, which encodes a small micro-RNA (miRNA), was undertaken in Australian and South African groups [58]. An increased risk of AT was demonstrated with the rs4919510 CC genotype. This miRNA can bind to a recognition sequence within the COL5A1 and other genes 3’-UTR and inhibits translation. It was the first non-coding gene to be associated with soft-tissue injuries and increased understanding of the complex mechanisms involved in the regulation of type V collagen production. A GWAS study investigating a large cohort, including samples of different ancestry, suggested that MIR608 rs4919510 showed moderate evidence for AT susceptibility—although not at the level of significance required for a GWAS study [29] (Table 2).
Table 2. Summary of genetic research and tendon injuries: Collagen types I and V.
Table 2. Summary of genetic research and tendon injuries: Collagen types I and V.
Collagen
Type
ReferenceCountryVariantPathologySubjectsControlsComments
Type I[46]South AfricaCOL1A1 rs1800012Achilles’ tendinopathy and rupture126125No significant association
[47]TurkeyCOL1A1 rs1800012Lateral epicondylitis (common extensor origin)183123No significant association
[58]SpainCOL1A1 rs1800012Patellar tendon injury150No difference between severity of injury and gene variants. No controls were included
[48]South Africa and Sweden (controls only)COL1A1 rs1800012Achilles’ tendon rupture41581Significant association of the TT genotype protecting against acute Achilles’ tendon rupture
[49]South Africa and UKCOL1A1 rs1107946
rs1800012
Achilles’ tendinopathy and rupture216193Significant association of the (G-T) haplotype protecting against Achilles tendinopathy and rupture
Type V[50]South AfricaCOL5A1 rs12722Achilles’ tendinopathy and rupture111129Significant protective association with variant in tendon disease
[59]ItalyCOL5A1 rs12722Bilateral quadriceps rupture90No controls but association with a polymorphism and tendon rupture
[60]ItalyCOL5A1 rs12722Bilateral quadriceps rupture10No controls but association with a polymorphism and tendon rupture (same finding as Galasso)
[58]SpainCOL5A1 rs12722Patellar tendon injury150No difference between severity of injury and gene variants. No controls were included
[55]South Africa, AustraliaCOL5A1 rs12722
COL5A1 rs3196378
Achilles’ tendinopathy and rupture178342Significant protective association with variant of COL5A1 rs12722 in tendon disease
In Australian cohort only, rs31996378 associated with tendinopathy
[57]South Africa, AustraliaCOL5A1 3’-UTR rs71746744, rs1134170, rs16399
MIR608 rs4919510
Achilles’ tendinopathy and rupture160342All variants within the untranslated region of the COL5A1 gene and MIR variant were associated with Achilles tendon disease
Green indicates an association and red indicates no association found.

5.2.4. Type XI Collagen

Type XI collagen is found in many structures including articular cartilage, bone, and muscle (Table 1). Variants in COL11A1 and COL11A2 interact with one another and with a COL5A1 3’-UTR variant to modulate the AT risk in South African and Australian cohorts [43] (Table 3).

5.2.5. Types XII, XIV, and XXVII Collagens

None of the investigated functional variants of COL12A1 or COL14A1 which encode the α-chains of types XII and XIV collagen have been associated with AT [59]. However, COL12A1 may be associated with acute Achilles tendon ruptures [59]. Saunders investigated several COL27A1 gene variants that encode Type XXVII collagen but could not identify any independent association with AT [60] (Table 1 and Table 3).

5.3. Proteoglycans

Proteoglycans (PGs) represent up to 5% of tendon dry weight. They consist of a core protein attached to one or more glycosaminoglycans (GAGs). The GAG’s negative charge binds water, which makes up 55–70% of the total tendon weight. PGs are found between the ECM’s collagenous structural components (Table 4). There are no reported genetic studies associated with tendon PG. However, ligament PG genetic variants have been investigated (see Section 6.2.2).

5.4. Glycoproteins

Glycoproteins are a large family of structurally and functionally diverse proteins to which a carbohydrate group(s) is covalently attached. Two important tendinous glycoproteins are tenascin-C and cartilage oligomeric matrix protein (COMP) (Table 4).
Table 3. Summary of genetic research and tendon injuries: Collagen types XI, XII, XIV, and XXVII.
Table 3. Summary of genetic research and tendon injuries: Collagen types XI, XII, XIV, and XXVII.
Collagen TypesReferenceCountryVariantPathologySubjectsControlsComments
Type XI[43]South Africa, AustraliaCOL11A1 rs1676486,
rs3753841
COL11A2 rs1799907
Achilles’ tendinopathy and rupture184338No significant independent associations found
Genes encoding structural and functionally related type XI (COL11A1 and COL11A2) and type V (COL5A1) collagens interact with one another to collectively modulate Achilles’ tendinopathy risk
Type XII[59]South Africa, AustraliaCOL12A1 rs240736, rs970547Achilles’ tendinopathy and rupture137131No significant associations found
There may be an association with Achilles tendon rupture
Type XIV[59]South Africa, AustraliaCOL14A1 rs4870723, rs1563392Achilles’ tendinopathy and rupture137131No significant associations found
Type XXVII[60]South Africa,
Australia
COL27A1 (several polymorphisms)Achilles’ tendinopathy and rupture178340No significant associations found among variants
Green indicates an association and red indicates no association found.
Table 4. Regulatory and structural components of tendons.
Table 4. Regulatory and structural components of tendons.
ComponentForm and TypesFunctionsAssociated Diseases
Proteoglycans (PGs)PGs
  • Found between extra-cellular matrix’s (ECM) collagenous structural components
  • PGs include Decorin, Lubricin, and Versican
  • Consist of a core protein attached to one or more glycosaminoglycans (GAGs)
  • Decorin (80%): roles include collagen fibrillogenesis and potentially enhancing strength by facilitating load transfer between discontinuous collagen fibrils. Most abundant in tendinous areas most subject to tension
  • Lubricin: Found close to tendon periphery and aids tendon gliding with lubrication
  • Versican: Found within the interfascicular matrix (IFM)
GlycoproteinsTenascin-C (TNC)
  • Protein is a hexamere, which binds to both ECM components and tenocyte surface receptors
  • Produced by TNC gene located close to the ABO gene on chromosome 9q34
  • TNC gene expression appears influenced by tension loading [61]
  • Important role in regulating the tenocytes need to interact with ECM components
  • Involved in cell adhesion and signalling, affecting cell proliferation and migration [62,63,64]
  • Appears to increase in the presence of tendon pathology [65]
  • Mutations within TNX gene (a tenascin family member) causes an autosomal recessive form of EDS from tenascin-X deficiency [66]
Thrombospondins (THBS)
  • Family of glycoproteins performing dynamic role within ECM
Cartilage oligomeric matrix protein (COMP)
  • Also known as thrombospondin V
  • Most abundant tendinous glycoprotein
  • Links to type I collagen and has five subunits allowing linkage between multiple fibrils
  • Found within the inter-fibrillar matrix [67] and tissues subject to significant loading, such as tendons, ligaments, cartilage, menisci, and intervertebral discs
Thrombospondin 2 (THBS2)
  • Mediates cell-to-cell and cell-to-matrix interactions
  • Involved in cell-to-cell adhesion and ECM communication
Thrombospondin type 1 domain–containing protein 7A
  • This protein regulates focal adhesions during angiogenic cell migration
  • Role debated
  • COMP-null knock out mice did not demonstrate any tendon abnormality [68]
  • Others reported in horses that COMP levels during growth corelate with mechanical strength at skeletal maturity [66]
  • Pseudo-achondroplasia is caused by a heterozygous mutation in the gene encoding COMP [16]
  • A COMP gene mutation can cause multiple epiphyseal dysplasia (MED)
Elastin and MicrofibrilsElastin (ELN)
  • Elastin provides elasticity to tendons, allowing them to stretch and return to their original state. Plays important load-bearing role in tendons and ligaments.
ELN rs2071307 variant has been associated with other multifactorial ECM conditions, such as aortic stenosis [69] and aortic aneurysm [70]
ELN rs2071307 and FBN2 rs331079 variants associate with aortic and intracranial aneurysms, respectively, causing ECM disruption
Fibrillin-1 abnormalities have been associated with Marfan’s syndrome and Fibrillin-2 have with Beal’s syndrome or congenital contractural arachnoldactyly [71,72]
Microfibrils, such as the glycoprotein, Fibrillin:
  • Found in ECM and become incorporated into insoluble microfibrils
  • Plays a role in early elastogenesis acting as a scaffold for elastin deposition [73]

5.4.1. Tenascin-C

Tenascin-C (TNC) plays an important role in regulating tenocytes that need to interact with ECM components (Table 4). The first research establishing a genetic association for a predisposition to Achilles tendon pathology was published in 2005 and implicated TNC [74]. A South African case-control association study reported that a Guanine-Thymine Dinucleotide repeat variant was associated with Achilles tendon injuries. A second study investigating different TNC variants (rs13321; rs2104772; rs1330363) found altered frequencies between cases and controls in Australian and South African populations—but did not reach statistical significance [75] (Table 5). However, the study did implicate a genetic region spanning both the TNC and the COL27A1 genes using haplotype analysis. More recently, the application of WES analyses assisted the identification of potential functional TNC variants implicating the TNC gene, specifically rs1061494 and the T-T haplotype (rs1061494-rs2104772) with increased risk of Achilles tendinopathy in a South African cohort [30]. This study therefore provided evidence that the risk susceptibility to Achilles’ tendinopathy is most likely within the TNC gene rather than within the COL27A1 gene locus.
Several TNC variants have been associated with rotator cuff injury [76,77]. Previously implicated TNC variants (rs1138545, rs72758637, rs7021589) have been replicated in a GWAS study using the UK biobank to be associated with an increased risk of rotator cuff injury [57,76]. Tashjian reported increased gene expression, for TNC (2.2×) in tears vs. controls [57] (Table 5).

5.4.2. COMP and Other Thrombospondins

COMP, also known as thrombospondin 5, is the most abundant tendinous glycoprotein. Thrombospondin 2 (THBS2) mediates cell-to-cell and cell-to-matrix interactions and is involved in cell-to-cell adhesion and ECM communication (Table 4). COMP (rs730079; rs28494505) and THBS2 (rs9505888; rs6422747) variants failed to show significant difference in Achilles tendon studies in Australian and South African cohorts [75]. No human genetic studies exist relating to the potential role of Thrombospondin 2 in tendon pathology, although its absence has been associated with connective tissue abnormalities in mice [76].
A GWAS study identified rs575224171 within the gene THSD7A encoding the endothelia protein thrombospondin type 1 domain, containing protein 7A, to be associated with increased risk of rotator cuff injury [57]. These authors hypothesised that gene variants may lead to poor rotator cuff angiogenesis, predisposing individuals towards increased tear risk. RNA sequencing analyses established a 2.6× decreased expression of this gene within rotator cuff tears vs. control tissues [57].

5.5. Elastin and Microfibrils

Elastin represents 1–10% of a tendon’s dry weight and is found in both the IFM and within the fascicles—especially around tenocytes. Microfibrils, such as the glycoprotein Fibrillin, are found in the ECM and become incorporated into insoluble microfibrils (Table 4). South African, Australian, and British cohorts did not find an association between the ELN rs2071307 variant and risk of developing Achilles’ tendon pathology [78,79]. However, the FBN2 rs331079 variant was associated with risk for Achilles’ tendon disease and ACL ruptures [78].
Table 5. Summary of genetic research and tendon injuries: Glycoproteins in the ECM and elastin and fibrillin.
Table 5. Summary of genetic research and tendon injuries: Glycoproteins in the ECM and elastin and fibrillin.
TypeProteinReferenceCountryVariantPathologySubjectsControlsComments
Glycoproteins in the ECMTenascin-C (TNC)[74]South Africa,
Australia
TNC—GT dinucleotide repeatsAchilles’ tendinopathy and rupture144127Association with the number of GT repeat polymorphism and the risk of tendinopathy and rupture
[60]South Africa,
Australia
TNC rs13321, rs2104772, rs1330363Achilles’ tendinopathy and rupture179339No significant associations found among variants
[80]SpainTNC rs2104772Patellar tendon injury150No difference between severity of injury and gene variants. No controls were included
Cartilage oligomeric matrix protein (COMP)[75]South Africa,
Australia
COMP rs730079, rs2849450
THBS2 rs9505888
Achilles’ tendinopathy and rupture178340No significant associations found among variants
Elastin and FibrillinElastin (ELN) and Fibrillin (FBN)[78]South Africa,
Australia
ELN rs2071307
FBN2 rs331079
Achilles’ tendinopathy and rupture135239No association with the ELN variant and tendon pathology
Significant association between the FBN2 gene variant and tendon disease
[81]UKELN rs2071307Achilles’ tendinopathy and rupture108131No association with the ELN variant and tendon pathology
[80]SpainELN rs2289630Patellar tendon injury150No difference between severity of injury and gene variants. No controls were included
Green indicates an association and red indicates no association found.

5.6. Tendon Development, Homeostasis, and Remodelling

Tendon ECM homeostasis and remodelling are maintained by complex enzyme systems. These include matrix metalloproteases (MMPs), ADAMTSs (a disintegrin and metalloproteinase with thrombospondin motifs) and ADAMs (a disintegrin and metalloproteinase), tissue inhibitors of MMPs (TIMPs), and growth factors like the transforming growth factor-ß (TGF-ß) families [82] (Table 6). Genetic research undertaken in this area of tendon development, homeostasis, and remodelling is summarised in Table 7.

5.6.1. MMPs, TIMPs, ADAMTSs, and ADAMs

The balance between MMPs and TIMPs is necessary to maintain tendon homeostasis and remodelling [82]. If intrinsic control of these systems is compromised by extraneous factors, the tendon’s ability to respond appropriately to loading will be affected, risking tendon disease (Table 6). No association was reported of ADAMTS2, ADAMTS5, ADAMTS14, and ADAM12 variants with Achilles’ tendon pathology in South African and Australian cohorts [83]. However, Raleigh found significant associations for MMP3 variants and AT (but not rupture) in a South African population [84]. In a British population, El Khoury found no associations in Achilles’ pathology groups overall—although subgroups did show some correlations [83]. Furthermore, El Khoury found a significant association with a variant within the TIMP2 gene rs478932 for Achilles’ tendon pathology in both a South African and Australian cohort. Different genotypes were overrepresented in the subject groups in each population [83]. Gibbon explored the MMP3 locus in an Australian cohort with AT and identified a 6A-G-C-G haplotype (rs3025058, rs679620, rs591058, rs650108) with reduced risk [85].
Table 6. Tendon development, homeostasis and remodelling.
Table 6. Tendon development, homeostasis and remodelling.
FamiliesFunctionsKey RegulatorsAssociated Diseases
Matrix metalloproteases (MMPs)
(23 family members)
Four subgroups:
  • Collagenases
  • Gelatinases
  • Stromelysins
  • Membrane-type
  • Broad proteolytic activity against collagen and other ECM compounds
  • MMP3: degrades different collagen types, proteoglycans, and fibronectin, and laminin
Fluoroquinolone antibiotics, which are associated with increased Achilles rupture risk [86], can affect MMP expression in human tendons [9,87]
A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs)
(20 family members)
  • Procollagen processing
  • ECM remodelling
  • ADAMTS2 and 14: regulate conversion of procollagen to collagen
  • ADAMTS5: cleaves proteoglycans, e.g., aggrecan
ADAMTS2 gene mutation causes EDS dermatosparaxis type
A disintegrin and metalloproteinase (ADAMs)
(19 family members)
  • Proteases
  • ADAM12: a marker of skeletal muscle regeneration and binds insulin growth factor binding protein-3 (IGFBP-3)
Tissue inhibitors of metalloproteases (TIMPs)
  • Inhibit MMPs activities
  • Balance between MMPs and TIMPs is necessary to maintain tendon homeostasis and remodelling
Transforming growth factor beta (TGF-ß) superfamily
  • ECM homeostasis and remodelling
  • TGF-ß1 and growth differentiation factor 5 (GDF-5), can increase Achilles tendon strength in animals [88].
  • Mechanical loading releases TGFß-1 and aids cell growth, proliferation, differentiation, and migration, as well as cell death (apoptosis). Involved in collagen and proteoglycan synthesis.
  • GDF-5 (also called CDMP-1 (cartilage-derived morphogenetic protein 1) or BMP-14 (bone morphogenic protein 14). Expressed in developing CNS and maintenance, development and repair of bones, cartilage, and various other musculoskeletal soft tissues, including tendons.
GDF-5 gene mutations (CDMP1) implicated in Hunter–Thompson type dwarfism and in Grebe Syndrome (characterized by short stature, extra digits, and short and deformed extremities) [89,90,91,92].
Bone morphogenetic proteins (BMPs) (20 family members)
  • Grouped into subfamilies of the TGF-ß superfamily
  • Growth factors and cytokines in many tissues in the body
Fibroblast growth factors (FGFs)
(22 family members)
  • Influence cell development and maturation by binding to receptors (FGFR) triggering intracellular events
Point mutation in FGFR3 can lead to achondroplasia.

5.6.2. Transforming Growth Factor-ß (TGF-ß) Superfamily

The TGF-ß superfamily has a similar role as the MMP/TIMP system in ECM homeostasis and remodelling (Table 6). Mechanotransduction is the process of converting mechanical forces into a cellular response. Tendon exposure to increased loading (within safe physiological limits) causes tenocytes to increase collagen synthesis and enhance tendon load resistance. It is the end goal of sensible incremental training [92]. The reverse occurs with inactivity. If a tendon’s tensile load is temporarily decreased, there is a reduction in secreted ECM structures including Type I collagen and COMP [93]. TGF-ß appears to be a major regulator of tendon development secondary to mechanical loading [94]. The exact mechanism of how loading activates the TGF-ß signalling pathway appears to involve an induction of scleraxis (Scx) and other markers, such as tenomodulin [95]. This promotes the synthesis and secretion into the ECM of collagen and other ECM components. TGF-ß and GDF-5 genes functional variants have been studied with an association established for GDF-5 rs143383, but not TGF-ß rs1800469, with risk of Achilles’ tendinopathy [96].

5.6.3. Bone Morphogenic Glycoproteins (BMP)

BMPs are grouped into subfamilies of the TGF-ß superfamily and function as growth factors or cytokines. Originally studied for their effect upon bone and cartilage formation, they are recognised to have a widespread function as signallers and regulators of many organs systems’ development [35]. A significant association was reported in a Brazilian mixed-injury cohort for the BMP4 variant (rs2761884) in tendinopathies [97].

5.6.4. Fibroblast Growth Factors (FGFs)

FGFs are required for normal development and cell maturation. They bind to receptors (FGFR), triggering intracellular events. Salles studied a group of Brazilian volleyball players with variously located tendinopathies. However, none of the investigated FGF3, FGF10, and FGFR1 variants were associated with altered risk for tendinopathy [97]. Similarly, no associations of the same genes with rotator cuff tears in American patients were reported [98]. Conversely, Motta found significant associations of the FGF3, FGF10, and FGFR1 variants with rotator cuff disease in large Brazilian case-control study [99].
Table 7. Summary of genetic research and tendon injuries: Development, homeostasis and remodelling.
Table 7. Summary of genetic research and tendon injuries: Development, homeostasis and remodelling.
FamiliesReferenceCountryVariantPathologySubjectsControlsComments
A disintegrin and metalloproteinase with thrombospondin motifs (ADAMTSs)
A disintegrin and metalloproteinase (ADAMs)
[83]South Africa, AustraliaADAMTS2 rs1054480
ADAMTS5 rs226794
ADAMTS14 rs4747096
ADAM12 rs4747096
Achilles’ tendinopathy and rupture173248No significant association with Achilles’ disease
ADAMTS14 associated with a later onset of disease
Matrix metalloproteases (MMPs)
and Tissue inhibitors of metalloproteases (TIMPs)
[84]South AfricanMMP3 rs591058, rs650108, rs591058Achilles’ tendinopathy and rupture11498Variants significantly associated with chronic tendinopathy
No increased risk of rupture with variants
Lowest risk of tendinopathy associated with combination of a MMP3 rs679620 and COL5A1 rs12722 variants
[81]UKMMP3 rs679620
TIMP2 rs4789932
Achilles’ tendinopathy and rupture118131MMP3 variant significantly associated with tendinopathy and rupture groups in males but not females or overall.
TIMP2 variants overrepresented in tendinopathy group
[83]South Africa, AustraliaTIMP2 rs4789932Achilles’ tendinopathy and rupture173248SNP significantly overrepresented in tendinopathy group
[85]AustraliaMMP3 rs679620, rs591058, rs650108, rs3025058Achilles’ tendinopathy79195An association was found with a 6A-G-C-G haplotype (rs3025058, rs679620, rs591058, rs650108) with reduced risk for Achilles tendinopathy
Transforming Growth Factor-ß Superfamily (TGFß)
Growth/differentiation factors (GDF)
[96]South Africa,
Australia
TGFB1 rs1800469
GDF5 rs143383
Achilles’ tendinopathy and rupture171238No associations with TGFB1 genotype variant
GDF5 variant associated with Achilles’ disease.
Bone morphogenetic proteins (BMPs)[97]BrazilBMP4 rs2761884Mixed—Achilles; Patellar; Rotator cuff; Hip abductors5286Significant association with BMP4 variant for tendinopathy
Fibroblast Growth Factors[97]BrazilFGF3 rs7932320, rs1893047, rs12574452, rs4631909, rs4980700
FGF10 rs1448037 rs900379, rs1011814, rs593307
FGFR1 rs13317
Mixed—Achilles; Patellar; Rotator cuff; Hip abductors5286No significant associations with tendinopathy
[98]USAFGF3
FGF10
FGFR1
Rotator Cuff tears1752595No significant associations
[99]BrazilFGF3 rs12574452
FGF10 rs11750845, rs1011814
FGFR1 rs13317
Rotator Cuff tears203207All showed significant associations with rotator cuff tears
Green indicates an association and red indicates no association found.

5.7. Cell Death (Apoptosis) and Inflammation in Tendons

Apoptosis is a natural phenomenon in many living tissues. In tendons, damaged tenocytes’ removal is facilitated by cytokine activity. Excessive tendon loading can increase apoptosis and affects the cell population’s abilities to respond effectively to exercise with secondary effects upon the ECM leading to tendon disease [100]. The role of inflammation in chronic tendinopathy has long been debated [101]. Early research utilising microscopic examination of tendinopathic tissue and biochemical analysis reported no evidence of the normal elements associated with ‘classical’ inflammation in chronic tendon injuries [102,103,104,105]. Animal work indicated that early tendinosis was associated with tenocyte stimulation rather than apoptosis and modulated by growth factors such as insulin-like growth factor 1 (IGF-1) [106]. However, the authors were unable to comment on the chronic effect of prolonged loading on cell survival. (Table 8).

5.7.1. Interleukins

Inflammatory pathways throughout the body involve numerous elements, interacting in a complex manner and resulting in gene expression alterations, apoptosis, and detrimental changes to the ECM. The protein family of interleukins are intimately involved in the inflammatory pathway. Interleukins are upregulated in early tendinopathy and involved in the inflammatory cascade and remodelling activities [114]. September reported interleukin gene-gene interactions with COL5A1 rs12772, suggesting that type V collagen may be regulated by certain inflammatory mediator proteins in the IL-1β-signalling pathway [115]. Altering the amount of type V collagen expression could impact α1(V) collagen chains and, thereby, the collagen tendon fibril diameter and ultimately tendon capacity (Table 9).

5.7.2. Caspases

Caspases are a family of protease enzymes that are integral to programmed cell death (apoptosis). The South African research group reported that two CASP8 genotypes had significant associations with AT [116] (Table 9).

5.7.3. Nitric Oxide Synthase (NOS) Enzymes

Nell found no association with gene variants for NOS2 and NOS3 and AT [115]. However, Brookes reported a reduced risk for AT with the NOS2 rs2779249 heterozygote variant, but no association with NOS2 rs2248814 [117] (Table 9).

5.8. Angiogenesis

Angiogenesis is the formation of new blood vessels from the existing vasculature. Tendons and ligaments have a poor blood supply and low metabolic rate. Consequently, their healing capacity is low [118]. Histopathological examination of chronic AT specimens shows marked increases in angiogenesis [110]. It is hypothesized that this is triggered by mechanical loading and designed to promote tendon remodelling. Further studies have identified increased levels of pro-angiogenic expression profiles and, specifically, vascular endothelial growth factor A after tenocyte mechanical loading [119,120,121]. Poorly regulated angiogenesis may lead to distortion of the neat parallel collagen fibril array in the tendon ECM. Increased levels of angiogenic associated proteins have been noted in both ruptured tendons and ligaments and including degenerative tendons [121,122,123]. Angiogenesis elements are placed centrally within the network of partners regulating key ECM components within tendon and ligament.
Several functional variants within the VEGFA (rs699947, rs1570360, and rs2010963) gene have been explored and a risk haplotype was implicated both in a (i) South African cohort and a (ii) combined South African and British cohort of mid portion chronic AT [124]. Specifically, the VEGFA A-G-G (rs699947 C/A–rs1570360 G/A–rs2010963 G/C) inferred haplotype was associated with increased risk of AT. This haplotype includes the collective alleles associated with decreased VEGFA gene transcription and a corresponding lower VEGFA plasma level [125]. Therefore, it is reasonable to hypothesise that these allele combinations would potentially contribute to limiting the capacity of the structure to regulate ECM remodelling within a hypovascular tendon [122]. The authors did not report an association with Achilles’ tendon risk for any of the variants explored in KDR (rs2071559 and rs1870377) [123] (Table 8).
Like other gene loci, differences in associations at the VEGFA and KDR loci have been noted in populations of different ancestry. One study reported no associations in two KDR polymorphisms (rs1870377; rs2071559) with AT in South African and UK cohorts [124] (Table 9).

5.9. Other Areas of Genetic Study in Tendons

5.9.1. ESRRB

Estrogen-related receptor beta (ERR-β) is a nuclear receptor encoded by ESRRB (Estrogen Related Receptor Beta) gene. Its function is unknown; however, a similar protein in mice plays an essential role in placental development. It appears to influence the expression of PPARGC1 and ESRR-inducing regulator muscle 1 (PERM1) in skeletal muscle. Motta identified two 2 SNPs in the ESRRB gene that were associated with rotator cuff disease [99]. Teerlink found a significant association for rotator cuff injury with an ESRRP rs17583842 variant [98] (Table 9).

5.9.2. Defensin ß1

Defensins form a family of microbicidal and cytotoxic peptides made by neutrophils. Defensin ß1 is encoded by the DFNB gene. It resists microbial organisms from attaching to epithelial surfaces. Motta found the DEFB-1 rs1800972 SNP to be associated with a preventive effect for rotator cuff tears [99] while Teerlink found no association with DEFB-1 [98] (Table 9).
Table 9. Summary of genetic research and tendon injuries: Inflammatory cascade, apoptosis, and others.
Table 9. Summary of genetic research and tendon injuries: Inflammatory cascade, apoptosis, and others.
TypeProteinReferenceCountryVariantPathologySubjectsControlsComments
Inflammatory Cascade
and Apoptosis
Interleukins (ILs)[116]South Africa,
Australia
IL1B rs1143627, rs16944
IL1RN rs2234663
IL6 rs1800795
Achilles tendinopathy and rupture175369No independent associations.
In combination with COL5A1 rs12722 these alleles had significant association with Achilles’ tendinopathy
Caspases (CASP)[115]South Africa, AustraliaCASP8 rs3834129, rs1045485Achilles tendinopathy and rupture166358Significant association with both polymorphisms
Nitric Oxide Synthases (NOS)[115]South Africa, AustraliaNOS2 rs2779249
NOS3 rs1799983
Achilles tendinopathy and rupture166358No significant associations
[117]UKiNOS rs2779249
iNOS rs2248814
Achilles tendinopathy and rupture132145Significant association (protective effect) with rs2779249
No association with rs2248814
AngiogenesisAngiogenic Factors[124]South Africa and
UK
VEGFA rs699947, rs1570360, rs2010963
KDR rs1870377, rs2071559
Achilles tendinopathy120
130
108
87
Significant association with all 3 VEGFA polymorphisms
No association with the KDR polymorphisms
OthersDefensins (DEF)[98]USADEFB1Rotator Cuff tears1752595No association
[99]BrazilDEFB1 rs1800972Rotator Cuff tears203207Significant association (protective effect)
Estrogen-related receptor ß (ESRRB)[98]USAESRRB rs17583842Rotator Cuff tears1752595Significantly associated with rotator cuff disease
[99]BrazilESRRB rs1676303, rs4903399Rotator Cuff tears203207Significantly associated with rotator cuff disease
Green indicates an association and red indicates no association found.

5.10. Ageing in Tendons

Diseased and damaged tendons increase with age. For instance, 11–37% develop tears in an ankle peroneal tendon during their lifetime [126]. Long exposure to mechanical stresses and the increasing inefficiency of tendon repair are implicated. It is a classic example of the Injury Causation model (Figure 1). As a result, performance and function are impaired [127]. Many age-related changes occur to tendons during their lifetime and differences are apparent in the way men and women respond to tendon loading [128]. Additional knowledge has been derived from animal research, especially equine studies and it has been proposed that the ability of pluripotent mesenchymal stem cells to differentiate into tenocytes reduces with time. Tendon stem cells become less numerous with age and their ability to differentiate and produce competent mature tenocytes reduces [129]. The senescence-inhibited gene within the tenocyte is downregulated [130]. The ageing tenocyte’s complement of produced proteins (proteome) is restricted [131]. The proteins affected include those with roles involved in cell survival and death, cytoskeletal changes, and antioxidant response.
The ability to repair damaged tendons is also affected by non-collagenous ECM protein turnover, including cytokines and various growth factors, and a disruption to the fine homeostatic mechanisms outlined in Section 5.6 [132]. Equine research has revealed that with age, the tendons’ protein turnover diminishes [133], glycosaminoglycans increase [134], the type III collagen increases proportionately [38], and collagen fibril diameter diminishes [135]. Changes occur to the IFM with reduced protein turnover and elasticity, increasing the risk of injury [136,137]. MMP activity reduces tendon strength [127]. There is additional evidence that age carries with it a reduced capacity to resolve inflammation [138]. In addition, degenerative human tendons can have altered responses to reactive oxygen species with age, and therefore oxidative stress may be an important pathway in tendinopathy development [139].
The progressive alteration in tendon function involves a complex interplay between many influences. This includes not only our inherited genetic material but temporal changes to gene expression regulation, including an array of epigenetic mechanisms [140]. The potential role of epigenetics will be elaborated upon in Section 9.

6. Ligament: Structure, Function, and Genetic Research

Ligaments span joints attaching at either end to bones. Like tendons, they are fibrous, dense connective tissues. They are designed to resist excessive load, control joint motion, prevent instability, and have a vital proprioceptive role on account of their rich innervation. They have a similar hierarchical structure to tendons. However, the degree of packing of the collagen is slightly different. Whereas tendons organise collagen fibres in an orderly, parallel orientation, ligament organisation is more random and less parallel. This allows ligaments to respond to tensile loads in different directions. The cellular elements tend to be more randomly distributed and rounder in shape than tendons. There is a higher percentage of proteoglycans and water and reduced percentage of collagen. The elastin content in ligament is higher than tendons.

6.1. Collagen

6.1.1. Type I Collagen

There has been considerable interest in the genetic architecture of the COL1A gene and susceptibility to tendon and ligament injuries. Differences in the genetic susceptibility to acute and chronic injuries have been linked to rs1107946 (−1997 G/T) and rs1800012 (+1245 G/T), within the COL1A1 gene. The rare TT genotype of the Sp1 binding site variant (rs1800012) was associated with decreased risk for acute injuries such as shoulder dislocations [141], ACL ruptures [47], and acute soft tissue ruptures [49]. A meta-analysis reported the association of the rs1800012 TT genotype with reduced risk for sports-related tendon and ligament injuries [142]. The same genotype has been associated with increased risk for intervertebral disc degeneration in the elderly and increased risk for lumbar disc disease in young military recruits [143,144]. The alternate rs1800012 GG genotype was reported to reduce risk for ACL ruptures sustained while skiing [145]. The rs1107946 variant, which is in linkage disequilibrium with rs1800012, has been independently associated with the risk of skiing-associated ACL ruptures [146]. Haplotype analyses with these two functional variants have been associated with ACL rupture risk in a Polish cohort [147]. The current theory proposes that these functional promotor variants work in concert to regulate COL1A1 expression [148].
However, several studies have failed to reflect an association between these COL1A1 variants and susceptibility to several musculoskeletal soft tissue injury phenotypes. This may result from insufficient power of the studies to detect the rare rs1800012 TT genotype in and may explain conflicting results when comparing data from larger combined analyses to that of smaller independent cohorts [50]. No associations were noted in the Chinese Yunnan Han ACL samples for the COL1A1 or COL5A1 locus [149]. However, the TT genotype of the COL1A1 Sp1 binding site polymorphism has been reported to be significantly underrepresented in South African participants with ACL ruptures [150] (Table 10).

6.1.2. Type V Collagen

As in AT research, variants within the 3’-UTR of the COL5A1 gene have been associated with ACL rupture susceptibility, specifically in females [152,153,154], and more recently with ligament injuries [155]. Laguette explored the intron 4-exon 5 region of COL5A1, which was previously implicated with ligament injuries in a canine model but found no significant associations in a South African ACL cohort [156]. The COL5A1 rs12722 C/T and COL5A1 rs13945 C/T polymorphisms were also associated with reduced ACL injury risk in male skiers [157]. Furthermore, Suijkerbuijk reported an association with ACL ruptures and COL5A1 rs12722 in a combined Swedish and South African cohort [158] (Table 11).

6.1.3. Types III and XII Collagen

Associations have been noted for variants in COL3A1 [151,152], and COL12A1 [149,159] with ACL rupture risk, while others reported no significant associations [147,161].

6.2. Regulatory and Structural Components of the Extracellular Matrix

6.2.1. Tenascin-C

Historically, there has been much interest in the associations between the TNC gene and its neighbouring genes with tendon and ligament injury susceptibility. Gibbon explored this region for ACL susceptibility. Variants within the TNC gene: rs2104772 and a TT haplotype (rs1061494 and rs2104772) were associated with ACL susceptibility using a tailored WES and bioinformatics approach [30]. However, no associations for the TNC locus were noted in a Polish cohort [162]. More functional research is required to understand the biological significance underpinning this genetic locus and tendon and ligament injury susceptibility.

6.2.2. Proteoglycans

Recent studies have investigated proteoglycans. Variants within their controlling genes have been implicated with susceptibility to ACL ruptures in independent cohorts from South Africa [163,164] and Poland [165] (Table 12).

6.2.3. MMPs

As in tendinopathy, the MMP locus (chr11q22) has been association with susceptibility to ACL ruptures. Posthumus demonstrated that MMP3 rs679620 variant may interact with several other MMP loci, MMP10 rs485055, MMP1 rs1799750, and MMP12 rs2276109, to collectively contribute to ACL rupture susceptibility in a South African cohort [167]. The MMP3 rs3025058 variant, which is tagged by rs679620, was independently associated with ACL ruptures in a Thai population [168]. No associations were noted when MMP1 rs1799750, MMP10 rs486055, and MMP12 rs2276109 variants were explored with ACL rupture susceptibility in a Polish cohort [169]. The MMP genes have been investigated with several different exercise-related phenotypes and conflicting associations have been noted. This suggest that there may be specific genetic signatures which are inherited together and underpin specific exercise-related phenotypes, which still require functional unravelling (Table 13).

6.2.4. Transforming Growth Factor Superfamily

Variants in several such genes controlling the TGF superfamily have been explored with an association to ACL rupture risk. These include variants within the TGF-β receptor III (TGFβR3) and the TGF-β induced (TGFβI) genes. An independent association of TGFBR3 rs1805113 G allele with a decreased risk of ACL injury has been described. Additionally, a genetic interval between TGFBR3 rs1805113-rs1805117 was associated with ACL injury risk in a South African cohort [158].
GDF5 plays a critical role in tendon and ligament repair. Variant analyses within GDF5 gene have shown conflicting risk associations with ACL injury and larger studies are required to understand the significance of this locus with ACL injury risk [170,171].

6.3. Signalling Factors

6.3.1. Interleukins

Investigation of interleukins have shown similar findings to tendinopathy. An inferred allele combination (IL1B, IL6, IL6R, and COL5A1) was associated with ACL rupture risk [158,172,173]. Differences were noted at the alleles implicated for the IL1RN rs2234663 and IL6 rs1800795 loci. The functional consequence of these genetic loci was subsequently explored [157]. Cells treated with either hrIL-β or hrTNF-α expressed altered levels of BGN mRNA (which encodes for the biglycan PG) and COL5A1 mRNA depending on their IL1B-high risk or IL1B-low genotype profiles. Evidence suggests that the inflammatory micro-environment together with an individual’s genetic profile can modulate ECM expression of tendon and ligament components and thereby potentially impact these structures’ functional capacity.

6.3.2. Caspases

Both Rahim and Seale have reported associations between caspase functional gene variants and ACL ruptures [174,175].
Table 13. Summary of genetic research and ligament injuries: Development, homeostasis and remodelling and inflammatory cascade and apoptosis.
Table 13. Summary of genetic research and ligament injuries: Development, homeostasis and remodelling and inflammatory cascade and apoptosis.
TypeProteinReferenceCountryVariantPathologySubjectsControlsComments
Development, Homeostasis, and RemodellingTGFβ
Growth Factor Superfamily
[156]South AfricaTGFB1 rs1442, TGFBR3 rs1805113, rs1805117ACL rupture249210Significant association
[170]South AfricaGDF5 rs1413383ACL rupture126214No significant association
[171]ChinaGDF5 rs1413383ACL rupture286500Significant association
Matrix metalloproteases (MMPs)[167]South AfricaMMP1 rs1799750
MMP3 rs679620
MMP10 rs486055
MMP12 rs2276109
ACL rupture129216Significant association
[168]ThailandMMP3 rs3025058, rs679620ACL rupture86100Significant association
[169]PolandMMP1 rs1799750
MMP10 rs486055
MMP12 rs2276109
ACL rupture228202Significant association
Inflammatory Cascade, and ApoptosisInterleukins (ILs)[173]PolandIL1B rs1143627, rs16944
IL6R rs2228145, IL6 rs1800795
ACL rupture229194Significant association
[158]South Africa and SwedenIL1B rs16944, IL6 rs1800795
IL6R rs2228145
ACL rupture79116Significant association
[176]South AfricaIL1B rs16944, IL6 rs1800795
IL6R rs2228145
ACL rupture234232Significant association
Caspases (CASP)[172]South AfricaCASP8 rs3834129ACL rupture234232Significant association
[175]South AfricaCASP8 rs383412, rs1045485, rs13113ACL rupture102116Significant association
Green indicates an association and red indicates no association found.

6.3.3. Angiogenesis

Independent and haplotype associations were noted for VEGFA functional variants (rs699947, rs1570360, and rs2010963) with ACL rupture susceptibility [172,174,177,178] including contrasting associations between ACL rupture and AT susceptibility. For example, the VEGFA rs699947 CC was associated with increased risk of non-contact ACL ruptures [174] but associated with a reduced risk of AT [124]. Similarly, the inferred haplotype, associated with increased VEGF production [125], was more often observed with an increased risk of ACL rupture whereas the low-VEGF producing haplotype was associated with a reduced risk of injury [174]. In contrast, the low-VEGF producing haplotype was associated with increased risk of tendinopathy [124]. Following a pathway-based approach, including DNA variants within the interleukin and the angiogenesis encoding genes, Rahim highlighted that VEGFA rs699947 CC, VEGFA rs2010963 GC, BMI, and age remain significant biological components in ACL rupture susceptibility [176].
Evidence suggests that a lower-level blood flow increase after running is associated with higher risk for developing AT in an age and sex-dependent manner [179]. Whereas, in the ACL model, overexpression of VEGFA may reduce the biomechanical strength of the tendon graft in the early stages of an ACL ligament reconstruction, whilst in the later stages of graft incorporation increased expression is essential [180].
Therefore, it does seem, that the “Goldilocks affect” is still plausible [115] suggesting that a finely tuned homeostatic feedback regulation of ECM components is required to maintain both tendon and ligament tissue integrity. Willard and Suijkerbuijk have shown functional evidence linking a genetic contribution, at key proteoglycan, interleukin, and collagen genes, to the expression of ECM components in a susceptibility model [158,164].
Further exploring the angiogenesis pathway and knowing the VEGF biological effects are mediated via its receptor kinase insert-domain receptor (KDR), Rahim showed that the inferred G-A KDR haplotype (rs2071559 A/G, rs1870377 A/T) was significantly associated with increased susceptibility to ACL ruptures [174]. It was suggested that the rs2071559 G allele alters a potential transcription factor binding site in the promoter region, thereby reducing KDR transcription [181] and the A allele of rs1870377 T/A was associated with reduced VEGF-binding efficiency [181]. Lulinska-Kuklik identified an association between VEGFA rs2010963 and ACL injuries in a Polish population, but not with VEGFA rs699947 or VEGFA rs1570360 [178]. These associations need to be repeated in larger ACL data sets. More recently, Feldmann investigated ACL injury risk in a combined cohort from Sweden, Poland, Australia, and South Africa, and further implicated the VEGFA rs201093 CC genotype and the VEGFA (rs699947 C/A, rs57036 G/A and rs2010963 G/C) A-A-G haplotype to be associated with reduced risk of an ACL injury [182]. In addition, the authors suggest that possibly variants in KDR are not associated with ACL risk susceptibility. The differences noted with KDR in the various populations may represent Type I statistical errors (Table 14).
More informative population specific genetic variants are required to be tested [183,184,185,186]. Observing the differences and similarities between these genetic association findings with tendon and ligament injury susceptibility may highlight different biological mechanisms underpinning the two injury models (acute vs. chronic) or it may be indicative of the differences between ligament and tendon molecular-functional correlations. Moreover, it may also be reflective that larger data sets are required to comprehensively screen a gene of interest across populations. It remains essential that the genetic associations are explored at a functional level to assess the biological significance of these variants in both acute and chronic injuries.

7. Epigenetics

Despite the growing number of genetic loci implicated in tendon and ligament injury susceptibility, a large unknown heritability component remains unresolved [24]. The impact of epigenetic regulation of the genome is an emerging area of research in understanding and deciphering the heritability of common complex phenotypes. Epigenetics was first described in 1968 [187]. It refers to heritable changes (from parent to child) which do not depend upon DNA sequence changes. DNA within every one of our cells is identical in content and sequencing with a few exceptions. What determines an individual cell’s function is the gene expression within it. Differential expression will determine, for example, if a cell acts as a tenocyte or osteoblast.
DNA must be accessible for transcription by RNA polymerase to be active. Genetic material transcription ‘visibility’ can be controlled by several mechanisms, which are important during development and differentiation of cellular components and tissues. Equally, they are important in influencing the way that mature tendons and ligaments respond to normal adaptation to loading and pathological disease evolution.
The main epigenetic mechanisms facilitating protein expression regulation, without altering the DNA sequence, involve chemical modifications to the genome and DNA, and the associated proteins. This occurs through either DNA methylation, histone modifications and the actions of non-coding RNAs (ncRNA). The latter is an RNA molecule produced by DNA, but not translated into protein. They regulate gene expression at both the transcriptional and post-transcriptional stages of protein synthesis. This type of RNA includes micro-RNAs. They influence and act alongside other epigenetic mechanisms to create ‘gene silencing’. Parts of epigenetic regulation can be inherited via the germline. Others result from a response to an individual’s environmental exposure, which can include but is not limited to lifestyle choices. Epigenetic mechanisms may play an important part in explaining the variation of phenotype in complex disease processes. This includes the lack of reproducibility in some linkage and association studies, observed twin discordance, and different ages of disease-onset when subjects possess the same genotype [188].

7.1. DNA Methylation

Some of the genes/gene families implicated in either tendon or ligament injury risk susceptibility have been associated with altered methylation status [189]. For example, a hypermethylation status, typically associated with reduced gene expression, of several CpG sites within the promoter regions of the MMP11 and ADAMTS4 genes but not TIMP2 was noted in pathological human patellar tendon samples compared to control tendon tissue samples [190,191]. The MMP11 and ADAMTS4 enzymes play an important role in ECM regulation and in particular PGs such as aggrecan. Enzyme reduction could result in substrate accumulation. An increased expression profile of large proteoglycans, such as aggrecan, was reported in pathological human patellar tendons [192,193].
The methylation status, together with the corresponding mRNA expression profiles of several MMPs (MMP1, −2, −3, −9, −13, and −14) and TIMP (TIMP1, 2 and 3) genes, was investigated using ruptured shoulder supraspinatus tendon tissue [194]. Significantly altered DNA methylation patterns for MMP1, MMP9, MMP13, TIMP2, and TIMP3 were described at the torn tendon edge compared to uninjured control tendon samples. An inverse correlation was noted between the overall methylation status of promotor sites within MMP1, −9, −13, and TIMP3, including the 5’-UTR of TIMP3, and the respective mRNA expression. Changes in the methylation pattern of some of these genes were partly influenced by age at surgery, sex, smoking habit, tear size, and/or duration of symptoms, which are some of the confounders previously implicated in injury susceptibility and/or impaired tendon healing [158].

7.2. RNA Interference

There has been increased interest in miRNAs and miRNA binding sites and their role in protein expression regulation at the post-translational level. In particular, the association of the putative hsa-miR-608 binding site within the 3’UTR region of COL5A1 gene with both AT and ACL rupture susceptibility [54,58].
The miRNA-29 family, for which direct/indirect targets include collagens, MMPs, integrins and some DNA methylases have been examined [195]. No significant differences in expression profile were noted in torn supraspinatus tendons versus uninjured tendon samples. However, there was an inverse correlation between the expression profiles of hsa-miR-29a-3p, hsa-miR-29b-3p and -5p and MMP2, MMP9 and MMP14 expression. hsa-miR-29a-3p and miR-29b-5p were inversely correlated with MMP1 expression [194]. This suggests that the miRNA-29 family contributes to regulating specific ECM components of rotator cuff tendons.
Several potential epigenetic candidate loci and regulators have been highlighted in both tendon and ligament biology. However, little has been explored in the context of tendon and ligament injury and susceptibility [196].

8. Discussion

This review has highlighted both overlaps and distinctions in the genetic loci implicated between tendon and ligament injuries, as well as for acute and chronic injuries of both. This underlines the subtleties within musculoskeletal soft tissue pathology. Underlying factors behind acute, subacute, and chronic pathology are likely to be very different. For acute injuries, extrinsic factors will probably dominate—irrespective of the genotype of the individual involved, e.g., an impact injury. More chronic problems (and those who fail to recover from acute injuries) are more likely to identify intrinsic factors, including genetic, as part of the underlying disease pathway.
It is important for researchers to avoid categorising all tendon and ligament issues as having a common pathology. Structural changes can affect different parts of the tendon and ligament. For example, the Achilles tendon can have problems arising from within both the main tendon and its calcaneal insertion. The anatomy, physiology, and biomechanics will vary according to site. Within the main tendon, changes can affect the paratenon or main tendon. The medially placed plantaris tendon can be the primary source of ‘Achilles’ pathology’. The tendon may suffer small splits and partial tears or, more commonly, tendon enlargement. An acute rupture needs to be categorised differently to a decade old swollen tendon. These different subsets of potential pathology highlight the importance of the care required when assembling cohorts for genetic research analysis.
It is becoming evident that tendon and ligament matrix remodelling is complex and, therefore, not surprising that genetic loci controlling both structural pathways and regulators of ECM homeostasis have been implicated in injury susceptibility. There appears to be a lag in progression in understanding the functional significance of most of these loci. Preliminary functional theories have been presented for COL1A1, COL5A1, and for the VEGFA loci. It is critical that (i) genetic loci are explored in large data sets, (ii) explored in multiple populations, (iii) and that the functional significances of these loci are explored at the cellular and tissue levels for us to (iv) start determining the clinical relevance of these genetic contributions to normal adaptation and injury susceptibility, recovery, and tissue capacity.
One reason for a lag in expression and functional analyses, is access to pathological tissue to explore gene-phenotype correlations and impact on these tissues of time on the “remodelling curve”. Tendons and ligaments are dynamic tissues capable of responding to changing environments and the capacity to be influenced by both environmental, genetic and non-genetic factors supporting and guiding ECM remodelling. The contributions to date of genetic research on tendon and ligament injury and disease have represented no more than the identification of small pieces in a huge jigsaw puzzle. We are still to fully understand the exact position, importance, and relationships of these pieces.
Most research to date has followed a case-control genetic association approach and have been relatively restricted in location and subject and control ethnicity. The need for larger, more highly powered studies is essential. The ability to replicate the findings of the early studies in more geographical locations and representing more diverse ethnic groupings is essential. The era of omics and high throughput technologies is well-established and there is a growing trend for its application in tendon and ligament injury susceptibility. These approaches are highlighting a few of the susceptibility loci identified in the case-control studies such as TNC and COL5A1. However, the large majority of these have not been identified in GWAS, whole genome sequencing (WGS), and WES analyses.
Poorly defined cases and controls have been a limitation of the hypothesis-free approaches to date. Moving forward, cases and controls recruited to genetic studies should be classified as carefully as possible based on the most appropriate clinical and imaging assessment tools. Exposure to other confounding risk factors, an injury profile of the full spectrum of tendon and ligament or any other connective tissue injuries, medication history, pain symptoms, flexibility and other relative measurements need careful documentation. Failure to do so will reduce the probability of identifying important genetic factors at various stages of the underlying pathology, as well as improving our understanding of the capacity of the tissues to resist, respond, and recover from load.
Future work should attempt to determine the relative contribution of different genetic factors on overall risk and the potential influence of epigenetic factors affecting gene expression. Once we have more puzzle pieces fitting, we should be in an improved position towards a golden opportunity of improving patients’ care, towards reducing and preventing musculoskeletal conditions [197]. The ‘holy grail’ of genetic research in this field is a better understanding and management of soft-tissue pathology utilising laboratory-based findings during clinical work. This may arise as part of a preventative programme recognising certain individuals increased susceptibility towards developing certain disorders. Equally, it might identify which patients should respond better to different treatment strategies or improve understanding of why certain management avenues have failed.
The ethical considerations of using such data in a clinical or sporting context must be considered. Within clinical professions, understanding the subtleties of such research results is more readily appreciated when interpreted in a medical context. However, the same does not necessarily apply within a sporting context—particularly in elite sport. Ideally, polygenic profiling of identified gene candidates and scientifically assigned weighting to their importance would allow athletes to be scored to determine relative risk of developing tendon or ligament injury. Preventative programmes could be instituted to minimise risk. However, at this stage, such profiling only identifies relative risk. It carries no certainty of freedom from, or the development of, certain problems. Ethical issues will occur if athletes are selected for, or more importantly denied, sporting opportunities based on such research. Already companies exist globally providing genetic profiling and major sports organisations have screened their athletes [198]. Our level of understanding as clinicians and biologists has not yet reached the point where such developments should be condoned.

9. Conclusions

The understanding of tendon and ligament structure, function and pathology and potential genetic influences has increased dramatically in the last decade. This review highlights overlaps and distinctions in the genetic loci implicated in tendon and ligament injuries. It also highlights the need for improved study designs, including well phenotyped participants, larger samples sets, increased utilisation of next generation sequencing technologies, and functional studies of implicated loci towards improved understanding of the molecular mechanisms of these genetic loci in injury biology and their clinical significance. This would facilitate steps towards improved management of the large numbers of problems presenting in health care settings.
Our present knowledge levels are still imprecise and subject to contradictions as new emerging research and technologies appear. Consequently, our ability to translate this information into meaningful patient interventions remains limited. However, the authors are confident that, with the present level of advances in research, the ability to fuse scientific work and clinical applications will emerge in the short- to medium-term.

Author Contributions

W.J.R. conceptualized and wrote the manuscript. A.V.S. and M.C. wrote parts of the manuscript and critically revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This review article did not require any funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

The authors would like to thank their many colleagues at The Universities of Cape Town and Northampton for their support over many years of research in this area. Additionally, we are indebted to our many colleagues throughout the world who have collaborated, encouraged, and advised in our research.

Conflicts of Interest

The authors declare that no conflict of interest exists.

References

  1. Jarvinen, T.A.; Kannus, P.; Maffulli, N.; Khan, K.M. Achilles tendon disorders: Etiology and epidemiology. Foot Ankle Clin. 2005, 10, 255–266. [Google Scholar] [CrossRef] [PubMed]
  2. Herzog, M.M.; Marshall, S.W.; Lund, J.L.; Pate, V.; Spang, J.T. Cost of Outpatient Arthroscopic Anterior Cruciate Ligament Reconstruction Among Commercially Insured Patients in the United States, 2005–2013. Orthop. J. Sports Med. 2017, 5, 2325967116684776. [Google Scholar] [CrossRef] [PubMed]
  3. Carmont, M.R.; Heaver, C.; Pradhan, A.; Mei-Dan, O.; Gravare Silbernagel, K. Surgical repair of the ruptured Achilles tendon: The cost-effectiveness of open versus percutaneous repair. Knee Surg. Sports Traumatol. Arthrosc. 2013, 21, 1361–1368. [Google Scholar] [CrossRef] [PubMed]
  4. Meeuwisse, W.H. Assessing causation in sport injury: A multifactorial model. Clin. J. Sport Med. 1994, 4, 166–170. [Google Scholar] [CrossRef]
  5. Bittencourt, N.F.N.; Meeuwisse, W.H.; Mendonca, L.D.; Nettel-Aguirre, A.; Ocarino, J.M.; Fonseca, S.T. Complex systems approach for sports injuries: Moving from risk factor identification to injury pattern recognition-narrative review and new concept. Br. J. Sports Med. 2016, 50, 1309–1314. [Google Scholar] [CrossRef] [Green Version]
  6. Gibbon, W.W.; Cooper, J.R.; Radcliffe, G.S. Sonographic incidence of tendon microtears in athletes with chronic Achilles tendinosis. Br. J. Sports Med. 1999, 33, 129–130. [Google Scholar] [CrossRef] [Green Version]
  7. Austin, J.C.; Honer, W.G. Psychiatric genetic counselling for parents of individuals affected with psychotic disorders: A pilot study. Early Interv. Psychiatry 2008, 2, 80–89. [Google Scholar] [CrossRef]
  8. Khaliq, Y.; Zhanel, G.G. Fluoroquinolone-associated tendinopathy: A critical review of the literature. Clin. Infect. Dis. 2003, 36, 1404–1410. [Google Scholar] [CrossRef] [Green Version]
  9. Tsai, W.C.; Hsu, C.C.; Chen, C.P.; Chang, H.N.; Wong, A.M.; Lin, M.S.; Pang, J.H. Ciprofloxacin up-regulates tendon cells to express matrix metalloproteinase-2 with degradation of type I collagen. J. Orthop. Res. 2011, 29, 67–73. [Google Scholar] [CrossRef]
  10. Tucker, R.; Collins, M. Athletic performance and risk of injury: Can genes explain all? Dialog. Cardiovasc. Med. 2012, 17, 31–39. [Google Scholar]
  11. Maes, H.H.M.; Beunen, G.P.; Vlietinck, R.F.; Neale, M.C.; Thomis, M.; VandenEynde, B.; Lysens, R.; Simons, J.; Derom, C.; Derom, R. Inheritance of physical fitness in 10-yr-old twins and their parents. Med. Sci. Sport Exer. 1996, 28, 1479–1491. [Google Scholar] [CrossRef] [PubMed]
  12. Battie, M.C.; Levalahti, E.; Videman, T.; Burton, K.; Kaprio, J. Heritability of lumbar flexibility and the role of disc degeneration and body weight. J. Appl. Physiol. 2008, 104, 379–385. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Hakim, A.J.; Cherkas, L.F.; Spector, T.D.; MacGregor, A.J. Genetic associations between frozen shoulder and tennis elbow: A female twin study. Rheumatology 2003, 42, 739–742. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Harvie, P.; Ostlere, S.J.; Teh, J.; McNally, E.G.; Clipsham, K.; Burston, B.J.; Pollard, T.C.; Carr, A.J. Genetic influences in the aetiology of tears of the rotator cuff. Sibling risk of a full-thickness tear. J. Bone Jt. Surg. Br. 2004, 86, 696–700. [Google Scholar] [CrossRef] [Green Version]
  15. Magnusson, K.; Turkiewicz, A.; Hughes, V.; Frobell, R.; Englund, M. High genetic contribution to anterior cruciate ligament rupture: Heritability ~69. Br. J. Sports Med. 2020, 55, 385–389. [Google Scholar] [CrossRef]
  16. Hecht, J.T.; Nelson, L.D.; Crowder, E.; Wang, Y.; Elder, F.F.; Harrison, W.R.; Francomano, C.A.; Prange, C.K.; Lennon, G.G.; Deere, M.; et al. Mutations in exon 17B of cartilage oligomeric matrix protein (COMP) cause pseudoachondroplasia. Nat. Genet. 1995, 10, 325–329. [Google Scholar] [CrossRef]
  17. Gajko-Galicka, A. Mutations in type I collagen genes resulting in osteogenesis imperfecta in humans. Acta Biochim. Pol. 2002, 49, 433–441. [Google Scholar] [CrossRef] [Green Version]
  18. Collins, M.; Raleigh, S.M. Genetic Risk Factors for Musculoskeletal Soft Tissue Injuries. In Genetics and Sports, 1st ed.; Collins, M., Ed.; Karger: Basel, Switzerland, 2009; pp. 136–149. [Google Scholar]
  19. Magnan, B.; Bondi, M.; Pierantoni, S.; Samaila, E. The pathogenesis of Achilles tendinopathy: A systematic review. Foot Ankle Surg. 2014, 20, 154–159. [Google Scholar] [CrossRef]
  20. Prockop, D.J.; Chu, M.L.; de Wet, W.; Myers, J.C.; Pihlajaniemi, T.; Ramirez, F.; Sippola, M. Mutations in osteogenesis imperfecta leading to the synthesis of abnormal type I procollagens. Ann. N. Y. Acad. Sci. 1985, 460, 289–297. [Google Scholar] [CrossRef]
  21. De Paepe, A.; Nuytinck, L.; Hausser, I.; Anton-Lamprecht, I.; Naeyaert, J.M. Mutations in the COL5A1 gene are causal in the Ehlers-Danlos syndromes I and II. Am. J. Hum. Genet. 1997, 60, 547–554. [Google Scholar]
  22. Judge, D.P.; Dietz, H.C. Marfan’s syndrome. Lancet 2005, 366, 1965–1976. [Google Scholar] [CrossRef]
  23. Dawn Teare, M.; Barrett, J.H. Genetic linkage studies. Lancet 2005, 366, 1036–1044. [Google Scholar] [CrossRef]
  24. Rahim, M.; Gibbon, A.; Collins, M.; September, A.V. Genetics of musculoskeletal soft tissue injuries: Current status, challenges, and future directions. In Sports, Exercise, and Nutritional Genomics; Academic Press: Cambridge, MA, USA, 2019; pp. 317–339. [Google Scholar] [CrossRef]
  25. Baird, A.E.; Carter, S.D.; Innes, J.F.; Ollier, W.; Short, A. Genome-wide association study identifies genomic regions of association for cruciate ligament rupture in Newfoundland dogs. Anim. Genet. 2014, 45, 542–549. [Google Scholar] [CrossRef] [PubMed]
  26. Baker, L.A.; Rosa, G.J.M.; Hao, Z.; Piazza, A.; Hoffman, C.; Binversie, E.E.; Sample, S.J.; Muir, P. Multivariate genome-wide association analysis identifies novel and relevant variants associated with anterior cruciate ligament rupture risk in the dog model. BMC Genet. 2018, 19, 39. [Google Scholar] [CrossRef] [PubMed]
  27. Baker, L.A.; Kirkpatrick, B.; Rosa, G.J.; Gianola, D.; Valente, B.; Sumner, J.P.; Baltzer, W.; Hao, Z.; Binversie, E.E.; Volstad, N.; et al. Genome-wide association analysis in dogs implicates 99 loci as risk variants for anterior cruciate ligament rupture. PLoS ONE 2017, 12, e0173810. [Google Scholar] [CrossRef] [PubMed]
  28. Kim, S.K.; Roos, T.R.; Roos, A.K.; Kleimeyer, J.P.; Ahmed, M.A.; Goodlin, G.T.; Fredericson, M.; Ioannidis, J.P.; Avins, A.L.; Dragoo, J.L. Genome-wide association screens for Achilles tendon and ACL tears and tendinopathy. PLoS ONE 2017, 12, e0170422. [Google Scholar] [CrossRef] [PubMed]
  29. Caso, E.; Maestro, A.; Sabiers, C.C.; Godino, M.; Caracuel, Z.; Pons, J.; Gonzalez, F.J.; Bautista, R.; Claros, M.G.; Caso-Onzain, J.; et al. Whole-exome sequencing analysis in twin sibling males with an anterior cruciate ligament rupture. Injury 2016, 47, S41–S50. [Google Scholar] [CrossRef]
  30. Gibbon, A.; Saunders, C.J.; Collins, M.; Gamieldien, J.; September, A.V. Defining the molecular signatures of Achilles tendinopathy and anterior cruciate ligament ruptures: A whole-exome sequencing approach. PLoS ONE 2018, 13, e0205860. [Google Scholar] [CrossRef] [Green Version]
  31. Sykaras, N.; Opperman, L.A. Bone morphogenetic proteins (BMPs): How do they function and what can they offer the clinician? J. Oral Sci. 2003, 45, 57–73. [Google Scholar] [CrossRef]
  32. Thorpe, C.T.; Screen, H.R. Tendon Structure and Composition. Adv. Exp. Med. Biol. 2016, 920, 3–10. [Google Scholar] [CrossRef]
  33. Spiesz, E.M.; Thorpe, C.T.; Chaudhry, S.; Riley, G.P.; Birch, H.L.; Clegg, P.D.; Screen, H.R. Tendon extracellular matrix damage, degradation and inflammation in response to in vitro overload exercise. J. Orthop. Res. 2015, 33, 889–897. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Thorpe, C.T.; Chaudhry, S.; Lei, I.I.; Varone, A.; Riley, G.P.; Birch, H.L.; Clegg, P.D.; Screen, H.R. Tendon overload results in alterations in cell shape and increased markers of inflammation and matrix degradation. Scand. J. Med. Sci. Sports 2015, 25, e381–e391. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Russo, V.; El Khatib, M.; Prencipe, G.; Citeroni, M.R.; Faydaver, M.; Mauro, A.; Berardinelli, P.; Cervero-Varona, A.; Haidar-Montes, A.A.; Turriani, M.; et al. Tendon Immune Regeneration: Insights on the Synergetic Role of Stem and Immune Cells during Tendon Regeneration. Cells 2022, 11, 434. [Google Scholar] [CrossRef] [PubMed]
  36. Snedeker, J.G.; Foolen, J. Tendon injury and repair—A perspective on the basic mechanisms of tendon disease and future clinical therapy. Acta Biomater. 2017, 63, 18–36. [Google Scholar] [CrossRef] [PubMed]
  37. Kadler, K.E.; Hojima, Y.; Prockop, D.J. Collagen fibrils in vitro grow from pointed tips in the C- to N-terminal direction. Biochem. J. 1990, 268, 339–343. [Google Scholar] [CrossRef] [Green Version]
  38. Birch, H.L.; Bailey, J.V.; Bailey, A.J.; Goodship, A.E. Age-related changes to the molecular and cellular components of equine flexor tendons. Equine Vet. J. 1999, 31, 391–396. [Google Scholar] [CrossRef]
  39. Malfait, F.; Wenstrup, R.J.; De Paepe, A. Clinical and genetic aspects of Ehlers-Danlos syndrome, classic type. Genet. Med. 2010, 12, 597–605. [Google Scholar] [CrossRef] [Green Version]
  40. Riley, G. The pathogenesis of tendinopathy. A molecular perspective. Rheumatology 2004, 43, 131–142. [Google Scholar] [CrossRef] [Green Version]
  41. September, A.V.; Schwellnus, M.P.; Collins, M. Tendon and ligament injuries: The genetic component. Br. J. Sports Med. 2007, 41, 241–246; discussion 246. [Google Scholar] [CrossRef]
  42. Mayne, R. Cartilage collagens. What is their function, and are they involved in articular disease? Arthritis Rheum. 1989, 32, 241–246. [Google Scholar] [CrossRef]
  43. Hay, M.; Patricios, J.; Collins, R.; Branfield, A.; Cook, J.; Handley, C.J.; September, A.V.; Posthumus, M.; Collins, M. Association of type XI collagen genes with chronic Achilles tendinopathy in independent populations from South Africa and Australia. Br. J. Sports Med. 2013, 47, 569–574. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Majava, M.; Hoornaert, K.P.; Bartholdi, D.; Bouma, M.C.; Bouman, K.; Carrera, M.; Devriendt, K.; Hurst, J.; Kitsos, G.; Niedrist, D.; et al. A report on 10 new patients with heterozygous mutations in the COL11A1 gene and a review of genotype-phenotype correlations in type XI collagenopathies. Am. J. Med. Genet. A 2007, 143, 258–264. [Google Scholar] [CrossRef] [PubMed]
  45. Banos, C.C.; Thomas, A.H.; Kuo, C.K. Collagen fibrillogenesis in tendon development: Current models and regulation of fibril assembly. Birth Defects Res. C Embryo Today 2008, 84, 228–244. [Google Scholar] [CrossRef] [PubMed]
  46. Van Esch, H.; Rosser, E.M.; Janssens, S.; Van Ingelghem, I.; Loeys, B.; Menten, B. Developmental delay and connective tissue disorder in four patients sharing a common microdeletion at 6q13-14. J. Med. Genet. 2010, 47, 717–720. [Google Scholar] [CrossRef] [PubMed]
  47. Posthumus, M.; September, A.V.; Schwellnus, M.P.; Collins, M. Investigation of the Sp1-binding site polymorphism within the COL1A1 gene in participants with Achilles tendon injuries and controls. J. Sci. Med. Sport 2009, 12, 184–189. [Google Scholar] [CrossRef] [PubMed]
  48. Erduran, M.; Altinisik, J.; Meric, G.; Ates, O.; Ulusal, A.E.; Akseki, D. Is Sp1 binding site polymorphism within COL1A1 gene associated with tennis elbow? Gene 2014, 537, 308–311. [Google Scholar] [CrossRef]
  49. Collins, M.; Posthumus, M.; Schwellnus, M.P. The COL1A1 gene and acute soft tissue ruptures. Br. J. Sports Med. 2010, 44, 1063–1064. [Google Scholar] [CrossRef]
  50. Gibbon, A.; Raleigh, S.M.; Ribbans, W.J.; Posthumus, M.; Collins, M.; September, A.V. Functional COL1A1 variants are associated with the risk of acute musculoskeletal soft tissue injuries. J. Orthop. Res. 2020, 38, 2290–2298. [Google Scholar] [CrossRef]
  51. Mokone, G.G.; Schwellnus, M.P.; Noakes, T.D.; Collins, M. The COL5A1 gene and Achilles tendon pathology. Scand. J. Med. Sci. Sports 2006, 16, 19–26. [Google Scholar] [CrossRef]
  52. Mazumder, B.; Seshadri, V.; Fox, P.L. Translational control by the 3′-UTR: The ends specify the means. Trends Biochem. Sci. 2003, 28, 91–98. [Google Scholar] [CrossRef]
  53. Xie, X.; Lu, J.; Kulbokas, E.J.; Golub, T.R.; Mootha, V.; Lindblad-Toh, K.; Lander, E.S.; Kellis, M. Systematic discovery of regulatory motifs in human promoters and 3′ UTRs by comparison of several mammals. Nature 2005, 434, 338–345. [Google Scholar] [CrossRef] [PubMed]
  54. Laguette, M.J.; Abrahams, Y.; Prince, S.; Collins, M. Sequence variants within the 3′-UTR of the COL5A1 gene alters mRNA stability: Implications for musculoskeletal soft tissue injuries. Matrix Biol. 2011, 30, 338–345. [Google Scholar] [CrossRef] [PubMed]
  55. Collins, M.; Posthumus, M. Type V collagen genotype and exercise-related phenotype relationships: A novel hypothesis. Exerc. Sport Sci. Rev. 2011, 39, 191–198. [Google Scholar] [CrossRef] [PubMed]
  56. September, A.V.; Cook, J.; Handley, C.J.; van der Merwe, L.; Schwellnus, M.P.; Collins, M. Variants within the COL5A1 gene are associated with Achilles tendinopathy in two populations. Br. J. Sports Med. 2009, 43, 357–365. [Google Scholar] [CrossRef]
  57. Tashjian, R.Z.; Kim, S.K.; Roche, M.D.; Jones, K.B.; Teerlink, C.C. Genetic variants associated with rotator cuff tearing utilizing multiple population-based genetic resources. J. Shoulder Elb. Surg. 2021, 30, 520–531. [Google Scholar] [CrossRef]
  58. Abrahams, Y.; Laguette, M.J.; Prince, S.; Collins, M. Polymorphisms within the COL5A1 3’-UTR that alters mRNA structure and the MIR608 gene are associated with Achilles tendinopathy. Ann. Hum. Genet. 2013, 77, 204–214. [Google Scholar] [CrossRef]
  59. September, A.V.; Posthumus, M.; van der Merwe, L.; Schwellnus, M.; Noakes, T.D.; Collins, M. The COL12A1 and COL14A1 genes and Achilles tendon injuries. Int. J. Sports Med. 2008, 29, 257–263. [Google Scholar] [CrossRef]
  60. Saunders, C.J.; van der Merwe, L.; Posthumus, M.; Cook, J.; Handley, C.J.; Collins, M.; September, A.V. Investigation of variants within the COL27A1 and TNC genes and Achilles tendinopathy in two populations. J. Orthop. Res. 2013, 31, 632–637. [Google Scholar] [CrossRef]
  61. Jarvinen, T.A.; Jozsa, L.; Kannus, P.; Jarvinen, T.L.; Hurme, T.; Kvist, M.; Pelto-Huikko, M.; Kalimo, H.; Jarvinen, M. Mechanical loading regulates the expression of tenascin-C in the myotendinous junction and tendon but does not induce de novo synthesis in the skeletal muscle. J. Cell Sci. 2003, 116, 857–866. [Google Scholar] [CrossRef] [Green Version]
  62. Erickson, H.P. Tenascin-C, tenascin-R and tenascin-X: A family of talented proteins in search of functions. Curr. Opin. Cell Biol. 1993, 5, 869–876. [Google Scholar] [CrossRef]
  63. Riley, G.P.; Harrall, R.L.; Cawston, T.E.; Hazleman, B.L.; Mackie, E.J. Tenascin-C and human tendon degeneration. Am. J. Pathol. 1996, 149, 933–943. [Google Scholar] [PubMed]
  64. Jones, P.L.; Crack, J.; Rabinovitch, M. Regulation of tenascin-C, a vascular smooth muscle cell survival factor that interacts with the alpha v beta 3 integrin to promote epidermal growth factor receptor phosphorylation and growth. J. Cell Biol. 1997, 139, 279–293. [Google Scholar] [CrossRef] [Green Version]
  65. Jarvinen, T.A.; Jozsa, L.; Kannus, P.; Jarvinen, T.L.; Kvist, M.; Hurme, T.; Isola, J.; Kalimo, H.; Jarvinen, M. Mechanical loading regulates tenascin-C expression in the osteotendinous junction. J. Cell Sci. 1999, 112 Pt 18, 3157–3166. [Google Scholar] [CrossRef] [PubMed]
  66. Smith, R.K.; Gerard, M.; Dowling, B.; Dart, A.J.; Birch, H.L.; Goodship, A.E. Correlation of cartilage oligomeric matrix protein (COMP) levels in equine tendon with mechanical properties: A proposed role for COMP in determining function-specific mechanical characteristics of locomotor tendons. Equine Vet. J. Suppl. 2002, 34, 241–244. [Google Scholar] [CrossRef] [PubMed]
  67. Sodersten, F.; Hultenby, K.; Heinegard, D.; Johnston, C.; Ekman, S. Immunolocalization of collagens (I and III) and cartilage oligomeric matrix protein in the normal and injured equine superficial digital flexor tendon. Connect. Tissue Res. 2013, 54, 62–69. [Google Scholar] [CrossRef] [Green Version]
  68. Svensson, L.; Aszodi, A.; Heinegard, D.; Hunziker, E.B.; Reinholt, F.P.; Fassler, R.; Oldberg, A. Cartilage oligomeric matrix protein-deficient mice have normal skeletal development. Mol. Cell. Biol. 2002, 22, 4366–4371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  69. Ellis, S.G.; Dushman-Ellis, S.; Luke, M.M.; Murugesan, G.; Kottke-Marchant, K.; Ellis, G.M.; Griffin, B.; Tuzcu, E.M.; Hazen, S. Pilot candidate gene analysis of patients >/= 60 years old with aortic stenosis involving a tricuspid aortic valve. Am. J. Cardiol. 2012, 110, 88–92. [Google Scholar] [CrossRef]
  70. Saracini, C.; Bolli, P.; Sticchi, E.; Pratesi, G.; Pulli, R.; Sofi, F.; Pratesi, C.; Gensini, G.F.; Abbate, R.; Giusti, B. Polymorphisms of genes involved in extracellular matrix remodeling and abdominal aortic aneurysm. J. Vasc. Surg. 2012, 55, 171–179.e172. [Google Scholar] [CrossRef] [Green Version]
  71. Gupta, P.A.; Putnam, E.A.; Carmical, S.G.; Kaitila, I.; Steinmann, B.; Child, A.; Danesino, C.; Metcalfe, K.; Berry, S.A.; Chen, E.; et al. Ten novel FBN2 mutations in congenital contractural arachnodactyly: Delineation of the molecular pathogenesis and clinical phenotype. Hum. Mutat. 2002, 19, 39–48. [Google Scholar] [CrossRef]
  72. Mellody, K.T.; Freeman, L.J.; Baldock, C.; Jowitt, T.A.; Siegler, V.; Raynal, B.D.; Cain, S.A.; Wess, T.J.; Shuttleworth, C.A.; Kielty, C.M. Marfan syndrome-causing mutations in fibrillin-1 result in gross morphological alterations and highlight the structural importance of the second hybrid domain. J. Biol. Chem. 2006, 281, 31854–31862. [Google Scholar] [CrossRef] [Green Version]
  73. Kielty, C.M.; Baldock, C.; Lee, D.; Rock, M.J.; Ashworth, J.L.; Shuttleworth, C.A. Fibrillin: From microfibril assembly to biomechanical function. Philos. Trans. R. Soc. Lond. B Biol. Sci. 2002, 357, 207–217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Mokone, G.G.; Gajjar, M.; September, A.V.; Schwellnus, M.P.; Greenberg, J.; Noakes, T.D.; Collins, M. The guanine-thymine dinucleotide repeat polymorphism within the tenascin-C gene is associated with achilles tendon injuries. Am. J. Sports Med. 2005, 33, 1016–1021. [Google Scholar] [CrossRef] [PubMed]
  75. Saunders, C.J.; Van Der Merwe, L.; Cook, J.; Handley, C.J.; Collins, M.; September, A.V. Variants within the COMP and THBS2 genes are not associated with Achilles tendinopathy in a case-control study of South African and Australian populations. J. Sports Sci. 2014, 32, 92–100. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Kluger, R.; Burgstaller, J.; Vogl, C.; Brem, G.; Skultety, M.; Mueller, S. Candidate gene approach identifies six SNPs in tenascin-C (TNC) associated with degenerative rotator cuff tears. J. Orthop. Res. 2017, 35, 894–901. [Google Scholar] [CrossRef] [PubMed]
  77. Figueiredo, E.A.; Loyola, L.C.; Belangero, P.S.; Campos Ribeiro-Dos-Santos, A.K.; Emanuel Batista Santos, S.; Cohen, C.; Wajnsztejn, A.; Martins de Oliveira, A.; Smith, M.C.; Pochini, A.C.; et al. Rotator Cuff Tear Susceptibility Is Associated With Variants in Genes Involved in Tendon Extracellular Matrix Homeostasis. J. Orthop. Res. 2020, 38, 192–201. [Google Scholar] [CrossRef] [PubMed]
  78. El Khoury, L.; Posthumus, M.; Collins, M.; van der Merwe, W.; Handley, C.; Cook, J.; Raleigh, S.M. ELN and FBN2 gene variants as risk factors for two sports-related musculoskeletal injuries. Int. J. Sports Med. 2015, 36, 333–337. [Google Scholar] [CrossRef] [Green Version]
  79. El Khoury, L.; Ribbans, W.J.; Raleigh, S.M. Investigating the role of ELN rs2071307 gene variant as a risk factor for Achilles Tendon Pathologies in a British Cohort. S. Afr. J. Sports Med. 2016, 28, 4–5. [Google Scholar]
  80. Pruna, R.; Artells, R.; Ribas, J.; Montoro, B.; Cos, F.; Munoz, C.; Rodas, G.; Maffulli, N. Single nucleotide polymorphisms associated with non-contact soft tissue injuries in elite professional soccer players: Influence on degree of injury and recovery time. BMC Musculoskelet. Disord. 2013, 14, 221. [Google Scholar] [CrossRef] [Green Version]
  81. El Khoury, L.; Ribbans, W.J.; Raleigh, S.M. MMP3 and TIMP2 gene variants as predisposing factors for Achilles tendon pathologies: Attempted replication study in a British case-control cohort. Meta Gene 2016, 9, 52–55. [Google Scholar] [CrossRef] [Green Version]
  82. Pasternak, B.; Aspenberg, P. Metalloproteinases and their inhibitors-diagnostic and therapeutic opportunities in orthopedics. Acta Orthop. 2009, 80, 693–703. [Google Scholar] [CrossRef]
  83. El Khoury, L.; Posthumus, M.; Collins, M.; Handley, C.J.; Cook, J.; Raleigh, S.M. Polymorphic variation within the ADAMTS2, ADAMTS14, ADAMTS5, ADAM12 and TIMP2 genes and the risk of Achilles tendon pathology: A genetic association study. J. Sci. Med. Sport 2013, 16, 493–498. [Google Scholar] [CrossRef] [PubMed]
  84. Raleigh, S.M.; van der Merwe, L.; Ribbans, W.J.; Smith, R.K.; Schwellnus, M.P.; Collins, M. Variants within the MMP3 gene are associated with Achilles tendinopathy: Possible interaction with the COL5A1 gene. Br. J. Sports Med. 2009, 43, 514–520. [Google Scholar] [CrossRef] [PubMed]
  85. Gibbon, A.; Hobbs, H.; van der Merwe, W.; Raleigh, S.M.; Cook, J.; Handley, C.J.; Posthumus, M.; Collins, M.; September, A.V. The MMP3 gene in musculoskeletal soft tissue injury risk profiling: A study in two independent sample groups. J. Sports Sci. 2017, 35, 655–662. [Google Scholar] [CrossRef] [PubMed]
  86. Kim, G.K. The Risk of Fluoroquinolone-induced Tendinopathy and Tendon Rupture: What Does The Clinician Need To Know? J. Clin. Aesthet. Derm. 2010, 3, 49–54. [Google Scholar]
  87. Corps, A.N.; Harrall, R.L.; Curry, V.A.; Hazleman, B.L.; Riley, G.P. Contrasting effects of fluoroquinolone antibiotics on the expression of the collagenases, matrix metalloproteinases (MMP)-1 and -13, in human tendon-derived cells. Rheumatology 2005, 44, 1514–1517. [Google Scholar] [CrossRef] [Green Version]
  88. Rickert, M.; Wang, H.; Wieloch, P.; Lorenz, H.; Steck, E.; Sabo, D.; Richter, W. Adenovirus-mediated gene transfer of growth and differentiation factor-5 into tenocytes and the healing rat Achilles tendon. Connect. Tissue Res. 2005, 46, 175–183. [Google Scholar] [CrossRef]
  89. Schwabe, G.C.; Turkmen, S.; Leschik, G.; Palanduz, S.; Stover, B.; Goecke, T.O.; Mundlos, S. Brachydactyly type C caused by a homozygous missense mutation in the prodomain of CDMP1. Am. J. Med. Genet. A 2004, 124, 356–363. [Google Scholar] [CrossRef]
  90. Szczaluba, K.; Hilbert, K.; Obersztyn, E.; Zabel, B.; Mazurczak, T.; Kozlowski, K. Du Pan syndrome phenotype caused by heterozygous pathogenic mutations in CDMP1 gene. Am. J. Med. Genet. A 2005, 138, 379–383. [Google Scholar] [CrossRef]
  91. Basit, S.; Naqvi, S.K.; Wasif, N.; Ali, G.; Ansar, M.; Ahmad, W. A novel insertion mutation in the cartilage-derived morphogenetic protein-1 (CDMP1) gene underlies Grebe-type chondrodysplasia in a consanguineous Pakistani family. BMC Med. Genet. 2008, 9, 102. [Google Scholar] [CrossRef] [Green Version]
  92. Buchanan, C.I.; Marsh, R.L. Effects of long-term exercise on the biomechanical properties of the Achilles tendon of guinea fowl. J. Appl. Physiol. 2001, 90, 164–171. [Google Scholar] [CrossRef]
  93. Sharir, A.; Zelzer, E. Tendon homeostasis: The right pull. Curr. Biol. 2011, 21, R472–R474. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Pryce, B.A.; Watson, S.S.; Murchison, N.D.; Staverosky, J.A.; Dunker, N.; Schweitzer, R. Recruitment and maintenance of tendon progenitors by TGFbeta signaling are essential for tendon formation. Development 2009, 136, 1351–1361. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Mendias, C.L.; Gumucio, J.P.; Bakhurin, K.I.; Lynch, E.B.; Brooks, S.V. Physiological loading of tendons induces scleraxis expression in epitenon fibroblasts. J. Orthop. Res. 2012, 30, 606–612. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Posthumus, M.; Collins, M.; Cook, J.; Handley, C.J.; Ribbans, W.J.; Smith, R.K.; Schwellnus, M.P.; Raleigh, S.M. Components of the transforming growth factor-beta family and the pathogenesis of human Achilles tendon pathology—A genetic association study. Rheumatology 2010, 49, 2090–2097. [Google Scholar] [CrossRef] [Green Version]
  97. Salles, J.I.; Amaral, M.V.; Aguiar, D.P.; Lira, D.A.; Quinelato, V.; Bonato, L.L.; Duarte, M.E.; Vieira, A.R.; Casado, P.L. BMP4 and FGF3 haplotypes increase the risk of tendinopathy in volleyball athletes. J. Sci. Med. Sport 2015, 18, 150–155. [Google Scholar] [CrossRef]
  98. Teerlink, C.C.; Cannon-Albright, L.A.; Tashjian, R.Z. Significant association of full-thickness rotator cuff tears and estrogen-related receptor-beta (ESRRB). J. Shoulder Elb. Surg. 2015, 24, e31–e35. [Google Scholar] [CrossRef]
  99. Motta Gda, R.; Amaral, M.V.; Rezende, E.; Pitta, R.; Vieira, T.C.; Duarte, M.E.; Vieira, A.R.; Casado, P.L. Evidence of genetic variations associated with rotator cuff disease. J. Shoulder Elb. Surg. 2014, 23, 227–235. [Google Scholar] [CrossRef]
  100. Pearce, C.J.; Ismail, M.; Calder, J.D. Is apoptosis the cause of noninsertional achilles tendinopathy? Am. J. Sports Med. 2009, 37, 2440–2444. [Google Scholar] [CrossRef]
  101. Khan, K.M.; Cook, J.L.; Kannus, P.; Maffulli, N.; Bonar, S.F. Time to abandon the “tendinitis” myth. BMJ 2002, 324, 626–627. [Google Scholar] [CrossRef]
  102. Alfredson, H.; Thorsen, K.; Lorentzon, R. In situ microdialysis in tendon tissue: High levels of glutamate, but not prostaglandin E2 in chronic Achilles tendon pain. Knee Surg. Sports Traumatol. Arthrosc. 1999, 7, 378–381. [Google Scholar] [CrossRef]
  103. Khan, K.M.; Cook, J.L.; Bonar, F.; Harcourt, P.; Astrom, M. Histopathology of common tendinopathies. Update and implications for clinical management. Sports Med. 1999, 27, 393–408. [Google Scholar] [CrossRef] [PubMed]
  104. Khan, K.M.; Cook, J. Overuse tendon injuries: Where does the pain come from. Sports Med. Arthrosc. Rev. 2000, 8, 17–31. [Google Scholar] [CrossRef]
  105. Khan, K.M.; Cook, J.L.; Maffulli, N.; Kannus, P. Where is the pain coming from in tendinopathy? It may be biochemical, not only structural, in origin. Br. J. Sports Med. 2000, 34, 81–83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Scott, A.; Cook, J.L.; Hart, D.A.; Walker, D.C.; Duronio, V.; Khan, K.M. Tenocyte responses to mechanical loading in vivo: A role for local insulin-like growth factor 1 signaling in early tendinosis in rats. Arthritis Rheum. 2007, 56, 871–881. [Google Scholar] [CrossRef] [PubMed]
  107. Vaughn, N.H.; Stepanyan, H.; Gallo, R.A.; Dhawan, A. Genetic Factors in Tendon Injury: A Systematic Review of the Literature. Orthop. J. Sports Med. 2017, 5, 2325967117724416. [Google Scholar] [CrossRef] [Green Version]
  108. Bokhari, A.R.; Murrell, G.A. The role of nitric oxide in tendon healing. J. Shoulder Elb. Surg. 2012, 21, 238–244. [Google Scholar] [CrossRef]
  109. Lin, J.H.; Wang, M.X.; Wei, A.; Zhu, W.; Diwan, A.D.; Murrell, G.A. Temporal expression of nitric oxide synthase isoforms in healing Achilles tendon. J. Orthop. Res. 2001, 19, 136–142. [Google Scholar] [CrossRef]
  110. Pufe, T.; Petersen, W.J.; Mentlein, R.; Tillmann, B.N. The role of vasculature and angiogenesis for the pathogenesis of degenerative tendons disease. Scand. J. Med. Sci. Sports 2005, 15, 211–222. [Google Scholar] [CrossRef]
  111. Evans, I. An Overview of VEGF-Mediated Signal Transduction. Methods Mol. Biol. 2015, 1332, 91–120. [Google Scholar] [CrossRef]
  112. Bao, P.; Kodra, A.; Tomic-Canic, M.; Golinko, M.S.; Ehrlich, H.P.; Brem, H. The role of vascular endothelial growth factor in wound healing. J. Surg. Res. 2009, 153, 347–358. [Google Scholar] [CrossRef] [Green Version]
  113. Ferrara, N.; Gerber, H.P.; LeCouter, J. The biology of VEGF and its receptors. Nat. Med. 2003, 9, 669–676. [Google Scholar] [CrossRef] [PubMed]
  114. Millar, N.L.; Silbernagel, K.G.; Thorborg, K.; Kirwan, P.D.; Galatz, L.M.; Abrams, G.D.; Murrell, G.A.C.; McInnes, I.B.; Rodeo, S.A. Tendinopathy. Nat. Rev. Dis. Primers 2021, 7, 1. [Google Scholar] [CrossRef] [PubMed]
  115. Nell, E.M.; van der Merwe, L.; Cook, J.; Handley, C.J.; Collins, M.; September, A.V. The apoptosis pathway and the genetic predisposition to Achilles tendinopathy. J. Orthop. Res. 2012, 30, 1719–1724. [Google Scholar] [CrossRef] [PubMed]
  116. September, A.V.; Nell, E.M.; O’Connell, K.; Cook, J.; Handley, C.J.; van der Merwe, L.; Schwellnus, M.; Collins, M. A pathway-based approach investigating the genes encoding interleukin-1beta, interleukin-6 and the interleukin-1 receptor antagonist provides new insight into the genetic susceptibility of Achilles tendinopathy. Br. J. Sports Med. 2011, 45, 1040–1047. [Google Scholar] [CrossRef] [PubMed]
  117. Brookes, C.; Ribbans, W.J.; El Khoury, L.Y.; Raleigh, S.M. Variability within the human iNOS gene and Achilles tendon injuries: Evidence for a heterozygous advantage effect. J. Sci. Med. Sport 2020, 23, 342–346. [Google Scholar] [CrossRef] [PubMed]
  118. O’Brien, M. Structure and metabolism of tendons. Scand. J. Med. Sci. Sports 1997, 7, 55–61. [Google Scholar] [CrossRef]
  119. Mousavizadeh, R.; Khosravi, S.; Behzad, H.; McCormack, R.G.; Duronio, V.; Scott, A. Cyclic strain alters the expression and release of angiogenic factors by human tendon cells. PLoS ONE 2014, 9, e97356. [Google Scholar] [CrossRef]
  120. Nakama, L.H.; King, K.B.; Abrahamsson, S.; Rempel, D.M. VEGF, VEGFR-1, and CTGF cell densities in tendon are increased with cyclical loading: An in vivo tendinopathy model. J. Orthop. Res. 2006, 24, 393–400. [Google Scholar] [CrossRef]
  121. Petersen, W.; Varoga, D.; Zantop, T.; Hassenpflug, J.; Mentlein, R.; Pufe, T. Cyclic strain influences the expression of the vascular endothelial growth factor (VEGF) and the hypoxia inducible factor 1 alpha (HIF-1alpha) in tendon fibroblasts. J. Orthop. Res. 2004, 22, 847–853. [Google Scholar] [CrossRef]
  122. Bray, R.C.; Leonard, C.A.; Salo, P.T. Correlation of healing capacity with vascular response in the anterior cruciate and medial collateral ligaments of the rabbit. J. Orthop. Res. 2003, 21, 1118–1123. [Google Scholar] [CrossRef]
  123. Beye, J.A.; Hart, D.A.; Bray, R.C.; McDougall, J.J.; Salo, P.T. Injury-induced changes in mRNA levels differ widely between anterior cruciate ligament and medial collateral ligament. Am. J. Sports Med. 2008, 36, 1337–1346. [Google Scholar] [CrossRef] [PubMed]
  124. Rahim, M.; El Khoury, L.Y.; Raleigh, S.M.; Ribbans, W.J.; Posthumus, M.; Collins, M.; September, A.V. Human Genetic Variation, Sport and Exercise Medicine, and Achilles Tendinopathy: Role for Angiogenesis-Associated Genes. OMICS 2016, 20, 520–527. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Lambrechts, D.; Storkebaum, E.; Morimoto, M.; Del-Favero, J.; Desmet, F.; Marklund, S.L.; Wyns, S.; Thijs, V.; Andersson, J.; van Marion, I.; et al. VEGF is a modifier of amyotrophic lateral sclerosis in mice and humans and protects motoneurons against ischemic death. Nat. Genet. 2003, 34, 383–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Sobel, M.; DiCarlo, E.F.; Bohne, W.H.; Collins, L. Longitudinal splitting of the peroneus brevis tendon: An anatomic and histologic study of cadaveric material. Foot Ankle 1991, 12, 165–170. [Google Scholar] [CrossRef] [PubMed]
  127. Dudhia, J.; Scott, C.M.; Draper, E.R.; Heinegard, D.; Pitsillides, A.A.; Smith, R.K. Aging enhances a mechanically-induced reduction in tendon strength by an active process involving matrix metalloproteinase activity. Aging Cell 2007, 6, 547–556. [Google Scholar] [CrossRef]
  128. Magnusson, S.P.; Hansen, M.; Langberg, H.; Miller, B.; Haraldsson, B.; Westh, E.K.; Koskinen, S.; Aagaard, P.; Kjaer, M. The adaptability of tendon to loading differs in men and women. Int. J. Exp. Pathol. 2007, 88, 237–240. [Google Scholar] [CrossRef]
  129. Smith, R.K.; Birch, H.L.; Goodman, S.; Heinegard, D.; Goodship, A.E. The influence of ageing and exercise on tendon growth and degeneration--hypotheses for the initiation and prevention of strain-induced tendinopathies. Comp. Biochem. Physiol. A Mol. Integr. Physiol. 2002, 133, 1039–1050. [Google Scholar] [CrossRef]
  130. Tsai, W.C.; Chang, H.N.; Yu, T.Y.; Chien, C.H.; Fu, L.F.; Liang, F.C.; Pang, J.H. Decreased proliferation of aging tenocytes is associated with down-regulation of cellular senescence-inhibited gene and up-regulation of p27. J. Orthop. Res. 2011, 29, 1598–1603. [Google Scholar] [CrossRef]
  131. Peffers, M.J.; Collins, J.; Loughlin, J.; Proctor, C.; Clegg, P.D. A proteomic analysis of chondrogenic, osteogenic and tenogenic constructs from ageing mesenchymal stem cells. Stem. Cell Res. 2016, 7, 133. [Google Scholar] [CrossRef] [Green Version]
  132. Lopez-Otin, C.; Blasco, M.A.; Partridge, L.; Serrano, M.; Kroemer, G. The hallmarks of aging. Cell 2013, 153, 1194–1217. [Google Scholar] [CrossRef] [Green Version]
  133. Thorpe, C.T.; Clegg, P.D.; Birch, H.L. A review of tendon injury: Why is the equine superficial digital flexor tendon most at risk? Equine Vet. J. 2010, 42, 174–180. [Google Scholar] [CrossRef] [PubMed]
  134. Thorpe, C.T.; Udeze, C.P.; Birch, H.L.; Clegg, P.D.; Screen, H.R. Capacity for sliding between tendon fascicles decreases with ageing in injury prone equine tendons: A possible mechanism for age-related tendinopathy? Eur. Cell Mater. 2013, 25, 48–60. [Google Scholar] [CrossRef] [PubMed]
  135. Parry, D.A.; Craig, A.S.; Barnes, G.R. Tendon and ligament from the horse: An ultrastructural study of collagen fibrils and elastic fibres as a function of age. Proc. R. Soc. Lond. B Biol. Sci. 1978, 203, 293–303. [Google Scholar] [CrossRef] [PubMed]
  136. Thorpe, C.T.; Udeze, C.P.; Birch, H.L.; Clegg, P.D.; Screen, H.R. Specialization of tendon mechanical properties results from interfascicular differences. J. R. Soc. Interface 2012, 9, 3108–3117. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  137. Thorpe, C.T.; Riley, G.P.; Birch, H.L.; Clegg, P.D.; Screen, H.R.C. Fascicles and the interfascicular matrix show decreased fatigue life with ageing in energy storing tendons. Acta Biomater. 2017, 56, 58–64. [Google Scholar] [CrossRef] [Green Version]
  138. Dakin, S.G.; Dudhia, J.; Werling, N.J.; Werling, D.; Abayasekara, D.R.; Smith, R.K. Inflamm-aging and arachadonic acid metabolite differences with stage of tendon disease. PLoS ONE 2012, 7, e48978. [Google Scholar] [CrossRef] [Green Version]
  139. Wang, M.X.; Wei, A.; Yuan, J.; Clippe, A.; Bernard, A.; Knoops, B.; Murrell, G.A. Antioxidant enzyme peroxiredoxin 5 is upregulated in degenerative human tendon. Biochem. Biophys. Res. Commun. 2001, 284, 667–673. [Google Scholar] [CrossRef]
  140. Riasat, K.; Bardell, D.; Goljanek-Whysall, K.; Clegg, P.D.; Peffers, M.J. Epigenetic mechanisms in Tendon Ageing. Br. Med. Bull. 2020, 135, 90–107. [Google Scholar] [CrossRef]
  141. Khoschnau, S.; Melhus, H.; Jacobson, A.; Rahme, H.; Bengtsson, H.; Ribom, E.; Grundberg, E.; Mallmin, H.; Michaelsson, K. Type I collagen alpha1 Sp1 polymorphism and the risk of cruciate ligament ruptures or shoulder dislocations. Am. J. Sports Med. 2008, 36, 2432–2436. [Google Scholar] [CrossRef]
  142. Wang, C.; Li, H.; Chen, K.; Wu, B.; Liu, H. Association of polymorphisms rs1800012 in COL1A1 with sports-related tendon and ligament injuries: A meta-analysis. Oncotarget 2017, 8, 27627–27634. [Google Scholar] [CrossRef] [Green Version]
  143. Pluijm, S.M.; van Essen, H.W.; Bravenboer, N.; Uitterlinden, A.G.; Smit, J.H.; Pols, H.A.; Lips, P. Collagen type I alpha1 Sp1 polymorphism, osteoporosis, and intervertebral disc degeneration in older men and women. Ann. Rheum. Dis. 2004, 63, 71–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  144. Tilkeridis, C.; Bei, T.; Garantziotis, S.; Stratakis, C.A. Association of a COL1A1 polymorphism with lumbar disc disease in young military recruits. J. Med. Genet. 2005, 42, e44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  145. Stepien-Slodkowska, M.; Ficek, K.; Eider, J.; Leonska-Duniec, A.; Maciejewska-Karlowska, A.; Sawczuk, M.; Zarebska, A.; Jastrzebski, Z.; Grenda, A.; Kotarska, K.; et al. The +1245g/t polymorphisms in the collagen type I alpha 1 (col1a1) gene in polish skiers with anterior cruciate ligament injury. Biol. Sport 2013, 30, 57–60. [Google Scholar] [CrossRef] [PubMed]
  146. Stepien-Slodkowska, M.; Ficek, K.; Zietek, P.; Kaczmarczyk, M.; Lubkowska, W.; Szark-Eckardt, M.; Cieszczyk, P. Is the Combination of COL1A1 Gene Polymorphisms a Marker of Injury Risk? J. Sport Rehabil. 2017, 26, 234–238. [Google Scholar] [CrossRef]
  147. Ficek, K.; Cieszczyk, P.; Kaczmarczyk, M.; Maciejewska-Karlowska, A.; Sawczuk, M.; Cholewinski, J.; Leonska-Duniec, A.; Stepien-Slodkowska, M.; Zarebska, A.; Stepto, N.K.; et al. Gene variants within the COL1A1 gene are associated with reduced anterior cruciate ligament injury in professional soccer players. J. Sci. Med. Sport 2013, 16, 396–400. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  148. Jin, H.; Evangelou, E.; Ioannidis, J.P.; Ralston, S.H. Polymorphisms in the 5′ flank of COL1A1 gene and osteoporosis: Meta-analysis of published studies. Osteoporos. Int. 2011, 22, 911–921. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Zhao, D.; Zhang, Q.; Lu, Q.; Hong, C.; Luo, T.; Duan, Q.; Shu, S.; Lv, J.; Zhao, W. Correlations Between the Genetic Variations in the COL1A1, COL5A1, COL12A1, and beta-fibrinogen Genes and Anterior Cruciate Ligament Injury in Chinese Patients(a). J. Athl. Train. 2020, 55, 515–521. [Google Scholar] [CrossRef] [Green Version]
  150. Posthumus, M.; September, A.V.; Keegan, M.; O’Cuinneagain, D.; Van der Merwe, W.; Schwellnus, M.P.; Collins, M. Genetic risk factors for anterior cruciate ligament ruptures: COL1A1 gene variant. Br. J. Sports Med. 2009, 43, 352–356. [Google Scholar] [CrossRef] [Green Version]
  151. Stepien-Slodkowska, M.; Ficek, K.; Maciejewska-Karlowska, A.; Sawczuk, M.; Zietek, P.; Krol, P.; Zmijewski, P.; Pokrywka, A.; Cieszczyk, P. Overrepresentation of the COL3A1 AA genotype in Polish skiers with anterior cruciate ligament injury. Biol. Sport 2015, 32, 143–147. [Google Scholar] [CrossRef] [Green Version]
  152. O’Connell, K.; Knight, H.; Ficek, K.; Leonska-Duniec, A.; Maciejewska-Karlowska, A.; Sawczuk, M.; Stepien-Slodkowska, M.; O’Cuinneagain, D.; van der Merwe, W.; Posthumus, M.; et al. Interactions between collagen gene variants and risk of anterior cruciate ligament rupture. Eur. J. Sport Sci. 2015, 15, 341–350. [Google Scholar] [CrossRef]
  153. Posthumus, M.; September, A.V.; O’Cuinneagain, D.; van der Merwe, W.; Schwellnus, M.P.; Collins, M. The COL5A1 gene is associated with increased risk of anterior cruciate ligament ruptures in female participants. Am. J. Sports Med. 2009, 37, 2234–2240. [Google Scholar] [CrossRef] [PubMed]
  154. Lulinska-Kuklik, E.; Rahim, M.; Domanska-Senderowska, D.; Ficek, K.; Michalowska-Sawczyn, M.; Moska, W.; Kaczmarczyk, M.; Brzezianski, M.; Brzezianska-Lasota, E.; Cieszczyk, P.; et al. Interactions between COL5A1 Gene and Risk of the Anterior Cruciate Ligament Rupture. J. Hum. Kinet. 2018, 62, 65–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Alvarez-Romero, J.; Laguette, M.N.; Seale, K.; Jacques, M.; Voisin, S.; Hiam, D.; Feller, J.A.; Tirosh, O.; Miyamoto-Mikami, E.; Kumagai, H.; et al. Genetic variants within the COL5A1 gene are associated with ligament injuries in physically active populations from Australia, South Africa, and Japan. Eur. J. Sport Sci. 2021, 1–10. [Google Scholar] [CrossRef] [PubMed]
  156. Laguette, M.N.; Barrow, K.; Firfirey, F.; Dlamini, S.; Saunders, C.J.; Dandara, C.; Gamieldien, J.; Collins, M.; September, A.V. Exploring new genetic variants within COL5A1 intron 4-exon 5 region and TGF-beta family with risk of anterior cruciate ligament ruptures. J. Orthop. Res. 2020, 38, 1856–1865. [Google Scholar] [CrossRef] [PubMed]
  157. Stepien-Slodkowska, M.; Ficek, K.; Kaczmarczyk, M.; Maciejewska-Karlowska, A.; Sawczuk, M.; Leonska-Duniec, A.; Stepinski, M.; Zietek, P.; Krol, P.; Chudecka, M.; et al. The Variants Within the COL5A1 Gene are Associated with Reduced Risk of Anterior Cruciate Ligament Injury in Skiers. J. Hum. Kinet. 2015, 45, 103–111. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  158. Suijkerbuijk, M.A.M.; Ponzetti, M.; Rahim, M.; Posthumus, M.; Hager, C.K.; Stattin, E.; Nilsson, K.G.; Teti, A.; Meuffels, D.E.; van der Eerden, B.J.C.; et al. Functional polymorphisms within the inflammatory pathway regulate expression of extracellular matrix components in a genetic risk dependent model for anterior cruciate ligament injuries. J. Sci. Med. Sport 2019, 22, 1219–1225. [Google Scholar] [CrossRef] [PubMed]
  159. Posthumus, M.; September, A.V.; O’Cuinneagain, D.; van der Merwe, W.; Schwellnus, M.P.; Collins, M. The association between the COL12A1 gene and anterior cruciate ligament ruptures. Br. J. Sports Med. 2010, 44, 1160–1165. [Google Scholar] [CrossRef] [PubMed]
  160. Ficek, K.; Stepien-Slodkowska, M.; Kaczmarczyk, M.; Maciejewska-Karlowska, A.; Sawczuk, M.; Cholewinski, J.; Leonska-Duniec, A.; Zarebska, A.; Cieszczyk, P.; Zmijewski, P. Does the A9285G Polymorphism in Collagen Type XII alpha1 Gene Associate with the Risk of Anterior Cruciate Ligament Ruptures? Balk. J. Med. Genet. 2014, 17, 41–46. [Google Scholar] [CrossRef] [Green Version]
  161. Lv, Z.T.; Wang, W.; Zhao, D.M.; Huang, J.M. COL12A1 rs970547 Polymorphism Does Not Alter Susceptibility to Anterior Cruciate Ligament Rupture: A Meta-Analysis. Front. Genet. 2021, 12, 665861. [Google Scholar] [CrossRef]
  162. Lulinska-Kuklik, E.; Laguette, M.N.; Moska, W.; Weber-Rajek, M.; Ficek, K.; Puchala, R.; Cieszczyk, P.; Sawczuk, M.; September, A.V.; Maciejewska-Skrendo, A. Are TNC gene variants associated with anterior cruciate ligament rupture susceptibility? J. Sci. Med. Sport 2019, 22, 408–412. [Google Scholar] [CrossRef] [Green Version]
  163. Mannion, S.; Mtintsilana, A.; Posthumus, M.; van der Merwe, W.; Hobbs, H.; Collins, M.; September, A.V. Genes encoding proteoglycans are associated with the risk of anterior cruciate ligament ruptures. Br. J. Sports Med. 2014, 48, 1640–1646. [Google Scholar] [CrossRef] [PubMed]
  164. Willard, K.; Laguette, M.N.; Alves de Souza Rios, L.; D’Alton, C.; Nel, M.; Prince, S.; Collins, M.; September, A.V. Altered expression of proteoglycan, collagen and growth factor genes in a TGF-beta1 stimulated genetic risk model for musculoskeletal soft tissue injuries. J. Sci. Med. Sport 2020, 23, 695–700. [Google Scholar] [CrossRef] [PubMed]
  165. Cieszczyk, P.; Willard, K.; Gronek, P.; Zmijewski, P.; Trybek, G.; Gronek, J.; Weber-Rajek, M.; Stastny, P.; Petr, M.; Lulinska-Kuklik, E.; et al. Are genes encoding proteoglycans really associated with the risk of anterior cruciate ligament rupture? Biol. Sport 2017, 34, 97–103. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  166. Willard, K.; Mannion, S.; Saunders, C.J.; Collins, M.; September, A.V. The interaction of polymorphisms in extracellular matrix genes and underlying miRNA motifs that modulate susceptibility to anterior cruciate ligament rupture. J. Sci. Med. Sport 2018, 21, 22–28. [Google Scholar] [CrossRef] [PubMed]
  167. Posthumus, M.; Collins, M.; van der Merwe, L.; O’Cuinneagain, D.; van der Merwe, W.; Ribbans, W.J.; Schwellnus, M.P.; Raleigh, S.M. Matrix metalloproteinase genes on chromosome 11q22 and the risk of anterior cruciate ligament (ACL) rupture. Scand. J. Med. Sci. Sports 2012, 22, 523–533. [Google Scholar] [CrossRef]
  168. Malila, S.; Yuktanandana, P.; Saowaprut, S.; Jiamjarasrangsi, W.; Honsawek, S. Association between matrix metalloproteinase-3 polymorphism and anterior cruciate ligament ruptures. Genet. Mol. Res. 2011, 10, 4158–4165. [Google Scholar] [CrossRef]
  169. Lulinska, E.; Gibbon, A.; Kaczmarczyk, M.; Maciejewska-Skrendo, A.; Ficek, K.; Leonska-Duniec, A.; Wilk, M.; Leznicka, K.; Michalowska-Sawczyn, M.; Huminska-Lisowska, K.; et al. Matrix Metalloproteinase Genes (MMP1, MMP10, MMP12) on Chromosome 11q22 and the Risk of Non-Contact Anterior Cruciate Ligament Ruptures. Genes 2020, 11, 766. [Google Scholar] [CrossRef]
  170. Raleigh, S.M.; Posthumus, M.; O’Cuinneagain, D.; van der Merwe, W.; Collins, M. The GDF5 gene and anterior cruciate ligament rupture. Int. J. Sports Med. 2013, 34, 364–367. [Google Scholar] [CrossRef]
  171. Chen, B.; Li, B.; Qi, Y.J.; Tie, K.; Chen, L.B. Association study between growth differentiation factor 5 polymorphism and non-contact anterior cruciate ligament rupture in Chinese Han population. Int. J. Clin. Exp. Med. 2015, 8, 22484–22490. [Google Scholar]
  172. Rahim, M.; Mannion, S.; Klug, B.; Hobbs, H.; van der Merwe, W.; Posthumus, M.; Collins, M.; September, A.V. Modulators of the extracellular matrix and risk of anterior cruciate ligament ruptures. J. Sci. Med. Sport 2017, 20, 152–158. [Google Scholar] [CrossRef]
  173. Lulinska-Kuklik, E.; Maculewicz, E.; Moska, W.; Ficek, K.; Kaczmarczyk, M.; Michalowska-Sawczyn, M.; Huminska-Lisowska, K.; Buryta, M.; Chycki, J.; Cieszczyk, P.; et al. Are IL1B, IL6 and IL6R Gene Variants Associated with Anterior Cruciate Ligament Rupture Susceptibility? J. Sports Sci. Med. 2019, 18, 137–145. [Google Scholar] [PubMed]
  174. Rahim, M.; Gibbon, A.; Hobbs, H.; van der Merwe, W.; Posthumus, M.; Collins, M.; September, A.V. The association of genes involved in the angiogenesis-associated signaling pathway with risk of anterior cruciate ligament rupture. J. Orthop. Res. 2014, 32, 1612–1618. [Google Scholar] [CrossRef] [PubMed]
  175. Seale, K.; Burger, M.; Posthumus, M.; Hager, C.K.; Stattin, E.; Nilsson, K.G.; Collins, M.; September, A.V. The Apoptosis Pathway and CASP8 Variants Conferring Risk for Acute and Overuse Musculoskeletal Injuries. J. Orthop. Res. 2020, 38, 680–688. [Google Scholar] [CrossRef] [PubMed]
  176. Rahim, M.; Lacerda, M.; Collins, M.; Posthumus, M.; September, A.V. Risk modelling further implicates the angiogenesis pathway in anterior cruciate ligament ruptures. Eur. J. Sport Sci. 2022, 22, 650–657. [Google Scholar] [CrossRef] [PubMed]
  177. Rahim, M.; Hobbs, H.; van der Merwe, W.; Posthumus, M.; Collins, M.; September, A.V. Investigation of angiogenesis genes with anterior cruciate ligament rupture risk in a South African population. J. Sports Sci. 2018, 36, 551–557. [Google Scholar] [CrossRef]
  178. Lulinska-Kuklik, E.; Leznicka, K.; Huminska-Lisowska, K.; Moska, W.; Michalowska-Sawczyn, M.; Ossowski, Z.; Maculewicz, E.; Cieszczyk, P.; Kaczmarczyk, M.; Ratkowski, W.; et al. The VEGFA gene and anterior cruciate ligament rupture risk in the Caucasian population. Biol. Sport 2019, 36, 3–8. [Google Scholar] [CrossRef]
  179. Wezenbeek, E.; Mahieu, N.; Willems, T.M.; Van Tiggelen, D.; De Muynck, M.; De Clercq, D.; Witvrouw, E. What does normal tendon structure look like? New insights into tissue characterization in the Achilles tendon. Scand. J. Med. Sci. Sports 2017, 27, 746–753. [Google Scholar] [CrossRef]
  180. Yoshikawa, T.; Tohyama, H.; Katsura, T.; Kondo, E.; Kotani, Y.; Matsumoto, H.; Toyama, Y.; Yasuda, K. Effects of local administration of vascular endothelial growth factor on mechanical characteristics of the semitendinosus tendon graft after anterior cruciate ligament reconstruction in sheep. Am. J. Sports Med. 2006, 34, 1918–1925. [Google Scholar] [CrossRef]
  181. Wang, Y.; Zheng, Y.; Zhang, W.; Yu, H.; Lou, K.; Zhang, Y.; Qin, Q.; Zhao, B.; Yang, Y.; Hui, R. Polymorphisms of KDR gene are associated with coronary heart disease. J. Am. Coll. Cardiol. 2007, 50, 760–767. [Google Scholar] [CrossRef]
  182. Feldmann, D.C.; Rahim, M.; Suijkerbuijk, M.A.M.; Laguette, M.N.; Cieszczyk, P.; Ficek, K.; Huminska-Lisowska, K.; Hager, C.K.; Stattin, E.; Nilsson, K.G.; et al. Investigation of multiple populations highlight VEGFA polymorphisms to modulate anterior cruciate ligament injury. J. Orthop. Res. 2021. [Google Scholar] [CrossRef]
  183. Patterson, N.; Petersen, D.C.; van der Ross, R.E.; Sudoyo, H.; Glashoff, R.H.; Marzuki, S.; Reich, D.; Hayes, V.M. Genetic structure of a unique admixed population: Implications for medical research. Hum. Mol. Genet. 2010, 19, 411–419. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  184. de Wit, E.; Delport, W.; Rugamika, C.E.; Meintjes, A.; Moller, M.; van Helden, P.D.; Seoighe, C.; Hoal, E.G. Genome-wide analysis of the structure of the South African Coloured Population in the Western Cape. Hum. Genet. 2010, 128, 145–153. [Google Scholar] [CrossRef] [PubMed]
  185. Greeff, J.M. Deconstructing Jaco: Genetic heritage of an Afrikaner. Ann. Hum. Genet. 2007, 71, 674–688. [Google Scholar] [CrossRef] [PubMed]
  186. Geldenhuys, G.; Glanzmann, B.; Lombard, D.; Boolay, S.; Carr, J.; Bardien, S. Identification of a common founder couple for 40 South African Afrikaner families with Parkinson’s disease. S. Afr. Med. J. 2014, 104, 413–419. [Google Scholar] [CrossRef] [Green Version]
  187. Waddington, C.H. Towards a theoretical biology. Nature 1968, 218, 525–527. [Google Scholar] [CrossRef]
  188. Petronis, A. Epigenetics as a unifying principle in the aetiology of complex traits and diseases. Nature 2010, 465, 721–727. [Google Scholar] [CrossRef]
  189. Thankam, F.G.; Boosani, C.S.; Dilisio, M.F.; Agrawal, D.K. Epigenetic mechanisms and implications in tendon inflammation (Review). Int. J. Mol. Med. 2019, 43, 3–14. [Google Scholar] [CrossRef] [Green Version]
  190. Rickaby, R.; El Khoury, L.Y.; Samiric, T.; Raleigh, S.M. Epigenetic Status of The Human MMP11 Gene Promoter is Altered in Patellar Tendinopathy. J. Sports Sci. Med. 2019, 18, 155–159. [Google Scholar]
  191. El Khoury, L.Y.; Rickaby, R.; Samiric, T.; Raleigh, S.M. Promoter methylation status of the TIMP2 and ADAMTS4 genes and patellar tendinopathy. J. Sci. Med. Sport 2018, 21, 378–382. [Google Scholar] [CrossRef] [Green Version]
  192. Parkinson, J.; Samiric, T.; Ilic, M.Z.; Cook, J.; Feller, J.A.; Handley, C.J. Change in proteoglycan metabolism is a characteristic of human patellar tendinopathy. Arthritis Rheum. 2010, 62, 3028–3035. [Google Scholar] [CrossRef]
  193. Attia, M.; Scott, A.; Carpentier, G.; Lian, O.; Van Kuppevelt, T.; Gossard, C.; Papy-Garcia, D.; Tassoni, M.C.; Martelly, I. Greater glycosaminoglycan content in human patellar tendon biopsies is associated with more pain and a lower VISA score. Br. J. Sports Med. 2014, 48, 469–475. [Google Scholar] [CrossRef] [PubMed]
  194. Leal, M.F.; Caires Dos Santos, L.; Martins de Oliveira, A.; Santoro Belangero, P.; Antonio Figueiredo, E.; Cohen, C.; de Seixas Alves, F.; Hiromi Yanaguizawa, W.; Vicente Andreoli, C.; de Castro Pochini, A.; et al. Epigenetic regulation of metalloproteinases and their inhibitors in rotator cuff tears. PLoS. ONE. 2017, 12, e0184141. [Google Scholar] [CrossRef] [PubMed]
  195. Slusarz, A.; Pulakat, L. The two faces of miR-29. J. Cardiovasc. Med. 2015, 16, 480–490. [Google Scholar] [CrossRef]
  196. van Wijnen, A.J.; Westendorf, J.J. Epigenetics as a New Frontier in Orthopedic Regenerative Medicine and Oncology. J. Orthop. Res. 2019, 37, 1465–1474. [Google Scholar] [CrossRef] [PubMed]
  197. September, A.; Rahim, M.; Collins, M. Towards an Understanding of the Genetics of Tendinopathy. Adv. Exp. Med. Biol. 2016, 920, 109–116. [Google Scholar] [CrossRef]
  198. Marsh, B. DNA clue to football injuries. The Sunday Times, 16 October 2011. [Google Scholar]
Figure 1. A hypothetical diagram showing the complex interaction of numerous genetic factors and extrinsic factors in determining an individual’s specific profile along the ‘reduced to increased risk (predisposed) spectrum’.
Figure 1. A hypothetical diagram showing the complex interaction of numerous genetic factors and extrinsic factors in determining an individual’s specific profile along the ‘reduced to increased risk (predisposed) spectrum’.
Life 12 00663 g001
Figure 2. The Jar Model. Accumulative risk factors for developing chronic tendon and ligament disease. Adapted from the ‘Jar Model’ of Jehannine Austin used in psychiatric genetic counselling (Austin, 2008) [7].
Figure 2. The Jar Model. Accumulative risk factors for developing chronic tendon and ligament disease. Adapted from the ‘Jar Model’ of Jehannine Austin used in psychiatric genetic counselling (Austin, 2008) [7].
Life 12 00663 g002
Figure 3. Hierarchical structure of the tendon.
Figure 3. Hierarchical structure of the tendon.
Life 12 00663 g003
Table 1. Principal tendon collagen types.
Table 1. Principal tendon collagen types.
Collagen TypeFunctionsAssociated Diseases
Type I
  • Major tendon in collagen—90%
  • Primary structural elements
  • Rare mutations within COL1A1 and COL1A2 genes, which encode for the α1 and α2 chains of type I collagen, respectively, cause osteogenesis imperfecta (OGI) [17]
Type III
  • Regulates type I collagen size (fibrillogenesis) [37]
  • Increases in ageing tendons [38]
  • Important role in flexibility and tissue strength
  • Active in early wound healing and, later, gradually replaced by type I collagen as the wound matures
  • COL3A1 mutations have been found to cause vascular type of Ehlers–Danlos syndrome (EDS) [39]
Type V
  • Similar to type III
  • Regulates fibril assembly and diameter and, thus, affects tendon mechanical properties [40]
  • Content increases with age and in degenerative conditions [41]
  • Common cause of EDS are mutations that inactivates one copy of COL5A1 (known as a haploinsufficiency) [39]
Type XI
  • Regulates type I and type II collagen fibrillogenesis by maintaining fibril spacing and diameter
  • Although predominately expressed together with type II collagen, type XI collagen shares structural and functional homology with type V collagen and expressed in developing tendons [42]
  • Proposed that genetic variants controlling type XI and V collagen production interact to regulate type I collagen fibril assembly [43]
  • Mutations in type XI collagen genes (COL11A1 and COL11A2) associated with Stickler syndrome, Marshall syndrome, fibrochondrogenesis, otospondylomegaepiphyseal dysplasia deafness, and Weissenbacher–Zweymüller syndrome [44]
Types XII and XIV
  • Type XII and XIV collagens are associated with type I collagen fibrils forming a molecular bridge between structural collagen and other ECM molecules [45]
  • Type XIV assists collagen fibrillogenesis regulation
  • Microdeletions on long arm of chromosome 6 containing COL12A1 gene cause developmental delay, mild dysmorphism and connective tissue laxity [46]
Table 8. Tendon disease: Inflammatory cascade, apoptosis, and other elements.
Table 8. Tendon disease: Inflammatory cascade, apoptosis, and other elements.
TypeElements or ActivityFunctionsKey Elements
Inflammatory Cascade and ApoptosisInterleukins (ILs)
  • Intimately involved in the inflammatory pathway
  • Interleukin-1ß (IL1B) influences cellular death, proliferation, and differentiation
  • Interleukin-1 receptor antagonist (ILRN) inhibits activities of both IL1B and interleukin-1α (IL1A)
  • Interleukin-6 (IL6) is found in both acute and chronic inflammation
Caspases (CASP)
  • Family of protease enzymes intimately involved in apoptosis
  • CASP8 is a pro-enzyme central to cell death regulation
Nitric Oxide Synthases (NOS)
  • Nitric oxide (NO), produced by NOS enzymes, is a free radical and important cellular signalling messenger with many functions including increased production of IL6 and Il8 [107]
  • Three NOS genes, NOS1, NOS2, and NOS3, encode for nNOS, iNOS and eNOS enzymes, respectively
  • These genes are designed to code for differing elements of the inflammatory cascade and control over cell death
  • Gene variations associated with alterations in susceptibility to tendinopathy [108]
  • NOS2 expression elevated 23-fold higher than controls within days of Achilles’ tendon injury
  • NO catalysed by the iNOS isoform of NOS induces apoptosis in inflammatory cells to eradicate cells from damaged area, preventing chronic inflammation and allowing remodelling to occur [109]
Angiogenesis
  • Marked increase in new blood vessels in chronic tendon disease [110]
  • Vascular endothelial growth factor A (VEGFA)
  • Kinase domain receptor (KDR)
  • Hypoxia inducible factor 1 subunit alpha (HIFIA)
  • Encoded by VEGFA, KDR, and HIF1A genes, respectively [111,112,113]
OthersEstrogen-related receptor beta (ERR-β)
  • Function unknown
  • May influence the expression of PPARGC1 and ESRR-inducing regulator muscle 1 (PERM1) in skeletal muscle
Defensins
  • Microbicidal and cytotoxic peptides made by neutrophils
Table 10. Summary of genetic research and ligament injuries: Collagen types I and III.
Table 10. Summary of genetic research and ligament injuries: Collagen types I and III.
Collagen TypesReferenceCountryVariantPathologySubjectsControlsComments
Type I[150]South AfricaCOL1A1 rs1800012ACL rupture117130Significant association
[147]PolandCOL1A1 rs1107946, rs1800012ACL rupture91143Significant association
[146]PolandCOL1A1 rs1107946, rs1800012ACL rupture138183Significant association
[142]Meta analysesCOL1A1 rs1800012Tendon and ligament injuries9331381Significant association
[149]Chinese Yunnan HanCOL1A1 rs1800012ACL rupture101110No significant association
[50]Combined population from SA, Sweden, Poland, FinlandCOL1A1 rs1800012ACL rupture and
Cruciate ligament rupture
1425407Significant association
Type III[151]PolandCOL3A1 rs1800255ACL rupture138183Significant association
[152]South Africa, PolandCOL3A1 rs1800255ACL rupture333378Significant association
Green indicates an association and red indicates no association found.
Table 11. Summary of genetic research and ligament injuries: Collagen types V and XII.
Table 11. Summary of genetic research and ligament injuries: Collagen types V and XII.
Collagen TypesReferenceCountryVariantPathologySubjectsControlsComments
Type V[153]South AfricaCOL5A1 rs12722, rs13946ACL rupture129216Significant association
[157]PolandCOL5A1 rs12722, rs13946ACL rupture138183Associated with reduced injury risk
[152]South Africa and PolandCOL5A1 rs12722ACL rupture333378Significant gene-gene association
[154]PolandCOL5A1 rs12722, rs13946ACL rupture134211Significant association
[158]South Africa and SwedenCOL5A1 rs12722ACL rupture98
79
116Significant association
[156]South AfricaCOL5A1 rs3922912, rs4841926ACL rupture249210No significant association
[149]Chinese Yunnan HanCOL5A1 rs12722, rs13946ACL rupture101110No significant association
[155]South Africa, Australia, JapanCOL5A1 rs12722, rs10628678ACL rupture and Ligament injury311592Significant association
Type XII[159]South AfricaCOL12A1 rs970547ACL rupture129216Significant association
[160]PolandCOL12A1 rs970547ACL rupture91143No significant association
[152]South Africa and PolandCOL12A1 rs970547ACL rupture333378Significant association
[149]Chinese Yunnan HanCOL12A1 rs970547, rs240736ACL rupture101110Significant association
Green indicates an association and red indicates no association found.
Table 12. Summary of genetic research and ligament injuries: Glycoproteins in the ECM and proteoglycans.
Table 12. Summary of genetic research and ligament injuries: Glycoproteins in the ECM and proteoglycans.
TypeProteinReferenceCountryVariantPathologySubjectsControlsComments
Glycoproteins in the ECMTenascin C (TNC)[30]South AfricaTNC rs1061494, rs1138545, rs2104772, rs1061495ACL rupture234232Significant association
[162]PolandTNC rs1330363, rs2104772, rs13321 ACL rupture229192No significant association
ProteoglycansAggrecan (ACAN)[163]South AfricaACAN rs2351491, rs1042631, rs1516797ACL rupture227234Significant association
[165]PolandACAN rs2351491 ACL rupture143229Significant association
Biglycan (BGN)[163]South AfricaBGN rs11264797, rs1126499, rs1042103ACL rupture227234Significant association
[165]PolandBGN rs11264797, rs1042103ACL rupture143229Significant association
[166]South AfricaBGN rs11264797, rs1042103ACL rupture227234Significant gene-gene interactions
Decorin (DCN)
Lumican (LUM)
Fibromodulin (FMOD)
[163]South AfricaDCN rs13312816, rs516115
LUM rs2268578
FMOD rs7543148
ACL rupture227234Significant association
Green indicates an association and red indicates no association found.
Table 14. Summary of genetic research and tendon injuries: Angiogenesis and fibrinogens.
Table 14. Summary of genetic research and tendon injuries: Angiogenesis and fibrinogens.
TypeProteinReferenceCountryVariantPathologySubjectsControlsComments
Angiogenic FactorsVascular endothelial growth factor receptor (KDR)
Vascular endothelial growth factor (VEGF)
Beta-nerve growth factor (NGF)
Hypoxia-inducible factor (HIF)
[174]South AfricaKDR rs2071559,
rs1870377
VEGFA rs699947,
rs1570360, rs2010963
NGFB rs6678788
HIF1A rs11549465
ACL rupture227126Significant association
[177]South AfricaKDR rs2071559,
rs1870377
VEGFA rs699947, rs1570360, rs2010963
ACL rupture98100Significant association
[176]South AfricaKDR rs2071559, rs1870377
VEGFA rs699947, rs1570360, rs2010963
ACL rupture234232Significant association
[178]PolandVEGFA rs699947, rs1570360, rs2010963ACL rupture222190Significant association for rs2010963
[182]Sweden, Poland, Australia and South Africa VEGFA rs699947, rs1570360, rs2010963 KDR rs2071559,
rs1870377
ACL rupture912765Significant association for rs201093 and the rs699947, rs57036 rs2010963 haplotype
FibrinogensFibrinogen beta chain (FGB)[149]Chinese Yunnan HanFGB rs1800789, rs1800790, rs1800791, rs2227389ACL rupture101110No significant association
Green indicates an association and red indicates no association found.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Ribbans, W.J.; September, A.V.; Collins, M. Tendon and Ligament Genetics: How Do They Contribute to Disease and Injury? A Narrative Review. Life 2022, 12, 663. https://doi.org/10.3390/life12050663

AMA Style

Ribbans WJ, September AV, Collins M. Tendon and Ligament Genetics: How Do They Contribute to Disease and Injury? A Narrative Review. Life. 2022; 12(5):663. https://doi.org/10.3390/life12050663

Chicago/Turabian Style

Ribbans, William J., Alison V. September, and Malcolm Collins. 2022. "Tendon and Ligament Genetics: How Do They Contribute to Disease and Injury? A Narrative Review" Life 12, no. 5: 663. https://doi.org/10.3390/life12050663

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop