Next Article in Journal
Beneficial Effects of Vitamins K and D3 on Redox Balance of Human Osteoblasts Cultured with Hydroxyapatite-Based Biomaterials
Next Article in Special Issue
The Adenosine A2B Receptor Drives Osteoclast-Mediated Bone Resorption in Hypoxic Microenvironments
Previous Article in Journal
The Potential Use of Metformin, Dipyridamole, N-Acetylcysteine and Statins as Adjunctive Therapy for Systemic Lupus Erythematosus
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Importance of the Knee Joint Meniscal Fibrocartilages as Stabilizing Weight Bearing Structures Providing Global Protection to Human Knee-Joint Tissues

by
James Melrose
1,2,3,4
1
Raymond Purves Bone and Joint Research Laboratory, Kolling Institute, Northern Sydney Local Health District, St. Leonards, NSW 2065, Australia
2
Graduate School of Biomedical Engineering, University of New South Wales, Sydney, NSW 2052, Australia
3
Sydney Medical School, Northern, University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
4
Faculty of Medicine and Health, University of Sydney, Royal North Shore Hospital, St. Leonards, NSW 2065, Australia
Cells 2019, 8(4), 324; https://doi.org/10.3390/cells8040324
Submission received: 21 March 2019 / Revised: 3 April 2019 / Accepted: 3 April 2019 / Published: 6 April 2019
(This article belongs to the Special Issue Pathways Contributing to Cartilage and Bone Destruction in Arthritis)

Abstract

:
The aim of this study was to review aspects of the pathobiology of the meniscus in health and disease and show how degeneration of the meniscus can contribute to deleterious changes in other knee joint components. The menisci, distinctive semilunar weight bearing fibrocartilages, provide knee joint stability, co-ordinating functional contributions from articular cartilage, ligaments/tendons, synovium, subchondral bone and infra-patellar fat pad during knee joint articulation. The meniscus contains metabolically active cell populations responsive to growth factors, chemokines and inflammatory cytokines such as interleukin-1 and tumour necrosis factor-alpha, resulting in the synthesis of matrix metalloproteases and A Disintegrin and Metalloprotease with ThromboSpondin type 1 repeats (ADAMTS)-4 and 5 which can degrade structural glycoproteins and proteoglycans leading to function-limiting changes in meniscal and other knee joint tissues. Such degradative changes are hall-marks of osteoarthritis (OA). No drugs are currently approved that change the natural course of OA and translate to long-term, clinically relevant benefits. For any pharmaceutical therapeutic intervention in OA to be effective, disease modifying drugs will have to be developed which actively modulate the many different cell types present in the knee to provide a global therapeutic. Many individual and combinatorial approaches are being developed to treat or replace degenerate menisci using 3D printing, bioscaffolds and hydrogel delivery systems for therapeutic drugs, growth factors and replacement progenitor cell populations recognising the central role the menisci play in knee joint health.

Graphical Abstract

1. Introduction

Osteoarthritis is responsible for a large and rapidly increasing global disease burden that is challenging health-care systems worldwide. The need for improved and rapid processes to develop new therapies, is obvious. Disease progression in osteoathritis, is marked by increasing pain and loss of joint function, however the development of osteoarthritis is usually slow, taking many years from its first mild symptoms to severe debilitating end-stage disease. Although progression to this end-state occurs in only a minority of patients with osteoarthritis and disease in many patients remains stable for many years, each year osteoarthritis led to ~160,000 total joint replacements in England and Wales and 492,000 in the USA in 2017 [1]. These numbers are expected to increase dramatically over the next two decades with global ageing trends effecting population demographics with a greater proportion of elderly OA prone patients becoming represented in the general population. Osteoarthritis is thus projected to become one of the leading musculoskeletal conditions by 2050.
The semilunar meniscal fibrocartilaginous menisci provide joint congruity to the curved weight bearing surfaces of the articular cartilages and collectively these structures provide weight bearing properties to the human knee joint [2] (Figure 1). The menisci are also designed to withstand shear forces and to withstand multidirectional stresses which are generated in with weight bearing and the torsional forces generated during normal knee articulation [3]. Complex collagenous fibrillar arrangements in the menisci are formed into lamellar structures and circumferential and radial tie bundles to provide the required material properties to this tissue [4,5]. Type I collagen is a major fibrillar component in the meniscus while type VI collagen forms pericellular collagenous structures around strings of meniscal cells and may have biomechanical sensory roles providing cell matrix feedback cues which allow the meniscal cells to regulate meniscal composition and tissue homeostasis. Several proteoglycans have been identified in the meniscus including aggrecan [6,7,8], versican [9,10], perlecan [11], decorin, biglycan, fibromodulin, lumican, keratocan [6,12,13,14] and lubricin (PRG4) [15,16,17]. These have roles in weight-bearing, regulation of collagen fibrillogenesis and lubrication of the meniscal surface.
The β-defensins are 2–6 kDa cationic peptides which bind to meniscal proteoglycans and have anti-bacterial, ant-fungal and anti-viral activities which have protective roles to play in human meniscal tissues. β-Defensins are significantly upregulated during the inflammatory conditions which are generated in the knee-joint during OA and act in concert with DAMP (damage associated molecular pattern) and PAMP (pathogen associated molecular pattern) receptors such as the Toll-like and NOD-like receptors of the innate immune system which identify lipopolysaccharide and peptidoglycan components on cell membranes of invading organisms [19,20] and this response protects meniscal tissues from microbial invasion [21]. Aquaporin1 (AQP1) is a specific water transport channel protein expressed by articular chondrocytes, meniscal cells and synoviocytes in the knee-joint which is upregulated during OA and RA [21]. This may represent a new molecular target in therapeutic procedures aimed at maintaining the hydration and functional viscoelastic hydrodynamic properties of knee joint tissues during these arthritic conditions [22].

2. Studies on Meniscal Tissues in Health and Disease

2.1. Development of High Resolution Imaging and Sensitive Biomechanical Methodologies for the Analysis of Knee Joint Tissues

Meniscal biology in health and disease [2,3,4,5] and novel synovial fluid biomarkers associated with meniscal pathology have been identified [23] including synovial fluid cytokine profiles in chronic meniscal tears of the knee [24]. Attempts have been made to correlate meniscal damage with the pathological status of knee joint tissues using a number of imaging techniques including radiography [25], μCT imaging and histology [26], semiquantitative imaging of biomarkers of knee OA progression [27,28], synchrotron-based X-ray μCT imaging of biomarkers [29] and by comparison of molecular biomarkers with magnetic resonance (MR)-based cartilage composition and knee joint morphology [30]. Phase-contrast μCT is an emerging imaging technique, which is able to resolve 3D micro-scale structures in tissue samples without the need for staining or sectioning but also complements conventional histology providing additional information on the fine structure of meniscal tissues. 3D-mapping of the joint space for the diagnosis of knee OA using high resolution computed tomography has compared radiographic, normal and degenerate meniscal pathology [31]. Bioluminescence and second harmonic generation imaging reveal dynamic changes in the inflammatory and collagen landscape in early OA [32]. Ex vivo quantitative multiparametric magnetic resonance imaging (MRI) mapping has been used to assess human meniscus degeneration [33]. Ex vivo quantitative multiparametric MRI mapping uses 3.0-T MRI using inversion-recovery (T1), spin-lock multi-gradient-echo (T1ρ), multi-spin-echo (T2) and multi-gradient-echo (T2* and UTE-T2*) sequences to determine relaxation times of quantitative MRI (qMRI) parameters to provide information on the structure of normal and degenerate meniscal tissues. Biomechanical properties of the murine meniscus surface using atomic force microscopy (AFM)-based nano-indentation methodology has been used to establish topographical structure-function relationships in pathological versus normal menisci [34]. Nanoindentation AFM techniques offer high spatial resolution and force sensitivity for the measurement of the mechanical properties of biomaterials and tissues. Laboratory-based X-ray phase-contrast μCT imaging has been applied to the imaging of mouse articular cartilage in models of OA and is a promising research tool for the identification of new OA therapeutic targets at higher spatial resolution than possible with MRI techniques offering the possibility of the detection of early cartilage lesions which if untreated will develop into advanced OA lesions [35]. With the difficulty of obtaining time on synchrotron facilities for imaging, laboratory based X-ray μCT scanners are being developed to provide images of comparable quality to synchrotron images [35].

2.2. Identification of Biomarkers of Degenerate Meniscal Pathology

Several prospective biomarkers have been correlated with various criteria of meniscal pathology including synovial fluid HMGB (high mobility group box)-1 levels [36,37], inflammatory cytokines and biomarkers of cartilage metabolism 8 years after ACL (anterior cruciate ligament) reconstruction in operated and contralateral knees [38], synovial chemokine expression [34], synovial fluid MMP activity in torn menisci [39] and transcriptomic analysis of synovial extracellular RNA conducted following knee trauma [40]. These studies have suggested probable roles for specific biomolecular markers in meniscal degeneration. Meniscal cells are responsive to the inflammatory cytokines IL-1α and TNFα in an ovine in-vitro model where zonal analyses can also be undertaken to compare the phenotypic characteristics of inner and outer zone meniscal cells [41,42]. This demonstrated the involvement of meniscal pathology in OA development and its contribution to global joint disease [20]. Meniscus induced cartilaginous damage has also been shown to result in anatomical progression of early-stage OA in a canine model [43] emphasising the importance of meniscal pathobiology in global knee joint integrity [3]. Furthermore, targeted mutation of NOV/CCN3 in mice disrupts joint homeostasis and can induce OA-like symptoms [44]. On the other hand enhanced E-cadherin and PPAR expression leads to increased cell proliferation in primary human meniscal cells and these are potential molecular targets for meniscal regeneration [45].
Proteomics approaches have identified meniscal markers associated with disease pathology [46,47,48,49]. Genetics studies have demonstrated that Hajdu Cheney syndrome mice display an altered B-cell allocation during immune responses and are sensitized to the development of OA [50,51]. In contrast, the articular cartilage of Ctsk(−/−) mice is protected from the development of OA but has associated cellular and molecular changes in subchondral bone and cartilage ECM [52]. Elf3 contributes to cartilage degradation in-vivo in a surgical model of post-traumatic OA [53]. Transcriptomics has compared menisci from patients with and without OA [54]. Wnt7a inhibits interleukin-1 beta (IL-1β) Induced catabolic gene expression and prevents articular cartilage damage in experimental OA [55], Wnt16 also antagonises excessive canonical Wnt activation and protects cartilage in OA [56]. Furthermore, inhibition of Wnt/β-catenin signaling ameliorates OA in a murine model of experimental OA [57]. Deletion of Runx2 in articular chondrocytes decelerates the progression of medial meniscal destabilised induction of OA in adult mice [58]. Screening for characteristic genes in OA induced by destabilization of the medial meniscus utilizing a bioinformatics type approach has identified genes associated with meniscal pathology [59].

2.3. Gene Profiling and Transcriptomics of Meniscal Tissue and Articular Cartilage Provide Insights into the Etiopathogenesis of OA and Identify Potential Therapeutic Targets

Gene profiling has established that traumatic and degenerative meniscal tears have different expression profiles [60] and that gene expression in human meniscal tears has limited association with early degenerative changes in knee articular cartilage [61]. Comprehensive expression analysis of microRNAs and mRNAs in synovial tissue in a mouse model of early post-traumatic OA [62] and regional gene expression analysis of multiple tissues has been undertaken [63]. Gene profiling of articular cartilage from knees with meniscal tears are distinctly different from knees with OA [64]. The presence of meniscal tears also alters the gene expression profile evident in anterior cruciate ligament injury [65]. These studies are insightful as to different forms of OA which develop following meniscal or ligament injury [66]. The use of animal meniscal models [43,67,68,69,70] have also contributed significantly to a greater understanding of the pathobiology of meniscal degeneration and traumatic meniscal injury and may aid in the formulation of therapeutic interventions on these meniscal tissues [71,72].
The underlying molecular changes underlying progression of OA are incompletely understood however some transcriptomic studies are now providing some insightful information into this disease process. Gene profiling of articular cartilage from knees with meniscal tears are distinctly different from knees with OA cartilage lesions but relatively intact menisci [64], the presence of meniscal tears also alters gene expression in anterior cruciate ligament injury [65]. These studies are insightful as to different forms of OA which develop following meniscal or ligament injury [66] and stress the importance of examination of meniscal gene expression in destabilised menisci directly to obtain gene expression data of relevance to meniscal disease processes.
Gene expression studies in cartilage of surgically induced OA in wild-type (WT) mice and Adamts5Δcat mice, in which ADAMTS-5 activity is ablated and thus aggrecan loss and cartilage erosion is inhibited has been undertaken to distinguish gene expression changes that are independent of ADAMTS-5 activity and cartilage breakdown. Gene expression in meniscal cartilage from the developing lesion in the destabilized medial meniscus and corresponding regions in sham-operated joints were compared using whole-genome microarrays [73]. Previously identified OA-related genes, including Ptgs2, Crlf1, and Inhba, and novel genes, such as Phdla2 and Il11, were up-regulated in both WT mice and Adamts5Δcat mice, indicating that these are regulated independently of ADAMTS-5 activity. Other genes, such as Col10a1, a marker of cartilage hypertrophy and terminal differentiation, and genes of the Wnt/β-catenin pathway however were dependent on ADAMTS-5 activity. Foxo4, and Xbp1 endoplasmic reticulum-stress cell death transcriptional networks were activated by meniscal destabilisation. Many degradative protease genes, including Mmp3, Capn2, and the novel cartilage proteases Prss46 and Klk8 were also upregulated by meniscal destabilisation and the development of OA changes. Identification of genes that act independently of ADAMTS-5 identify these as genes which may initiate meniscal degeneration in OA and are prospective new therapeutic targets. Thus a complex picture of meniscal degradomics is emerging in OA.
Integrative epigenomics, transcriptomics and proteomics of patient chondrocytes has been used to identify cartilage genes and pathways involved in OA [74]. Examination of genes and transcription pathways that mark OA progression in isolated primary chondrocytes from paired intact versus degraded articular cartilage from knee-joint replacement donors has been undertaken. Genome-wide DNA methylation, RNA sequencing, and quantitative proteomics has been applied to these samples. Forty nine differentially regulated genes have been identified in intact versus degraded cartilage, 16 of these genes had not previously been implicated in OA progression. Three genes were particularly strongly identified across all analytic platforms used, namely AQP1, COL1A1 and CLEC3B and an increased distribution of these proteins confirmed in OA cartilage using immunohistochemistry [74]. Integrated pathway analysis showed the involvement of ECM degradation, collagen catabolism and angiogenesis as important components of disease progression [74]. The growth of blood vessels and nerves are closely linked processes that share common regulatory mechanisms [75]. Histology of OA and normal cartilage has also demonstrated greater blood vessel ingrowth in tissues with more advanced OA [74].
As already noted, three genes have been identified strongly associated with OA cartilage, aquaporin-1 (AQP1), tetranectin (CLEC3B) and COL1A1 [74]. AQP1 facilitates water transport across biological membranes. In OA cartilage increased cartilage hydration leads to chondrocyte swelling which has been suggested to contribute to OA development [76]. AQP1 is also overexpressed in the meniscus in a rat model of OA [22], and its levels are elevated in degenerate human articular cartilage specimens compared to normal intact cartilage [77]. Tetranectin has also been previously shown to be upregulated in OA [78,79]. Collagens are the main fibrillar structural components of cartilage, disruption of collagen biosynthesis and elevated degradation of type-I collagen by MMPs have important roles in the progression of OA [80,81]. A search of Drugbank [82] for existing drugs approved for human use for the treatment of OA, identified Acetazolamide, a carbonic anhydrase inhibitor diuretic agent which inhibits AQP1 and Tenecteplase, a thromobolytic agent which binds tetranectin [74] as candidates for further evaluation. However it remains to be established how effective these compounds will be as therapeutic agents targeting the degenerate meniscus before any assessment can be made as to whether they will have a global impact on the knee-joint as a therapeutic agents for the treatment of OA.

3. Current Approaches in Meniscal Repair

The potential of mesenchymal stem cells (MSCs) for tissue repair became evident in 2003 [83] when adult stem cells isolated from bone marrow were used in a caprine model where the medial meniscus was completely removed and the anterior cruciate ligament (ACL) was excised. Marked regeneration of the medial meniscus was evident in MSC treated joints, and degeneration of the articular cartilage, osteophytic remodeling, and subchondral sclerosis were reduced compared to joints treated with vehicle alone without cells, however there was no evidence of repair of the ACL in any of the joints. This study clearly showed that local delivery of adult MSCs to injured joints stimulated regeneration of meniscal tissue and retarded the progressive destruction of other tissue in the knee joint which is normally seen in animal meniscectomy models of OA [84,85,86,87,88].
Many reviews have emphasized the great potential of MSCs for meniscal repair applications [25,31,36,40,42,43,45,71]. MSCs have been employed in numerous strategies aimed to promote repair of meniscal lacerations [23] and defects in the avascular inner meniscal zone [39,89,90,91] or in meniscal replacement in meniscectomised animals [83]. A number of animal models have been employed in these studies including goats [83], rabbits [92,93], minipigs [69,94], sheep [84,85,86,87,88,95] and rats [26,29,63,96]. MSCs have been sourced from bone marrow [95,97,98,99,100,101], adipose tissues [98,99,102,103] and synovium [62,94,98,103]. The utility of a number of bioscaffolds seeded with therapeutic cells have also been examined to promote repair of inner zone meniscal defects (reviewed in [18]). These scaffolds include a stem cell/collagen-derived scaffold [91], polyurethane [104] and a scaffold-free tissue-engineered construct derived from allogeneic synovial MSCs [98] besides a number of conventional tissue engineering synthetic resorbale bioscaffolds [105]. Fibrin-glue has also been evaluated as a cell delivery vehicle for meniscal repair [89,97,103,106]. Examination of the cell directive properties of MSCs over meniscal cells co-cultured in scaffold free micromass pellet cultures has demonstrated a significant increase in cellular proliferation, type I and II collagen and aggrecan production [105]. Three-dimensional coculture of meniscal cells and MSCs in collagen Type I hydrogels have also demonstrated positive responses in terms of matrix production conducive to meniscal repair processes [107]. Evaluation of effects on cell shape, matrix production, and mechanical performance of neo-tissues produced by such 3D MSC: meniscal cell co-cultures has shown positive effects of potential application in the repair of meniscal defects [31,108]. Direct cell-cell contact between MSCs and meniscal cells is not required to elicit such positive responses since MSC conditioned media can elicit similar responses in meniscal cells due to trophic factors transferred from the MSCs by paracrine effects. Transfer of material from MSCs can also be undertaken by microvesicles (exosomes) and these are of clinical potential as a means of directing positive therapeutic responses in resident meniscal cell populations [109,110,111].
The meniscus is a tissue which has very significant physical demands placed on it within the knee joint to protect and stabilise other knee joint tissues which provide knee joint articulation and weight bearing. Artificial scaffolds developed for partial or total meniscal replacement still do not totally reproduce the material properties of native meniscal tissues and further developments will be required to perfect this technology [112,113,114]. Stereolithography [115] (3D printing) of individualised menisci or meniscal segments with bioscaffolds which can be seeded with autologous stem cells [108] to eventually synthesize a replacement meniscal structure represents a promising advancement in this area of meniscal repair.
Knowledge of biological glues and tissue adhesives for the attachment of meniscal flaps or tears have undergone significant improvements in recent years. Tissue adhesives currently used in surgical applications, reviewed in [18,105] include the topical skin adhesive Dermabond® (2-octyl cyanoacrylate), fibrin glue for pulmonary leaks, and the recently developed TissuGlu® (an FDA approved urethane-based adhesive) for abdominoplasty surgery [116,117]. Tissue adhesives inspired by high performance adhesive marine compounds which firmly attach mussels [118] and barnacles [119] to marine substrates are promising tissue adhesives. An interesting biological glue produced by the Australian native frog species Notaden benetii (also known as the crucifix toad or holycross frog) is a skin exudate secreted by the female frog during mating and ensures extended sexual union by the male for efficient egg fertilization. This frog glue [120,121] provides superior adhesive properties to fibrin glue, is non-immunogenic and has been successfully trialled in the re-attachment of a bucket handle tear and in rotator cuff repair surgery, a re-engineered development of this promising polymer deserves to be undertaken [122,123]. This colourful digging toad occurs in the semi-arid grasslands and black soil plains of central inland New South Wales and the interior of southern Queensland, west of the Great Dividing Range in Australia. The use of bioadhesive or hydrogels as delivery systems for therapeutic cells, drugs or antibiotics is another innovative development in repair biology but has yet to be applied in meniscal repair procedures [124,125,126,127,128].
Moderate physical exercise has been examined as a means of positively regulating beneficial chondroprotective and anti-inflammatory genes in human knee joint tissues as a means of therapeutic intervention during inflammatory osteoarthritic conditions in the knee-joint [129]. Such an approach has delivered some beneficial effects in type B synoviocytes which suggests that moderate exercise may delay the onset of degenerative changes in knee-joint tissues in OA and RA [130]. Moderate physical exercise has also been shown to improve the lubrication of articular cartilage and joint function in ageing rats through an upregulation in lubricin expression [129]. Excessive joint loading also down-regulates lubricin expression and decreases the functional properties of knee-joint tissues in an ovine meniscectomy model [131,132]. A virgin-olive oil supplemented diet in combination with moderate physical exercise has been shown to ameliorate degenerative effects on knee-joint tissues in an ACL transection model of OA in rats [133]. This treatment upregulates lubricin expression which has a beneficial effect on knee-joint function, a recent study has suggested that lubricin can regulate knee-joint inflammation. These findings therefore suggest that this simple dietary modification may be a useful medical therapy to use in combination with moderate exercise in man to prevent osteoarthritic disease progression not only preserving the articular cartilage and improving its lubricative properties and thus joint articulation but may also be globally beneficial to all knee-joint tissues [133].

4. Conclusions

The menisci are major cartilaginous structures in the human knee with essential roles in the stabilization and weight bearing of this structure. A healthy meniscus is therefore a high priority to ensure the health of other knee joint tissues. A realization of the inter-dependance of all knee joint tissues to provide an efficient articulatory and weight bearing structure indicates that for any therapeutic measure to meet with success in the treatment of OA or RA it must be beneficial to all of the varied cell populations in the human knee. The large number of therapeutic measures being developed to specifically halt further degeneration, stimulate regeneration or replace meniscal tissues points to the central importance of the meniscus to knee joint function. Without a therapeutic measure effectively addressing all these parameters in OA there is little likelihood of a successful outcome in this most debilitating of conditions. Moderate exercise has been shown to be beneficial to knee joint tissues and dietary adjustment to include virgin olive oil has further beneficial effects in terms of a lowering of knee joint inflammation. With knee OA predicted to become a leading musculoskeletal condition within the next two decades there is an extreme need for effective measures to be developed and all avenues of therapy need careful evaluation.

Author Contributions

J.M. conceived wrote, edited and submitted the final manuscript version.

Funding

This research received no external funding.

Conflicts of Interest

The author declares no conflict of interest.

Abbreviations

AAOSAmerican Academy of Sciences
ADAMSA Disintegrin and Metalloproteinase
ADAMTSA Disintegrin And Metalloprotease with ThromboSpondin type 1 repeats
ADAMTS Cat∆5A Disintegrin And Metalloprotease with ThromboSpondin type 1 repeat-5 devoid of its catalytic domain
AFMAtomic force microscopy
ACLAnterior cruciate ligament
AQP1Aquaporin-1
Capn2Calpain-2
CCNacronym term derived from the first three members of the gene families, CYR61(cysteine-rich angiogenic protein 61 or CCN1), CTGF (connective tissue growth factor or CCN2), and NOV (nephroblastom-overexpressed or CCN3)
CLEC3BTetranectin
CRLF1Cytokine receptor-like factor 1
CtskCathepsin K
µCTMicro computed tomography
ElfE74-like factor-1 [ets, (erythroblast transformation-specific) domain transcription factor]
ECMExtracellular matrix
Foxo4Forkhead box protein O4
HMBGHigh mobility group box-1
IL-1αInterleukin-1 alpha
IL11Interleukin-11
INHBAInhibin, beta A
Klk8Kallikrein-8
MMPMatrix metalloprotease
MSCsMesenchymal stem (stromal) cells
NODNucleotide oligomerization domain
NOVNephroblastom-overexpressed or CCN3
OAOsteoarthritis
PHDLA2Pleckstrin homology-like domain, family A, member 2
PPARPeroxisome proliferator-activated receptor
PRG4Proteoglycan 4 (lubricin)
Prss46Serine protease 46
PTGS2Prostaglandin-endoperoxide synthase-2 (cyclooxygenase-2, COX-2)
RARheumatoid arthritis
RNARibonucleic acid
3Dthree dimensional
TNF-αTumour necrosis alpha
Tp53Tumour protein 53
Wnta condensation of terms describing the Winged and Int transcription factor morphogens

References

  1. Anonymous, American Academy of Surgeons (AAOS). Available online: https://www.aaos.org/annual report/ (accessed on 21 March 2019).
  2. Fox, A.J.; Wanivenhaus, F.; Burge, A.J.; Warren, R.F.; Rodeo, S.A. The human meniscus: A review of anatomy, function, injury, and advances in treatment. Clin. Anat. 2015, 28, 269–287. [Google Scholar] [CrossRef] [PubMed]
  3. Cook, J.L.; Kuroki, K.; Stoker, A.M.; Monibi, F.A.; Roller, B.L. Meniscal biology in health and disease. Connect. Tissue Res. 2017, 58, 225–237. [Google Scholar] [CrossRef] [PubMed]
  4. Andrews, S.H.J.; Adesida, A.B.; Abusara, Z.; Shrive, N.G. Current concepts on structure-function relationships in the menisci. Connect. Tissue Res. 2017, 58, 271–281. [Google Scholar] [CrossRef] [PubMed]
  5. Makris, E.A.; Hadidi, P.; Athanasiou, K.A. The knee meniscus: Structure-function, pathophysiology, current repair techniques, and prospects for regeneration. Biomaterials 2011, 32, 7411–7431. [Google Scholar] [CrossRef]
  6. McAlinden, A.; Dudhia, J.; Bolton, M.C.; Lorenzo, P.; Heinegard, D.; Bayliss, M.T. Age-related changes in the synthesis and mRNA expression of decorin and aggrecan in human meniscus and articular cartilage. Osteoarthr. Cartil. 2001, 9, 33–41. [Google Scholar] [CrossRef] [PubMed]
  7. Melrose, J.; Smith, S.; Cake, M.; Read, R.; Whitelock, J. Comparative spatial and temporal localisation of perlecan, aggrecan and type I, II and IV collagen in the ovine meniscus: An ageing study. Histochem. Cell Biol. 2005, 124, 225–235. [Google Scholar] [CrossRef]
  8. Valiyaveettil, M.; Mort, J.S.; McDevitt, C.A. The concentration, gene expression, and spatial distribution of aggrecan in canine articular cartilage, meniscus, and anterior and posterior cruciate ligaments: A new molecular distinction between hyaline cartilage and fibrocartilage in the knee joint. Connect. Tissue Res. 2005, 46, 83–91. [Google Scholar] [CrossRef]
  9. Endo, J.; Sasho, T.; Akagi, R.; Muramatsu, Y.; Watanabe, A.; Akatsu, Y.; Fukawa, T.; Tahara, M.; Yamaguchi, S. Comparative Analysis of Gene Expression between Cartilage and Menisci in Early-Phase Osteoarthritis of the Knee-An Animal Model Study. J. Knee Surg. 2018, 31, 664–669. [Google Scholar] [CrossRef] [PubMed]
  10. Son, M.; Levenston, M.E. Discrimination of meniscal cell phenotypes using gene expression profiles. Eur. Cells Mater. 2012, 23, 195–208. [Google Scholar] [CrossRef]
  11. Smith, S.M.; Shu, C.; Melrose, J. Comparative immunolocalisation of perlecan with collagen II and aggrecan in human foetal, newborn and adult ovine joint tissues demonstrates perlecan as an early developmental chondrogenic marker. Histochem. Cell Biol. 2010, 134, 251–263. [Google Scholar] [CrossRef]
  12. Kavanagh, E.; Ashhurst, D.E. Distribution of biglycan and decorin in collateral and cruciate ligaments and menisci of the rabbit knee joint. J. Histochem. Cytochem. 2001, 49, 877–885. [Google Scholar] [CrossRef] [PubMed]
  13. Melrose, J.; Fuller, E.S.; Roughley, P.J.; Smith, M.M.; Kerr, B.; Hughes, C.E.; Caterson, B.; Little, C.B. Fragmentation of decorin, biglycan, lumican and keratocan is elevated in degenerate human meniscus, knee and hip articular cartilages compared with age-matched macroscopically normal and control tissues. Arthritis Res. Ther. 2008, 10, R79. [Google Scholar] [CrossRef] [PubMed]
  14. Roughley, P.J.; White, R.J. The dermatan sulfate proteoglycans of the adult human meniscus. J. Orthop. Res. 1992, 10, 631–637. [Google Scholar] [CrossRef]
  15. Musumeci, G.; Loreto, C.; Carnazza, M.L.; Cardile, V.; Leonardi, R. Acute injury affects lubricin expression in knee menisci: An immunohistochemical study. Ann. Anat. 2013, 195, 151–158. [Google Scholar] [CrossRef]
  16. Musumeci, G.; Trovato, F.M.; Loreto, C.; Leonardi, R.; Szychlinska, M.A.; Castorina, S.; Mobasheri, A. Lubricin expression in human osteoarthritic knee meniscus and synovial fluid: A morphological, immunohistochemical and biochemical study. Acta Histochem. 2014, 116, 965–972. [Google Scholar] [CrossRef]
  17. Zhang, D.; Cheriyan, T.; Martin, S.D.; Schmid, T.M.; Spector, M. Lubricin Distribution in the Menisci and Labra of Human Osteoarthritic Joints. Cartilage 2012, 3, 165–172. [Google Scholar] [CrossRef] [PubMed]
  18. Melrose, J. Novel Approaches in Meniscal Preservation, Replacement and Repair Utilizing Mesenchymal Stem Cells, New Generation Bioscaffolds, Hydrogels and Biological Adhesives as Cell Delivery Vehicles. In The Athletic Knee—Function, Pathology & Management; Abdulwaba, T., Ed.; InTech Open Publishers: London, UK, 2018. [Google Scholar]
  19. Melrose, J. Glycans Provide Molecular Recognition Motifs Which Regulate Endoplasmic Protein Folding, Transport, Lysosomal Targeting, and are Used by Pattern Recognition Receptors in Pathogen Surveyance and Innate Immunity. In Advances in Medicine and Biology; Berhardt, L.V., Ed.; Nova Publishers: New York, NY, USA, 2017; Volume 114, pp. 110–168. [Google Scholar]
  20. Melrose, J.; Fuller, E.S.; Little, C.B. The biology of meniscal pathology in osteoarthritis and its contribution to joint disease: Beyond simple mechanics. Connect. Tissue Res. 2017, 58, 282–294. [Google Scholar] [CrossRef] [PubMed]
  21. Musumeci, G.; Carnazza, M.L.; Leonardi, R.; Loreto, C. Expression of β-defensin-4 in “an in vivo and ex vivo model” of human osteoarthritic knee meniscus. Knee Surg. Sports Traumatol. Arthrosc. 2012, 20, 216–222. [Google Scholar] [CrossRef]
  22. Musumeci, G.; Leonardi, R.; Carnazza, M.L.; Cardile, V.; Pichler, K.; Weinberg, A.M.; Loreto, C. Aquaporin 1 (AQP1) expression in experimentally induced osteoarthritic knee menisci: An in vivo and in vitro study. Tissue Cell 2013, 45, 145–152. [Google Scholar] [CrossRef] [PubMed]
  23. Roller, B.L.; Monibi, F.; Stoker, A.M.; Bal, B.S.; Cook, J.L. Identification of Novel Synovial Fluid Biomarkers Associated with Meniscal Pathology. J. Knee Surg. 2016, 29, 47–62. [Google Scholar] [CrossRef]
  24. Bigoni, M.; Turati, M.; Sacerdote, P.; Gaddi, D.; Piatti, M.; Castelnuovo, A.; Franchi, S.; Gandolla, M.; Pedrocchi, A.; Omeljaniuk, R.J.; et al. Characterization of synovial fluid cytokine profiles in chronic meniscal tear of the knee. J. Orthop. Res. 2017, 35, 340–346. [Google Scholar] [CrossRef]
  25. Monibi, F.; Roller, B.L.; Stoker, A.; Garner, B.; Bal, S.; Cook, J.L. Identification of Synovial Fluid Biomarkers for Knee Osteoarthritis and Correlation with Radiographic Assessment. J. Knee Surg. 2016, 29, 242–247. [Google Scholar] [CrossRef]
  26. Bagi, C.M.; Zakur, D.E.; Berryman, E.; Andresen, C.J.; Wilkie, D. Correlation between muCT imaging, histology and functional capacity of the osteoarthritic knee in the rat model of osteoarthritis. J. Transl. Med. 2015, 13, 276. [Google Scholar] [CrossRef]
  27. Collins, J.E.; Losina, E.; Nevitt, M.C.; Roemer, F.W.; Guermazi, A.; Lynch, J.A.; Katz, J.N.; Kent Kwoh, C.; Kraus, V.B.; Hunter, D.J. Semiquantitative Imaging Biomarkers of Knee Osteoarthritis Progression: Data From the Foundation for the National Institutes of Health Osteoarthritis Biomarkers Consortium. Arthritis Rheumatol. 2016, 68, 2422–2431. [Google Scholar] [CrossRef]
  28. Roemer, F.W.; Guermazi, A.; Collins, J.E.; Losina, E.; Nevitt, M.C.; Lynch, J.A.; Katz, J.N.; Kwoh, C.K.; Kraus, V.B.; Hunter, D.J. Semi-quantitative MRI biomarkers of knee osteoarthritis progression in the FNIH biomarkers consortium cohort—Methodologic aspects and definition of change. BMC Musculoskelet. Disord. 2016, 17, 466. [Google Scholar] [CrossRef]
  29. Donato, S.; Pacile, S.; Colombo, F.; Garrovo, C.; Monego, S.D.; Macor, P.; Tromba, G.; Biffi, S. Meniscal Ossicles as micro-CT Imaging Biomarker in a Rodent Model of Antigen-Induced Arthritis: A Synchrotron-Based X-ray Pilot Study. Sci. Rep. 2017, 7, 7544. [Google Scholar] [CrossRef]
  30. Joseph, G.B.; Nevitt, M.C.; McCulloch, C.E.; Neumann, J.; Lynch, J.A.; Heilmeier, U.; Lane, N.E.; Link, T.M. Associations between molecular biomarkers and MR-based cartilage composition and knee joint morphology: Data from the Osteoarthritis Initiative. Osteoarthr. Cartil. 2018, 26, 1070–1077. [Google Scholar] [CrossRef]
  31. Mezlini-Gharsallah, H.; Youssef, R.; Uk, S.; Laredo, J.D.; Chappard, C. Three-dimensional mapping of the joint space for the diagnosis of knee osteoarthritis based on high resolution computed tomography: Comparison with radiographic, outerbridge, and meniscal classifications. J. Orthop. Res. 2018, 36, 2380–2391. [Google Scholar] [CrossRef]
  32. Mingalone, C.K.H.; Liu, Z.; Hollander, J.M.; Garvey, K.D.; Gibson, A.L.; Banks, R.E.; Zhang, M.; McAlindon, T.E.; Nielsen, H.C.; Georgakoudi, I.; et al. Bioluminescence and second harmonic generation imaging reveal dynamic changes in the inflammatory and collagen landscape in early osteoarthritis. Lab. Investig. 2018, 98, 656–669. [Google Scholar] [CrossRef]
  33. Nebelung, S.; Tingart, M.; Pufe, T.; Kuhl, C.; Jahr, H.; Truhn, D. Ex vivo quantitative multiparametric MRI mapping of human meniscus degeneration. Skeletal. Radiol. 2016, 45, 1649–1660. [Google Scholar] [CrossRef]
  34. Nair, A.; Gan, J.; Bush-Joseph, C.; Verma, N.; Tetreault, M.W.; Saha, K.; Margulis, A.; Fogg, L.; Scanzello, C.R. Synovial chemokine expression and relationship with knee symptoms in patients with meniscal tears. Osteoarthr. Cartil. 2015, 23, 1158–1164. [Google Scholar] [CrossRef]
  35. Marenzana, M.; Hagen, C.K.; Borges, P.D.; Endrizzi, M.; Szafraniec, M.B.; Vincent, T.L.; Rigon, L.; Arfelli, F.; Menk, R.H.; Olivo, A. Synchrotron- and laboratory-based X-ray phase-contrast imaging for imaging mouse articular cartilage in the absence of radiopaque contrast agents. Philos. Trans. A Math. Phys. Eng. Sci. 2014, 372. [Google Scholar] [CrossRef]
  36. Ke, X.; Jin, G.; Yang, Y.; Cao, X.; Fang, R.; Feng, X.; Lei, B. Synovial Fluid HMGB-1 Levels are Associated with Osteoarthritis Severity. Clin. Lab. 2015, 61, 809–818. [Google Scholar] [CrossRef]
  37. Li, Z.C.; Cheng, G.Q.; Hu, K.Z.; Li, M.Q.; Zang, W.P.; Dong, Y.Q.; Wang, W.L.; Liu, Z.D. Correlation of synovial fluid HMGB-1 levels with radiographic severity of knee osteoarthritis. Clin. Investig. Med. 2011, 34, E298. [Google Scholar] [CrossRef]
  38. Ahlen, M.; Roshani, L.; Liden, M.; Struglics, A.; Rostgard-Christensen, L.; Kartus, J. Inflammatory cytokines and biomarkers of cartilage metabolism 8 years after anterior cruciate ligament reconstruction: Results from operated and contralateral knees. Am. J. Sports Med. 2015, 43, 1460–1466. [Google Scholar] [CrossRef]
  39. Carter, T.E.; Taylor, K.A.; Spritzer, C.E.; Utturkar, G.M.; Taylor, D.C.; Moorman, C.T., 3rd; Garrett, W.E.; Guilak, F.; McNulty, A.L.; DeFrate, L.E. In vivo cartilage strain increases following medial meniscal tear and correlates with synovial fluid matrix metalloproteinase activity. J. Biomech. 2015, 48, 1461–1468. [Google Scholar] [CrossRef]
  40. Griswold, A.J.; Perez, J.; Nuytemans, K.; Strong, T.A.; Wang, L.; Vance, D.D.; Ennis, H.; Smith, M.K.; Best, T.M.; Vance, J.M.; et al. Transcriptomic analysis of synovial extracellular RNA following knee trauma: A pilot study. J. Orthop. Res. 2018, 36, 1659–1665. [Google Scholar] [CrossRef]
  41. Fuller, E.; Little, C.B.; Melrose, J. Interleukin-1α induces focal degradation of biglycan and tissue degeneration in an in-vitro ovine meniscal model. Exp. Mol. Pathol. 2016, 101, 214–220. [Google Scholar] [CrossRef]
  42. Fuller, E.S.; Smith, M.M.; Little, C.B.; Melrose, J. Zonal differences in meniscus matrix turnover and cytokine response. Osteoarthr. Cartil. 2012, 20, 49–59. [Google Scholar] [CrossRef]
  43. Kahn, D.; Mittelstaedt, D.; Matyas, J.; Qu, X.; Lee, J.H.; Badar, F.; Les, C.; Zhuang, Z.; Xia, Y. Meniscus Induced Cartilaginous Damage and Non-linear Gross Anatomical Progression of Early-stage Osteoarthritis in a Canine Model. Open Orthop. J. 2016, 10, 690–705. [Google Scholar] [CrossRef]
  44. Roddy, K.A.; Boulter, C.A. Targeted mutation of NOV/CCN3 in mice disrupts joint homeostasis and causes osteoarthritis-like disease. Osteoarthr. Cartil. 2015, 23, 607–615. [Google Scholar] [CrossRef] [PubMed]
  45. Pillai, M.M.; Elakkiya, V.; Gopinathan, J.; Sabarinath, C.; Shanthakumari, S.; Sahanand, K.S.; Dinakar Rai, B.K.; Bhattacharyya, A.; Selvakumar, R. A combination of biomolecules enhances expression of E-cadherin and peroxisome proliferator-activated receptor gene leading to increased cell proliferation in primary human meniscal cells: An in vitro study. Cytotechnology 2016, 68, 1747–1761. [Google Scholar] [CrossRef]
  46. Folkesson, E.; Turkiewicz, A.; Englund, M.; Onnerfjord, P. Differential protein expression in human knee articular cartilage and medial meniscus using two different proteomic methods: A pilot analysis. BMC Musculoskelet. Disord. 2018, 19, 416. [Google Scholar] [CrossRef] [PubMed]
  47. Liao, W.; Li, Z.; Li, T.; Zhang, Q.; Zhang, H.; Wang, X. Proteomic analysis of synovial fluid in osteoarthritis using SWATHmass spectrometry. Mol. Med. Rep. 2018, 17, 2827–2836. [Google Scholar] [PubMed]
  48. Liao, W.; Li, Z.; Zhang, H.; Li, J.; Wang, K.; Yang, Y. Proteomic analysis of synovial fluid as an analytical tool to detect candidate biomarkers for knee osteoarthritis. Int. J. Clin. Exp. Pathol. 2015, 8, 9975–9989. [Google Scholar]
  49. Roller, B.L.; Monibi, F.; Stoker, A.M.; Bal, B.S.; Stannard, J.P.; Cook, J.L. Characterization of Meniscal Pathology Using Molecular and Proteomic Analyses. J. Knee Surg. 2015, 28, 496–505. [Google Scholar] [CrossRef] [PubMed]
  50. Yu, J.; Zanotti, S.; Schilling, L.; Schoenherr, C.; Economides, A.N.; Sanjay, A.; Canalis, E. Induction of the Hajdu-Cheney Syndrome Mutation in CD19 B Cells in Mice Alters B-Cell Allocation but Not Skeletal Homeostasis. Am. J. Pathol. 2018, 188, 1430–1446. [Google Scholar] [CrossRef]
  51. Zanotti, S.; Yu, J.; Bridgewater, D.; Wolf, J.M.; Canalis, E. Mice harboring a Hajdu Cheney Syndrome mutation are sensitized to osteoarthritis. Bone 2018, 114, 198–205. [Google Scholar] [CrossRef]
  52. Soki, F.N.; Yoshida, R.; Paglia, D.N.; Duong, L.T.; Hansen, M.F.; Drissi, H. Articular cartilage protection in Ctsk(-/-) mice is associated with cellular and molecular changes in subchondral bone and cartilage matrix. J. Cell. Physiol. 2018, 233, 8666–8676. [Google Scholar] [CrossRef]
  53. Wondimu, E.B.; Culley, K.L.; Quinn, J.; Chang, J.; Dragomir, C.L.; Plumb, D.A.; Goldring, M.B.; Otero, M. Elf3 Contributes to Cartilage Degradation in vivo in a Surgical Model of Post-Traumatic Osteoarthritis. Sci. Rep. 2018, 8, 6438. [Google Scholar] [CrossRef]
  54. Brophy, R.H.; Zhang, B.; Cai, L.; Wright, R.W.; Sandell, L.J.; Rai, M.F. Transcriptome comparison of meniscus from patients with and without osteoarthritis. Osteoarthr. Cartil. 2018, 26, 422–432. [Google Scholar] [CrossRef] [PubMed]
  55. Gibson, A.L.; Hui Mingalone, C.K.; Foote, A.T.; Uchimura, T.; Zhang, M.; Zeng, L. Wnt7a Inhibits IL-1β Induced Catabolic Gene Expression and Prevents Articular Cartilage Damage in Experimental Osteoarthritis. Sci. Rep. 2017, 7, 41823. [Google Scholar] [CrossRef] [PubMed]
  56. Nalesso, G.; Thomas, B.L.; Sherwood, J.C.; Yu, J.; Addimanda, O.; Eldridge, S.E.; Thorup, A.S.; Dale, L.; Schett, G.; Zwerina, J.; et al. WNT16 antagonises excessive canonical WNT activation and protects cartilage in osteoarthritis. Ann. Rheum. Dis. 2017, 76, 218–226. [Google Scholar] [CrossRef]
  57. Lietman, C.; Wu, B.; Lechner, S.; Shinar, A.; Sehgal, M.; Rossomacha, E.; Datta, P.; Sharma, A.; Gandhi, R.; Kapoor, M.; et al. Inhibition of Wnt/β-catenin signaling ameliorates osteoarthritis in a murine model of experimental osteoarthritis. JCI Insight 2018, 3. [Google Scholar] [CrossRef] [PubMed]
  58. Liao, L.; Zhang, S.; Gu, J.; Takarada, T.; Yoneda, Y.; Huang, J.; Zhao, L.; Oh, C.D.; Li, J.; Wang, B.; et al. Deletion of Runx2 in Articular Chondrocytes Decelerates the Progression of DMM-Induced Osteoarthritis in Adult Mice. Sci. Rep. 2017, 7, 2371. [Google Scholar] [CrossRef]
  59. Yang, X.; Zhao, J.; He, Y.; Huangfu, X. Screening for characteristic genes in osteoarthritis induced by destabilization of the medial meniscus utilizing bioinformatics approach. J. Musculoskelet. Neuronal. Interact. 2014, 14, 343–348. [Google Scholar]
  60. Brophy, R.H.; Sandell, L.J.; Rai, M.F. Traumatic and Degenerative Meniscus Tears Have Different Gene Expression Signatures. Am. J. Sports Med. 2017, 45, 114–120. [Google Scholar] [CrossRef]
  61. Brophy, R.H.; Sandell, L.J.; Cheverud, J.M.; Rai, M.F. Gene expression in human meniscal tears has limited association with early degenerative changes in knee articular cartilage. Connect. Tissue Res. 2017, 58, 295–304. [Google Scholar] [CrossRef]
  62. Kung, L.H.W.; Ravi, V.; Rowley, L.; Bell, K.M.; Little, C.B.; Bateman, J.F. Comprehensive Expression Analysis of microRNAs and mRNAs in Synovial Tissue from a Mouse Model of Early Post-Traumatic Osteoarthritis. Sci. Rep. 2017, 7, 17701. [Google Scholar] [CrossRef]
  63. Salazar-Noratto, G.E.; De Nijs, N.; Stevens, H.Y.; Gibson, G.; Guldberg, R.E. Regional gene expression analysis of multiple tissues in an experimental animal model of post-traumatic osteoarthritis. Osteoarthr. Cartil. 2019, 27, 294–303. [Google Scholar] [CrossRef]
  64. Rai, M.F.; Tycksen, E.D.; Cai, L.; Yu, J.; Wright, R.W.; Brophy, R.H. Distinct Degenerative Phenotype of Articular Cartilage from Knees with Meniscus Tear Compared to Knees with Osteoarthritis. Osteoarthr. Cartil. 2019. [Google Scholar] [CrossRef]
  65. Brophy, R.H.; Rothermich, M.A.; Tycksen, E.D.; Cai, L.; Rai, M.F. Presence of meniscus tear alters gene expression profile of anterior cruciate ligament tears. J. Orthop. Res. 2018, 36, 2612–2621. [Google Scholar] [CrossRef] [PubMed]
  66. Rai, M.F.; Brophy, R.H.; Sandell, L.J. Osteoarthritis following meniscus and ligament injury: Insights from translational studies and animal models. Curr. Opin. Rheumatol. 2019, 31, 70–79. [Google Scholar] [CrossRef] [PubMed]
  67. Cruz, R.; Ramirez, C.; Rojas, O.I.; Casas-Mejia, O.; Kouri, J.B.; Vega-Lopez, M.A. Menisectomized miniature Vietnamese pigs develop articular cartilage pathology resembling osteoarthritis. Pathol. Res. Pract. 2015, 211, 829–838. [Google Scholar] [CrossRef]
  68. Hiyama, K.; Muneta, T.; Koga, H.; Sekiya, I.; Tsuji, K. Meniscal regeneration after resection of the anterior half of the medial meniscus in mice. J. Orthop. Res. 2017, 35, 1958–1965. [Google Scholar] [CrossRef] [PubMed]
  69. Kreinest, M.; Reisig, G.; Strobel, P.; Dinter, D.; Attenberger, U.; Lipp, P.; Schwarz, M. A Porcine Animal Model for Early Meniscal Degeneration—Analysis of Histology, Gene Expression and Magnetic Resonance Imaging Six Months after Resection of the Anterior Cruciate Ligament. PLoS ONE 2016, 11, e0159331. [Google Scholar] [CrossRef] [PubMed]
  70. Rai, M.F.; Duan, X.; Quirk, J.D.; Holguin, N.; Schmidt, E.J.; Chinzei, N.; Silva, M.J.; Sandell, L.J. Post-Traumatic Osteoarthritis in Mice Following Mechanical Injury to the Synovial Joint. Sci. Rep. 2017, 7, 45223. [Google Scholar] [CrossRef] [PubMed]
  71. Rai, M.F.; McNulty, A.L. Meniscus beyond mechanics: Using biology to advance our understanding of meniscus injury and treatment. Connect. Tissue Res. 2017, 58, 221–224. [Google Scholar] [CrossRef]
  72. Bateman, J.F.; Rowley, L.; Belluoccio, D.; Chan, B.; Bell, K.; Fosang, A.J.; Little, C.B. Transcriptomics of wild-type mice and mice lacking ADAMTS-5 activity identifies genes involved in osteoarthritis initiation and cartilage destruction. Arthritis Rheumatol. 2013, 65, 1547–1560. [Google Scholar] [CrossRef]
  73. Steinberg, J.; Ritchie, G.R.S.; Roumeliotis, T.I.; Jayasuriya, R.L.; Clark, M.J.; Brooks, R.A.; Binch, A.L.A.; Shah, K.M.; Coyle, R.; Pardo, M.; et al. Integrative epigenomics, transcriptomics and proteomics of patient chondrocytes reveal genes and pathways involved in osteoarthritis. Sci. Rep. 2017, 7, 8935. [Google Scholar] [CrossRef]
  74. Mapp, P.I.; Walsh, D.A. Mechanisms and targets of angiogenesis and nerve growth in osteoarthritis. Nat. Rev. Rheumatol. 2012, 8, 390–398. [Google Scholar] [CrossRef]
  75. Bush, P.G.; Hall, A.C. The volume and morphology of chondrocytes within non-degenerate and degenerate human articular cartilage. Osteoarthr. Cartil. 2003, 11, 242–251. [Google Scholar] [CrossRef]
  76. Geyer, M.; Grassel, S.; Straub, R.H.; Schett, G.; Dinser, R.; Grifka, J.; Gay, S.; Neumann, E.; Muller-Ladner, U. Differential transcriptome analysis of intraarticular lesional vs intact cartilage reveals new candidate genes in osteoarthritis pathophysiology. Osteoarthr. Cartil. 2009, 17, 328–335. [Google Scholar] [CrossRef] [PubMed]
  77. Karlsson, C.; Dehne, T.; Lindahl, A.; Brittberg, M.; Pruss, A.; Sittinger, M.; Ringe, J. Genome-wide expression profiling reveals new candidate genes associated with osteoarthritis. Osteoarthr. Cartil. 2010, 18, 581–592. [Google Scholar] [CrossRef] [PubMed]
  78. Valdes, A.M.; Hart, D.J.; Jones, K.A.; Surdulescu, G.; Swarbrick, P.; Doyle, D.V.; Schafer, A.J.; Spector, T.D. Association study of candidate genes for the prevalence and progression of knee osteoarthritis. Arthritis Rheumatol. 2004, 50, 2497–2507. [Google Scholar] [CrossRef] [PubMed]
  79. Tchetina, E.V. Developmental mechanisms in articular cartilage degradation in osteoarthritis. Arthritis 2011, 2011, 683970. [Google Scholar] [CrossRef] [PubMed]
  80. Xia, B.; Di, C.; Zhang, J.; Hu, S.; Jin, H.; Tong, P. Osteoarthritis pathogenesis: A review of molecular mechanisms. Calcif. Tissue Int. 2014, 95, 495–505. [Google Scholar] [CrossRef]
  81. Law, V.; Knox, C.; Djoumbou, Y.; Jewison, T.; Guo, A.C.; Liu, Y.; Maciejewski, A.; Arndt, D.; Wilson, M.; Neveu, V.; et al. DrugBank 4.0: Shedding new light on drug metabolism. Nucleic Acids Res. 2014, 42, D1091–D1097. [Google Scholar] [CrossRef]
  82. Murphy, J.M.; Fink, D.J.; Hunziker, E.B.; Barry, F.P. Stem cell therapy in a caprine model of osteoarthritis. Arthritis Rheumatol. 2003, 48, 3464–3474. [Google Scholar] [CrossRef]
  83. Appleyard, R.C.; Burkhardt, D.; Ghosh, P.; Read, R.; Cake, M.; Swain, M.V.; Murrell, G.A. Topographical analysis of the structural, biochemical and dynamic biomechanical properties of cartilage in an ovine model of osteoarthritis. Osteoarthr. Cartil. 2003, 11, 65–77. [Google Scholar] [CrossRef]
  84. Little, C.; Smith, S.; Ghosh, P.; Bellenger, C. Histomorphological and immunohistochemical evaluation of joint changes in a model of osteoarthritis induced by lateral meniscectomy in sheep. J. Rheumatol. 1997, 24, 2199–2209. [Google Scholar] [PubMed]
  85. Oakley, S.P.; Lassere, M.N.; Portek, I.; Szomor, Z.; Ghosh, P.; Kirkham, B.W.; Murrell, G.A.; Wulf, S.; Appleyard, R.C. Biomechanical, histologic and macroscopic assessment of articular cartilage in a sheep model of osteoarthritis. Osteoarthr. Cartil. 2004, 12, 667–679. [Google Scholar] [CrossRef] [PubMed]
  86. Smith, M.M.; Cake, M.A.; Ghosh, P.; Schiavinato, A.; Read, R.A.; Little, C.B. Significant synovial pathology in a meniscectomy model of osteoarthritis: Modification by intra-articular hyaluronan therapy. Rheumatology 2008, 47, 1172–1178. [Google Scholar] [CrossRef]
  87. Young, A.A.; Smith, M.M.; Smith, S.M.; Cake, M.A.; Ghosh, P.; Read, R.A.; Melrose, J.; Sonnabend, D.H.; Roughley, P.J.; Little, C.B. Regional assessment of articular cartilage gene expression and small proteoglycan metabolism in an animal model of osteoarthritis. Arthritis Res. Ther. 2005, 7, R852–R861. [Google Scholar] [CrossRef]
  88. Dutton, A.Q.; Choong, P.F.; Goh, J.C.; Lee, E.H.; Hui, J.H. Enhancement of meniscal repair in the avascular zone using mesenchymal stem cells in a porcine model. J. Bone Jt. Surg. Br. 2010, 92, 169–175. [Google Scholar] [CrossRef]
  89. Grogan, S.P.; Pauli, C.; Lotz, M.K.; D’Lima, D.D. Relevance of meniscal cell regional phenotype to tissue engineering. Connect. Tissue Res. 2017, 58, 259–270. [Google Scholar] [CrossRef] [PubMed]
  90. Pabbruwe, M.B.; Kafienah, W.; Tarlton, J.F.; Mistry, S.; Fox, D.J.; Hollander, A.P. Repair of meniscal cartilage white zone tears using a stem cell/collagen-scaffold implant. Biomaterials 2010, 31, 2583–2591. [Google Scholar] [CrossRef]
  91. Angele, P.; Johnstone, B.; Kujat, R.; Zellner, J.; Nerlich, M.; Goldberg, V.; Yoo, J. Stem cell based tissue engineering for meniscus repair. J. Biomed. Mater. Res. A 2008, 85, 445–455. [Google Scholar] [CrossRef] [PubMed]
  92. Zellner, J.; Hierl, K.; Mueller, M.; Pfeifer, C.; Berner, A.; Dienstknecht, T.; Krutsch, W.; Geis, S.; Gehmert, S.; Kujat, R.; et al. Stem cell-based tissue-engineering for treatment of meniscal tears in the avascular zone. J. Biomed. Mater. Res. B Appl. Biomater. 2013, 101, 1133–1142. [Google Scholar] [CrossRef]
  93. Nakagawa, Y.; Muneta, T.; Kondo, S.; Mizuno, M.; Takakuda, K.; Ichinose, S.; Tabuchi, T.; Koga, H.; Tsuji, K.; Sekiya, I. Synovial mesenchymal stem cells promote healing after meniscal repair in microminipigs. Osteoarthr. Cartil. 2015, 23, 1007–1017. [Google Scholar] [CrossRef]
  94. Desando, G.; Giavaresi, G.; Cavallo, C.; Bartolotti, I.; Sartoni, F.; Nicoli Aldini, N.; Martini, L.; Parrilli, A.; Mariani, E.; Fini, M.; et al. Autologous bone marrow concentrate in a sheep model of osteoarthritis: New perspectives for cartilage and meniscus repair. Tissue Eng. Part. C Methods 2016, 22, 608–619. [Google Scholar] [CrossRef] [PubMed]
  95. Yuan, X.; Wei, Y.; Villasante, A.; Ng, J.J.D.; Arkonac, D.E.; Chao, P.G.; Vunjak-Novakovic, G. Stem cell delivery in tissue-specific hydrogel enabled meniscal repair in an orthotopic rat model. Biomaterials 2017, 132, 59–71. [Google Scholar] [CrossRef]
  96. Ferris, D.; Frisbie, D.; Kisiday, J.; McIlwraith, C.W. In vivo healing of meniscal lacerations using bone marrow-derived mesenchymal stem cells and fibrin glue. Stem Cells Int. 2012, 2012, 691605. [Google Scholar] [CrossRef] [PubMed]
  97. Korpershoek, J.V.; de Windt, T.S.; Hagmeijer, M.H.; Vonk, L.A.; Saris, D.B. Cell-Based Meniscus Repair and Regeneration: At the Brink of Clinical Translation? A Systematic Review of Preclinical Studies. Orthop. J. Sports Med. 2017, 5. [Google Scholar] [CrossRef] [PubMed]
  98. Pak, J.; Lee, J.H.; Park, K.S.; Jeon, J.H.; Lee, S.H. Potential use of mesenchymal stem cells in human meniscal repair: Current insights. Open Access J. Sports Med. 2017, 8, 33–38. [Google Scholar] [CrossRef]
  99. Whitehouse, M.R.; Howells, N.R.; Parry, M.C.; Austin, E.; Kafienah, W.; Brady, K.; Goodship, A.E.; Eldridge, J.D.; Blom, A.W.; Hollander, A.P. Repair of Torn Avascular Meniscal Cartilage Using Undifferentiated Autologous Mesenchymal Stem Cells: From In Vitro Optimization to a First-in-Human Study. Stem Cells Transl. Med. 2017, 6, 1237–1248. [Google Scholar] [CrossRef]
  100. Zellner, J.; Pattappa, G.; Koch, M.; Lang, S.; Weber, J.; Pfeifer, C.G.; Mueller, M.B.; Kujat, R.; Nerlich, M.; Angele, P. Autologous mesenchymal stem cells or meniscal cells: What is the best cell source for regenerative meniscus treatment in an early osteoarthritis situation? Stem Cell Res. Ther. 2017, 8, 225. [Google Scholar] [CrossRef] [PubMed]
  101. Ruiz-Iban, M.A.; Diaz-Heredia, J.; Garcia-Gomez, I.; Gonzalez-Lizan, F.; Elias-Martin, E.; Abraira, V. The effect of the addition of adipose-derived mesenchymal stem cells to a meniscal repair in the avascular zone: An experimental study in rabbits. Arthroscopy 2011, 27, 1688–1696. [Google Scholar] [CrossRef] [PubMed]
  102. Yu, H.; Adesida, A.B.; Jomha, N.M. Meniscus repair using mesenchymal stem cells—A comprehensive review. Stem Cell Res. Ther. 2015, 6, 86. [Google Scholar] [CrossRef] [PubMed]
  103. Koch, M.; Achatz, F.P.; Lang, S.; Pfeifer, C.G.; Pattappa, G.; Kujat, R.; Nerlich, M.; Angele, P.; Zellner, J. Tissue Engineering of Large Full-Size Meniscus Defects by a Polyurethane Scaffold: Accelerated Regeneration by Mesenchymal Stromal Cells. Stem Cells Int. 2018, 2018, 8207071. [Google Scholar] [CrossRef]
  104. Melrose, J. Strategies in regenerative medicine for intervertebral disc repair using mesenchymal stem cells and bioscaffolds. Regen. Med. 2016, 11, 705–724. [Google Scholar] [CrossRef] [PubMed]
  105. Bochynska, A.I.; Hannink, G.; Grijpma, D.W.; Buma, P. Tissue adhesives for meniscus tear repair: An overview of current advances and prospects for future clinical solutions. J. Mater. Sci. Mater. Med. 2016, 27, 85. [Google Scholar] [CrossRef] [PubMed]
  106. Kremer, A.; Ribitsch, I.; Reboredo, J.; Durr, J.; Egerbacher, M.; Jenner, F.; Walles, H. Three-Dimensional Coculture of Meniscal Cells and Mesenchymal Stem Cells in Collagen Type I Hydrogel on a Small Intestinal Matrix-A Pilot Study Toward Equine Meniscus Tissue Engineering. Tissue Eng. Part A 2017, 23, 390–402. [Google Scholar] [CrossRef] [PubMed]
  107. Zhang, Z.Z.; Wang, S.J.; Zhang, J.Y.; Jiang, W.B.; Huang, A.B.; Qi, Y.S.; Ding, J.X.; Chen, X.S.; Jiang, D.; Yu, J.K. 3D-Printed Poly(epsilon-caprolactone) Scaffold Augmented With Mesenchymal Stem Cells for Total Meniscal Substitution: A 12- and 24-Week Animal Study in a Rabbit Model. Am. J. Sports Med. 2017, 45, 1497–1511. [Google Scholar] [CrossRef]
  108. Giebel, B.; Kordelas, L.; Borger, V. Clinical potential of mesenchymal stem/stromal cell-derived extracellular vesicles. Stem Cell Investig. 2017, 4, 84. [Google Scholar] [CrossRef]
  109. Phinney, D.G.; Pittenger, M.F. Concise Review: MSC-Derived Exosomes for Cell-Free Therapy. Stem Cells 2017, 35, 851–858. [Google Scholar] [CrossRef]
  110. Surman, M.; Drozdz, A.; Stepien, E.; Przybylo, M. Extracellular vesicles as drug delivery systems -methods of production and potential therapeutic applications. Curr. Pharm. Des. 2019. [Google Scholar] [CrossRef]
  111. Ghodbane, S.A.; Patel, J.M.; Brzezinski, A.; Lu, T.M.; Gatt, C.J.; Dunn, M.G. Biomechanical characterization of a novel collagen-hyaluronan infused 3D-printed polymeric device for partial meniscus replacement. J. Biomed. Mater. Res. B Appl. Biomater. 2019. [Google Scholar] [CrossRef] [PubMed]
  112. Iulian, A.; Dan, L.; Camelia, T.; Claudia, M.; Sebastian, G. Synthetic Materials for Osteochondral Tissue Engineering. Adv. Exp. Med. Biol. 2018, 1058, 31–52. [Google Scholar]
  113. Shimomura, K.; Hamamoto, S.; Hart, D.A.; Yoshikawa, H.; Nakamura, N. Meniscal repair and regeneration: Current strategies and future perspectives. J. Clin. Orthop. Trauma 2018, 9, 247–253. [Google Scholar] [CrossRef] [PubMed]
  114. Van Bochove, B.; Hannink, G.; Buma, P.; Grijpma, D.W. Preparation of Designed Poly(trimethylene carbonate) Meniscus Implants by Stereolithography: Challenges in Stereolithography. Macromol. Biosci. 2016, 16, 1853–1863. [Google Scholar] [CrossRef]
  115. Gilbert, T.W.; Badylak, S.F.; Beckman, E.J.; Clower, D.M.; Rubin, J.P. Prevention of seroma formation with TissuGlu(R) surgical adhesive in a canine abdominoplasty model: Long term clinical and histologic studies. J. Plast. Reconstr. Aesthet. Surg. 2013, 66, 414–422. [Google Scholar] [CrossRef] [PubMed]
  116. Walgenbach, K.J.; Bannasch, H.; Kalthoff, S.; Rubin, J.P. Randomized, prospective study of TissuGlu(R) surgical adhesive in the management of wound drainage following abdominoplasty. Aesthet. Plast. Surg. 2012, 36, 491–496. [Google Scholar] [CrossRef] [PubMed]
  117. Cha, H.J.; Hwang, D.S.; Lim, S. Development of bioadhesives from marine mussels. Biotechnol. J. 2008, 3, 631–638. [Google Scholar] [CrossRef] [PubMed]
  118. Favi, P.; Yi, S.; Lenaghan, S.; Xia, L.; Zhang, M. Inspiration from the natural world: From bio-adhesives to bio-inspire adhesives. J. Adhes. Sci. Technol. 2014, 28, 290–319. [Google Scholar] [CrossRef]
  119. Graham, L. An Adhesive Secreted by Australian Frogs of the Genus Notaden. In Biological Adhesives; Smith, A., Ed.; Springer: Cham, Switzerland, 2016. [Google Scholar]
  120. Graham, L.D.; Glattauer, V.; Li, D.; Tyler, M.J.; Ramshaw, J.A. The adhesive skin exudate of Notaden bennetti frogs (Anura: Limnodynastidae) has similarities to the prey capture glue of Euperipatoides sp. velvet worms (Onychophora: Peripatopsidae). Comp. Biochem. Physiol. B Biochem. Mol. Biol. 2013, 165, 250–259. [Google Scholar] [CrossRef] [PubMed]
  121. Brennan, M.J.; Hollingshead, S.E.; Wilker, J.J.; Liu, J.C. Critical factors for the bulk adhesion of engineered elastomeric proteins. R. Soc. Open Sci. 2018, 5. [Google Scholar] [CrossRef] [PubMed]
  122. Moaresifar, K.; Azizian, S.; Hadjizadeh, A. Nano/Biomimetic Tissue Adhesives Development: From Research to Clinical Application. Polym. Rev. 2016, 56, 329–361. [Google Scholar] [CrossRef]
  123. Baek, P.; Voorhaar, L.; Barker, D.; Travas-Sejdic, J. Molecular Approach to Conjugated Polymers with Biomimetic Properties. Acc. Chem. Res. 2018, 51, 1581–1589. [Google Scholar] [CrossRef]
  124. Lee, Y.B.; Kim, S.J.; Kim, E.M.; Byun, H.; Chang, H.K.; Park, J.; Choi, Y.S.; Shin, H. Microcontact printing of polydopamine on thermally expandable hydrogels for controlled cell adhesion and delivery of geometrically defined microtissues. Acta Biomater. 2017, 61, 75–87. [Google Scholar] [CrossRef]
  125. Wei, Z.; Gerecht, S. A self-healing hydrogel as an injectable instructive carrier for cellular morphogenesis. Biomaterials 2018, 185, 86–96. [Google Scholar] [CrossRef]
  126. Wollenberg, A.L.; O’Shea, T.M.; Kim, J.H.; Czechanski, A.; Reinholdt, L.G.; Sofroniew, M.V.; Deming, T.J. Injectable polypeptide hydrogels via methionine modification for neural stem cell delivery. Biomaterials 2018, 178, 527–545. [Google Scholar] [CrossRef] [PubMed]
  127. Yoon, Y.E.; Im, B.G.; Kim, J.S.; Jang, J.H. Multifunctional Self-Adhesive Fibrous Layered Matrix (FiLM) for Tissue Glues and Therapeutic Carriers. Biomacromolecules 2017, 18, 127–140. [Google Scholar] [CrossRef] [PubMed]
  128. Musumeci, G.; Castrogiovanni, P.; Trovato, F.M.; Imbesi, R.; Giunta, S.; Szychlinska, M.A.; Loreto, C.; Castorina, S.; Mobasheri, A. Physical activity ameliorates cartilage degeneration in a rat model of aging: A study on lubricin expression. Scand. J. Med. Sci. Sports 2015, 25, e222–e230. [Google Scholar] [CrossRef]
  129. Castrogiovanni, P.; Di Rosa, M.; Ravalli, S.; Castorina, A.; Guglielmino, C.; Imbesi, R.; Vecchio, M.; Drago, F.; Szychlinska, M.A.; Musumeci, G. Moderate Physical Activity as a Prevention Method for Knee Osteoarthritis and the Role of Synoviocytes as Biological Key. Int. J. Mol. Sci. 2019, 20, 511. [Google Scholar] [CrossRef] [PubMed]
  130. Young, A.A.; Appleyard, R.C.; Smith, M.M.; Melrose, J.; Little, C.B. Dynamic biomechanics correlate with histopathology in human tibial cartilage: A preliminary study. Clin. Orthop. Relat. Res. 2007, 462, 212–220. [Google Scholar] [CrossRef] [PubMed]
  131. Young, A.A.; McLennan, S.; Smith, M.M.; Smith, S.M.; Cake, M.A.; Read, R.A.; Melrose, J.; Sonnabend, D.H.; Flannery, C.R.; Little, C.B. Proteoglycan 4 downregulation in a sheep meniscectomy model of early osteoarthritis. Arthritis Res. Ther. 2006, 8, R41. [Google Scholar] [CrossRef] [PubMed]
  132. Musumeci, G.; Trovato, F.M.; Pichler, K.; Weinberg, A.M.; Loreto, C.; Castrogiovanni, P. Extra-virgin olive oil diet and mild physical activity prevent cartilage degeneration in an osteoarthritis model: An in vivo and in vitro study on lubricin expression. J. Nutr. Biochem. 2013, 24, 2064–2075. [Google Scholar] [CrossRef] [PubMed]
  133. Das, N.; Schmidt, T.A.; Krawetz, R.J.; Dufour, A. Proteoglycan 4: From Mere Lubricant to Regulator of Tissue Homeostasis and Inflammation: Does proteoglycan 4 have the ability to buffer the inflammatory response? BioEssays 2019, 41, e1800166. [Google Scholar] [CrossRef]
Figure 1. Knee anatomy showing the location of the knee joint menisci (a, b) with a radial tear arrowed (c) and the collagen fibre organisation of the menisci in vertical sections through lateral (d) and medial menisci (e) which equip these as weight bearing stabilising structures in the knee joint. Radial tie bundles and collagen fibrillar arrangements in the surface and basal meniscal laminas are also shown (fh). The inner zone meniscal cells are cartilaginous whereas the outer zone meniscal cells are more fibrocartilaginous (i). This is clearly evident in the focal localisation of the HS-proteoglycan perlecan which is a chondrogenic marker [10]. Figure modified from [18] with permission of InTech Open Publishers: London. Figure copyright J. Melrose.
Figure 1. Knee anatomy showing the location of the knee joint menisci (a, b) with a radial tear arrowed (c) and the collagen fibre organisation of the menisci in vertical sections through lateral (d) and medial menisci (e) which equip these as weight bearing stabilising structures in the knee joint. Radial tie bundles and collagen fibrillar arrangements in the surface and basal meniscal laminas are also shown (fh). The inner zone meniscal cells are cartilaginous whereas the outer zone meniscal cells are more fibrocartilaginous (i). This is clearly evident in the focal localisation of the HS-proteoglycan perlecan which is a chondrogenic marker [10]. Figure modified from [18] with permission of InTech Open Publishers: London. Figure copyright J. Melrose.
Cells 08 00324 g001

Share and Cite

MDPI and ACS Style

Melrose, J. The Importance of the Knee Joint Meniscal Fibrocartilages as Stabilizing Weight Bearing Structures Providing Global Protection to Human Knee-Joint Tissues. Cells 2019, 8, 324. https://doi.org/10.3390/cells8040324

AMA Style

Melrose J. The Importance of the Knee Joint Meniscal Fibrocartilages as Stabilizing Weight Bearing Structures Providing Global Protection to Human Knee-Joint Tissues. Cells. 2019; 8(4):324. https://doi.org/10.3390/cells8040324

Chicago/Turabian Style

Melrose, James. 2019. "The Importance of the Knee Joint Meniscal Fibrocartilages as Stabilizing Weight Bearing Structures Providing Global Protection to Human Knee-Joint Tissues" Cells 8, no. 4: 324. https://doi.org/10.3390/cells8040324

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