Next Article in Journal
Muscle Cell Insulin Resistance Is Attenuated by Rosmarinic Acid: Elucidating the Mechanisms Involved
Next Article in Special Issue
Extracellular Matrix Disorganization and Sarcolemmal Alterations in COL6-Related Myopathy Patients with New Variants of COL6 Genes
Previous Article in Journal
Uridine Alleviates Sepsis-Induced Acute Lung Injury by Inhibiting Ferroptosis of Macrophage
Previous Article in Special Issue
A Schematic Approach to Defining the Prevalence of COL VI Variants in Five Years of Next-Generation Sequencing
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Collagen VI in the Musculoskeletal System

by
Alberto Di Martino
1,2,
Matilde Cescon
3,
Claudio D’Agostino
1,2,
Francesco Schilardi
1,2,
Patrizia Sabatelli
4,5,
Luciano Merlini
2,* and
Cesare Faldini
1,2
1
I Orthopedic and Traumatology Department, IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
2
Department of Biomedical and Neuromotor Science, DIBINEM, University of Bologna, 40136 Bologna, Italy
3
Department of Molecular Medicine, University of Padova, 35131 Padova, Italy
4
Unit of Bologna, CNR-Institute of Molecular Genetics “Luigi Luca Cavalli-Sforza”, 40136 Bologna, Italy
5
IRCCS Istituto Ortopedico Rizzoli, 40136 Bologna, Italy
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(6), 5095; https://doi.org/10.3390/ijms24065095
Submission received: 2 February 2023 / Revised: 26 February 2023 / Accepted: 1 March 2023 / Published: 7 March 2023
(This article belongs to the Special Issue Collagen VI-Related Myopathies—COL6-RMs)

Abstract

:
Collagen VI exerts several functions in the tissues in which it is expressed, including mechanical roles, cytoprotective functions with the inhibition of apoptosis and oxidative damage, and the promotion of tumor growth and progression by the regulation of cell differentiation and autophagic mechanisms. Mutations in the genes encoding collagen VI main chains, COL6A1, COL6A2 and COL6A3, are responsible for a spectrum of congenital muscular disorders, namely Ullrich congenital muscular dystrophy (UCMD), Bethlem myopathy (BM) and myosclerosis myopathy (MM), which show a variable combination of muscle wasting and weakness, joint contractures, distal laxity, and respiratory compromise. No effective therapeutic strategy is available so far for these diseases; moreover, the effects of collagen VI mutations on other tissues is poorly investigated. The aim of this review is to outline the role of collagen VI in the musculoskeletal system and to give an update about the tissue-specific functions revealed by studies on animal models and from patients’ derived samples in order to fill the knowledge gap between scientists and the clinicians who daily manage patients affected by collagen VI-related myopathies.

1. Introduction

The musculoskeletal system has the primary functions of supporting the body, allowing motion, and protecting vital organs [1]. It is composed of the skeleton’s bones, cartilage, tendons, ligaments, and other connective tissue that supports and connects the different counterparts constituting the joints. Motion around the joints is powered by skeletal muscles, connected to neighboring bony structures, able to transduce the force generated by muscle contraction. Through contraction, muscles allow the motion of bones attached to them by tendons. While bones provide support to the body, joints ensure the connections of contiguous bones; the cartilage at that level prevents the bone ends from rubbing directly onto each other, while ligaments connect bones and contribute to articular stabilization [1].
The word “connective tissue” refers to the tissue that supports and ties tissues and organs. It consists of fibroblasts and their surrounding extracellular matrix (ECM). The ECM is crucial for cell adhesion, stability, and regeneration in different tissues [1]. An important role is played by the different types of collagens allowing the formation of a structure that supports a remarkably wide range of tissues. Among the different types of collagens typically involved in the formation of the ECM (including types I, II, III, VI, IX, and XI), collagen VI (colVI) is expressed as a ubiquitous ECM protein in the stroma, forming a microfibrillar network associated with the basement membrane [2].
ColVI exerts several functions in the tissues in which it is expressed, ranging from mechanical roles, which are typical of collagen components of the ECM, to more specific cytoprotective functions, including the inhibition of apoptosis and oxidative damage, the promotion of tumor growth and progression, and the regulation of cell differentiation and autophagic mechanisms; moreover, it is involved in the maintenance of cell stemness [3]. ColVI monomer is made by the assembly of three different chains, α1(VI), α2(VI), and α3(VI), the latter alternatively substituted by α5(VI) or α6(VI) in humans, in a tissue-specific manner [4,5]. Mutations in the genes encoding its main chains, COL6A1, COL6A2 and COL6A3, are responsible for the spectrum of congenital muscular disorders, namely Ullrich congenital muscular dystrophy (UCMD), Bethlem myopathy (BM), and myosclerosis myopathy (MM), showing a variable combination of muscle wasting and weakness, joint contractures, distal laxity, and respiratory compromise [6,7,8]. These diseases differ in terms of clinical appearance and severity of the disorder. UCMD has a more severe pattern, and it is characterized by early-onset weakness and proximal contractures, as well as by pronounced distal joint hyperlaxity. Patients in most severe cases do not achieve ambulation, or it can be achieved only for a limited period of time [9]. BM has milder manifestations; patients usually ambulate well in adulthood, albeit the typical joint contractures are prominent [9]. MM is characterized by early-onset widespread and progressive muscle and joint contractures, paralleled by “woody” and atrophic muscle consistency, causing a severe limitation of motion [8]. Despite the depth of our knowledge about these diseases, no effective therapeutic strategy has been developed so far [10]; moreover, the effects of colVI mutations on other tissues have been poorly investigated so far.
The aim of this review is to outline the role of colVI in the musculoskeletal system and to give an update about the tissue-specific functions revealed by studies on animal models and from samples of patients affected by the above-mentioned diseases. The main objective is to fill the knowledge gap between scientists and the clinicians who daily manage patients affected by collagen VI-related myopathies, as well as to provide an easy consultation guide and a source to plan and drive further research on this topic.

2. Collagen VI Expression, Assembly, and Secretion

ColVI formation requires the harmonic expression of different genes. Six independent genes were found conserved between mice and humans that are responsible for the expression of colVI α-chains. By mapping the human genome, the genes have been located on three chromosomes: the COL6A1 and COL6A2 genes coding for α1(VI) and α2(VI), respectively, are located on chromosome 21q22.3 [9]; on chromosome 2q37 lies the COL6A3 gene; and three other alternative chains of α3(VI) are transcribed by the COL6A4, COL6A5, and COL6A6 genes that are mapped on chromosome 3q22.1 [4]. The human COL6A4 gene, due to chromosome inversion, transcribes for noncoding pseudogenes [4].
In the N- and C-terminal globular regions of all colVI chains, a von Willebrand factor type A (vWF-A) module flanks the triple-helix domain of 335–336 amino acid residues. The α1(VI) and α2(VI) chains are 130–150 kDa in size, with one N-terminal and two C-terminal vWF-A modules (N1, and C1 and C2, respectively) [11]. The α3(VI), α5(VI) and α6(VI) chains are considerably larger, ranging from approximately 220 kDa to over 300 kDa. Despite structural similarities at the N-terminal end, these show two C-terminal vWF-A modules (C1 and C2) followed by distinctive regions (C3, C4, and C5). The C3 domain includes a proline-rich sequence; C4 is a fibronectin-type-III domain; and C5 is a Kunitz-like domain [12].
Several alternatively spliced variants that involve some of the vWF-A modules and the N-domains have been described in mouse tissues and human cells [13,14]. Most frequently in humans, the alternative splicing of α2(VI) can produce three different protein variants, known as α2C2, α2C2a and α2C2a′ [15].
Early studies described colVI as being constituted by the α1(VI), α2(VI), and α3(VI) chains, which assemble into a triple-helix monomer (≈500 kDa) [16,17,18,19,20,21]. The short triple-helix formation is favored by the presence of Gly-X-Y triplets; the presence of proline in the Y position, potentially modified to hydroxyproline [22], is able to confer most of the stability to the triple helix [23]. A 1:1:1 stochiometric ratio for the α1(VI), α2(VI), and α3(VI) chains in forming a heterotrimeric monomer is strongly supported by in vitro studies performed by Lamandé et al. [24] in a particular cell line expressing high levels of mRNA encoding for α1(VI) and α2(VI), but not α3(VI). As a result, the cell line did not produce a colVI triple helix, demonstrating that α3(VI) expression was essential for the assembly of a colVI monomer [24]. Recently, Gara et al. [4] identified three novel chains, α4(VI), α5(VI), and α6(VI), that share a similar structure to the α3(VI). These polypeptides allow the variable assembly of colVI triple-helix monomers as α1–α2–αX (where αX could alternatively be α3, α4, α5, or α6) [4].
Prior to secretion, colVI goes through a rigorous multistep intracellular assembly process into larger aggregates [25]. The intracellular assembly involves the formation of antiparallel dimers (≈1000 kDa), which assemble into tetramers (≈2000 kDa), both stabilized by disulfide bonds [26]. Finally, these tetramers are released into the extracellular space, where they link end-to-end via non-covalent interactions to create the peculiar 105 nm beaded microfilaments visible by electron microscopy [21,27].
Globular domains of colVI not only contribute to monomer assembly [28] and further to dimer [25] and tetramer [28] association and stabilization but are also pivotal in granting heterotypic interactions with several ECM molecules in the extracellular environment [29,30,31], although triple helical regions also mediate some of these interactions [32,33]. The major colVI interactors among ECM proteins include hyaluronan and heparin [34], fibrillar collagens such as collagen I [12], collagen II [31], basement membrane-associated collagen IV [29], fibronectin [35] and the proteoglycans biglycan, decorin [36], and perlecan [32] (a wider description is provided in Cescon et al., 2015 [3]). Nonetheless, the colVI superstructure is supposed to favor the engagement of specific membrane protein receptors. While some of these were experimentally identified, the search for the colVI receptors transducing its signal intracellularly is still an open field of research.

3. Collagen VI in Tissues Relevant for the Musculoskeletal System

The ubiquitous expression of colVI and its interaction with ECM constituents suggest that it plays a key role in the repair, development, and homeostasis processes in a multitude of tissues [37,38].
With relevance for the musculoskeletal system, current knowledge about the role of colVI in skeletal muscle, bone, tendon, cartilage, and ligament tissue will be presented in subsequent sections. A schematic description is also provided in Figure 1 and Figure 2.

4. Skeletal Muscle

4.1. General Aspects

Muscle tissue is a highly specialized contractile tissue found in the human body in three types: smooth, cardiac, and skeletal [1].
Skeletal muscle is composed of muscle cells and myoblasts that, during development, fuse into a multinucleated muscle fiber. Thanks to the arrangement of sarcomeres, this muscle fiber can contract and shorten along a longitudinal axis. Muscle fibers are surrounded by a layer of connective tissue, called the endomysium. Muscle fibers associated in muscle bundles are enclosed by a further layer of connective tissue, the perimysium, while a third layer, called the epimysium, enwraps the whole muscle structure and is in continuity with the deep fascia, separating muscles from other surrounding tissues. These layers of connective tissues, beyond their containment role, are pivotal elements in the transmission of skeletal muscle force produced by myofiber contraction [39].
ColVI is a major component of the skeletal muscle ECM; it is diffusely expressed in the different layers of muscle connective tissue, including the endomysium, perimysium, epimysium, and deep fascia [40]. As visualized by the electron microscope immunogold technique, colVI microfilaments distribute close to the collagen and fibronectin fibrils [41] of the muscle interstitium and form a dense network in the reticular lamina of the muscle fibers, connecting the basal lamina with the fibrillar collagen of the ECM [30,41]. Interestingly, by using single-chain-specific antibodies, the α3 chain appears ubiquitous, the α6 chain is less expressed and mainly interstitial, while the α5 chain is selectively expressed at the myotendinous junctions. The restricted and differential distribution pattern of the α5 and α6 chains, with respect to their homologue α3 chain, implies that different colVI isoforms may have specific roles in specialized muscle ECM structures. A unique critical role of α3 chain-based heterotrimers is at the level of the muscle fiber basement membrane [42], where, by interacting with ECM components and sarcolemmal receptors, they may contribute to muscle fiber stability and integrity during contraction.
Interstitial fibroblasts are the major cell type responsible for the deposition of colVI [43], whereas myogenic cells, which produce factors that can influence collagen secretion, do not express it [44], with the exception of quiescent satellite cells that express colVI until they become activated [45]. Interestingly, colVI was recently described as being part of the highly specialized ECM within the synaptic cleft of the neuromuscular junction (NMJ) [46].
The crucial role of colVI in skeletal muscle is emphasized by the fact that mutations in the genes encoding its chains have a causative role in several forms of inherited human muscle diseases, including BM, UCMD, and MM [8,47,48] as previously described.

4.2. Animal Models

Initial studies aiming at understanding the role of colVI in vivo led to the production of the first murine model with inactivation of the Col6a1 gene, the Col6a1−/− mouse, in which the ablation of the major α1(VI) chain results in the complete absence of colVI secretion in all the tissues studied so far [49]. Analyses performed on the Col6a1 null mice revealed a cytoprotective role of colVI since the ablation of the protein triggered spontaneous apoptosis in muscle fibers that were associated with latent mitochondrial dysfunction and organelle alterations [50]. In the colVI knockout muscle, an increased opening of the permeability transition pore (PTP) in the mitochondrial inner membrane was highlighted as causative for the dystrophic phenotype [27,51] and became a major successfully targeted downstream defect by the therapeutical approaches tested thus far [51,52]. Mitochondrial alterations were paralleled by increased oxidative damage and reactive oxygen species (ROS) production [53]. Therapeutic approaches were successfully designed, targeting the increased level of ROS activity found in colVI null muscles and depending on the raised activity of mono-amino-oxidase (MAO), an enzyme located on the external mitochondrial membrane and responsible for myofibrillar protein oxidation. MAO inhibition, by its inhibitor pargyline, was tested in mice and was able to ameliorate the muscle phenotype [53].
Subsequent studies demonstrated that the occurrence of altered mitochondria in Col6a1−/− muscle is due to defects in the regulation of the autophagic pathway and that autophagy flux reactivation by pharmacological, genetic, or nutritional approaches is able to rescue the occurrence of dysfunctional organelles, apoptosis, and muscle strength [54,55,56]. Interestingly, the same features, namely motility impairment, altered organelle ultrastructure, and defective autophagy were also described in a novel mutant colVI knockout zebrafish, throwing light on the role of colVI during embryonic development and rendering the mentioned features as candidate targets in high-throughput drug screening approaches [57].
ColVI was found to have a role in the niche of muscle stem cells. The colVI genes are not only expressed by isolated quiescent (but not activated) satellite cells but are also overexpressed in skeletal muscle upon injury, triggered by cardiotoxin. In this condition, Urciuolo and colleagues demonstrated that Col6a1−/− mice had impaired satellite cell self-renewal capabilities and defective muscle regeneration [45]. In this context, a role for colVI in regulating muscle stiffness was unraveled by demonstrating a lower stiffness in Col6a1−/− muscles at a level that both in vivo as well as in vitro, when recreated by engineered structures, promotes satellite cell differentiation rather than sustaining self-renewal [45].
The in vivo function of colVI at the NMJ was also investigated in adult Col6a1−/− mice. Fluorophore-coupled α-bungarotoxin (αBT), which specifically binds acetylcholine receptors (AChRs), allowed monitoring of the morphology of post-synaptic boutons on different muscles, showing greater fragmentation than wild-type, a typical hallmark of denervation or aging and altered expression level for a panel of NMJ-enriched proteins at the post-synaptic apparatus [46]. This was paralleled by an increase in the expression of the fetal Chrng and activity-dependent Chrna genes, but not Chrne, indicating NMJ changes and an abnormal neuromuscular transmission, further demonstrated by electrophysiological analysis [46]. Of note, studies performed on colVI knockout zebrafish demonstrated a reduced motor axon elongation and branching within the myotome at both 24 and 48 h of development, thus revealing an early defect in muscle innervation and, in turn, impairing mutant fish motility [57].
Despite the severe effects of colVI ablation on muscle fibers and cell-survival pathways like apoptosis and autophagy, the specific cell receptors that mediate these effects in muscle from colVI are still unclear [50,54]. Early in vitro studies identified several cell surface receptors that are capable of binding colVI, including α1β1, α2β1, α3β1, α10β1, and αvβ3 integrins [58,59,60], as well as the chondroitin sulfate proteoglycan-4 (CSPG4; also known as NG2) [61,62,63] and, more recently, the anthrax toxin receptor 2 (Antxr2/CMG2) [64]. However, how colVI extracellular signals are transduced within muscle fibers remains unknown.

4.3. Clinical Studies

In patients affected by BM, symptoms differ from the onset of the disease. Patients with prenatal-onset BM usually show decreased fetal movements, congenital torticollis or bilateral clubfoot. Hypotonia and delayed motor milestones are the clinical typical presentation of neonatal-onset BM. More often, patients with early-childhood-onset BM present limb-girdle weakness or joint contractures. Sometimes in adult-onset BM, contractures can define the clinical presentation and overshadow muscle weakness [65]. Patients present systemic muscular atrophy and weakness affecting proximal muscles more than distal ones and extensors more than flexors. Contractures of the interphalangeal joints of the last four fingers as well as flexion contractures of the elbow and ankles are the most common clinical symptoms [66].
Otherwise, patients affected by UCMD manifest proximal joint contractures, striking distal hyperextensibility, and protruding calcanei [67,68]. Contractures of proximal joints, congenital torticollis, and delayed motor developmental milestones are other common clinical presentations [69]. Muscular weakening and atrophy are systemic and gradually progressive. Hyperextensibility of distal appendicular joints and flexor muscle weakness are common clinical signs [70].
Alterations in skeletal muscles underlie the typical clinical symptoms of patients affected by BM and UCMD. Different research groups [27,50,51,52], through the study of muscle biopsies and myoblast cultures, assessed the role of a Ca2+-mediated mitochondrial dysfunction in the pathogenesis of muscle fiber death. Functional and ultrastructural mitochondrial abnormalities are due to improper opening of the permeability transition pore, a mitochondrial inner membrane channel [52]. Moreover, the inadequate removal of defective mitochondria and the increased apoptosis amplify the damage [27].
This evidence laid the foundation for the latest clinical trials, and aiming at the downstream targets of genetic defects is still a major focus in colVI-RM therapeutic approaches.
In one of the latest clinical trials, Merlini and colleagues [10,71] demonstrated how these typical mitochondrial alterations could be controlled using Cyclosporin A, an immunosuppressant that desensitizes the permeability transition pore independently of calcineurin inhibition [72]. In accordance with studies in mice, patients’ myoblasts were also found to be more sensitive to oxidative damage in cultures, and the inhibition of MAO activity by pargyline significantly reduced both ROS accumulation and mitochondrial dysfunction [73].
Moreover, colVI deficiency in mice results in defective autophagy flux regulation and sustained activation of the Akt–mTOR pathway under fasting conditions, as well as reduced levels of Beclin 1 and BNIP3, which are essential effectors in autophagy initiation. Indeed, muscle biopsies from UCMD and Bethlem myopathy patients exhibited a significant drop in Beclin 1 and BNIP3 protein levels, demonstrating a failure in autophagy regulation in collagen VI-related myopathies [54]. Such an aspect was also approached in a clinical trial aiming at testing a low-protein diet for the reactivation of autophagy in BM/UCMD patients, leading to increased autophagic markers and reduced apoptosis in patients’ biopsies, with evidence of benefits for muscles and metabolic modulation towards an improved mitochondrial homeostasis, as well as to the identification of blood leukocytes as a worthy noninvasive tool to monitor autophagy in patients [74].
Clinical observations also highlighted the relevance for the role of colVI in satellite cells and at the NMJ. Indeed, a defect in the regenerative potential of satellite cells upon the lack of colVI is consistent with our study [75] and other works [76] on UCMD muscle biopsies, which reported prominent but abnormal regeneration in UCMD muscle biopsies. Moreover, muscle biopsies isolated from patients with different colVI gene mutations and clinical phenotypes ranging from typical BM to severe UCMD, showed significantly greater colVI and utrophin expression at the level of NMJs than in unaffected control biopsies. This finding is consistent with the Col6a1−/− mice abnormality. Moreover, UCMD biopsies revealed elevated mRNA levels of the CHRNG and CHRNA genes (which code for the acetylcholine receptor subunits) when compared with unaffected control biopsies, similarly to Col6a1−/− mice [46].

5. Bone

5.1. General Aspects

ColVI is an important bone matrix protein [77] that is linked to bone remodeling and bone development [78,79]. Its deficiency alters the configuration and the shape of osteoblasts in both the inner periosteal layer and the cancellous bone, which affects the maintenance of bone mass because osteoblasts require a regular morphology to work properly [77]. Bone forms the skeleton—the structure that gives shape to the body, supports its weight, and facilitates locomotion. Bone is also able to react elastically to mechanical forces. If it breaks, and under the appropriate conditions, it heals without leaving scars, regaining its original shape [1]. Bone is a highly dynamic tissue whose formation and structural integrity are regulated by specialized cell types: osteoblasts regulate the anabolic processes that lead to new bone formation and closely collaborate with osteoclasts, which have the catabolic role of resorbing bones. This role-play is at the basis of the bone turnover, which is the process that replaces and renews micro-damaged bone tissue from impact or aging [80]. An imbalance in bone turnover can lead to osteopetrosis (an excess of bone matrix) or osteopenia (insufficient bone matrix). In both cases, the bone is fragile and at risk of impending fractures [81,82]. The most prevalent type of collagen in bone is type I collagen, which is made up of two α1(I) and one α2(I) chains joined together to form a triple-helix structure. The clinical manifestations of individuals with mutations in type I collagen chains or in the enzymes that control collagen maturation demonstrate the significance of type I collagen in bone, as in the case of brittle bone disease, also known as osteogenesis imperfecta [83]. Collagen I is an important regulator of osteoblast differentiation and a guide for the calcification of bone tissue [84]. In this scenario, colVI appears to be involved in ECM structural stabilization via direct interactions with other macromolecules, including collagen I. In the initial stages of IL-4-induced mineralization, colVI plays a role in controlling collagen I expression [85]. In normal human periosteal osteoblast-like cells (named SaM-1 cells), interleukin-4 increased collagen I and osteocalcin accumulation and caused mineralization [86]. ColVI was reported to be expressed by SaM-1 cells. The microenvironment of osteoblasts and their surrounding matrix would be mediated by colVI, which is produced by osteoblasts [77]. The rhIL-4 used in the studies [87,88] increased the expression of α1(VI) and α2(VI) collagen mRNA and the deposition of colVI in the cell layer, suggesting that an accumulation of colVI in the surrounding ECM may modulate the upregulation of collagen I mRNA in rhIL-4 treated SaM-1 cells [87,88]. In the early stages of osteoblastic differentiation, colVI may act as a matrix-derived inducer of integrin-mediated collagen I expression [85].

5.2. Animal Models

Several researchers investigated the hypothesis that the absence of colVI affects bone formation and age-related alterations.
After an initial observation that colVI deficiency led to delayed ossification in one-month-old mice and general decreased bone mineral density in adults [89], Izu et al. [77] studied how osteoblasts and bone structure react upon colVI depletion in four-week-old Col6a1−/− and wild-type mice. According to 3D-μCT images, the secondary trabecular bone in the metaphyseal area of the femur showed a sparse pattern due to the colVI deficit. The levels of bone volume in cancellous bone decreased by approximately 30% in Col6a1−/− mice [77]. Histologically, the effects of colVI deficit on the morphology of osteoblasts were investigated. In cancellous bone from wild-type mice, osteoblastic cells were seen with a cuboidal form in a single layer on the surface of the trabecular bone at hematoxylin and eosin staining. These cells also showed an ovoid nucleus filled with cytoplasm, which is a common characteristic of osteoblastic cells secreting bone matrix. Conversely, in Col6a1−/− mice, osteoblasts on the surface of the trabecular bone displayed irregular cellular borders, and the shape of the osteoblasts in the cortical bone was flattened. Moreover, osteoblasts appeared chaotically organized, and the border between these and the bone matrix was severely disrupted, which shifted from having a straight look in wild-type to a wavy appearance in knockout mice [77]. The study also looked at the bone’s dynamic metabolic characteristics. Based on the results of the investigation utilizing calcein double labeling, bone formation parameters were investigated. Although not statistically significant, the mean values of the rates of mineral apposition and bone formation were lower in Col6a1−/− than in wild-type mice [77].
The knee joints of the Col6a1−/− mice were another subject of the earliest in vivo investigation conducted by Christensen et al. [78] that looked at the role of colVI in bone structure and physical characteristics. By 3D-μCT performed in the trabecular regions of the tibial epiphysis, they found that, compared with wild-type mice whose trabecular bone volume increased noticeably with age, the trabecular bone volume of Col6a1−/− mice remained smaller and steady. Connectivity density decreased noticeably as Col6a1−/− mice aged, while it remained consistently greater in wild-type mice. Bone tissue density increased only 4% with skeletal maturity from 2 to 9 months in Col6a1−/− mice, whereas in wild-type mice bone tissue density increased 19% [78]. Bone volume and bone tissue density were measured by 3D-μCT at the tibial metaphysis. Both displayed a statistically significant main effect of age (but not genotype) and a statistically significant age-by-genotype interaction effect. In contrast to wild-type mice, whose bone volume had an increase of 77% from 2 to 9 months, the bone volume of Col6a1−/− mice volume did not alter appreciably overtime. Metaphyseal tissue density in wild-type mice had an increase of 34% compared with 7% in Col6a1−/− mice [78]. Finally, Col6a1−/− mice did not gain any mineral content with age [78]. All these findings clearly indicated that substantial skeletal disorders are linked to colVI deficiency.
It is well established that mechanical loading is critical for bone development and maturation [78]. For these reasons, due to the myopathy of Col6a1−/− mice, inappropriate knee loading may exist and may affect the properties of trabecular bone [49]. The variations between Col6a1−/− and wild-type trabecular bone formations closely reflect the pattern of alterations discovered in the research on limited locomotor modes [78], mechanically-altered loading [90], or disuse [91].
More recently, Pham and colleagues [83] used 3D-μCT to measure femoral and vertebral bone mass in a different model, the Col6a2−/− mouse. ColVI expression in bone was evaluated using immunofluorescence labeling in wild-type and Col6a2−/− mouse bones and was found to be strong at the level of the hypertrophic cartilage layer next to newly produced bone in the primary ossification center in one-month-old wild-type mice, while absent in the knockout ones. Col6a2−/− mice were marginally smaller than their age- and sex-matched wild-type counterparts, with DEXA measurement revealing a reduced whole-body bone mineral density (BMD) compared with wild-type [83], in agreement with findings previously published by Alexopulos and colleagues in the Col6a1−/− mice [89]. To determine how Col6a2−/− mouse bones were affected by colVI deficiency, isolated femora and vertebrae (L3) were dissected and µCT analyzed. Both the femur and the L3 vertebrae of three-month-old Col6a2−/− mice showed significantly reduced BMD, reduced bone volume upon the whole tissue volume (BV/TV), smaller numbers of trabeculae, and higher trabecular spacing compared with wild-type mice of the same age [83]. These variations persisted with age and were still visible in six-month-old mice [83]. In mice lacking Col6a2, cortical bone was not affected. When the middle diaphyseal cortical femur was examined for modifications, micro-CT examinations revealed no glaring differences in the cortical morphology. Quantification of the cortical area revealed no appreciable differences in the diameter of the diaphysis, in medullary diameter, in BV/TV, cortical thickness, cortical porosity, or BMD between the Col6a2−/− and wild-type controls [83].
The skeletal phenotype of mice deficient in α2(VI) protein was further analyzed, demonstrating how colVI governs the balance between bone formation and resorption to regulate trabecular bone mass in both femur and spine. Col6a2−/− mice exhibit enhanced osteoclastogenesis but no change in bone formation characteristics. Dynamic histomorphometry using a double fluorochrome labeling technique was used to identify the cellular basis for the reduced bone mass phenotype observed in Col6a2−/− mice. There were no significant differences in mineral apposition rate, mineralizing perimeter, or bone formation rate between Col6a2−/− and wild-type mice [83]. The osteoblast-expressed genes osteopontin (Spp1) and osteocalcin (Bglap) had identical relative mRNA expression levels in wild-type and Col6a2−/−-derived cells. Using fluorescence-based staining to examine osteoclastogenesis levels in bones, Col6a2−/− animals exhibited significantly more positive staining than wild-type mice in the number of osteoclasts per trabecular length. The expression of an osteoclast-associated Ig-like receptor and Cathepsin K revealed that the relative levels of osteoclast-expressed genes were substantially higher in the Col6a2−/− bones [83].
The dysregulation of bone remodeling pathways was revealed by RNA sequencing analysis that was performed on bones from wild-type and Col6a2−/− mice, supporting an enhanced osteoclastogenesis and a potential connection to TNF-α.
The quantity of myeloid osteoclast precursors was not affected in Col6a2−/− mice, but the response to TNF-α was increased in osteoclast precursors of Col6a2−/− mice. TNF-α was found to be an upstream regulator in Col6a2−/− cells by further analysis of the RNA sequencing data. Pham and colleagues [83] treated wild-type osteoclast progenitors with or without conditioned media from wild-type or Col6a2−/− BMSCs (bone marrow stroma cells) to see if TNF-α activity was altered. TNF-α response was more effective in BMSCs treated with conditioned medium from Col6a2−/− cells than in the other sets, indicating that TNF-α plays a role in the hyperactive osteoclastogenesis reported in Col6a2−/− mice [83].
In vitro, α2(VI) directly bound to TNF-α and inhibited its ability to cause osteoclastogenesis in osteoclast precursors exposed to TNF-α: a dose-dependent response was observed in recombinant human COL6A2 fragment binding when the experiment was conducted on TNF-α-coated plates. If an osteoclast precursor cell-line was treated with either RANKL or TNF-α to enhance osteoclastogenesis or with colVI, it was demonstrated that TNF-α increased RANKL-induced osteoclastogenesis and colVI decreased osteoclast quantity and differentiation in RANKL and TNF-α-induced cells. ColVI binds to TNF-α and inhibits its capacity to promote osteoclastogenesis. Therefore, when Col6a2 was depleted, TNF-α was not trapped in the ECM and was free to boost RANKL’s effects on osteoclastogenesis [83].

5.3. Clinical Studies

In the literature to date, there are few clinical studies focusing on bone in patients affected by colVI-RM. However, osteopenia is a commonly reported finding in patients with BM or UCMD, as well as bone deformities (e.g., high-arched palate, congenital hip dislocation, protruding calcanei, torticollis, and kyphotic spine deformity) [92]. As early as Ullrich’s initial description, osteopenia was found in several cases [68,93].
In the study conducted by Toni and colleagues [92], patients with BM and UCDM underwent dual-energy X-ray absorptiometry (DXA). By calculating the T-score, the analysis showed that the bone mineral content is greatly reduced in these patients, especially in men.
Finally, Philippe et al. [94] analyzed bone mineral density in a group of adult patients with muscular dystrophies and noted greatly reduced values compared with the general population, especially in wheelchair-bound patients. Although those patients were affected by different syndromes, their clinical conditions and disabilities are similar to those affected by BM and UCDM.

6. Tendon

6.1. General Aspects

Tendons are bands of fibrous connective tissue that connect a muscle to a bone, allowing the transmission of force generated at the muscle level. Each muscle is characterized by at least two tendons, one proximal and one distal. The transition area between muscle and tendon tissue is called the myotendinous junction, while the area of insertion at bone level is called the osteotendinous junction [1].
Tendons transfer mechanical stresses from muscle to bone; these are composed of fibroblast columns (tenocytes) with collagen fibrils serving as the primary structural component. The majority of fibrils are composed of collagen type I, but they also contain glycoproteins, proteoglycans, collagen types III, V, VI, XII, and XIV, as well as other substances that aid in the process of fibrillogenesis. Fibrils are arranged uniaxially into fibers; tenocytes and fibers join together to form fascicles, which are then linked together by connective tissue sheaths to form the mature, weight-bearing tendon [95,96,97].
ColVI is found in the interfibrillar tendon’s extracellular matrix (ECM), where it acts as a link among the coarser collagen fibrils. In addition, it is enriched in the tenocyte’s pericellular matrix (PCM), where it forms a microfibrillar scaffold critical for cell behavior and function [98]. Because of its cell–matrix and matrix–matrix interactions, colVI acts as a critical regulator of matrix signals [29,99]. Moreover, it influences collagen fibril construction and tendon function. ColVI may function as a “bridge” between cells and the ECM, playing important roles in cell mechanosensation and cell surface growth factor modulation [100].
Patients suffering from collagen VI-related myopathies have axial and proximal joint contractures, as well as distal joint hypermobility, indicating that tendon structure and function are compromised.

6.2. Animal Models

The functional roles of colVI in tendons were investigated using a mouse model with a specific deletion of the Col6a1 gene. The flexor digitorum longus (FDL) tendons of Col6Ia1−/− and wild-type mice were dissected at P1, P7, P16, and P30. ColVI was immuno-localized in wild-type FDL tendons on days P1, P7, P16, and P30 of development. Its reactivity was increased in the pericellular area of tendon fibroblasts, localizing within and around tendon fibroblasts at the level of the fibroblast processes, suggesting a role for colVI in tendon formation at the fibroblast–matrix interface.
The morphological characteristics of Col6a1−/− and wild-type growing tendons were studied using TEM of FDL tendons at P1 and P30. At an early stage of tendon formation, wild-type tendon fibroblasts were physiologically structured and produced well-defined fibers (P1). Tendon fibroblasts aided in the formation of micro-domains in which fibers were assembled; this arrangement was disrupted in P1 Col6a1−/− tendons, displaying altered fibroblast cell shape and organization. This resulted in disordered micro-domain organization and disruption of tendons fibers structure in Col6a1−/− tendons. Of note, micro-domain structure and fiber organization remained abnormal as tendon development progressed (P30).
Smaller and less structured fibers were present in Col6a1−/− tendons than in wild-type. Thus, it is thought, that col VI plays a key role in tendon development by maintaining cell shape and fiber organization.
At time P1, no differences were evident in the tendons of col6a1−/− and wild-types. Conversely, at P30, structural and fibrillar organization differences were shown in Col6a1−/− pericellular area [101].
Fibrillar structure and fibrillar diameter variation at the pericellular region and regions displaced from the cells to the fiber’s center were examined. Fibrils in Col6a1−/− tendons had a smaller diameter than those in wild-type tendons in both areas; in wild-type tendons, on the other hand, fibrils of larger diameter and uniform distribution were present. Thus, colVI deficiency resulted in an alteration in fibrillar assembly in the pericellular region and the following fibrillar growth, leading to less tendon thickness in Col6a1−/− mice. This lower tendon thickness obviously affects the biomechanical properties of the tendon, resulting in lower tensile strength and less stiffness of the tendon in Col6a1−/− mice compared with wild-type [101].
The study of a colVI null mouse model demonstrated faulty regulation of tendon fibrillogenesis. There was a disruption in cell and extracellular micro-domain organization in the absence of colVI [101]. The breakdown of micro-domain structure seen in Col6a1−/− tendons was linked to abnormal fiber development within these micro-domains. Finally, in the absence of colVI, there was abnormal fibril assembly in the pericellular area as well as abnormally small diameter fibril assembly throughout the tendon [101]. The abnormal structure of the tendon fibrillar matrix was linked to altered function, lower tensile strength, and stiffness in the absence of colVI [101]. Lack of adequate muscle stimulation in patients with BM and UCMD could play a role in the abnormalities found in tendons, both structurally and functionally [101].
Of note, the role of colVI in tendons was also confirmed in the Col6a3hm/hm mouse model, designed by Pan and colleagues [102] in order to ablate specifically the production of the α3(VI) and resulting in the almost complete absence of colVI tetramers secretion in tissues and by fibroblasts in vitro [102]. Similar to what was observed in Col6a1−/− mice, colVI absence determined an alteration in the assembly of collagen I fibrils that appeared highly dishomogeneous in terms of cross-sectional diameters, with a prevalence of smaller fibers, compared with wild-type tendons. This was particularly evident in the pericellular region of tendon fibroblasts, supporting the idea of a role for colVI in orchestrating the assembly of collagen fibrils within fibroblasts’ pericellular micro-domains [102]. Whether these alterations are directly mediated by the absence of colVI in the extracellular space or by the modulation of other ECM proteins found to be altered in their expression in colVI null tenocytes [101,102] remains to be defined.

6.3. Clinical Studies

Patients with collagen VI-related myopathies have axial and proximal joint contractures, as well as distal joint hypermobility, indicating that tendon function is involved in addition to muscle weakness. Therefore, in addition to intensive physiotherapy aimed at stretching and recovery of joint ROM, orthopedic surgery plays a key role in recovering mobility and improving quality of life, with surgeries such as Achilles tendon lengthening. Available clinical studies are anecdotical but give a clear insight of the pathology of these diseases [10].
Antoniel et al. [98] examined biopsies of the pedidium tendon from a UCMD patient with a heterozygous COL6A1 mutation and of the piriformis tendon from a BM patient with heterozygous COL6A2 mutation who had a femur fracture surgeryand compared them with two biopsies of similar tendons from controls who had surgery for other reasons. In the control cultures, the colVI network appeared well arranged and attached along the cell surface, thus ensuring cell attachment to the substrate. The UCMD and BM cultures showed a disordered colVI network and limited interaction with cellular processes [98]. The controls exhibited an even distribution of colVI filaments along tendon fibroblast processes, whereas in the BM and UCMD cultures, colVI exhibited mostly intercellular distribution. In comparison to the filamentous arrangement of the controls in long-term patient cultures, colVI organization appeared spotty [98]. ColVI organization was altered in tendon cultures from both BM and UCMD patients using immunofluorescence. In fact, colVI formed evident clumps in the ECM of patients’ cells and did not appear to be connected with the cell surface. The immunofluorescence pattern was validated by rotary shadowing analysis, which revealed that colVI aggregates were made up of tangled microfilaments that were hardly linked to the cell processes [98].
According to previous studies on muscle and cutaneous cultures, changes in colVI expression differ between BM and UCMD. In BM, there are changes in protein organization, detected by rotary shading and electron microscopy, while in UCMD, there is an absence or reduced production of colVI. Consequently, the severity of colVI abnormalities observed in BM tendon fibroblast cultures could indicate that colVI mutations affect protein organization differently depending on the tissue [98].
In another study [103], peroneal tendon biopsies were taken from two healthy patients (17 and 21 year of age) during foot surgery, and from a previously genetically described UCMD patient during a talar-calcaneal extra-articular fusion (Grice procedure). Ultrastructurally, the tendons of the controls had tenocytes with long cellular processes and aligned and well-packed collagen fibers. The PCM of tenocytes contained collagen fibrils that were tightly connected to the cell surface. In the tendons of the patients, on the other hand, tenocytes showed reduced cellular processes and typical features of necrotic cells, with an abnormal accumulation of microfibrillar/reticular material in the PCM. Cross-sectioning of the collagen fibrils showed a smaller mean fibrillar diameter than in the controls. The effect of colVI deficiency on tendon matrix organization in the normal and UCMD patient tendon cultures was investigated using immunofluorescence microscopy. In the UCMD tenocyte culture, colVI was significantly reduced and weakly linked to the cell surface. In tendon sections and tenocyte culture from the UCMD patients, colVI was extremely low, in agreement with the low levels and interrupted network of colVI observed at the skeletal muscle level [103].
The BM/UCMD tenocyte cultures not only were important to describe in vitro alterations and ultrastructural differences in collagen deposition and ECM organization but also to unravel more mechanistic insight related to how colVI impact in this cell type is mediated intracellularly. Indeed, a study conducted in tenocyte cultures from healthy patients’ biopsies revealed that CSPG4/NG2, a major transmembrane proteoglycan, is the receptor able to mediate cell contact to the secreted colVI and to modulate cell migration [104]. The same colVI–NG2 axis appeared disrupted in tenocytes derived from the BM and UCMD patients bearing mutations in different colVI genes, showing impaired migration upon in vitro scratch wound assays, thereby highlighting a further relevance in injury response [98].
ColVI deficiency negatively affects tendon matrix organization both in vivo and in vitro, similar to the changes observed in Ehlers–Danlos syndrome, suggesting a similar pathophysiological mechanism [105,106].
Reduced load, disuse, and sarcopenia due to aging were believed to cause the fibrillar abnormalities. The presence of alterations in the ECM of patient cultures reduced the importance of muscle dysfunction as a determinant of the tendon phenotype [104,107,108].

7. Cartilage

7.1. General Aspects

Cartilage surrounds the articular surfaces and reduces friction, allowing for painless movement of synovial joints. It is made up of chondrocytes that are integrated in an ECM that is made up of a macromolecular framework and water. Its response to trauma is very limited due to its avascular, aneural, and hypocellular structure. Chondrocytes are primarily responsible for articular cartilage homeostasis. Collagen, thanks in part to its interaction with proteoglycans and interstitial fluid components, ensures the mechanical properties of cartilage [109,110].
ColVI is a prominent component of the PCM compartment in articular cartilage, which is important in the physiology of the synovial joint and has an indirect role in the mechanical environment of chondrocytes [89]. ColVI deficiency can lead to osteoarthritis progression due to either loss of PCM characteristics.
Actually, the amount of colVI in the articular cartilage is estimated to be only 1% of the total collagen species [111]. Here, it is mostly found in the PCM of adult cartilage, where it helps chondrocyte adhesion and integrity [3]. PCM completely surrounds articular cartilage cells and governs the biomechanical, biophysical, and biochemical signals sensed by the chondrocyte. PCM plays an important role in either protecting cells or acting as a filter or transducer of physical signals in the ECM, possibly through an interaction of colVI with integrins or other cell surface receptors. Many ECM components and the cell membrane have a high affinity for colVI, as it is essential in mediating cell–matrix and intermolecular interactions in a variety of tissues and cell cultures. In articular cartilage, colVI interacts with hyaluronan, decorin, and fibronectin to form a network that connects the chondrocyte to the PCM [89]. It is thought to play a role in cell anchoring and matrix cell signaling and is most likely a contact between the hard interterritorial cartilage matrix and the chondrocyte [112]. As a result, colVI serves a dual purpose in chondron integrity preservation: (I) it stabilizes collagens and proteoglycans of the pericellular microenvironment, by connecting to the radial collagen network; (II) it ensures that the PCM and cell nucleus are securely attached, thanks to specific cell surface receptors mediating its interaction with chondrocytes [111].
Interestingly, increased colVI was observed in osteoarthritic cartilage [113], where the PMC derangement is expected to have an impact on chondrocyte metabolic activity [114,115]. The type VI in the milieu could possibly play a role in the creation of chondrocyte clusters, as the PMC is thought to be a key factor in chondrocyte proliferation, which is one of the hallmarks of osteoarthritic cartilage [112].

7.2. Animal Models

Several studies found new evidence of severe articular cartilage alterations in Col6a1−/− mice [89,116].
A reduction in cartilage stiffness, despite a structurally intact PCM, was observed in the absence of colVI. Furthermore, colVI deficiency results in chondrocyte swelling and affects osmotic signal transmission, thus highlighting the role that collagen VI plays in the transmission of mechanical and osmotic stresses from the ECM to the PCM and then to the chondrocytes [116].
Alexopoulos et al. [89] used Col6a1−/− animals to see if a lack of colVI affects PCM formation and biomechanical function, as well as the mechanical environment of chondrocytes during hip joint loading. In this study [89], mice lacking colVI showed no signs of disease and had similar survival to wild-type mice. The articular cartilage of wild-types had pericellular staining for colVI, which was absent in knockout mice. In knockout mice, the semi-quantitative histologic study of cartilage deterioration indicated significant age-dependent osteoarthritic changes. Col6a1−/− mice exhibited cartilage with linearly elastic PCM and a significantly lower Young’s modulus of chondrons than wild-type [89]. As the Col6a1−/ mice aged, they developed osteoarthritis more quickly [89]. These findings imply that early changes in the mechanical properties of the PCM may be related to the progression of osteoarthritis [89].
Another study looked at colVI role in the morphology and physical characteristics of cartilage in the knee joint of Col6a1−/− mice. Col6a1−/− mice showed delayed or reduced cartilage deterioration with age compared with Col6a1+/+ mice, unlike the hip joint, but significantly greater and earlier osteophyte formation. Physical characteristics of articular cartilage (elastic modulus, coefficient of friction, roughness) did not differ consistently between genotypes. Thus, the lack of colVI on synovial joint health and function may depend on the specific site [78].
Not only limb-related articular compartments but also intervertebral disks (IVD) were the object of studies in ovine and murine samples [117]. IVDs are fibrocartilage cushions located between the respective superior and inferior facets of the vertebral articular processes as well as through the joints of the vertebral bodies. The inner nucleus pulposus, the outer annulus fibrosus, and the cartilaginous endplates that anchor the discs to adjacent vertebrae are the three major components of the IVD [118,119]. The first is a gel-like tissue in the center of the IVD, accounting for much of the spine’s strength and flexibility and composed mostly of collagen and proteoglycans. It is surrounded by the annulus fibrosus, containing an inner and an outer portion differing primarily in their collagen composition and morphology of ECM-secreting cells [120]. Anulus fibrosus is organized in 15 to 25 lamellae, interspersed with proteoglycans, glycoproteins, elastic fibers, and connective tissue cells. Lamellae are linked together by translamellar cross-bridges, and the number of these bridges per unit area strikes a compromise between strength and flexibility [121].
By immunofluorescence on intervertebral disc tissue sections, colVI was found in the PCM close to the disc cell borders in the outer and inner anulus fibrosus and nucleus pulposus. ColVI exhibited a diffuse distribution from the cell margin to the edge of the chondrons surrounding the disc. ColVI also joined strings of cells in the outer fibrous annulus situated along the primary axis of the type I collagen fiber bundles laid in the annular lamellae and was also an important component of translamellar cross-bridge networks in ovine and murine IVDs. Translamellar cross-bridge abundance in lumbar ovine IVDs also showed age- and spinal level-dependent trends. In addition, the relative size of the transverse bridges and their numbers decreased significantly with advancing age [117].
Of note, articular alterations were reported also in zebrafish in the absence of colVI [57]. In zebrafish, colVI was found to be highly expressed, starting from 48 h post-fertilization and still detectable at six days post-fertilization at the level of palatoquadrate and ceratohyals cartilages [122]. Of note, development of craniofacial cartilaginous compartments is particularly conserved from fish to humans [123] and in accordance to what was described in the mouse, colVI was found during cartilage morphogenesis to stain PMC of chondrocytes and perichondrium [122]. When colVI is lacking, the arrangement of ceratohyal angles appear to vary in an age-dependent manner, from narrower angles in the embryos to larger in the adult, in comparison to the wild-type condition. While the presence of an alteration is relevant for clinical considerations, such a contrasting phenotype dependent on the developmental stage is in line with the start of eating behavior that in zebrafish occurs approximately seven days post-fertilization, therefore potentially being an effect of the differential force and functioning exerted by muscles and tendons in the colVI knockout context [57].

7.3. Clinical Studies

Despite the reduced number of specific research studies on cartilage with a major relevance for BM and UCMD patients, the presence of hyperlaxity in distal joints was one of the first recognized diagnostic features for UCMD patients [68] and, although to a minor extent, present in some BM patients [124,125] as well. This became useful in a differential diagnosis with Emery–Dreifuss muscular dystrophy [126].
By the way several reports were focused on human studies aimed at understanding the role of colVI in osteoarthritis.
Pullig and colleagues [127] studied the expression of colVI in normal and osteoarthritis knee cartilage. They showed an increased expression of colVI in osteoarthritis with a different distribution of colVI among different cartilage zones (superficial, central, and deep) compared with normal cartilage, thus suggesting a potential role of colVI in the pathology development [127].
A similar result was found by Nugent et al. [128]; they showed a more widespread diffusion of colVI in osteoarthritis cartilage. Chondrocytes showed upregulation of colVI as a response to altered endoplasmic reticulum function present in the osteoarthritis [128].
Recent approaches for cartilage repair and regeneration have examined the abilities of soluble colVI to stimulate the proliferation of chondrocyte cultures [129]. This may open the way toward using soluble colVI as a useful biologic booster to the proliferation of scarce sources of chondrocytes and subsequent autologous implantation (ACI) [129].

8. Ligament

8.1. General Aspects

Ligaments are strong structures composed of dense fibrous connective tissue that connect two bones or two parts of the same bone together with a stabilizing function [1].
Ligaments share many properties with tendons, and little is known about the molecular differences between them. Recent studies have focused attention on the developmental and differentiation at the bases of ligaments and tendons [130].
As previously described in tendons, Sardone et al. showed that colVI is associated both in vivo and in vitro with the cell membrane of ligament fibroblasts, extending along with their processes and forming a fibrillar PCM [131].
With relevance for colVI in ligaments, the posterior longitudinal ligament and the ligamentum flavum were the specific object of some studies on their pathological deterioration involving ECM alteration [132,133]. The first one runs along the posterior aspect of the vertebral bodies inside the vertebral canal, from the body of the axis to the sacrum [134]. The ligament is composed of longitudinal fibers: the denser fibers are deeper and span one vertebra while the superficial fibers span three to four vertebrae [135]. Fibers are wider at the intervertebral spaces and are more adherent to the annulus fibrosus of the intervertebral discs than to the vertebral body. The ligamentum flavum is a two-layered structure that bridges the interlaminar gap between consecutive segments. It begins on the ventral side of the suprajacent lamina and inserts on the superior edge of the subjacent lamina. The ligamentum flavum owes its name to the particular yellowish appearance due to the high concentration of elastin and other collagen fibers, including colVI.
In contrast with the relatively rich literature regarding the composition and the biomechanical proprieties of the intervertebral disc, the posterior longitudinal ligament and the ligamentum flavum need further investigations to characterize the presence and the role of colVI.

8.2. Animal Models

There are few studies on this subject in the literature, indicating a topic that still has potential for new research. It may be thought that colVI plays a similar role in tendons and ligaments due to their similar composition [130].

8.3. Clinical Studies

Sardone et al. [131] studied the anterior cruciate ligament (ACL) and observed that the distribution of colVI at the cell surface depended on the expression of NG2 proteoglycan, a receptor of colVI. They also found how treatment of ACL fibroblast cultures with an anti-NG2 antibody altered the colVI distribution; thus, NG2 proteoglycan mediated the organization of colVI in the PMC. In vitro, mechanical stresses reduced NG2 proteoglycan levels, altering the binding between colVI and the cell surface and modified the cell cycle. They suggested that ligament injury altered the cell–ECM interaction, leading to poor fibroblast self-renewal capabilities and, thus, poor ligament regeneration [131].
An interesting study of Kawahara and colleagues focused on an increase in colVI in the pathological ligamentum flavum. They demonstrated by morphological and quantitative methods that colVI is a major component of cicatricial and hyalinization thickening of the ligamenta flava following rupture of the normal elastic fiber framework [132].
In controls, colVI was detected immunohistochemically in the collagen among elastic fibers and was weakly colored. Elastic fibers were swollen and degenerated near the hypertrophic edge and the collagenous matrix was expanded and hyalinized. ColVI was abundantly and densely marked in hyalinized tissue, mostly among loosely packed thin collagen fibers and around granular elastic fibrils.
Disruption of elastic fibers and hyalinization appears to be linked to colVI in thickened fibrous ligamenta flava [132].
Another interesting study was conducted to clarify the role of colVI in the ossification of the posterior longitudinal ligament of the spine (OPLL) and the ossification of the ligamentum flavum (OLF) [136]. A variety of possible risk factors have been demonstrated to be implicated in OPLL and OLF by epidemiologic studies, including sex, trauma, hormonal imbalance, dietary habits, and diabetes [133,137,138]; however, the genetic background is considered to be a predominant factor in the etiology of these diseases [139,140,141]. Tanaka et al. recently showed that the locus of susceptibility to OPLL was accurately directed to COL6A1 by a genome-wide linkage and by linkage disequilibrium studies, and seven single nucleotide polymorphisms (SNPs) of COL6A1 were associated with OPLL [142]. Among these seven SNPs, the intron32 [143] was not only the most significantly associated with OPLL but also associated with diffuse idiopathic skeletal hyperostosis in Japan [142,144].

9. Conclusions

The current review aimed at filling the knowledge gap between scientists and clinicians through a transdisciplinary approach to the role of colVI in the musculoskeletal system, giving insights to its tissue-specific functions; current evidence does not show clear guidelines on the management of patients affected by collagen VI-related myopathies.
However, a better understanding of the role of colVI and its interactions to cells in the musculoskeletal tissues could be the key to correct patient-centered management and to pose the basis to a new molecular approach to therapeutics. UCMD, BM, and MM represent a challenging condition for both the patient and the clinicians. Therefore, it is important to create a multidisciplinary group of professionals capable of focusing on the patient’s needs and, above all, be able to communicate in a more effective fashion.
More recent animal models, including zebrafish, could help researchers to improve the relatively poor literature on bone, ligament, and cartilage tissues available so far. Furthermore, in the near future, we trust that this model could be applied at a preclinical stage to discover new therapeutic targets among the downstream of the genetic defects, allowing tests for new drugs. Physiopathology shows that at an early stage, clinical management should focus on intensive physiotherapy aimed at relieving joint contractures through muscle trophism strengthening exercises and tendon stretching. Orthopedic surgeons may contribute by correcting the severe deformities of the spine and extremities, typical of these patients, and through the performance of less invasive approaches to promote a rapid recovery and more effective post-surgical care.
The present study summarized the updated research progress in the area and gave an overview to support further studies and identify potential novel therapeutic strategies and targets in the management of patients affected by collagen VI-related myopathies.

Author Contributions

Conceptualization, C.D., F.S., A.D.M. and L.M.; methodology, A.D.M. and C.F.; validation, L.M., M.C. and P.S.; investigation, C.D. and F.S.; writing—original draft preparation, F.S. and C.D.; writing—review and editing, C.D., A.D.M. and M.C.; supervision, A.D.M., L.M., P.S. and C.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kapandji, I.A. The physiology of the joints. Churchill Livingstone 1977, 2, 186–189. [Google Scholar]
  2. Hessle, H.; Engvall, E. Type VI collagen. Studies on its localization, structure, and biosynthetic form with monoclonal antibodies. J. Biol. Chem. 1984, 259, 3955–3961. [Google Scholar] [CrossRef]
  3. Cescon, M.; Gattazzo, F.; Chen, P.; Bonaldo, P. Collagen VI at a glance. J. Cell Sci. 2015, 128, 3525–3531. [Google Scholar] [CrossRef] [Green Version]
  4. Gara, S.K.; Grumati, P.; Urciuolo, A.; Bonaldo, P.; Kobbe, B.; Koch, M.; Paulsson, M.; Wagener, R. Three novel collagen VI chains with high homology to the α3 chain. J. Biol. Chem. 2008, 283, 10658–10670. [Google Scholar] [CrossRef] [Green Version]
  5. Fitzgerald, J.; Rich, C.; Zhou, F.H.; Hansen, U. Three novel collagen VI chains, alpha4(VI), alpha5(VI), and alpha6(VI). J. Biol. Chem. 2008, 283, 20170–20180. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Caria, F.; Cescon, M.; Gualandi, F.; Pichiecchio, A.; Rossi, R.; Rimessi, P.; Piccinelli, S.C.; Cassarino, S.G.; Gregorio, I.; Galvagni, A.; et al. Autosomal recessive Bethlem myopathy. Neurology 2009, 73, 1883–1891. [Google Scholar] [CrossRef]
  7. Briñas, L.; Richard, P.; Quijano-Roy, S.; Gartioux, C.; Ledeuil, C.; Lacène, E.; Makri, S.; Ferreiro, A.; Maugenre, S.; Topaloglu, H. Early onset collagen VI myopathies: Genetic and clinical correlations. Ann. Neurol. 2010, 68, 511–520. [Google Scholar] [CrossRef] [Green Version]
  8. Merlini, L.; Martoni, E.; Grumati, P.; Sabatelli, P.; Squarzoni, S.; Urciuolo, A.; Ferlini, A.; Gualandi, F.; Bonaldo, P. Autosomal recessive myosclerosis myopathy is a collagen VI disorder. Neurology 2008, 71, 1245–1253. [Google Scholar] [CrossRef]
  9. Lampe, A.K.; Bushby, K.M.D. Collagen VI related muscle disorders. J. Med. Genet. 2005, 42, 673–685. [Google Scholar] [CrossRef] [Green Version]
  10. Merlini, L.; Bernardi, P. Therapy of collagen VI-related myopathies (Bethlem and Ullrich). Neurother. J. Am. Soc. Exp. Neurother. 2008, 5, 613–618. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Bonaldo, P.; Russo, V.; Bucciotti, F.; Bressan, G.M.; Colombatti, A. α1 chain of chick type VI collagen: The complete cDNA sequence reveals a hybrid molecule made of one short collagen and three von Willebrand factor type A-like domains. J. Biol. Chem. 1989, 264, 5575–5580. [Google Scholar] [CrossRef] [PubMed]
  12. Bonaldo, P.; Russo, V.; Bucciotti, F.; Doliana, R.; Colombatti, A. Structural and functional features of the alpha 3 chain indicate a bridging role for chicken collagen VI in connective tissues. Biochemistry 1990, 29, 1245–1254. [Google Scholar] [CrossRef] [PubMed]
  13. Doliana, R.; Bonaldo, P.; Colombatti, A. «Multiple forms of chicken alpha 3(VI) collagen chain generated by alternative splicing in type A repeated domains. J. Cell Biol. 1990, 111, 2197–2205. [Google Scholar] [CrossRef] [PubMed]
  14. Dziadek, M.; Kazenwadel, J.S.; Hendrey, J.A.; Pan, T.-C.; Zhang, R.-Z.; Chu, M.-L. Alternative splicing of transcripts for the alpha3 chain of mouse collagen VI: Identification of an abundant isoform lacking domains N7–N10 in mouse and human. Matrix Biol. 2002, 21, 227–241. [Google Scholar] [CrossRef]
  15. Saitta, B.; Stokes, D.G.; Vissing, H.; Timpl, R.; Chu, M.L. Alternative splicing of the human alpha 2 (VI) collagen gene generates multiple mRNA transcripts which predict three protein variants with distinct carboxyl termini. J. Biol. Chem. 1990, 265, 6473–6480. [Google Scholar] [CrossRef] [PubMed]
  16. Engel, J.; Furthmayr, H.; Odermatt, E.; von der Mark, H.; Aumailley, M.; Fleischmajer, R.; Timpl, R. Structure and Macromolecular Organization of Type VI Collagena. Ann. N. Y. Acad. Sci. 1985, 460, 25–37. [Google Scholar] [CrossRef]
  17. Colombatti, A.; Mucignat, M.T.; Bonaldo, P. Secretion and Matrix Assembly of Recombinant Type VI Collagen*. J. Biol. Chem. 1995, 270, 13105–13111. [Google Scholar] [CrossRef] [Green Version]
  18. Colombatti, A.; Bonaldo, P.; Ainger, K.; Bressan, G.M.; Volpin, D. Biosynthesis of chick type VI collagen. I. Intracellular assembly and molecular structure. J. Biol. Chem. 1987, 262, 14454–14460. [Google Scholar] [CrossRef]
  19. Colombatti, A.; Bonaldo, P. Biosynthesis of chick type VI collagen. II. Processing and secretion in fibroblasts and smooth muscle cells. J. Biol. Chem. 1987, 262, 14461–14466. [Google Scholar] [CrossRef]
  20. Chu, M.L.; Pan, T.C.; Conway, D.; Saitta, B.; Stokes, D.; Kuo, H.J.; Glanville, R.W.; Timpl, R.; Mann, K.; Deutzmann, R. The Structure of Type VI Collagen a. Ann. N. Y. Acad. Sci. 1990, 580, 55–63. [Google Scholar] [CrossRef]
  21. Knupp, C.; Pinali, C.; Munro, P.M.; Gruber, H.E.; Sherratt, M.J.; Baldock, C.; Squire, J.M. Structural correlation between collagen VI microfibrils and collagen VI banded aggregates. J. Struct. Biol. 2006, 154, 312–326. [Google Scholar] [CrossRef] [PubMed]
  22. Lamandé, S.R.; Mörgelin, M.; Selan, C.; Jöbsis, G.J.; Baas, F.; Bateman, J.F. Kinked collagen VI tetramers and reduced microfibril formation as a result of Bethlem myopathy and introduced triple helical glycine mutations. J. Biol. Chem. 2002, 277, 1949–1956. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Persikov, A.V.; Ramshaw, J.A.; Kirkpatrick, A.; Brodsky, B. Amino acid propensities for the collagen triple-helix. Biochemistry 2000, 39, 14960–14967. [Google Scholar] [CrossRef] [PubMed]
  24. Lamandé, S.; Sigalas, E.; Pan, T.; Chu, M.; Dziadek, M.; Timpl, R.; Bateman, J. The Role of the α3(VI) Chain in Collagen VI Assembly: Expression of an α3(vi) chain lacking n-terminal modules n10–n7 restores collagen vi assembly, secretion, and matrix deposition in an α3(vi)-deficient cell line. J. Biol. Chem. 1998, 273, 7423–7430. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Furthmayr, H.; Wiedemann, H.; Timpl, R.; Odermatt, E.; Engel, J. Electron-microscopical approach to a structural model of intima collagen. Biochem. J. 1983, 211, 303–311. [Google Scholar] [CrossRef]
  26. Ball, S.; Bella, J.; Kielty, C.; Shuttleworth, A. Structural basis of type VI collagen dimer formation. J. Biol. Chem. 2003, 278, 15326–15332. [Google Scholar] [CrossRef] [Green Version]
  27. Bernardi, P.; Bonaldo, P. Dysfunction of Mitochondria and Sarcoplasmic Reticulum in the Pathogenesis of Collagen VI Muscular Dystrophies. Ann. N. Y. Acad. Sci. 2008, 1147, 303–311. [Google Scholar] [CrossRef]
  28. Fitzgerald, J.; Mörgelin, M.; Selan, C.; Wiberg, C.; Keene, D.; Lamandé, S.; Bateman, J. The N-terminal N5 Subdomain of the α3(VI) Chain Is Important for Collagen VI Microfibril Formation*. J. Biol. Chem. 2001, 276, 187–193. [Google Scholar] [CrossRef] [Green Version]
  29. Kuo, H.-J.; Maslen, C.L.; Keene, D.R.; Glanville, R.W. Type VI collagen anchors endothelial basement membranes by interacting with type IV collagen. J. Biol. Chem. 1997, 272, 26522–26529. [Google Scholar] [CrossRef] [Green Version]
  30. Sabatelli, P.; Bonaldo, P.; Lattanzi, G.; Braghetta, P.; Bergamin, N.; Capanni, C.; Mattioli, E.; Columbaro, M.; Ognibene, A.; Pepe, G.; et al. Collagen VI deficiency affects the organization of fibronectin in the extracellular matrix of cultured fibroblasts. Matrix Biol. 2001, 20, 475–486. [Google Scholar] [CrossRef] [PubMed]
  31. Bidanset, D.J.; Guidry, C.; Rosenberg, L.C.; Choi, H.U.; Timpl, R.; Hook, M. Binding of the proteoglycan decorin to collagen type VI. J. Biol. Chem. 1992, 267, 5250–5256. [Google Scholar] [CrossRef] [PubMed]
  32. Tillet, E.; Wiedemann, H.; Golbik, R.; Pan, T.C.; Zhang, R.Z.; Mann, K.; Chu, M.L.; Timpl, R. Recombinant expression and structural and binding properties of α1(VI) and α2(VI) chains of human collagen type VI. Eur. J. Biochem. 1994, 221, 177–187. [Google Scholar] [CrossRef]
  33. Hansen, U.; Allen, J.M.; White, R.; Moscibrocki, C.; Bruckner, P.; Bateman, J.F.; Fitzgerald, J. WARP Interacts with Collagen VI-Containing Microfibrils in the Pericellular Matrix of Human Chondrocytes. PLoS ONE 2012, 7, e52793. [Google Scholar] [CrossRef] [Green Version]
  34. Specks, U.; Mayer, U.; Nischt, R.; Spissinger, T.; Mann, K.; Timpl, R.; Engel, J.; Chu, M.L. Structure of recombinant N-terminal globule of type VI collagen alpha 3 chain and its binding to heparin and hyaluronan. EMBO J. 1992, 11, 4281–4290. [Google Scholar] [CrossRef] [PubMed]
  35. Fitch, J.M.; Birk, D.E.; Linsenmayer, C.; Linsenmayer, T.F. Stromal assemblies containing collagen types IV and VI and fibronectin in the developing embryonic avian cornea. Dev. Biol. 1991, 144, 379–391. [Google Scholar] [CrossRef] [PubMed]
  36. Wiberg, C.; Hedbom, E.; Khairullina, A.; Lamandé, S.R.; Oldberg, A.; Timpl, R.; Mörgelin, M.; Heinegård, D. Biglycan and decorin bind close to the n-terminal region of the collagen VI triple helix. J. Biol. Chem. 2001, 276, 18947–18952. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  37. Maraldi, N.M.; Sabatelli, P.; Columbaro, M.; Zamparelli, A.; Manzoli, F.A.; Bernardi, P.; Bonaldo, P.; Merlini, L. Collagen VI myopathies: From the animal model to the clinical trial. Adv. Enzyme Regul. 2009, 49, 197–211. [Google Scholar] [CrossRef]
  38. Bönnemann, C.G. The collagen VI-related myopathies: Muscle meets its matrix. Nat. Rev. Neurol. 2011, 7, 379–390. [Google Scholar] [CrossRef] [Green Version]
  39. Turrina, A.; Martínez-González, M.A.; Stecco, C. The muscular force transmission system: Role of the intramuscular connective tissue. J. Bodyw. Mov. Ther. 2013, 17, 95–102. [Google Scholar] [CrossRef]
  40. Csapo, R.; Gumpenberger, M.; Wessner, B. Skeletal Muscle Extracellular Matrix—What Do We Know About Its Composition, Regulation, and Physiological Roles? A Narrative Review. Front. Physiol. 2020, 11, 253. [Google Scholar] [CrossRef] [Green Version]
  41. Gara, S.K.; Grumati, P.; Squarzoni, S.; Sabatelli, P.; Urciuolo, A.; Bonaldo, P.; Paulsson, M.; Wagener, R. Differential and restricted expression of novel collagen VI chains in mouse. Matrix Biol. J. Int. Soc. Matrix Biol. 2011, 30, 248–257. [Google Scholar] [CrossRef]
  42. Sabatelli, P.; Gualandi, F.; Gara, S.K.; Grumati, P.; Zamparelli, A.; Martoni, E.; Pellegrini, C.; Merlini, L.; Ferlini, A.; Bonaldo, P.; et al. Expression of collagen VI α5 and α6 chains in human muscle and in Duchenne muscular dystrophy-related muscle fibrosis. Matrix Biol. J. Int. Soc. Matrix Biol. 2012, 31, 187–196. [Google Scholar] [CrossRef] [Green Version]
  43. Braghetta, P.; Ferrari, A.; Fabbro, C.; Bizzotto, D.; Volpin, D.; Bonaldo, P.; Bressan, G.M. An enhancer required for transcription of the Col6a1 gene in muscle connective tissue is induced by signals released from muscle cells. Exp. Cell Res. 2008, 314, 3508–3518. [Google Scholar] [CrossRef]
  44. Zou, Y.; Zhang, R.-Z.; Sabatelli, P.; Chu, M.-L.; Bönnemann, C.G. Muscle Interstitial Fibroblasts Are the Main Source of Collagen VI Synthesis in Skeletal Muscle: Implications for Congenital Muscular Dystrophy Types Ullrich and Bethlem. J. Neuropathol. Exp. Neurol. 2008, 67, 144–154. [Google Scholar] [CrossRef] [Green Version]
  45. Urciuolo, A.; Quarta, M.; Morbidoni, V.; Gattazzo, F.; Molon, S.; Grumati, P.; Montemurro, F.; Tedesco, F.S.; Blaauw, B.; Cossu, G.; et al. Collagen VI regulates satellite cell self-renewal and muscle regeneration. Nat. Commun. 2013, 4, 1964. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Cescon, M.; Gregorio, I.; Eiber, N.; Borgia, D.; Fusto, A.; Sabatelli, P.; Scorzeto, M.; Megighian, A.; Pegoraro, E.; Hashemolhosseini, S. Collagen VI is required for the structural and functional integrity of the neuromuscular junction. Acta Neuropathol. 2018, 136, 483–499. [Google Scholar] [CrossRef]
  47. Jöbsis, G.J.; Keizers, H.; Vreijling, J.P.; de Visser, M.; Speer, M.C.; Wolterman, R.A.; Baas, F.; Bolhuis, P.A. Type VI collagen mutations in Bethlem myopathy, an autosomal dominant myopathy with contractures. Nat. Genet. 1996, 14, 113–115. [Google Scholar] [CrossRef] [PubMed]
  48. Vanegas, O.C.; Bertini, E.; Zhang, R.Z.; Petrini, S.; Minosse, C.; Sabatelli, P.; Giusti, B.; Chu, M.L.; Pepe, G. Ullrich scleroatonic muscular dystrophy is caused by recessive mutations in collagen type VI. Proc. Natl. Acad. Sci. USA 2001, 98, 7516–7521. [Google Scholar] [CrossRef] [Green Version]
  49. Bonaldo, P.; Braghetta, P.; Zanetti, M.; Piccolo, S.; Volpin, D.; Bressan, G.M. Collagen VI deficiency induces early onset myopathy in the mouse: An animal model for Bethlem myopathy. Hum. Mol. Genet. 1998, 7, 2135–2140. [Google Scholar] [CrossRef] [PubMed]
  50. Irwin, W.A.; Bergamin, N.; Sabatelli, P.; Reggiani, C.; Megighian, A.; Merlini, L.; Braghetta, P.; Columbaro, M.; Volpin, D.; Bressan, G.M.; et al. Mitochondrial dysfunction and apoptosis in myopathic mice with collagen VI deficiency. Nat. Genet. 2003, 35, 367–371. [Google Scholar] [CrossRef]
  51. Angelin, A.; Tiepolo, T.; Sabatelli, P.; Grumati, P.; Bergamin, N.; Golfieri, C.; Mattioli, E.; Gualandi, F.; Ferlini, A.; Merlini, L.; et al. Mitochondrial dysfunction in the pathogenesis of Ullrich congenital muscular dystrophy and prospective therapy with cyclosporins. Proc. Natl. Acad. Sci. USA 2007, 104, 991–996. [Google Scholar] [CrossRef] [Green Version]
  52. Bernardi, P.; Bonaldo, P. Mitochondrial dysfunction and defective autophagy in the pathogenesis of collagen VI muscular dystrophies. Cold Spring Harb. Perspect. Biol. 2013, 5, a011387. [Google Scholar] [CrossRef] [Green Version]
  53. Menazza, S.; Blaauw, B.; Tiepolo, T.; Toniolo, L.; Braghetta, P.; Spolaore, B.; Reggiani, C.; Di Lisa, F.; Bonaldo, P.; Canton, M. Oxidative stress by monoamine oxidases is causally involved in myofiber damage in muscular dystrophy. Hum. Mol. Genet. 2010, 19, 4207–4215. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Grumati, P.; Coletto, L.; Sabatelli, P.; Cescon, M.; Angelin, A.; Bertaggia, E.; Blaauw, B.; Urciuolo, A.; Tiepolo, T.; Merlini, L.; et al. Autophagy is defective in collagen VI muscular dystrophies, and its reactivation rescues myofiber degeneration. Nat. Med. 2010, 16, 1313–1320. [Google Scholar] [CrossRef]
  55. Chrisam, M.; Pirozzi, M.; Castagnaro, S.; Blaauw, B.; Polishchuck, R.; Cecconi, F.; Grumati, P.; Bonaldo, P. Reactivation of autophagy by spermidine ameliorates the myopathic defects of collagen VI-null mice. Autophagy 2015, 11, 2142–2152. [Google Scholar] [CrossRef] [PubMed]
  56. Metti, S.; Gambarotto, L.; Chrisam, M.; La Spina, M.; Baraldo, M.; Braghetta, P.; Blaauw, B.; Bonaldo, P. Corrigendum: The Polyphenol Pterostilbene Ameliorates the Myopathic Phenotype of Collagen VI Deficient Mice via Autophagy Induction. Front. Cell Dev. Biol. 2021, 8, 580933. [Google Scholar] [CrossRef]
  57. Tonelotto, V.; Consorti, C.; Facchinello, N.; Trapani, V.; Sabatelli, P.; Giraudo, C.; Spizzotin, M.; Cescon, M.; Bertolucci, C.; Bonaldo, P. Collagen VI ablation in zebrafish causes neuromuscular defects during developmental and adult stages. Matrix Biol. J. Int. Soc. Matrix Biol. 2022, 112, 39–61. [Google Scholar] [CrossRef] [PubMed]
  58. Doane, K.J.; Yang, G.; Birk, D.E. Corneal cell-matrix interactions: Type VI Collagen promotes adhesion and spreading of corneal fibroblasts. Exp. Cell Res. 1992, 200, 490–499. [Google Scholar] [CrossRef]
  59. Pfaff, M.; Aumailley, M.; Specks, U.; Knolle, J.; Zerwes, H.G.; Timpl, R. Integrin and Arg-Gly-Asp Dependence of Cell Adhesion to the Native and Unfolded Triple Helix of Collagen Type VI. Exp. Cell Res. 1993, 206, 167–176. [Google Scholar] [CrossRef]
  60. Tulla, M.; Pentikäinen, O.T.; Viitasalo, T.; Käpylä, J.; Impola, U.; Nykvist, P.; Nissinen, L.; Johnson, M.S.; Heino, J. Selective binding of collagen subtypes by integrin α1I, α2I, and α10I domains. J. Biol. Chem. 2001, 276, 48206–48212. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Marcelino, J.; McDevitt, C.A. Attachment of articular cartilage chondrocytes to the tissue form of type VI collagen. Biochim. Biophys. Acta BBA-Protein Struct. Mol. Enzymol. 1995, 1249, 180–188. [Google Scholar] [CrossRef]
  62. Petrini, S.; Tessa, A.; Stallcup, W.B.; Sabatelli, P.; Pescatori, M.; Giusti, B.; Carrozzo, R.; Verardo, M.; Bergamin, N.; Columbaro, M.; et al. Altered expression of the MCSP/NG2 chondroitin sulfate proteoglycan in collagen VI deficiency. Mol. Cell. Neurosci. 2005, 30, 408–417. [Google Scholar] [CrossRef] [PubMed]
  63. Doane, K.J.; Howell, S.J.; Birk, D.E. Identification and functional characterization of two type VI collagen receptors, alpha 3 beta 1 integrin and NG2, during avian corneal stromal development. Invest. Ophthalmol. Vis. Sci. 1998, 39, 263–275. [Google Scholar] [PubMed]
  64. Bürgi, J.; Kunz, B.; Abrami, L.; Deuquet, J.; Piersigilli, A.; Scholl-Bürgi, S.; Lausch, E.; Unger, S.; Superti-Furga, A.; van der Goot, F.G. CMG2/ANTXR2 regulates extracellular collagen VI which accumulates in hyaline fibromatosis syndrome. Nat. Commun. 2017, 8, 15861. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Jöbsis, G.J.; Boers, J.M.; Barth, P.G.; de Visser, M. Bethlem myopathy: A slowly progressive congenital muscular dystrophy with contractures. Brain J. Neurol. 1999, 122, 649–655. [Google Scholar] [CrossRef] [Green Version]
  66. Merlini, L.; Morandi, L.; Granata, C.; Ballestrazzi, A. Bethlem myopathy: Early-onset benign autosomal dominant myopathy with contractures. Description of two new families. Neuromuscul. Disord. NMD 1994, 4, 503–511. [Google Scholar] [CrossRef]
  67. Ullrich, O. Kongenitale, atonisch-sklerotische Muskeldystrophie. Monatsschr Kinderheilkd 1930, 47, 502–510. [Google Scholar]
  68. Ullrich, O. Kongenitale, atonisch-sklerotische Muskeldystrophie, ein weiterer Typus der heredodegenerativen Erkrankungen des neuromuskulären Systems. Z. Für Gesamte Neurol. Psychiatr. 1930, 126, 171–201. [Google Scholar] [CrossRef]
  69. Nonaka, I.; Une, Y.; Ishihara, T.; Miyoshino, S.; Nakashima, T.; Sugita, H. A clinical and histological study of Ullrich’s disease (congenital atonic-sclerotic muscular dystrophy). Neuropediatrics 1981, 12, 197–208. [Google Scholar] [CrossRef] [PubMed]
  70. Voit, T. Congenital muscular dystrophies: 1997 update. Brain Dev. 1998, 20, 65–74. [Google Scholar] [CrossRef]
  71. Merlini, L.; Angelin, A.; Tiepolo, T.; Braghetta, P.; Sabatelli, P.; Zamparelli, A.; Ferlini, A.; Maraldi, N.M.; Bonaldo, P.; Bernardi, P. Cyclosporin A corrects mitochondrial dysfunction and muscle apoptosis in patients with collagen VI myopathies. Proc. Natl. Acad. Sci. USA 2008, 105, 5225–5229. [Google Scholar] [CrossRef] [Green Version]
  72. Merlini, L.; Sabatelli, P.; Armaroli, A.; Gnudi, S.; Angelin, A.; Grumati, P.; Michelini, M.E.; Franchella, A.; Gualandi, F.; Bertini, E.; et al. Cyclosporine A in Ullrich congenital muscular dystrophy: Long-term results. Oxid. Med. Cell. Longev. 2011, 2011, 139194. [Google Scholar] [CrossRef]
  73. Sorato, E.; Menazza, S.; Zulian, A.; Sabatelli, P.; Gualandi, F.; Merlini, L.; Bonaldo, P.; Canton, M.; Bernardi, P.; Di Lisa, F. Monoamine oxidase inhibition prevents mitochondrial dysfunction and apoptosis in myoblasts from patients with collagen VI myopathies. Free Radic. Biol. Med. 2014, 75, 40–47. [Google Scholar] [CrossRef] [PubMed]
  74. Castagnaro, S.; Pellegrini, C.; Pellegrini, M.; Chrisam, M.; Sabatelli, P.; Toni, S.; Grumati, P.; Ripamonti, C.; Pratelli, L.; Maraldi, N.M.; et al. Autophagy activation in COL6 myopathic patients by a low-protein-diet pilot trial. Autophagy 2016, 12, 2484–2495. [Google Scholar] [CrossRef] [Green Version]
  75. Sabatelli, P.; Merlini, L.; Di Martino, A.; Cenni, V.; Faldini, C. Early Morphological Changes of the Rectus Femoris Muscle and Deep Fascia in Ullrich Congenital Muscular Dystrophy. Int. J. Environ. Res. Public. Health 2022, 19, 1252. [Google Scholar] [CrossRef]
  76. Higuchi, I.; Horikiri, T.; Niiyama, T.; Suehara, M.; Shiraishi, T.; Hu, J.; Uchida, Y.; Saito, A.; Nakagawa, M.; Arimura, K.; et al. Pathological characteristics of skeletal muscle in Ullrich’s disease with collagen VI deficiency. Neuromuscul. Disord. 2003, 13, 310–316. [Google Scholar] [CrossRef] [PubMed]
  77. Izu, Y.; Ezura, Y.; Mizoguchi, F.; Kawamata, A.; Nakamoto, T.; Nakashima, K.; Hayata, T.; Hemmi, H.; Bonaldo, P.; Noda, M. Type VI collagen deficiency induces osteopenia with distortion of osteoblastic cell morphology. Tissue Cell 2012, 44, 1–6. [Google Scholar] [CrossRef]
  78. Christensen, S.E.; Coles, J.M.; Zelenski, N.A.; Furman, B.D.; Leddy, H.A.; Zauscher, S.; Bonaldo, P.; Guilak, F. Altered Trabecular Bone Structure and Delayed Cartilage Degeneration in the Knees of Collagen VI Null Mice. PLoS ONE 2012, 7, e33397. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Keene, D.R.; Sakai, L.Y.; Burgeson, R.E. Human bone contains type III collagen, type VI collagen, and fibrillin: Type III collagen is present on specific fibers that may mediate attachment of tendons, ligaments, and periosteum to calcified bone cortex. J. Histochem. Cytochem. Off. J. Histochem. Soc. 1991, 39, 59–69. [Google Scholar] [CrossRef]
  80. Harada, S.; Rodan, G.A. Control of osteoblast function and regulation of bone mass. Nature 2003, 423, 349–355. [Google Scholar] [CrossRef]
  81. Stark, Z.; Savarirayan, R. Osteopetrosis. Orphanet J. Rare Dis. 2009, 4, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Cummings, S.R.; Melton, L.J. Epidemiology and outcomes of osteoporotic fractures. Lancet Lond. Engl. 2002, 359, 1761–1767. [Google Scholar] [CrossRef] [PubMed]
  83. Pham, H.T.; Kram, V.; Dar, Q.-A.; Komori, T.; Ji, Y.; Mohassel, P.; Rooney, J.; Li, L.; Kilts, T.; Bonnemann, C.; et al. Collagen VIα2 chain deficiency causes trabecular bone loss by potentially promoting osteoclast differentiation through enhanced TNFα signaling. Sci. Rep. 2020, 10, 13749. [Google Scholar] [CrossRef] [PubMed]
  84. Shi, S.; Kirk, M.; Kahn, A.J. The role of type I collagen in the regulation of the osteoblast pheno type. J. Bone Miner. Res. 1996, 11, 1139–1145. [Google Scholar] [CrossRef]
  85. Harumiya, S.; Gibson, M.A.; Koshihara, Y. Antisense suppression of collagen VI synthesis results in reduced expression of collagen I in normal human osteoblast-like cells. Biosci. Biotechnol. Biochem. 2002, 66, 2743–2747. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Ueno, K.; Katayama, T.; Miyamoto, T.; Koshihara, Y. Interleukin-4 enhances in vitro mineralization in human osteoblast-like cells. Biochem. Biophys. Res. Commun. 1992, 189, 1521–1526. [Google Scholar] [CrossRef]
  87. Ishibashi, H.; Harumiya, S.; Koshihara, Y. Involvement of type VI collagen in interlekin-4-induced mineralization by human osteoblast-like cells in vitro. Biochem. Biophys. Acta 1999, 1472, 153–164. [Google Scholar] [CrossRef]
  88. Ishibashi, H.; Karube, S.; Yamakawa, A.; Koshihara, Y. Interleukin-4 stimulates pro-alpha 1(VI) collagen gene expression in cultured human osteoblast-like cells. Biochem. Biophys. Res. Commun. 1995, 211, 727–734. [Google Scholar] [CrossRef]
  89. Alexopoulos, L.G.; Youn, I.; Bonaldo, P.; Guilak, F. Developmental and osteoarthritic changes in Col6a1-knockout mice: Biomechanics of type VI collagen in the cartilage pericellular matrix. Arthritis Rheum. Off. J. Am. Coll. Rheumatol. 2009, 60, 771–779. [Google Scholar] [CrossRef] [Green Version]
  90. Giesen, E.B.W.; Ding, M.; Dalstra, M.; van Eijden, T.M.G.J. Changed Morphology and Mechanical Properties of Cancellous Bone in the Mandibular Condyles of Edentate People. J. Dent. Res. 2004, 83, 255–259. [Google Scholar] [CrossRef]
  91. Damrongrungruang, T.; Kuroda, S.; Kondo, H.; Aoki, K.; Ohya, K.; Kasugai, S. A Simple Murine Model for Immobilization Osteopenia. Clin. Orthop. Relat. Res. 2004, 425, 244–251. [Google Scholar] [CrossRef] [PubMed]
  92. Toni, S.; Morandi, R.; Busacchi, M.; Tardini, L.; Merlini, L.; Battistini, N.C.; Pellegrini, M. Nutritional status evaluation in patients affected by bethlem myopathy and ullrich congenital muscular dystrophy. Front. Aging Neurosci. 2014, 6, 315. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Bönnemann, C.G. The collagen VI-related myopathies Ullrich congenital muscular dystrophy and Bethlem myopathy. Handb. Clin. Neurol. 2011, 101, 81–96. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Philippe, V.; Pruna, L.; Fattah, M.A.; Pascal, V.; Kaminsky, P. Decreased bone mineral density in adult patients with muscular dystrophy. Jt. Bone Spine 2011, 78, 651–652. [Google Scholar] [CrossRef] [PubMed]
  95. Screen, H.R.; Berk, D.E.; Kadler, K.E.; Ramirez, F.; Young, M.F. Tendon functional extracellular matrix. J. Orthop. Res. 2015, 33, 793–799. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Kastelic, J.; Galeski, A.; Baer, E. The multicomposite structure of tendon. Connect. Tissue Res. 1978, 6, 11–23. [Google Scholar] [CrossRef] [PubMed]
  97. Zhang, G.; Young, B.B.; Ezura, Y.; Favata, M.; Soslowsky, L.J.; Chakravarti, S.; Birk, D.E. Development of tendon structure and function: Regulation of collagen fibrillogenesis. J. Musculoskelet. Neuronal Interact. 2005, 5, 5–21. [Google Scholar] [PubMed]
  98. Antoniel, M.; Traina, F.; Merlini, L.; Andrenacci, D.; Tigani, D.; Santi, S.; Cenni, V.; Sabatelli, P.; Faldini, C.; Squarzoni, S. Tendon Extracellular Matrix Remodeling and Defective Cell Polarization in the Presence of Collagen VI Mutations. Cells 2020, 9, 409. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  99. Kielty, C.M.; Whittaker, S.P.; Grant, M.E.; Shuttleworth, C.A. Type VI collagen microfibrils: Evidence for a structural association with hyaluronan. J. Cell Biol. 1992, 118, 979–990. [Google Scholar] [CrossRef] [Green Version]
  100. Leiphart, R.J.; Pham, H.; Harvey, T.; Komori, T.; Kilts, T.M.; Shetye, S.S.; Weiss, S.N.; Adams, S.M.; Birk, D.E.; Soslowsky, L.J.; et al. Coordinate roles for collagen VI and biglycan in regulating tendon collagen fibril structure and function. Matrix Biol. Plus 2022, 13, 100099. [Google Scholar] [CrossRef]
  101. Izu, Y.; Ansorge, H.L.; Zhang, G.; Soslowsky, L.; Bonaldo, P.; Chu, M.-L.; Birk, D. Dysfunctional tendon collagen fibrillogenesis in collagen VI null mice. Matrix Biol. J. Int. Soc. Matrix Biol. 2011, 30, 53–61. [Google Scholar] [CrossRef] [Green Version]
  102. Pan, T.C.; Zhang, R.Z.; Markova, D.; Arita, M.; Zhang, Y.; Bogdanovich, S.; Khurana, T.S.; Bönnemann, C.G.; Birk, D.E.; Chu, M.L. COL6A3 protein deficiency in mice leads to muscle and tendon defects similar to human collagen VI congenital muscular dystrophy. J. Biol. Chem. 2013, 288, 14320–14331. [Google Scholar] [CrossRef] [Green Version]
  103. Tagliavini, F.; Pellegrini, C.; Sardone, F.; Squarzoni, S.; Paulsson, M.; Wagener, R.; Gualandi, F.; Trabanelli, C.; Ferlini, A.; Merlini, L.; et al. Defective collagen VI α6 chain expression in the skeletal muscle of patients with collagen VI-related myopathies. Biochim. Biophys. Acta 2014, 1842, 1604–1612. [Google Scholar] [CrossRef] [Green Version]
  104. Sardone, F.; Traina, F.; Bondi, A.; Merlini, L.; Santi, S.; Maraldi, N.M.; Faldini, C.; Sabatelli, P. Tendon Extracellular Matrix Alterations in Ullrich Congenital Muscular Dystrophy. Front. Aging Neurosci. 2016, 8, 131. [Google Scholar] [CrossRef] [Green Version]
  105. Kobayasi, T. Abnormality of dermal collagen fibrils in Ehlers Danlos syndrome. Anticipation of the abnormality for the inherited hypermobile disorders. Eur. J. Dermatol. EJD 2004, 14, 221–229. [Google Scholar] [PubMed]
  106. Sun, M.; Connizzo, B.K.; Adams, S.M.; Freedman, B.R.; Wenstrup, R.J.; Soslowsky, L.J.; Birk, D.E. Targeted Deletion of Collagen V in Tendons and Ligaments Results in a Classic Ehlers-Danlos Syndrome Joint Phenotype. Am. J. Pathol. 2015, 185, 1436–1447. [Google Scholar] [CrossRef] [Green Version]
  107. Heinemeier, K.M.; Kjaer, M. In vivo investigation of tendon responses to mechanical loading. J. Musculoskelet. Neuronal Interact. 2011, 11, 115–123. [Google Scholar]
  108. Narici, M.V.; Maganaris, C.N. Plasticity of the muscle-tendon complex with disuse and aging. Exerc. Sport Sci. Rev. 2007, 35, 126–134. [Google Scholar] [CrossRef] [PubMed]
  109. Tatari, H. The structure, physiology, and biomechanics of articular cartilage: Injury and repair. Acta Orthop. Traumatol. Turc. 2007, 41, 1–5. [Google Scholar] [PubMed]
  110. Cohen, N.P.; Foster, R.J.; Mow, V.C. Composition and dynamics of articular cartilage: Structure, function, and maintaining healthy state. J. Orthop. Sports Phys. Ther. 1998, 28, 203–215. [Google Scholar] [CrossRef] [PubMed]
  111. Horikawa, O.; Nakajima, H.; Kikuchi, T.; Ichimura, S.; Yamada, H.; Fujikawa, K.; Toyama, Y. Distribution of type VI collagen in chondrocyte microenvironment: Study of chondrons isolated from human normal and degenerative articular cartilage and cultured chondrocytes. J. Orthop. Sci. Off. J. Jpn. Orthop. Assoc. 2004, 9, 29–36. [Google Scholar] [CrossRef]
  112. Söder, S.; Hambach, L.; Lissner, R.; Kirchner, T.; Aigner, T. Ultrastructural localization of type VI collagen in normal adult and osteoarthritic human articular cartilage. Osteoarthr. Cartil. 2002, 10, 464–470. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Ronzière, M.C.; Ricard-Blum, S.; Tiollier, J.; Hartmann, D.J.; Garrone, R.; Herbage, D. Comparative analysis of collagens solubilized from human foetal, and normal and osteoarthritic adult articular cartilage, with emphasis on type VI collagen. Biochim. Biophys. Acta 1990, 1038, 222–230. [Google Scholar] [CrossRef] [PubMed]
  114. Mankin, H.J.; Lippiello, L. Biochemical and metabolic abnormalities in articular cartilage from osteo-arthritic human hips. J. Bone Jt. Surg. Am. 1970, 52, 424–434. [Google Scholar] [CrossRef]
  115. Lee, D.A.; Bentley, G.; Archer, C.W. The control of cell division in articular chondrocytes. Osteoarthr. Cartil. 1993, 1, 137–146. [Google Scholar] [CrossRef]
  116. Zelenski, N.A.; Leddy, H.A.; Sanchez-Adams, J.; Zhang, J.; Bonaldo, P.; Liedtke, W.; Guilak, F. Type VI collagen regulates pericellular matrix properties, chondrocyte swelling, and mechanotransduction in mouse articular cartilage. Arthritis Rheumatol. 2015, 67, 1286–1294. [Google Scholar] [CrossRef] [Green Version]
  117. Hayes, A.J.; Shu, C.C.; Lord, M.S.; Little, C.B.; Whitelock, J.M.; Melrose, J. Pericellular colocalisation and interactive properties of type VI collagen and perlecan in the intervertebral disc. Eur. Cell. Mater. 2016, 32, 40–57. [Google Scholar] [CrossRef] [Green Version]
  118. Berg, E.J.; Ashurst, J.V. Anatomy, Back, Cauda Equina. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022; Available online: http://www.ncbi.nlm.nih.gov/books/NBK513251 (accessed on 18 August 2022).
  119. Huang, Y.-C.; Hu, Y.; Li, Z.; Luk, K.D.K. Biomaterials for intervertebral disc regeneration: Current status and looming challenges. J. Tissue Eng. Regen. Med. 2018, 12, 2188–2202. [Google Scholar] [CrossRef]
  120. van Uden, S.; Silva-Correia, J.; Oliveira, J.M.; Reis, R.L. Current strategies for treatment of intervertebral disc degeneration: Substitution and regeneration possibilities. Biomater. Res. 2017, 21, 22. [Google Scholar] [CrossRef] [Green Version]
  121. Waxenbaum, J.A.; Reddy, V.; Futterman, B. Anatomy, Back, Intervertebral Discs. In StatPearls; StatPearls Publishing: Treasure Island, FL, USA, 2022; Available online: http://www.ncbi.nlm.nih.gov/books/NBK470583 (accessed on 18 August 2022).
  122. Tonelotto, V.; Trapani, V.; Bretaud, S.; Heumüller, S.E.; Wagener, R.; Ruggiero, F.; Bonaldo, P. Spatio-temporal expression and distribution of collagen VI during zebrafish development. Sci. Rep. 2019, 9, 19851. [Google Scholar] [CrossRef] [Green Version]
  123. Mork, L.; Crump, G. Zebrafish Craniofacial Development: A Window into Early Patterning. Curr. Top. Dev. Biol. 2015, 115, 235–269. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Sasaki, T.; Hohenester, E.; Zhang, R.; Gotta, S.; Speer, M.; Tandan, R.; Timpl, R.; Chu, M. A Bethlem myopathy Gly to Glu mutation in the von Willebrand factor A domain N2 of the collagen alpha3(VI) chain interferes with protein folding. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 2000, 14, 761–768. [Google Scholar] [CrossRef] [Green Version]
  125. TPan, C.; Zhang, R.-Z.; Sudano, D.G.; Marie, S.K.; Bönnemann, C.G.; Chu, M.-L. New molecular mechanism for Ullrich congenital muscular dystrophy: A heterozygous in-frame deletion in the COL6A1 gene causes a severe phenotype. Am. J. Hum. Genet. 2003, 73, 355–369. [Google Scholar] [CrossRef] [Green Version]
  126. Demir, E.; Ferreiro, A.; Sabatelli, P.; Allamand, V.; Makri, S.; Echenne, B.; Maraldi, M.; Merlini, L.; Topaloglu, H.; Guicheney, P. Collagen VI status and clinical severity in Ullrich congenital muscular dystrophy: Phenotype analysis of 11 families linked to the COL6 loci. Neuropediatrics 2004, 35, 103–112. [Google Scholar] [CrossRef]
  127. Pullig, O.; Weseloh, G.; Swoboda, B. Expression of type VI collagen in normal and osteoarthritic human cartilage. Osteoarthr. Cartil. 1999, 7, 191–202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Nugent, A.E.; Speicher, D.M.; Gradisar, I.; McBurney, D.L.; Baraga, A.; Doane, K.J.; Horton, W.E., Jr. Advanced Osteoarthritis in Humans Is Associated With Altered Collagen VI Expression and Upregulation of ER-stress Markers Grp78 and Bag-1. J. Histochem. Cytochem. 2009, 57, 923–931. [Google Scholar] [CrossRef] [Green Version]
  129. Smeriglio, P.; Dhulipala, L.; Lai, J.H.; Goodman, S.B.; Dragoo, J.L.; Smith, R.L.; Maloney, W.J.; Yang, F.; Bhutani, N. Collagen VI Enhances Cartilage Tissue Generation by Stimulating Chondrocyte Proliferation. Tissue Eng. Part A 2015, 21, 840–849. [Google Scholar] [CrossRef]
  130. Bobzin, L.; Roberts, R.R.; Chen, H.-J.; Crump, J.G.; Merrill, A.E. Development and maintenance of tendons and ligaments. Dev. Camb. Engl. 2021, 148, dev186916. [Google Scholar] [CrossRef]
  131. Sardone, F.; Traina, F.; Tagliavini, F.; Pellegrini, C.; Merlini, L.; Squarzoni, S.; Santi, S.; Neri, S.; Faldini, C.; Maraldi, N.; et al. Effect of mechanical strain on the collagen VI pericellular matrix in anterior cruciate ligament fibroblasts. J. Cell. Physiol. 2014, 229, 878–886. [Google Scholar] [CrossRef]
  132. Kawahara, E.; Oda, Y.; Katsuda, S.; Nakanishi, I.; Aoyama, K.; Tomita, K. Microfilamentous type VI collagen in the hyalinized stroma of the hypertrophied ligamentum flavum. Virchows Arch. A Pathol. Anat. Histopathol. 1991, 419, 373–380. [Google Scholar] [CrossRef]
  133. Inamasu, J.; Guiot, B.H.; Sachs, D.C. Ossification of the posterior longitudinal ligament: An update on its biology, epidemiology, and natural history. Neurosurgery 2006, 58, 1027–1039. [Google Scholar] [CrossRef] [PubMed]
  134. Loughenbury, P.R.; Wadhwani, S.; Soames, R.W. The posterior longitudinal ligament and peridural (epidural) membrane. Clin. Anat. N. 2006, 19, 487–492. [Google Scholar] [CrossRef]
  135. Putz, R. Anatomic-functional viewpoints in treatment of injuries of the spine. Langenbecks Arch. Chir. Suppl. Kongressband Dtsch. Ges. Chir. Kongr. 1992, 1992, 256–262. [Google Scholar]
  136. Kong, Q.; Ma, X.; Li, F.; Guo, Z.; Qi, Q.; Li, W.; Yuan, H.-f.; Wang, Z.; Chen, Z. COL6A1 polymorphisms associated with ossification of the ligamentum flavum and ossification of the posterior longitudinal ligament. Spine 2007, 32, 2834–2838. [Google Scholar] [CrossRef]
  137. Okamoto, K.; Kobashi, G.; Washio, M.; Sasaki, S.; Yokoyama, T.; Miyake, Y.; Sakamoto, N.; Ohta, K.; Inaba, Y.; Tanaka, H. Dietary habits and risk of ossification of the posterior longitudinal ligaments of the spine (OPLL); findings from a case-control study in Japan. J. Bone Miner. Metab. 2004, 22, 612–617. [Google Scholar] [CrossRef] [PubMed]
  138. Kobashi, G.; Washio, M.; Okamoto, K.; Sasaki, S.; Yokoyama, T.; Miyake, Y.; Sakamoto, N.; Ohta, K.; Inaba, Y.; Tanaka, H. High body mass index after age 20 and diabetes mellitus are independent risk factors for ossification of the posterior longitudinal ligament of the spine in Japanese subjects: A case-control study in multiple hospitals. Spine 2004, 29, 1006–1010. [Google Scholar] [CrossRef]
  139. Matsunaga, S.; Yamaguchi, M.; Hayashi, K.; Sakou, T. Genetic analysis of ossification of the posterior longitudinal ligament. Spine 1999, 24, 937–938; discussion 939. [Google Scholar] [CrossRef]
  140. Taketomi, E.; Sakou, T.; Matsunaga, S.; Yamaguchi, M. Family study of a twin with ossification of the posterior longitudinal ligament in the cervical spine. Spine 1992, 17, S55–S56. [Google Scholar] [CrossRef]
  141. Sakou, T.; Taketomi, E.; Matsunaga, S.; Yamaguchi, M.; Sonoda, S.; Yashiki, S. Genetic study of ossification of the posterior longitudinal ligament in the cervical spine with human leukocyte antigen haplotype. Spine 1991, 16, 1249–1252. [Google Scholar] [CrossRef]
  142. Tanaka, T.; Ikari, K.; Furushima, K.; Okada, A.; Tanaka, H.; Furukawa, K.-I.; Yoshida, K.; Ikeda, T.; Ikegawa, S.; Hunt, S.C.; et al. Genomewide linkage and linkage disequilibrium analyses identify COL6A1, on chromosome 21, as the locus for ossification of the posterior longitudinal ligament of the spine. Am. J. Hum. Genet. 2003, 73, 812–822. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Epstein, N.E. Ossification of the yellow ligament and spondylosis and/or ossification of the posterior longitudinal ligament of the thoracic and lumbar spine. J. Spinal Disord. 1999, 12, 250–256. [Google Scholar] [CrossRef] [PubMed]
  144. Tsukahara, S.; Miyazawa, N.; Akagawa, H.; Forejtova, S.; Pavelka, K.; Tanaka, T.; Toh, S.; Tajima, A.; Akiyama, I.; Inoue, I. COL6A1, the candidate gene for ossification of the posterior longitudinal ligament, is associated with diffuse idiopathic skeletal hyperostosis in Japanese. Spine 2005, 30, 2321–2324. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The role of collagen VI in different tissues: Skeletal Muscle, Bone, Tendon, Cartilage and Neuromuscular Junction (NMJ).
Figure 1. The role of collagen VI in different tissues: Skeletal Muscle, Bone, Tendon, Cartilage and Neuromuscular Junction (NMJ).
Ijms 24 05095 g001
Figure 2. The role of collagen VI in different tissues: Intervertebral disk and Ligament.
Figure 2. The role of collagen VI in different tissues: Intervertebral disk and Ligament.
Ijms 24 05095 g002
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Di Martino, A.; Cescon, M.; D’Agostino, C.; Schilardi, F.; Sabatelli, P.; Merlini, L.; Faldini, C. Collagen VI in the Musculoskeletal System. Int. J. Mol. Sci. 2023, 24, 5095. https://doi.org/10.3390/ijms24065095

AMA Style

Di Martino A, Cescon M, D’Agostino C, Schilardi F, Sabatelli P, Merlini L, Faldini C. Collagen VI in the Musculoskeletal System. International Journal of Molecular Sciences. 2023; 24(6):5095. https://doi.org/10.3390/ijms24065095

Chicago/Turabian Style

Di Martino, Alberto, Matilde Cescon, Claudio D’Agostino, Francesco Schilardi, Patrizia Sabatelli, Luciano Merlini, and Cesare Faldini. 2023. "Collagen VI in the Musculoskeletal System" International Journal of Molecular Sciences 24, no. 6: 5095. https://doi.org/10.3390/ijms24065095

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