Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 87
1.
Acta Biomater ; 180: 279-294, 2024 May.
Article En | MEDLINE | ID: mdl-38604466

The myotendinous junction (MTJ) is a vulnerable region at the interface of skeletal muscle and tendon that forms an integrated mechanical unit. This study presents a technique for the spatially restrictive co-culture of human embryonic stem cell (hESC)-derived skeletal myocytes and primary tenocytes for two-dimensional modeling of the MTJ. Micropatterned lanes of extracellular matrix and a 2-well culture chamber define the initial regions of occupation. On day 1, both lines occupy less than 20 % of the initially vacant interstitial zone, referred to henceforth as the junction. Myocyte-tenocyte interdigitations are observed by day 7. Immunocytochemistry reveals enhanced organization and alignment of patterned myocyte and tenocyte features, as well as differential expression of multiple MTJ markers. On day 24, electrically stimulated junction myocytes demonstrate negative contractile strains, while positive tensile strains are exhibited by mechanically passive tenocytes at the junction. Unpatterned tenocytes distal to the junction experience significantly decreased strains in comparison to cells at the interface. Unpatterned myocytes have impaired organization and uncoordinated contractile behavior. These findings suggest that this platform is capable of inducing myocyte-tenocyte junction formation and mechanical coupling similar to the native MTJ, showing transduction of force across the cell-cell interface. STATEMENT OF SIGNIFICANCE: The myotendinous junction (MTJ) is an integrated structure that transduces force across the muscle-tendon boundary, making the region vulnerable to strain injury. Despite the clinical relevance, previous in vitro models of the MTJ lack the structure and mechanical accuracy of the native tissue and have difficulty transmitting force across the cell-cell interface. This study demonstrates an in vitro model of the MTJ, using spatially restrictive cues to inform human myocyte-tenocyte interactions and architecture. The model expressed MTJ markers and developed anisotropic myocyte-tenocyte integrations that resemble the native tissue and allow for force transduction from contracting myocytes to passive tenocyte regions. As such, this study presents a system capable of investigating development, injury, and pathology in the human MTJ.


Tendons , Tenocytes , Tissue Engineering , Humans , Tendons/cytology , Tendons/physiology , Tissue Engineering/methods , Tenocytes/cytology , Tenocytes/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Muscle Fibers, Skeletal/physiology , Models, Biological , Coculture Techniques , Muscle, Skeletal/cytology , Muscle, Skeletal/physiology , Myotendinous Junction
2.
Sci Rep ; 14(1): 9495, 2024 04 25.
Article En | MEDLINE | ID: mdl-38664570

The biological mechanisms regulating tenocyte differentiation and morphological maturation have not been well-established, partly due to the lack of reliable in vitro systems that produce highly aligned collagenous tissues. In this study, we developed a scaffold-free, three-dimensional (3D) tendon culture system using mouse tendon cells in a differentially adherent growth channel. Transforming Growth Factor-ß (TGFß) signaling is involved in various biological processes in the tendon, regulating tendon cell fate, recruitment and maintenance of tenocytes, and matrix organization. This known function of TGFß signaling in tendon prompted us to utilize TGFß1 to induce tendon-like structures in 3D tendon constructs. TGFß1 treatment promoted a tendon-like structure in the peripheral layer of the constructs characterized by increased thickness with a gradual decrease in cell density and highly aligned collagen matrix. TGFß1 also enhanced cell proliferation, matrix production, and morphological maturation of cells in the peripheral layer compared to vehicle treatment. TGFß1 treatment also induced early tenogenic differentiation and resulted in sufficient mechanical integrity, allowing biomechanical testing. The current study suggests that this scaffold-free 3D tendon cell culture system could be an in vitro platform to investigate underlying biological mechanisms that regulate tenogenic cell differentiation and matrix organization.


Cell Differentiation , Cell Proliferation , Tendons , Tenocytes , Transforming Growth Factor beta1 , Animals , Transforming Growth Factor beta1/pharmacology , Transforming Growth Factor beta1/metabolism , Tendons/cytology , Tendons/metabolism , Mice , Cell Differentiation/drug effects , Tenocytes/metabolism , Tenocytes/cytology , Cell Proliferation/drug effects , Cell Culture Techniques, Three Dimensional/methods , Cells, Cultured , Cell Culture Techniques/methods , Extracellular Matrix/metabolism , Collagen/metabolism , Tissue Engineering/methods
3.
J Biomech Eng ; 146(9)2024 Sep 01.
Article En | MEDLINE | ID: mdl-38529730

Tendinopathy is a leading cause of mobility issues. Currently, the cell-matrix interactions involved in the development of tendinopathy are not fully understood. In vitro tendon models provide a unique tool for addressing this knowledge gap as they permit fine control over biochemical, micromechanical, and structural aspects of the local environment to explore cell-matrix interactions. In this study, direct-write, near-field electrospinning of gelatin solution was implemented to fabricate micron-scale fibrous scaffolds that mimic native collagen fiber size and orientation. The stiffness of these fibrous scaffolds was found to be controllable between 1 MPa and 8 MPa using different crosslinking methods (EDC, DHT, DHT+EDC) or through altering the duration of crosslinking with EDC (1 h to 24 h). EDC crosslinking provided the greatest fiber stability, surviving up to 3 weeks in vitro. Differences in stiffness resulted in phenotypic changes for equine tenocytes with low stiffness fibers (∼1 MPa) promoting an elongated nuclear aspect ratio while those on high stiffness fibers (∼8 MPa) were rounded. High stiffness fibers resulted in the upregulation of matrix metalloproteinase (MMPs) and proteoglycans (possible indicators for tendinopathy) relative to low stiffness fibers. These results demonstrate the feasibility of direct-written gelatin scaffolds as tendon in vitro models and provide evidence that matrix mechanical properties may be crucial factors in cell-matrix interactions during tendinopathy formation.


Gelatin , Tenocytes , Tissue Scaffolds , Gelatin/chemistry , Animals , Horses , Tenocytes/cytology , Tenocytes/metabolism , Tissue Scaffolds/chemistry , Mechanical Phenomena , Gene Expression Regulation , Cell Shape , Biomechanical Phenomena
4.
Stem Cell Rev Rep ; 20(4): 1040-1059, 2024 May.
Article En | MEDLINE | ID: mdl-38396222

Tissue fibrosis following tendon injury is a major clinical problem due to the increased risk of re-injury and limited treatment options; however, its mechanism remains unclear. Evidence suggests that insufficient resolution of inflammation contributes to fibrotic healing by disrupting tenocyte activity, with the NF-κB pathway being identified as a potential mediator. Equine embryonic stem cell (ESC) derived tenocytes may offer a potential cell-based therapy to improve tendon regeneration, but how they respond to an inflammatory environment is largely unknown. Our findings reveal for the first time that, unlike adult tenocytes, ESC-tenocytes are unaffected by IFN-γ, TNFα, and IL-1ß stimulation; producing minimal changes to tendon-associated gene expression and generating 3-D collagen gel constructs indistinguishable from unstimulated controls. Inflammatory pathway analysis found these inflammatory cytokines failed to activate NF-κB in the ESC-tenocytes. However, NF-κB could be activated to induce changes in gene expression following stimulation with NF-κB pharmaceutical activators. Transcriptomic analysis revealed differences between cytokine and NF-κB signalling components between adult and ESC-tenocytes, which may contribute to the mechanism by which ESC-tenocytes escape inflammatory stimuli. Further investigation of these molecular mechanisms will help guide novel therapies to reduce fibrosis and encourage superior tendon healing.


Cytokines , Embryonic Stem Cells , NF-kappa B , Tenocytes , Animals , Horses , Tenocytes/cytology , Tenocytes/metabolism , Tenocytes/drug effects , Cytokines/metabolism , NF-kappa B/metabolism , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/drug effects , Signal Transduction/drug effects , Inflammation/pathology , Inflammation/metabolism , Cells, Cultured , Tendons/cytology
5.
Int J Mol Sci ; 22(22)2021 Nov 16.
Article En | MEDLINE | ID: mdl-34830254

Healing of ruptured tendons remains a clinical challenge because of its slow progress and relatively weak mechanical force at an early stage. Extracellular vesicles (EVs) derived from mesenchymal stem cells (MSCs) have therapeutic potential for tissue regeneration. In this study, we isolated EVs from adipose-derived stem cells (ADSCs) and evaluated their ability to promote tendon regeneration. Our results indicated that ADSC-EVs significantly enhanced the proliferation and migration of tenocytes in vitro. To further study the roles of ADSC-EVs in tendon regeneration, ADSC-EVs were used in Achilles tendon repair in rabbits. The mechanical strength, histology, and protein expression in the injured tendon tissues significantly improved 4 weeks after ADSC-EV treatment. Decorin and biglycan were significantly upregulated in comparison to the untreated controls. In summary, ADSC-EVs stimulated the proliferation and migration of tenocytes and improved the mechanical strength of repaired tendons, suggesting that ADSC-EV treatment is a potential highly potent therapeutic strategy for tendon injuries.


Achilles Tendon/injuries , Adipocytes/metabolism , Extracellular Vesicles/transplantation , Stem Cells/chemistry , Tendon Injuries/therapy , Wound Healing/physiology , Adipocytes/cytology , Adipose Tissue/cytology , Adipose Tissue/metabolism , Animals , Biglycan/genetics , Biglycan/metabolism , Biomarkers/metabolism , Cell Differentiation , Cell Movement , Cell Proliferation , Decorin/genetics , Decorin/metabolism , Extracellular Vesicles/chemistry , Female , Gene Expression , Rabbits , Stem Cells/cytology , Stem Cells/metabolism , Tendon Injuries/metabolism , Tendon Injuries/pathology , Tenocytes/cytology , Tenocytes/metabolism , Treatment Outcome
6.
Int J Mol Sci ; 22(20)2021 Oct 18.
Article En | MEDLINE | ID: mdl-34681872

To recreate the in vivo niche for tendon tissue engineering in vitro, the characteristics of tendon tissue underlines the use of biochemical and biophysical cues during tenocyte culture. Herein, we prepare core-sheath nanofibers with polycaprolactone (PCL) sheath for mechanical support and hyaluronic acid (HA)/platelet-rich plasma (PRP) core for growth factor delivery. Three types of core-sheath nanofiber membrane scaffolds (CSNMS), consisting of random HA-PCL nanofibers (Random), random HA/PRP-PCL nanofibers (Random+) or aligned HA/PRP-PCL (Align+) nanofibers, were used to study response of rabbit tenocytes to biochemical (PRP) and biophysical (fiber alignment) stimulation. The core-sheath structures as well as other pertinent properties of CSNMS have been characterized, with Align+ showing the best mechanical properties. The unidirectional growth of tenocytes, as induced by aligned fiber topography, was confirmed from cell morphology and cytoskeleton expression. The combined effects of PRP and fiber alignment in Align+ CSNMS lead to enhanced cell proliferation rates, as well as upregulated gene expression and marker protein synthesis. Another biophysical cue on tenocytes was introduced by dynamic culture of tenocyte-seeded Align+ in a bioreactor with cyclic tension stimulation. Augmented by this biophysical beacon from mechanical loading, dynamic cell culture could shorten the time for tendon maturation in vitro, with improved cell proliferation rates and tenogenic phenotype maintenance, compared to static culture. Therefore, we successfully demonstrate how combined use of biochemical/topographical cues as well as mechanical stimulation could ameliorate cellular response of tenocytes in CSNMS, which can provide a functional in vitro environmental niche for tendon tissue engineering.


Nanofibers/chemistry , Platelet-Rich Plasma/chemistry , Tendons , Tenocytes , Tissue Scaffolds/chemistry , Animals , Cell Culture Techniques/instrumentation , Cell Culture Techniques/methods , Cell Proliferation , Collagen/genetics , Collagen/metabolism , Elastic Modulus , Hyaluronic Acid/chemistry , Materials Testing , Microscopy, Electron, Scanning , Polyesters/chemistry , Rabbits , Tenocytes/cytology , Tenocytes/physiology , Thermogravimetry , Tissue Engineering
7.
Nat Commun ; 12(1): 5012, 2021 08 18.
Article En | MEDLINE | ID: mdl-34408142

Tendon self-renewal is a rare occurrence because of the poor vascularization of this tissue; therefore, reconstructive surgery using autologous tendon is often performed in severe injury cases. However, the post-surgery re-injury rate is relatively high, and the collection of autologous tendons leads to muscle weakness, resulting in prolonged rehabilitation. Here, we introduce an induced pluripotent stem cell (iPSC)-based technology to develop a therapeutic option for tendon injury. First, we derived tenocytes from human iPSCs by recapitulating the normal progression of step-wise narrowing fate decisions in vertebrate embryos. We used single-cell RNA sequencing to analyze the developmental trajectory of iPSC-derived tenocytes. We demonstrated that iPSC-tenocyte grafting contributed to motor function recovery after Achilles tendon injury in rats via engraftment and paracrine effects. The biomechanical strength of regenerated tendons was comparable to that of healthy tendons. We suggest that iPSC-tenocytes will provide a therapeutic option for tendon injury.


Achilles Tendon/injuries , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/transplantation , Tendon Injuries/therapy , Tenocytes/cytology , Tenocytes/transplantation , Achilles Tendon/cytology , Achilles Tendon/physiopathology , Animals , Cell Self Renewal , Cell- and Tissue-Based Therapy , Cells, Cultured , Humans , Male , Rats , Rats, Inbred F344 , Recovery of Function , Tendon Injuries/physiopathology
8.
Differentiation ; 120: 1-9, 2021.
Article En | MEDLINE | ID: mdl-34062407

Tendon injuries are common and account for up to 50% of musculoskeletal injuries in the United States. The poor healing nature of the tendon is attributed to poor vascularization and cellular composition. In the absence of FDA-approved growth factors for tendon repair, engineering strategies using bioactive factors, donor cells, and delivery matrices to promote tendon repair and regeneration are being explored. Growth factor alternatives in the form of small molecules, donor cells, and progenitors offer several advantages and enhance the tendon healing response. Small drug molecules and peptides offer stability over growth factors that are known to suffer from relatively short biological half-lives. The primary focus of this study was to assess the ability of the exendin-4 (Ex-4) peptide, a glucagon-like peptide 1 (GLP-1) receptor agonist, to induce tenocyte differentiation in bone marrow-derived human mesenchymal stem cells (hMSCs). We treated hMSCs with varied doses of Ex-4 in culture media to evaluate proliferation and tendonogenic differentiation. A 20 nM Ex-4 concentration was optimal for promoting cell proliferation and tendonogenic differentiation. Tendonogenic differentiation of hMSCs was evaluated via gene expression profile, immunofluorescence, and biochemical analyses. Collectively, the levels of tendon-related transcription factors (Mkx and Scx) and extracellular matrix (Col-I, Dcn, Bgn, and Tnc) genes and proteins were elevated compared to media without Ex-4 and other controls including insulin and IGF-1 treatments. The tendonogenic factor Ex-4 in conjunction with hMSCs appear to enhance tendon regeneration.


Cell Differentiation , Exenatide/pharmacology , Incretins/pharmacology , Mesenchymal Stem Cells/drug effects , Tenocytes/metabolism , Biglycan/metabolism , Cell Proliferation , Cells, Cultured , Collagen Type I/metabolism , Decorin/metabolism , Humans , Insulin/pharmacology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Tenascin/metabolism , Tenocytes/cytology
9.
Int J Mol Sci ; 22(9)2021 May 06.
Article En | MEDLINE | ID: mdl-34066472

The mechanosensitive gene tenomodulin (Tnmd) is implicated in tendon maturation and repair. However, the mechanism by which mechanical loading regulates Tnmd's expression and its role in tenocyte migration is yet to be defined. Here, we show that Tnmd and migration were upregulated in uniaxial cyclic stress-stimulated tenocytes. The knockdown of Tnmd reduced cell migration in the presence and absence of mechanical loading, suggesting that Tnmd is involved in tenocyte migration. Moreover, the treatment of stress-stimulated tenocytes with the actin inhibitor latrunculin (Lat A), histone acetyltransferase inhibitor anacardic acid (ANA), or histone demethylases inhibitor GSK-J4 suppressed Tnmd expression and tenocyte migration. These results show that actin stress fiber formation and chromatin decondensation regulates Tnmd expression, which might then regulate tenocyte migration. Thus, this study proposes the involvement of the actin and chromatin mechanotransduction pathway in the regulation of Tnmd and reveals a novel role of Tnmd in tenocyte migration. The identification of Tnmd function in tenocyte migration provides insight into the molecular mechanisms involved in Tnmd-mediated tendon repair.


Actins/metabolism , Cell Movement , Chromatin Assembly and Disassembly , Membrane Proteins/metabolism , Stress, Mechanical , Tenocytes/cytology , Tenocytes/metabolism , Animals , Cells, Cultured , Chromatin/metabolism , Membrane Proteins/genetics , Rats, Sprague-Dawley , Stress Fibers/metabolism , Up-Regulation/genetics
10.
J Biol Chem ; 297(1): 100819, 2021 07.
Article En | MEDLINE | ID: mdl-34029590

Collagen-derived hydroxyproline (Hyp)-containing peptides have a variety of biological effects on cells. These bioactive collagen peptides are locally generated by the degradation of endogenous collagen in response to injury. However, no comprehensive study has yet explored the functional links between Hyp-containing peptides and cellular behavior. Here, we show that the dipeptide prolyl-4-hydroxyproline (Pro-Hyp) exhibits pronounced effects on mouse tendon cells. Pro-Hyp promotes differentiation/maturation of tendon cells with modulation of lineage-specific factors and induces significant chemotactic activity in vitro. In addition, Pro-Hyp has profound effects on cell proliferation, with significantly upregulated extracellular signal-regulated kinase phosphorylation and extracellular matrix production and increased type I collagen network organization. Using proteomics, we have predicted molecular transport, cellular assembly and organization, and cellular movement as potential linked-network pathways that could be altered in response to Pro-Hyp. Mechanistically, cells treated with Pro-Hyp demonstrate increased directional persistence and significantly increased directed motility and migration velocity. They are accompanied by elongated lamellipodial protrusions with increased levels of active ß1-integrin-containing focal contacts, as well as reorganization of thicker peripheral F-actin fibrils. Pro-Hyp-mediated chemotactic activity is significantly reduced (p < 0.001) in cells treated with the mitogen-activated protein kinase kinase 1/2 inhibitor PD98059 or the α5ß1-integrin antagonist ATN-161. Furthermore, ATN-161 significantly inhibits uptake of Pro-Hyp into adult tenocytes. Thus, our findings document the molecular basis of the functional benefits of the Pro-Hyp dipeptide in cellular behavior. These dynamic properties of collagen-derived Pro-Hyp dipeptide could lead the way to its application in translational medicine.


Cell Movement/drug effects , Dipeptides/pharmacology , Homeostasis/drug effects , Integrin beta1/metabolism , Pseudopodia/metabolism , Tendons/cytology , Aging , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Collagen Type I/genetics , Collagen Type I/metabolism , Extracellular Matrix/drug effects , Extracellular Matrix/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Mice , Pseudopodia/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Stem Cells/cytology , Stem Cells/drug effects , Stem Cells/metabolism , Tenocytes/cytology , Tenocytes/drug effects , Up-Regulation/drug effects
11.
FASEB J ; 35(6): e21618, 2021 06.
Article En | MEDLINE | ID: mdl-33982337

Tendons are dense connective tissues that transmit muscle forces to the skeleton. After adult injury, healing potential is generally poor and dominated by scar formation. Although the immune response is a key feature of healing, the specific immune cells and signals that drive tendon healing have not been fully defined. In particular, the immune regulators underlying tendon regeneration are almost completely unknown due to a paucity of tendon regeneration models. Using a mouse model of neonatal tendon regeneration, we screened for immune-related markers and identified upregulation of several genes associated with inflammation, macrophage chemotaxis, and TGFß signaling after injury. Depletion of macrophages using AP20187 treatment of MaFIA mice resulted in impaired functional healing, reduced cell proliferation, reduced ScxGFP+ neo-tendon formation, and altered tendon gene expression. Collectively, these results show that inflammation is a key component of neonatal tendon regeneration and demonstrate a requirement for macrophages in effective functional healing.


Cell Proliferation , Inflammation/therapy , Macrophages/immunology , Regeneration , Tendon Injuries/therapy , Tenocytes/cytology , Wound Healing , Animals , Animals, Newborn , Disease Models, Animal , Female , Inflammation/immunology , Inflammation/pathology , Male , Mice , Tendon Injuries/immunology , Tendon Injuries/pathology , Tenocytes/physiology
12.
Dev Growth Differ ; 63(1): 38-46, 2021 Jan.
Article En | MEDLINE | ID: mdl-33270251

Human pluripotent stem cells (PSCs) are used as a platform for therapeutic purposes such as cell transplantation therapy and drug discovery. Another motivation for studying PSCs is to understand human embryogenesis and development. All cell types that make up the body tissues develop through defined trajectories during embryogenesis. For example, paraxial mesoderm is considered to differentiate into several cell types including skeletal muscle cells, chondrocytes, osteocytes, dermal fibroblasts, and tenocytes. Tenocytes are fibroblast cells that constitute the tendon. The step-wise narrowing fate decisions of paraxial mesoderm in the embryo have been modeled in vitro using PSCs; however, deriving tenocytes from human-induced PSCs and their application in cell therapy have long been challenging. PSC-derived tenocytes can be used for a source of cell transplantation to treat a damaged or ruptured tendon due to injury, disorder, or aging. In this review, we discuss the latest research findings on the use of PSCs for studying the biology of tenocyte development and their application in therapeutic settings.


Pluripotent Stem Cells/cytology , Tenocytes/cytology , Cell Differentiation , Humans
13.
PLoS One ; 15(12): e0242640, 2020.
Article En | MEDLINE | ID: mdl-33373386

To examine the differential mechanobiological responses of specific resident tendon cells, we developed an in vivo model of whole-body irradiation followed by injection of either tendon stem/progenitor cells (TSCs) expressing green fluorescent protein (GFP-TSCs) or mature tenocytes expressing GFP (GFP-TNCs) into the patellar tendons of wild type C57 mice. Injected mice were subjected to short term (3 weeks) treadmill running, specifically moderate treadmill running (MTR) and intensive treadmill running (ITR). In MTR mice, both GFP-TSC and GFP-TNC injected tendons maintained normal cell morphology with elevated expression of tendon related markers collagen I and tenomodulin. In ITR mice injected with GFP-TNCs, cells also maintained an elongated shape similar to the shape found in normal/untreated control mice, as well as elevated expression of tendon related markers. However, ITR mice injected with GFP-TSCs showed abnormal changes, such as cell morphology transitioning to a round shape, elevated chondrogenic differentiation, and increased gene expression of non-tenocyte related genes LPL, Runx-2, and SOX-9. Increased gene expression data was supported by immunostaining showing elevated expression of SOX-9, Runx-2, and PPARγ. This study provides evidence that while MTR maintains tendon homeostasis by promoting the differentiation of TSCs into TNCs, ITR causes the onset of tendinopathy development by inducing non-tenocyte differentiation of TSCs, which may eventually lead to the formation of non-tendinous tissues in tendon tissue after long term mechanical overloading conditions on the tendon.


Chondrocytes/cytology , Stem Cells/cytology , Tendinopathy/pathology , Tendons/pathology , Tenocytes/cytology , Animals , Biomarkers/metabolism , Cell Differentiation , Cell Shape , Cell Tracking , Chondrocytes/metabolism , Collagen Type I/genetics , Collagen Type I/metabolism , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Exercise Test , Female , Gene Expression Regulation , Genes, Reporter , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Lipoprotein Lipase/genetics , Lipoprotein Lipase/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , PPAR gamma/genetics , PPAR gamma/metabolism , Physical Conditioning, Animal/adverse effects , Running , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/metabolism , Stem Cells/metabolism , Tendinopathy/etiology , Tendinopathy/genetics , Tendinopathy/metabolism , Tendons/metabolism , Tenocytes/metabolism
14.
Cells ; 9(12)2020 12 08.
Article En | MEDLINE | ID: mdl-33302563

Mechanotransduction is the ability of cells to translate mechanical stimuli into biochemical signals that can ultimately influence gene expression, cell morphology and cell fate. Tenocytes are responsible for tendon mechanical adaptation converting mechanical stimuli imposed during mechanical loading, thus affecting extracellular matrix homeostasis. Since we previously demonstrated that MD-Tissue, an injectable collagen-based medical compound containing swine-derived collagen as the main component, is able to affect tenocyte properties, the aim of this study was to analyze whether the effects triggered by MD-Tissue were based on mechanotransduction-related mechanisms. For this purpose, MD-Tissue was used to coat Petri dishes and cytochalasin B was used to deprive tenocytes of mechanical stimulation mediated by the actin cytoskeleton. Cell morphology, migration, collagen turnover pathways and the expression of key mechanosensors were analyzed by morphological and molecular methods. Our findings confirm that MD-Tissue affects collagen turnover pathways and favors cell migration and show that the MD-Tissue-induced effect represents a mechanical input involving the mechanotransduction machinery. Overall, MD-Tissue, acting as a mechanical scaffold, could represent an effective medical device for a novel therapeutic, regenerative and rehabilitative approach to favor tendon healing in tendinopathies.


Collagen/chemistry , Stress, Mechanical , Tenocytes/metabolism , Actin Cytoskeleton , Aged , Animals , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Movement/drug effects , Cells, Cultured , Collagen Type I/metabolism , Cytochalasin B/pharmacology , Female , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Focal Adhesions/metabolism , Humans , Male , Middle Aged , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/genetics , Procollagen-Lysine, 2-Oxoglutarate 5-Dioxygenase/metabolism , Swine , Tenocytes/cytology , Tenocytes/drug effects , Tenocytes/pathology , Transcription Factors/genetics , Transcription Factors/metabolism , Vinculin/genetics , Vinculin/metabolism
15.
Int J Mol Sci ; 21(22)2020 Nov 18.
Article En | MEDLINE | ID: mdl-33218011

Defining the best combination of cells and biomaterials is a key challenge for the development of tendon tissue engineering (TE) strategies. Adipose-derived stem cells (ASCs) are ideal candidates for this purpose. In addition, controlled cell-based products adherent to good manufacturing practice (GMP) are required for their clinical scale-up. With this aim, in this study, ASC 3D bioprinting and GMP-compliant tenogenic differentiation were investigated. In detail, primary human ASCs were embedded within a nanofibrillar-cellulose/alginate bioink and 3D-bioprinted into multi-layered square-grid matrices. Bioink viscoelastic properties and scaffold ultrastructural morphology were analyzed by rheology and scanning electron microscopy (SEM). The optimal cell concentration for printing among 3, 6 and 9 × 106 ASC/mL was evaluated in terms of cell viability. ASC morphology was characterized by SEM and F-actin immunostaining. Tenogenic differentiation ability was then evaluated in terms of cell viability, morphology and expression of scleraxis and collagen type III by biochemical induction using BMP-12, TGF-ß3, CTGF and ascorbic acid supplementation (TENO). Pro-inflammatory cytokine release was also assessed. Bioprinted ASCs showed high viability and survival and exhibited a tenocyte-like phenotype after biochemical induction, with no inflammatory response to the bioink. In conclusion, we report a first proof of concept for the clinical scale-up of ASC 3D bioprinting for tendon TE.


Adipose Tissue/metabolism , Bioprinting , Cell Differentiation/drug effects , Culture Media , Printing, Three-Dimensional , Stem Cells/metabolism , Tenocytes/metabolism , Adipose Tissue/cytology , Cell Culture Techniques , Culture Media/chemistry , Culture Media/pharmacology , Humans , Stem Cells/cytology , Tenocytes/cytology
16.
Int J Mol Sci ; 21(21)2020 Oct 26.
Article En | MEDLINE | ID: mdl-33114764

Rotator cuff tears (RCTs) and rotator cuff disease (RCD) are important causes of disability in middle-aged individuals affected by nontraumatic shoulder dysfunctions. Our previous studies have demonstrated that four different hyaluronic acid preparations (HAPs), including Artrosulfur® hyaluronic acid (HA) (Alfakjn S.r.l., Garlasco, Italy), may exert a protective effect in human RCT-derived tendon cells undergoing oxidative stress damage. Recently, methylsulfonylmethane (MSM) (Barentz, Paderno Dugnano, Italy) has proven to have anti-inflammatory properties and to cause pain relief in patients affected by tendinopathies. This study aims at evaluating three preparations (Artrosulfur® HA, MSM, and Artrosulfur® MSM + HA) in the recovery from hydrogen peroxide-induced oxidative stress damage in human tenocyte. Cell proliferation, Lactate Dehydrogenase (LDH) release, and inducible nitric oxide synthases (iNOS) and prostaglandin E2 (PGE2) modulation were investigated. In parallel, expression of metalloproteinases 2 (MMP2) and 14 (MMP14) and collagen types I and III were also examined. Results demonstrate that Artrosulfur® MSM + HA improves cell escape from oxidative stress by decreasing cytotoxicity and by reducing iNOS and PGE2 secretion. Furthermore, it differentially modulates MMP2 and MMP14 levels and enhances collagen III expression after 24 h, proteins globally related to rapid acceleration of the extracellular matrix (ECM) remodelling and thus tendon healing. By improving the anti-cytotoxic effect of HA, the supplementation of MSM may represent a feasible strategy to ameliorate cuff tendinopathies.


Anti-Inflammatory Agents/pharmacology , Dimethyl Sulfoxide/pharmacology , Hyaluronic Acid/pharmacology , Hydrogen Peroxide/adverse effects , Rotator Cuff Injuries/metabolism , Sulfones/pharmacology , Tenocytes/cytology , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Dinoprostone/metabolism , Drug Synergism , Humans , L-Lactate Dehydrogenase/metabolism , Matrix Metalloproteinase 14/metabolism , Matrix Metalloproteinase 2/metabolism , Nitric Oxide Synthase Type II/metabolism , Oxidative Stress/drug effects , Rotator Cuff Injuries/drug therapy , Tenocytes/drug effects , Tenocytes/metabolism
17.
Sci Rep ; 10(1): 13939, 2020 09 03.
Article En | MEDLINE | ID: mdl-32883960

Tendinopathy accounts for over 30% of primary care consultations and represents a growing healthcare challenge in an active and increasingly ageing population. Recognising critical cells involved in tendinopathy is essential in developing therapeutics to meet this challenge. Tendon cells are heterogenous and sparsely distributed in a dense collagen matrix; limiting previous methods to investigate cell characteristics ex vivo. We applied next generation CITE-sequencing; combining surface proteomics with in-depth, unbiased gene expression analysis of > 6400 single cells ex vivo from 11 chronically tendinopathic and 8 healthy human tendons. Immunohistochemistry validated the single cell findings. For the first time we show that human tendon harbours at least five distinct COL1A1/2 expressing tenocyte populations in addition to endothelial cells, T-cells, and monocytes. These consist of KRT7/SCX+ cells expressing microfibril associated genes, PTX3+ cells co-expressing high levels of pro-inflammatory markers, APOD+ fibro-adipogenic progenitors, TPPP3/PRG4+ chondrogenic cells, and ITGA7+ smooth muscle-mesenchymal cells. Surface proteomic analysis identified markers by which these sub-classes could be isolated and targeted in future. Chronic tendinopathy was associated with increased expression of pro-inflammatory markers PTX3, CXCL1, CXCL6, CXCL8, and PDPN by microfibril associated tenocytes. Diseased endothelium had increased expression of chemokine and alarmin genes including IL33.


Single-Cell Analysis/methods , Stromal Cells/cytology , Tendons/cytology , Tendons/pathology , Adipogenesis/physiology , Adult , Antigens, CD/genetics , Antigens, CD/metabolism , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Female , Gene Expression Profiling , Humans , Integrin alpha Chains/genetics , Male , Middle Aged , Proteomics/methods , Stromal Cells/pathology , Tenocytes/cytology , Tenocytes/metabolism , Tenocytes/pathology , Young Adult
18.
Int J Mol Sci ; 21(16)2020 Aug 17.
Article En | MEDLINE | ID: mdl-32824547

Mesenchymal stem cells derived from human bone marrow (hBM-MSCs) are utilized in tendon tissue-engineering protocols while extra-embryonic cord-derived, including from Wharton's Jelly (hWJ-MSCs), are emerging as useful alternatives. To explore the tenogenic responsiveness of hBM-MSCs and hWJ-MSCs to human Growth Differentiation Factor 5 (hGDF-5) we supplemented each at doses of 1, 10, and 100 ng/mL of hGDF-5 and determined proliferation, morphology and time-dependent expression of tenogenic markers. We evaluated the expression of collagen types 1 (COL1A1) and 3 (COL3A1), Decorin (DCN), Scleraxis-A (SCX-A), Tenascin-C (TNC) and Tenomodulin (TNMD) noting the earliest and largest increase with 100 ng/mL. With 100 ng/mL, hBM-MSCs showed up-regulation of SCX-A (1.7-fold) at Day 1, TNC (1.3-fold) and TNMD (12-fold) at Day 8. hWJ-MSCs, at the same dose, showed up-regulation of COL1A1 (3-fold), DCN (2.7-fold), SCX-A (3.8-fold) and TNC (2.3-fold) after three days of culture. hWJ-MSCs also showed larger proliferation rate and marked aggregation into a tubular-shaped system at Day 7 (with 100 ng/mL of hGDF-5). Simultaneous to this, we explored the expression of pro-inflammatory (IL-6, TNF, IL-12A, IL-1ß) and anti-inflammatory (IL-10, TGF-ß1) cytokines across for both cell types. hBM-MSCs exhibited a better balance of pro-inflammatory and anti-inflammatory cytokines up-regulating IL-1ß (11-fold) and IL-10 (10-fold) at Day 8; hWJ-MSCs, had a slight expression of IL-12A (1.5-fold), but a greater up-regulation of IL-10 (2.5-fold). Type 1 collagen and tenomodulin proteins, detected by immunofluorescence, confirming the greater protein expression when 100 ng/mL were supplemented. In the same conditions, both cell types showed specific alignment and shape modification with a length/width ratio increase, suggesting their response in activating tenogenic commitment events, and they both potential use in 3D in vitro tissue-engineering protocols.


Bone Marrow Cells/metabolism , Growth Differentiation Factor 5/pharmacology , Mesenchymal Stem Cells/metabolism , Tenocytes/metabolism , Adult , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Cell Differentiation , Cells, Cultured , Collagen/genetics , Collagen/metabolism , Decorin/genetics , Decorin/metabolism , Female , Humans , Interleukins/genetics , Interleukins/metabolism , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Tenascin/genetics , Tenascin/metabolism , Tenocytes/cytology , Umbilical Cord/cytology
19.
Int J Mol Sci ; 21(12)2020 Jun 14.
Article En | MEDLINE | ID: mdl-32545914

Tendinopathy is a rare but serious complication of quinolone therapy. Risk factors associated with quinolone-induced tendon disorders include chronic kidney disease accompanied by the accumulation of uremic toxins. Hence, the present study explored the effects of the representative uremic toxins phenylacetic acid (PAA) and quinolinic acid (QA), both alone and in combination with ciprofloxacin (CPX), on human tenocytes in vitro. Tenocytes incubated with uremic toxins +/- CPX were investigated for metabolic activity, vitality, expression of the dominant extracellular tendon matrix (ECM) protein type I collagen, cell-matrix receptor ß1-integrin, proinflammatory interleukin (IL)-1ß, and the ECM-degrading enzyme matrix metalloproteinase (MMP)-1. CPX, when administered at high concentrations (100 mM), suppressed tenocyte metabolism after 8 h exposure and at therapeutic concentrations after 72 h exposure. PAA reduced tenocyte metabolism only after 72 h exposure to very high doses and when combined with CPX. QA, when administered alone, led to scarcely any cytotoxic effect. Combinations of CPX with PAA or QA did not cause greater cytotoxicity than incubation with CPX alone. Gene expression of the pro-inflammatory cytokine IL-1ß was reduced by CPX but up-regulated by PAA and QA. Protein levels of type I collagen decreased in response to high CPX doses, whereas PAA and QA did not affect its synthesis significantly. MMP-1 mRNA levels were increased by CPX. This effect became more pronounced in the form of a synergism following exposure to a combination of CPX and PAA. CPX was more tenotoxic than the uremic toxins PAA and QA, which showed only distinct suppressive effects.


Ciprofloxacin/adverse effects , Interleukin-1beta/genetics , Phenylacetates/adverse effects , Quinolinic Acid/adverse effects , Tenocytes/cytology , Adult , Cell Survival/drug effects , Cells, Cultured , Collagen Type I/metabolism , Female , Gene Expression Regulation/drug effects , Humans , Male , Matrix Metalloproteinase 1/genetics , Tenocytes/drug effects , Tenocytes/metabolism
20.
J Vet Med Sci ; 82(7): 948-954, 2020 Jul 31.
Article En | MEDLINE | ID: mdl-32418945

The lateral cytoplasmic processes of tenocytes extend to form three-dimensional network surrounding collagen fibers. It is unknown whether connections between two cytoplasmic processes involve overlapping of the processes or merely surface contact. In this study, the two-dimensional and three-dimensional structure of tenocytes in the Achilles tendons of the newly hatched chicks were studied using transmission electron microscopy and serial block face-scanning electron microscopy. Observation of the two-dimensional structures revealed various forms of cellular connections, including connections between the cytoplasmic processes of adjacent tenocytes and between the cytoplasmic process of tenocytes and fibroblasts. Three-dimensional observation showed spike-like cytoplasmic processes extending from one tenocyte that interlocked with cytoplasmic processes from other tenocytes. Cytoplasmic processes from each tenocyte within the chick tendons interlocked to ensure a tight cell-to-cell connection around growing collagen fibers. A cellular network formed by these cytoplasmic processes surrounds each collagen fiber. Cell-cell junctions, which were suggested to be gap junctions, observed at sites of cytoplasmic process overlap most likely represent the major route for communication between tenocytes associated with fibroblasts, enabling vital signals important for maintaining the cell and tendon integrity to be transmitted.


Achilles Tendon/ultrastructure , Fibroblasts/ultrastructure , Tenocytes/ultrastructure , Achilles Tendon/cytology , Animals , Animals, Newborn , Chickens , Cytoplasmic Structures , Extracellular Matrix/ultrastructure , Fibroblasts/cytology , Gap Junctions , Microscopy, Electron, Scanning/veterinary , Microscopy, Electron, Transmission/veterinary , Tenocytes/cytology
...