Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
Mediators Inflamm ; 2024: 5273198, 2024.
Article in English | MEDLINE | ID: mdl-39108992

ABSTRACT

Tendinopathy is one of the most frequent musculoskeletal disorders characterized by sustained tissue inflammation and oxidative stress, accompanied by extracellular matrix remodeling. Patients suffering from this pathology frequently experience pain, swelling, stiffness, and muscle weakness. Current pharmacological interventions are based on nonsteroidal anti-inflammatory drugs; however, the effectiveness of these strategies remains ambiguous. Accumulating evidence supports that oral supplementation of natural compounds can provide preventive, and possibly curative, effects. Vitamin C (Vit C), collagen peptides (Coll), resveratrol (Res), and astaxanthin (Asx) were reported to be endowed with potential beneficial effects based on their anti-inflammatory and antioxidant activities. Here, we analyzed the efficacy of a novel combination of these compounds (Mix) in counteracting proinflammatory (IL-1ß) and prooxidant (H2O2) stimuli in human tenocytes. We demonstrated that Mix significantly impairs IL-6-induced IL-1ß secretion, NF-κB nuclear translocation, and MMP-2 production; notably, a synergistic effect of Mix over the single compounds could be observed. Moreover, Mix was able to significantly counteract H2O2-triggered ROS production. Together, these results point out that Mix, a novel combination of Vit C, Coll, Resv, and Asx, significantly impairs proinflammatory and prooxidant stimuli in tenocytes, mechanisms that contribute to the onset of tendinopathies.


Subject(s)
Anti-Inflammatory Agents , Antioxidants , Ascorbic Acid , Collagen , Resveratrol , Tendinopathy , Tenocytes , Xanthophylls , Humans , Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Resveratrol/pharmacology , Antioxidants/pharmacology , Xanthophylls/pharmacology , Xanthophylls/therapeutic use , Tendinopathy/drug therapy , Tendinopathy/metabolism , Collagen/metabolism , Anti-Inflammatory Agents/pharmacology , Tenocytes/metabolism , Tenocytes/drug effects , Interleukin-1beta/metabolism , Peptides/chemistry , Peptides/pharmacology , Hydrogen Peroxide/metabolism , Stilbenes/pharmacology , Stilbenes/therapeutic use , Reactive Oxygen Species/metabolism , NF-kappa B/metabolism , Cells, Cultured , Oxidative Stress/drug effects
2.
J Biomech Eng ; 146(9)2024 09 01.
Article in English | MEDLINE | ID: mdl-38529730

ABSTRACT

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.


Subject(s)
Gelatin , Tenocytes , Tissue Scaffolds , Gelatin/chemistry , Animals , Horses , Tenocytes/cytology , Tenocytes/metabolism , Tissue Scaffolds/chemistry , Mechanical Phenomena , Gene Expression Regulation , Cell Shape , Biomechanical Phenomena
3.
BMC Musculoskelet Disord ; 25(1): 561, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030590

ABSTRACT

BACKGROUND: Tendons are important dense fibrous structures connecting muscle to bone, and tendon stem cells (TDSCs) affect their repair and regeneration. The role of TDSC-derived exosomes (TDSC-Exos) is still being unexplored; therefore, this study aimed to investigate the protective effect of TDSC-Exos on tenocytes. METHODS: The TDSCs and tenocytes were all derived from Sprague Dawley (SD) rats. The expression of positive and negative markers of TDSCs were detected by flow cytometry, and the multi-differentiation ability was also detected to identify TDSCs. Exos were derived from TDSCs using ultracentrifugation; furthermore, Exos enriched with microRNA(miR)-377-3p were generated from TDSCs stably overexpressing miR-377-3p after transfection, identified with transmission electron microscopy (TEM), western blot and PKH26 staining assay. Moreover, the cell functions of tenocytes were evaluated by MTT, EdU, transwell, and flow cytometry. Dual luciferase reporter and RNA pull-down assays were used to verify the binding sites of miR-337-3p and caspase3 (CASP3) predicted by Targetscan. RESULTS: Exos (miR-337-3p) were taken up by tenocytes, and promoted the proliferation, migration, and invasion and suppressed the apoptosis of tenocytes in a dose-dependent manner. Bioinformatics analysis showed that CASP3 was a target of miR-377-3p, which was further verified by luciferase and RNA pull-down assays. Moreover, over-expressed CASP3 reversed the effects of Exos (miR-337-3p) on cell functions of tenocytes. CONCLUSIONS: Our findings suggest that Exos derived from miR-337-3p over-expressing TDSCs could potentially protect against tenocyte apoptosis by regulating CASP3. This novel therapeutic approach holds promise for the treatment of tendon injury, offering a glimmer of hope for improved patient outcomes.


Subject(s)
Apoptosis , Caspase 3 , Exosomes , MicroRNAs , Rats, Sprague-Dawley , Stem Cells , Tendons , Tenocytes , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Exosomes/metabolism , Exosomes/genetics , Apoptosis/physiology , Rats , Caspase 3/metabolism , Caspase 3/genetics , Tenocytes/metabolism , Stem Cells/metabolism , Tendons/metabolism , Tendons/cytology , Cell Proliferation/physiology , Cells, Cultured , Male , Tendon Injuries/genetics , Tendon Injuries/metabolism , Tendon Injuries/pathology , Cell Movement
4.
Int J Mol Sci ; 25(14)2024 Jul 11.
Article in English | MEDLINE | ID: mdl-39062855

ABSTRACT

Inflammation is a driving force of tendinopathy. The oxidation of phospholipids by free radicals is a consequence of inflammatory reactions and is an important indicator of tissue damage. Here, we have studied the impact of oxidized phospholipids (OxPAPC) on the function of human tenocytes. We observed that treatment with OxPAPC did not alter the morphology, growth and capacity to produce collagen in healthy or diseased tenocytes. However, since OxPAPC is a known modulator of the function of immune cells, we analyzed whether OxPAPC-treated immune cells might influence the fate of tenocytes. Co-culture of tenocytes with immature, monocyte-derived dendritic cells treated with OxPAPC (Ox-DCs) was found to enhance the proliferation of tenocytes, particularly those from diseased tendons. Using transcriptional profiling of Ox-DCs, we identified amphiregulin (AREG), a ligand for EGFR, as a possible mediator of this proliferation enhancing effect, which we could confirm using recombinant AREG. Of note, diseased tenocytes were found to express higher levels of EGFR compared to tenocytes isolated from healthy donors and show a stronger proliferative response upon co-culture with Ox-DCs, as well as AREG treatment. In summary, we identify an AREG-EGFR axis as a mediator of a DC-tenocyte crosstalk, leading to increased tenocyte proliferation and possibly tendon regeneration.


Subject(s)
Amphiregulin , Cell Proliferation , Coculture Techniques , Dendritic Cells , Oxidation-Reduction , Phospholipids , Tenocytes , Humans , Dendritic Cells/metabolism , Dendritic Cells/drug effects , Amphiregulin/metabolism , Amphiregulin/genetics , Cell Proliferation/drug effects , Tenocytes/metabolism , Tenocytes/cytology , Tenocytes/drug effects , Phospholipids/metabolism , ErbB Receptors/metabolism , Cells, Cultured , Male , Female , Middle Aged
5.
Eur J Cell Biol ; 103(3): 151445, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39024989

ABSTRACT

Allogeneic serum and tissue-specific extracellular matrix have been shown to maintain permanently differentiated cell phenotype in culture. This is of particular importance for human tenocytes, a cell population that readily loses its function during ex vivo culture. With these in mind, herein we extracted human tenocytes using either foetal bovine serum or human serum, cultured them in the absence and presence of carrageenan and Ficoll®, the most widely used macromolecular crowding agents (to induce tissue-specific extracellular matrix deposition), and assessed cellular function, via metabolic activity, viability, proliferation and immunofluorescence for collagen related molecules, non-collagenous molecules and transmembrane molecules. At day 7, longest time point assessed, neither carrageenan nor Ficoll® significantly affected metabolic activity, viability and proliferation in either serum and human serum significantly increased metabolic activity and proliferation. At day 7, in the absence of macromolecular crowding, cells in human serum deposited significantly lower collagen type VI, biglycan, versican and tenomodulin than cells in foetal bovine serum. Interestingly, at day 7, in comparison to the no macromolecular crowding group, carrageenan in foetal bovine serum induced the highest effect, as judged by the highest number of significantly increased molecules (collagen type I, collagen type IV, collagen type V, collagen type VI, transforming growth factor ß1, matrix metalloproteinase 14, lumican, versican, scleraxis and integrin α2ß1). These data, although contradict previous observations where human serum outperformed foetal bovine serum, at the same time, support the use of foetal bovine serum in the development of cell-based medicines.


Subject(s)
Tenocytes , Humans , Tenocytes/metabolism , Tenocytes/cytology , Cells, Cultured , Cell Proliferation , Animals , Serum/metabolism , Serum/chemistry , Cattle , Carrageenan/pharmacology , Ficoll , Extracellular Matrix/metabolism
6.
Acta Biomater ; 180: 279-294, 2024 05.
Article in English | MEDLINE | ID: mdl-38604466

ABSTRACT

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.


Subject(s)
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
7.
J ISAKOS ; 9(4): 603-608, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38754838

ABSTRACT

OBJECTIVES: Autologous tenocyte implantation (OrthoATI™) therapy has demonstrated efficacy in treating patients with tendinopathy at various anatomical sites. This study evaluates the effect of patient age, gender, and tendon biopsy site on morphology, growth, and gene expression of autologous tendon cells used to treat chronic tendinopathy. METHODS: Patients undergoing OrthoATI™ for tendinopathies between 2020 and 2022 were initially treated by biopsies taken from patella tendon (PT) or palmaris longus tendon (PL). The biopsies were sent to a Good Manufacturing Practice (GMP) cell laboratory where tendon cells were isolated, cultured, and expanded for four to six weeks. Cell morphology was assessed using phase contrast microscopy. Droplet digital PCR (ddPCR) was utilized for gene expression analysis. Dichotomous results were compared between groups using x2 or Fisher's exact tests with no adjustment for multiple comparisons. The nonparametric Mann-Whitney U and Kruskal-Wallis tests were utilized for the sex and age (<35y, 35-44y, 45-54y, >55y) analyses, respectively. All analyses were performed using IBM SPSS v27, and a two-tailed P-value of <0.05 was considered statistically significant. RESULTS: 149 patients were included in the analysis. The PT was biopsied in 63 patients, and PL in 86 patients. There were no observer effects for age and gender between the PT and PL groups. There was no statistical significance between the PT and PL tendons for cell morphology, average cell population doubling time (PDT) (PT 83.9 vs PL 82.7 â€‹h, p â€‹= â€‹0.482), cellular yield (PT 16.2 vs PL 15.2 â€‹× â€‹106, p â€‹= â€‹0.099), and cell viability (PT 98.7 vs PL 99.0%, p â€‹= â€‹0.277). Additionally, ddPCR analyses showed no statistical significance found in tenogenic gene expression, including collagen type I (COL1, p â€‹= â€‹0.86), tenomodulin (TNMD, p â€‹= â€‹0.837) and scleraxis (SCX, p â€‹= â€‹0.331) between PT- and PL-derived tendon cells. An age stratification analysis found no effect on growth and gene expression. COL1 was found to be higher in males when compared to females (P â€‹< â€‹0.001), but otherwise no difference was seen in growth and gene expression in the gender analysis. No postbiopsy clinical complications were reported for either group. CONCLUSION: This study has shown that the growth and bioactivities of tendon cells from tendon biopsies for OrthoATI™ are not affected by tendon donor site and age. LEVEL OF EVIDENCE: IV.


Subject(s)
Tendinopathy , Tenocytes , Transplantation, Autologous , Humans , Tendinopathy/therapy , Female , Male , Middle Aged , Adult , Tenocytes/metabolism , Transplantation, Autologous/methods , Age Factors , Chronic Disease , Tendons , Transplant Donor Site , Biopsy/methods , Sex Factors
8.
Stem Cell Rev Rep ; 20(4): 1040-1059, 2024 May.
Article in English | MEDLINE | ID: mdl-38396222

ABSTRACT

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.


Subject(s)
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
9.
Am J Sports Med ; 52(2): 406-422, 2024 02.
Article in English | MEDLINE | ID: mdl-38193194

ABSTRACT

BACKGROUND: Tendons have limited regenerative potential, so healing of ruptured tendon tissue requires a prolonged period, and the prognosis is suboptimal. Although stem cell transplantation-based approaches show promise for accelerating tendon repair, the resultant therapeutic efficacy remains unsatisfactory. HYPOTHESIS: The transplantation of stem cells preassembled as 3-dimensional spheroids achieves a superior therapeutic outcome compared with the transplantation of single-cell suspensions. STUDY DESIGN: Controlled laboratory study. METHODS: Adipose-derived stem cells (ADSCs) were assembled as spheroids using a methylcellulose hydrogel system. The secretome of ADSC suspensions or spheroids was collected and utilized to treat tenocytes and macrophages to evaluate their therapeutic potential and investigate the mechanisms underlying their effects. RNA sequencing was performed to investigate the global difference in gene expression between ADSC suspensions and spheroids in an in vitro inflammatory microenvironment. For the in vivo experiment, rabbits that underwent Achilles tendon transection, followed by stump suturing, were randomly assigned to 1 of 3 groups: intratendinous injection of saline, rabbit ADSCs as conventional single-cell suspensions, or preassembled ADSC spheroids. The tendons were harvested for biomechanical testing and histological analysis at 4 weeks postoperatively. RESULTS: Our in vitro results demonstrated that the secretome of ADSCs assembled as spheroids exhibited enhanced modulatory activity in (1) tenocyte proliferation (P = .015) and migration (P = .001) by activating extracellular signal-regulated kinase (ERK) signaling and (2) the suppression of the secretion of interleukin-6 (P = .005) and interleukin-1α (P = .042) by M1 macrophages via the COX-2/PGE2/EP4 signaling axis. Gene expression profiling of cells exposed to an inflammatory milieu revealed significantly enriched terms that were associated with the immune response, cytokines, and tissue remodeling in preassembled ADSC spheroids. Ex vivo fluorescence imaging revealed that the engraftment efficiency of ADSCs in the form of spheroids was higher than that of ADSCs in single-cell suspensions (P = .003). Furthermore, the transplantation of ADSC spheroids showed superior therapeutic effects in promoting the healing of sutured stumps, as evidenced by improvements in the tensile strength (P = .019) and fiber alignment (P < .001) of the repaired tendons. CONCLUSION: The assembly of ADSCs as spheroids significantly advanced their potential to harness tenocytes and macrophages. As a proof of concept, this study clearly demonstrates the effectiveness of using ADSC spheroids to promote tendon regeneration. CLINICAL RELEVANCE: The present study lays a foundation for future clinical applications of stem cell spheroid-based therapy for the management of tendon injuries.


Subject(s)
Achilles Tendon , Tendon Injuries , Animals , Rabbits , Achilles Tendon/pathology , Tenocytes , Adipose Tissue/pathology , Tendon Injuries/surgery , Macrophages/pathology , Stem Cells/physiology , Cell Proliferation
10.
Biomater Adv ; 163: 213950, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38972278

ABSTRACT

Traditional tendon engineering using cell-loaded scaffold has limited application potential due to the need of autologous cells. We hypothesize that potent mechanical loading can efficiently induce in situ Achilles tendon regeneration in a rabbit model by using a cell-free porous composite scaffold. In this study, melt-spinning was used to fabricate PGA (polyglycolic acid) and PLA (polylactic acid) filament fibers as well as non-woven PGA fibers. The PLA/PGA (4:2) filament fibers were further braided into a hybrid yarn,which was knitted into a PLA/PGA tubular mesh with potent mechanical property for sustaining natural tendon strain. The results showed that a complete cross-section of Achilles tendon created a model of full mechanical loading on the bridging scaffold, which could efficiently induce in situ tendon regeneration by promoting host cell infiltration, matrix production and tissue remodeling. Histologically, mechanical loading assisted in forming parallel aligned collagen fibers and tenocytes in a fashion similar to those of native tendon. Transmission electron microscope further demonstrated that mechanical strain induced collagen fibril development by increasing fibril diameter and forming bipolar structure, which resulted in enhanced mechanical properties. Interestingly, the synergistic effect between mechanical loading and hyaluronic acid modification was also observed on the induced tenogenic differentiation of infiltrated host fibroblasts. In conclusion, potent mechanical loading is the key inductive microenvironment for in situ tendon regeneration for this polymer-based composite scaffold with proper matrix modification, which may serve as a universal scaffold product for tendon regeneration.


Subject(s)
Achilles Tendon , Polyesters , Regeneration , Tissue Engineering , Tissue Scaffolds , Animals , Rabbits , Tissue Scaffolds/chemistry , Tissue Engineering/methods , Polyesters/chemistry , Tenocytes , Polymers/chemistry , Biocompatible Materials/chemistry , Stress, Mechanical
11.
Sci Rep ; 14(1): 9495, 2024 04 25.
Article in English | MEDLINE | ID: mdl-38664570

ABSTRACT

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.


Subject(s)
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
12.
PLoS One ; 19(8): e0309301, 2024.
Article in English | MEDLINE | ID: mdl-39172877

ABSTRACT

Tendinopathy is a common age-related disease which causes significant morbidity for both human athletes and performance horses. In the latter, the superficial digital flexor tendon is an excellent model for human tendinopathies because it is a functional homologue of the human Achilles tendon and a primary site of injuries with strong similarities to the human disease. Corticosteroids have been previously used clinically to treat tendinopathic inflammation, but they upregulate the p53-p21 axis with concomitant reductions in cell proliferation and collagen synthesis in human tenocytes. This phenotype is consistent with the induction of cellular senescence in vitro and in vivo and probably represents an important clinical barrier to their effective use. Because of the many differences in senescence mechanisms between species, this study aimed to evaluate these mechanisms after corticosteroid treatment in equine tenocytes. Exposure to clinically reflective levels of dexamethasone for 48 hours drove equine tenocytes into steroid induced senescence (SIS). This was characterised by permanent growth arrest and upregulation of p53, the cyclin dependent kinase inhibitors p21waf and p16ink4a as well as the matrix degrading enzymes MMP1, MMP2 and MMP13. SIS also induced a distinctive equine senescence associated secretory phenotype (eSASP) characterised by enhanced secretion of IL-8 and MCP-1. Preincubation with resveratrol or the potent SIRT1 activator SRT1720 prevented SIS in equine tenocytes, while treatment with the non-SIRT1 activating resveratrol analogue V29 was equally protective against SIS, consistent with a novel, as yet uncharacterised SIRT1-indendent mechanism which has relevance for the development of future preventative and therapeutic strategies.


Subject(s)
Cellular Senescence , Dexamethasone , Sirtuin 1 , Tenocytes , Animals , Horses , Sirtuin 1/metabolism , Cellular Senescence/drug effects , Tenocytes/drug effects , Tenocytes/metabolism , Dexamethasone/pharmacology , Resveratrol/pharmacology , Cell Proliferation/drug effects , Tumor Suppressor Protein p53/metabolism , Tendinopathy/metabolism , Tendinopathy/pathology , Tendinopathy/drug therapy , Cells, Cultured , Tendons/drug effects , Tendons/cytology , Tendons/metabolism
13.
Tissue Cell ; 88: 102392, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38643674

ABSTRACT

The effect of interleukin-38 (IL-38), a recently identified member of the IL-1 family with potential applications in various inflammation-related conditions, on ER stress has not been explored. Furthermore, its role in obesity-associated tendinopathy has not been investigated. In this study, human primary tenocytes were treated with palmitate (200 or 400 µM) and palmitate plus IL-38 (0-50 ng/mL) for 24 h. Western blotting was used to assess ER stress and tendinopathogenic markers in tenocytes. Monodansylcadaverine (MDC) staining was used to evaluate autophagosomes. Apoptosis was determined by cell viability assays, caspase 3 activity assays and TUNEL assays. Cell migration was evaluated by a cell scratch assay. Small interfering (si) RNA transfection was used for target gene silencing. Treatment of tenocytes with IL-38 attenuated apoptosis, restored the balance between MMPs and TIMP-1, and alleviated ER stress under palmitate conditions. IL-38 treatment enhanced AMPK phosphorylation and promoted the expression of autophagy markers related to LC3 conversion, p62 degradation, and autophagosome formation in cultured tenocytes. The effects of IL-38 on ER stress, apoptosis, and MMP-9, MMP-13, and TIMP-1 expression in palmitate-treated tenocytes were abrogated by AMPK siRNA or 3-methyladenine (3MA). These results suggest that IL-38 alleviates ER stress through the AMPK/autophagy pathway, thereby reducing apoptosis and preventing extracellular matrix (ECM) degradation in tenocytes under hyperlipidemic conditions. This study provides a promising therapeutic avenue for treating obesity-related tendinopathy using an endogenous compound such as IL-38.


Subject(s)
Apoptosis , Autophagy , Endoplasmic Reticulum Stress , Obesity , Tendinopathy , Tenocytes , Humans , Autophagy/drug effects , Tendinopathy/pathology , Tendinopathy/metabolism , Tendinopathy/drug therapy , Obesity/metabolism , Obesity/pathology , Apoptosis/drug effects , Tenocytes/metabolism , Tenocytes/drug effects , Endoplasmic Reticulum Stress/drug effects , AMP-Activated Protein Kinases/metabolism , Interleukins/metabolism , Cell Movement/drug effects
14.
Biochem Pharmacol ; 226: 116370, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38880359

ABSTRACT

Tendon injuries typically display limited reparative capacity, often resulting in suboptimal outcomes and an elevated risk of recurrence or rupture. While cytokines of the IL-6 family are primarily recognised for their inflammatory properties, they also have multifaceted roles in tissue regeneration and repair. Despite this, studies examining the association between IL-6 family cytokines and tendon repair remained scarce. gp130, a type of glycoprotein, functions as a co-receptor for all cytokines in the IL-6 family. Its role is to assist in the transmission of signals following the binding of ligands to receptors. RCGD423 is a gp130 modulator. Phosphorylation of residue Y759 of gp130 recruits SHP2 and SOCS3 and inhibits activation of the STAT3 pathway. In our study, RCGD423 stimulated the formation of homologous dimers of gp130 and the phosphorylation of Y759 residues without the involvement of IL-6 and IL-6R. Subsequently, the phosphorylated residues recruited SHP2, activating the downstream ERK and AKT pathways. These mechanisms ultimately promoted the migration ability of tenocytes and matrix synthesis, especially collagen I. Moreover, RCGD423 also demonstrated significant improvements in collagen content, alignment of collagen fibres, and biological and biomechanical function in a rat Achilles tendon injury model. In summary, we demonstrated a promising gp130 modulator (RCGD423) that could potentially enhance tendon injury repair by redirecting downstream signalling of IL-6, suggesting its potential therapeutic application for tendon injuries.


Subject(s)
Achilles Tendon , Cell Movement , Cytokine Receptor gp130 , Protein Tyrosine Phosphatase, Non-Receptor Type 11 , Proto-Oncogene Proteins c-akt , Rats, Sprague-Dawley , Tenocytes , Animals , Protein Tyrosine Phosphatase, Non-Receptor Type 11/metabolism , Cytokine Receptor gp130/metabolism , Achilles Tendon/metabolism , Achilles Tendon/injuries , Achilles Tendon/drug effects , Cell Movement/drug effects , Cell Movement/physiology , Rats , Proto-Oncogene Proteins c-akt/metabolism , Tenocytes/metabolism , Tenocytes/drug effects , Tenocytes/physiology , Collagen/metabolism , Signal Transduction/drug effects , Signal Transduction/physiology , Male , MAP Kinase Signaling System/drug effects , MAP Kinase Signaling System/physiology , Tendon Injuries/metabolism , Tendon Injuries/drug therapy
15.
Sci Rep ; 14(1): 13540, 2024 06 12.
Article in English | MEDLINE | ID: mdl-38866832

ABSTRACT

Mast cells are immune cells minimally present in normal tendon tissue. The increased abundance of mast cells in tendinopathy biopsies and at the sites of tendon injury suggests an unexplored role of this cell population in overuse tendon injuries. Mast cells are particularly present in tendon biopsies from patients with more chronic symptom duration and a history of intensive mechanical loading. This study, therefore, examined the cross talk between mast cells and human tendon cells in either static or mechanically active conditions in order to explore the potential mechanistic roles of mast cells in overuse tendon injuries. A coculture of isolated human tenocytes and mast cells (HMC-1) combined with Flexcell Tension System for cyclic stretching of tenocytes was used. Additionally, human tenocytes were exposed to agonists and antagonists of substance P (SP) receptors. Mast cell degranulation was assessed by measuring ß-hexosaminidase activity. Transwell and cell adhesion assays were used to evaluate mast cell migration and binding to tendon extracellular matrix components (collagen and fibronectin), respectively. Gene expressions were analyzed using real time qRT-PCR. Our results indicate that mechanical stimulation of human tenocytes leads to release of SP which, in turn, activates mast cells through the Mas-related G-protein-coupled receptor X2 (MRGPRX2). The degranulation and migration of mast cells in response to MRGPRX2 activation subsequently cause human tenocytes to increase their expression of inflammatory factors, matrix proteins and matrix metalloproteinase enzymes. These observations may be important in understanding the mechanisms by which tendons become tendinopathic in response to repetitive mechanical stimulation.


Subject(s)
Mast Cells , Receptors, G-Protein-Coupled , Receptors, Neuropeptide , Substance P , Tendons , Tenocytes , Humans , Substance P/metabolism , Substance P/pharmacology , Mast Cells/metabolism , Tenocytes/metabolism , Receptors, G-Protein-Coupled/metabolism , Receptors, Neuropeptide/metabolism , Receptors, Neuropeptide/genetics , Tendons/metabolism , Tendons/pathology , Nerve Tissue Proteins/metabolism , Nerve Tissue Proteins/genetics , Cell Degranulation , Tendinopathy/metabolism , Tendinopathy/pathology , Inflammation/metabolism , Inflammation/pathology , Male , Coculture Techniques , Cells, Cultured , Adult , Cell Movement
16.
J Am Vet Med Assoc ; 262(S1): S61-S72, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38547589

ABSTRACT

OBJECTIVE: The study objectives were to 1) determine the mesenchymal stem cell (MSC) surface expression of major histocompatibility complex (MHC) class I and transcriptome-wide gene expression changes following IL-1ß + TGF-ß2 dual licensing and 2) evaluate if IL-1ß + TGF-ß2 dual-licensed MSCs had a greater ability to positively modulate tenocyte function compared to naive MSCs. SAMPLE: Equine bone marrow-derived MSCs from 6 donors and equine superficial digital flexor tenocytes from 3 donors. METHODS: Experiments were performed in vitro. Flow cytometry and bulk RNA sequencing were utilized to determine naive and dual-licensed MSC phenotype and transcriptome-wide changes in gene expression. Conditioned media were generated from MSCs and utilized in tenocyte cell culture assays as a method to determine the effect of MSC paracrine factors on tenocyte function. RESULTS: Dual-licensed MSCs have a reduced expression of MHC class I and exhibit enrichment in functional pathways associated with the extracellular matrix, cell signaling, and tissue development. Additionally, dual-licensed MSC-conditioned media significantly improved in vitro tenocyte migration and metabolism to a greater degree than naive MSC-conditioned media. In tenocytes exposed to IL-1ß, dual-licensed conditioned media also positively modulated tenocyte gene expression. CLINICAL RELEVANCE: Our data indicate that conditioned media containing paracrine factors secreted from dual-licensed MSCs significantly modulates in vitro tenocyte function, which may confer benefits in vivo to healing tendons following injury. Additionally, due to reduced MHC class I expression in dual-licensed MSCs, this technique may also provide an avenue to provide an effective "off-the-shelf" allogenic source of MSCs.


Subject(s)
Interleukin-1beta , Mesenchymal Stem Cells , Tenocytes , Transforming Growth Factor beta2 , Animals , Mesenchymal Stem Cells/metabolism , Horses , Interleukin-1beta/metabolism , Interleukin-1beta/genetics , Transforming Growth Factor beta2/metabolism , Transforming Growth Factor beta2/genetics , Tenocytes/metabolism , Cell Movement/drug effects , Gene Expression Regulation/drug effects , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class I/genetics , Cells, Cultured
17.
Vet J ; 306: 106159, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38849026

ABSTRACT

Local anaesthetics (LAs) can have detrimental effects on rat, bovine, canine, and human tendon tissues and cells. Currently, there has been no available data on the impact of these drugs on equine tenocytes. Even if LA injection for managing painful tendon conditions in horses is limited, it is usually used via intra-articular, intrasynovial, perineural, and intrathecal as well as for lameness examinations. In this in vitro study, the cytotoxic effects of LAs, including lidocaine, mepivacaine, and bupivacaine on equine tenocytes, in the presence and absence of platelet rich plasma (PRP), were investigated. PRP accelerates tissue healing and can exert cytoprotective effects on different cell types exposed to different stressful conditions, including drugs. Results indicated that the exposure to LAs significantly reduced tenocytes viability in dose- and time-dependent manners while PRP was able to counteract their cytotoxic effects. Furthermore, microscopy and flow cytometry analyses revealed apoptosis and necrosis in equine tenocytes exposed to these drugs, that were both reduced when PRP was in the medium. These findings highlight the importance of considering the tenocyte toxicity associated with intrathecal and intraneural LA injections, as they might affect tenocytes or reduce the efficacy of associated therapies. Moreover, this study also highlights the protective effects of PRP, which could make LA injections safer.


Subject(s)
Anesthetics, Local , Bupivacaine , Lidocaine , Mepivacaine , Platelet-Rich Plasma , Tenocytes , Animals , Horses , Tenocytes/drug effects , Anesthetics, Local/pharmacology , Anesthetics, Local/toxicity , Bupivacaine/toxicity , Lidocaine/pharmacology , Cell Survival/drug effects , Cells, Cultured , Apoptosis/drug effects
18.
Aging (Albany NY) ; 16(3): 2702-2714, 2024 02 02.
Article in English | MEDLINE | ID: mdl-38309291

ABSTRACT

OBJECTIVE: Tendinopathy is influenced by multiple factors, including chronic inflammation and aging. Senescent cells exhibit characteristics such as the secretion of matrix-degrading enzymes and pro-inflammatory cytokines, collectively known as senescence-associated secretory phenotypes (SASPs). Many of these SASP cytokines and enzymes are implicated in the pathogenesis of tendinopathy. MicroRNA-146a (miR-146a) blocks senescence by targeting interleukin-1ß (IL-1ß) receptor-associated kinase 4 (IRAK-4) and TNF receptor-associated factor 6 (TRAF6), thus inhibiting NF-κB activity. The aims of this study were to (1) investigate miR-146a expression in tendinopathic tendons and (2) evaluate the role of miR-146a in countering senescence and SASPs in tendinopathic tenocytes. METHODS: MiR-146a expression was assessed in human long head biceps (LHB) and rat tendinopathic tendons by in situ hybridization. MiR-146a over-expression in rat primary tendinopathic tenocytes was achieved by lentiviral vector-mediated precursor miR-146a transfer (LVmiR-146a). Expression of various senescence-related markers was analyzed by quantitative reverse transcription polymerase chain reaction (qRT-PCR), immunoblotting and immunofluorescence. MiR-146a expression showed a negative correlation with the severity of tendinopathy in human and rat tendinopathic tendons (p<0.001). RESULTS: Tendinopathic tenocyte transfectants overexpressing miR-146a exhibited downregulation of various senescence and SASP markers, as well as the target molecules IRAK-4 and TRAF6, and the inflammatory mediator phospho-NF-κB. Additionally, these cells showed enhanced nuclear staining of high mobility group box 1 (HMGB1) compared to LVmiR-scramble-transduced controls in response to IL-1ß stimulation. CONCLUSIONS: We demonstrate that miR-146a expression is negatively correlated with the progression of tendinopathy. Moreover, its overexpression protects tendinopathic tenocytes from SASPs and senescence through the IRAK-4/TRAF6/NF-kB pathway.


Subject(s)
MicroRNAs , Tendinopathy , Animals , Humans , Rats , Cytokines/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Senescence-Associated Secretory Phenotype , Tendinopathy/genetics , Tenocytes/metabolism , TNF Receptor-Associated Factor 6/metabolism
19.
Int J Biol Macromol ; 273(Pt 2): 133193, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38885859

ABSTRACT

A major problem after tendon injury is adhesion formation to the surrounding tissue leading to a limited range of motion. A viable strategy to reduce adhesion extent is the use of physical barriers that limit the contact between the tendon and the adjacent tissue. The purpose of this study was to fabricate an electrospun bilayered tube of hyaluronic acid/polyethylene oxide (HA/PEO) and biodegradable DegraPol® (DP) to improve the anti-adhesive effect of the implant in a rabbit Achilles tendon full laceration model compared to a pure DP tube. Additionally, the attachment of rabbit tenocytes on pure DP and HA/PEO containing scaffolds was tested and Scanning Electron Microscopy, Fourier-transform Infrared Spectroscopy, Differential Scanning Calorimetry, Water Contact Angle measurements, and testing of mechanical properties were used to characterize the scaffolds. In vivo assessment after three weeks showed that the implant containing a second HA/PEO layer significantly reduced adhesion extent reaching levels comparable to native tendons, compared with a pure DP implant that reduced adhesion formation only by 20 %. Tenocytes were able to attach to and migrate into every scaffold, but cell number was reduced over two weeks. Implants containing HA/PEO showed better mechanical properties than pure DP tubes and with the ability to entirely reduce adhesion extent makes this implant a promising candidate for clinical application in tendon repair.


Subject(s)
Hyaluronic Acid , Polyethylene Glycols , Tissue Scaffolds , Animals , Hyaluronic Acid/chemistry , Hyaluronic Acid/pharmacology , Rabbits , Polyethylene Glycols/chemistry , Tissue Scaffolds/chemistry , Tenocytes/drug effects , Tenocytes/metabolism , Achilles Tendon/drug effects , Tendon Injuries/therapy , Cell Adhesion/drug effects , Tissue Adhesions/prevention & control , Tendons/drug effects , Tissue Engineering/methods , Biocompatible Materials/chemistry , Biocompatible Materials/pharmacology , Polyesters/chemistry , Polyurethanes
20.
Cell Rep ; 43(4): 114049, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38573853

ABSTRACT

Heterotopic ossification (HO) is a challenging condition that occurs after musculoskeletal injury and is characterized by the formation of bone in non-skeletal tissues. While the effect of HO on blood vessels is well established, little is known about its impact on lymphatic vessels. Here, we use a mouse model of traumatic HO to investigate the relationship between HO and lymphatic vessels. We show that injury triggers lymphangiogenesis at the injury site, which is associated with elevated vascular endothelial growth factor C (VEGF-C) levels. Through single-cell transcriptomic analyses, we identify mesenchymal progenitor cells and tenocytes as sources of Vegfc. We demonstrate by lineage tracing that Vegfc-expressing cells undergo osteochondral differentiation and contribute to the formation of HO. Last, we show that Vegfc haploinsufficiency results in a nearly 50% reduction in lymphangiogenesis and HO formation. These findings shed light on the complex mechanisms underlying HO formation and its impact on lymphatic vessels.


Subject(s)
Lymphangiogenesis , Mesenchymal Stem Cells , Ossification, Heterotopic , Vascular Endothelial Growth Factor C , Animals , Ossification, Heterotopic/metabolism , Ossification, Heterotopic/pathology , Ossification, Heterotopic/genetics , Vascular Endothelial Growth Factor C/metabolism , Vascular Endothelial Growth Factor C/genetics , Mice , Mesenchymal Stem Cells/metabolism , Lymphatic Vessels/metabolism , Lymphatic Vessels/pathology , Cell Differentiation , Tenocytes/metabolism , Osteogenesis , Haploinsufficiency , Mice, Inbred C57BL , Disease Models, Animal , Male
SELECTION OF CITATIONS
SEARCH DETAIL