RESUMO
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.
Assuntos
Anti-Inflamatórios , Antioxidantes , Ácido Ascórbico , Colágeno , Resveratrol , Tendinopatia , Tenócitos , Xantofilas , Humanos , Ácido Ascórbico/farmacologia , Ácido Ascórbico/uso terapêutico , Resveratrol/farmacologia , Antioxidantes/farmacologia , Xantofilas/farmacologia , Xantofilas/uso terapêutico , Tendinopatia/tratamento farmacológico , Tendinopatia/metabolismo , Colágeno/metabolismo , Anti-Inflamatórios/farmacologia , Tenócitos/metabolismo , Tenócitos/efeitos dos fármacos , Interleucina-1beta/metabolismo , Peptídeos/química , Peptídeos/farmacologia , Peróxido de Hidrogênio/metabolismo , Estilbenos/farmacologia , Estilbenos/uso terapêutico , Espécies Reativas de Oxigênio/metabolismo , NF-kappa B/metabolismo , Células Cultivadas , Estresse Oxidativo/efeitos dos fármacosRESUMO
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.
Assuntos
Senescência Celular , Dexametasona , Sirtuína 1 , Tenócitos , Animais , Cavalos , Sirtuína 1/metabolismo , Senescência Celular/efeitos dos fármacos , Tenócitos/efeitos dos fármacos , Tenócitos/metabolismo , Dexametasona/farmacologia , Resveratrol/farmacologia , Proliferação de Células/efeitos dos fármacos , Proteína Supressora de Tumor p53/metabolismo , Tendinopatia/metabolismo , Tendinopatia/patologia , Tendinopatia/tratamento farmacológico , Células Cultivadas , Tendões/efeitos dos fármacos , Tendões/citologia , Tendões/metabolismoRESUMO
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.
Assuntos
Anfirregulina , Proliferação de Células , Técnicas de Cocultura , Células Dendríticas , Oxirredução , Fosfolipídeos , Tenócitos , Humanos , Células Dendríticas/metabolismo , Células Dendríticas/efeitos dos fármacos , Anfirregulina/metabolismo , Anfirregulina/genética , Proliferação de Células/efeitos dos fármacos , Tenócitos/metabolismo , Tenócitos/citologia , Tenócitos/efeitos dos fármacos , Fosfolipídeos/metabolismo , Receptores ErbB/metabolismo , Células Cultivadas , Masculino , Feminino , Pessoa de Meia-IdadeRESUMO
Postoperative tissue adhesion and poor tendon healing are major clinical problems associated with tendon surgery. To avoid postoperative adhesion and promote tendon healing, we developed and synthesized a membrane to wrap the surgical site after tendon suturing. The bilayer-structured porous membrane comprised an outer layer [1,4-butanediol diglycidyl ether cross-linked with carboxymethyl cellulose (CX)] and an inner layer [1,4-butanediol diglycidyl ether cross-linked with Bletilla striata polysaccharides and carboxymethyl cellulose (CXB)]. The morphology, chemical functional groups, and membrane structure were determined. In vitro experiments revealed that the CX/CXB membrane demonstrated good biosafety and biodegradability, promoted tenocyte proliferation and migration, and exhibited low cell attachment and anti-inflammatory effects. Furthermore, in in vivo animal study, the CX/CXB membrane effectively reduced postoperative tendon-peripheral tissue adhesion and improved tendon repair, downregulating inflammatory cytokines in the tendon tissue at the surgical site, which ultimately increased tendon strength by 54% after 4 weeks.
Assuntos
Tendão do Calcâneo , Carboximetilcelulose Sódica , Polissacarídeos , Animais , Aderências Teciduais/prevenção & controle , Polissacarídeos/química , Polissacarídeos/farmacologia , Carboximetilcelulose Sódica/química , Carboximetilcelulose Sódica/farmacologia , Tendão do Calcâneo/efeitos dos fármacos , Tendão do Calcâneo/cirurgia , Tendão do Calcâneo/lesões , Orchidaceae/química , Membranas Artificiais , Ratos , Cicatrização/efeitos dos fármacos , Tenócitos/efeitos dos fármacos , Tenócitos/metabolismo , Proliferação de Células/efeitos dos fármacos , Masculino , Ratos Sprague-DawleyRESUMO
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.
Assuntos
Tendão do Calcâneo , Movimento Celular , Receptor gp130 de Citocina , Proteína Tirosina Fosfatase não Receptora Tipo 11 , Proteínas Proto-Oncogênicas c-akt , Ratos Sprague-Dawley , Tenócitos , Animais , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Receptor gp130 de Citocina/metabolismo , Tendão do Calcâneo/metabolismo , Tendão do Calcâneo/lesões , Tendão do Calcâneo/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Ratos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tenócitos/metabolismo , Tenócitos/efeitos dos fármacos , Tenócitos/fisiologia , Colágeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Masculino , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/fisiologia , Traumatismos dos Tendões/metabolismo , Traumatismos dos Tendões/tratamento farmacológicoRESUMO
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.
Assuntos
Ácido Hialurônico , Polietilenoglicóis , Alicerces Teciduais , Animais , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Coelhos , Polietilenoglicóis/química , Alicerces Teciduais/química , Tenócitos/efeitos dos fármacos , Tenócitos/metabolismo , Tendão do Calcâneo/efeitos dos fármacos , Traumatismos dos Tendões/terapia , Adesão Celular/efeitos dos fármacos , Aderências Teciduais/prevenção & controle , Tendões/efeitos dos fármacos , Engenharia Tecidual/métodos , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Poliésteres/química , PoliuretanosRESUMO
PURPOSE: Intra-articular administration of tranexamic acid (TXA) in orthopaedic arthroplasty and arthroscopic procedures has become increasingly common over the past decade. However, several recent reports have shown that TXA has the potential to be cytotoxic to cartilage, tendon and synovium. Our aim was to review the literature for evidence of toxic effects from TXA exposure to intra-articular tissue. METHODS: A scoping review methodology was used to search for studies assessing the toxic effects of TXA exposure to intra-articular tissues. MEDLINE, EMBASE, SCOPUS and The Cochrane Library were searched. Relevant information was extracted and synthesis of the retrieved data followed a basic content analytical approach. RESULTS: A total of 15 laboratory studies were retrieved. No clinical studies reporting a toxic effect of TXA on intra-articular tissue were identified in our search. Studies were analysed according to species of origin, tissue of origin and study setting (in vitro, ex vivo, or in vivo). There was increasing cytotoxicity to chondrocytes, tenocytes, synoviocytes and periosteum-derived cells with TXA concentrations beyond 20 mg/ml. Monolayer cell cultures appear more susceptible to TXA exposure, than three-dimensional and explant culture models. In vivo studies have not demonstrated a major toxic effect. CONCLUSIONS: Current evidence suggests a dose-dependent toxic effect on cartilage, tendon, and synovial tissue. Concentrations of 20 mg/ml or less are expected to be safe. There is a significant body of evidence to suggest the need for caution with intraarticular administration of TXA. There is a need for further human clinical trials in order to clarify the long-term safety of TXA topical application.
Assuntos
Antifibrinolíticos/efeitos adversos , Artroscopia , Condrócitos/efeitos dos fármacos , Sinoviócitos/efeitos dos fármacos , Tenócitos/efeitos dos fármacos , Ácido Tranexâmico/efeitos adversos , Antifibrinolíticos/administração & dosagem , Humanos , Injeções Intra-Articulares , Periósteo/efeitos dos fármacos , Ácido Tranexâmico/administração & dosagemRESUMO
Gradual wear and tear can cause a local inflammatory response in tendons. The trauma and inflammatory reaction eventually impair the biomechanical properties of the tendon. In this study, we prepared lactoferrin-immobilized, heparin-anchored, poly(lactic-co-glycolic acid) nanoparticles (LF/Hep-PLGA NPs) and evaluated their in vitro anti-inflammatory effects on interleukin-1ß (IL-1ß)-treated tenocytes and in vivo tendon healing effects in a rat model of Achilles tendinitis. Long-term LF-deliverable NPs (LF/Hep-PLGA NPs) remarkably decreased mRNA levels of pro-inflammatory factors [cyclooxygenase-2 (COX-2), IL-1ß, matrix metalloproteinase-3 (MMP-3), MMP-13, IL-6, and tumor necrosis factor-α (TNF-α)] and increased mRNA levels of anti-inflammatory cytokines (IL-4 and IL-10) in both IL-1ß-treated tenocytes and the Achilles tendons of a collagenase-induced Achilles tendinitis rat model. Interestingly, anti-inflammatory LF/Hep-PLGA NPs greatly enhanced collagen content, mRNA levels of tenogenic markers [collagen type I (COL1A1), decorin (DCN), tenascin-C (TNC)], and biomechanical properties such as tendon stiffness and tensile strength. These results suggest that anti-inflammatory LF/Hep-PLGA NPs are effective at restoring tendons in Achilles tendinitis.
Assuntos
Tendão do Calcâneo/efeitos dos fármacos , Anti-Inflamatórios/administração & dosagem , Heparina/administração & dosagem , Lactoferrina/administração & dosagem , Nanopartículas/administração & dosagem , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/administração & dosagem , Tendinopatia/tratamento farmacológico , Tendão do Calcâneo/metabolismo , Tendão do Calcâneo/patologia , Tendão do Calcâneo/fisiologia , Animais , Anti-Inflamatórios/química , Colágeno/metabolismo , Citocinas/genética , Modelos Animais de Doenças , Heparina/química , Lactoferrina/química , Masculino , Nanopartículas/química , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Ratos Sprague-Dawley , Tendinopatia/genética , Tendinopatia/metabolismo , Tendinopatia/patologia , Tenócitos/efeitos dos fármacos , Resistência à TraçãoRESUMO
Various therapeutic effects of mesenchymal stem cells (MSCs) have been reported. However, the rapid clearance of these cells in vivo, difficulties in identifying their therapeutic mechanism of action, and insufficient production levels remain to be resolved. We investigated whether a pioglitazone pre-treatment of MSCs (Pio-MSCs) would stimulate the proliferation of co-cultured tenocytes. Pioglitazone increased the proliferation of MSCs and enhanced the secretion of VEGF (vascular endothelial growth factor) and collagen in these cells. We then examined the effects of Pio-MSCs on tenocytes using an indirect transwell culture system. A significant increase in tenocyte proliferation and cell cycle progression was observed in these co-cultures. Significant increases were observed in wound scratch closure by tenocytes from a Pio-MSC co-culture. Pio-MSCs also enhanced the secretion of collagen from tenocytes. A higher mRNA level of collagen type 1 (Col 1) and type 3 (Col 3), scleraxis (Scx), and tenascin C (TnC) was found in the tenocytes in Pio-MSC co-cultures compared with monocultured cells or tenocytes cultured with non-treated MSCs. Our results indicate that pioglitazone enhances the therapeutic effects of MSCs on tendon repair.
Assuntos
Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Pioglitazona/farmacologia , Animais , Ciclo Celular/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Citometria de Fluxo , Immunoblotting , Masculino , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Tenócitos/efeitos dos fármacos , Tenócitos/metabolismoRESUMO
Corticosteroids are commonly used for pain control in rotator cuff tear. Deregulated NF-κB activation is a hallmark of chronic inflammatory diseases and has been responsible for the pathogenesis of rotator cuff tear. The Dexamethasone(DEXA) is a synthetic corticosteroid. The purpose of this study was to examine the exact effect of dexamethasone on NF-κB signaling in rotator cuff tear. We measured NF-κB expression in four groups: control, TNF-α-treated, DEXA-treated, and combined treatment with TNF-α and DEXA. Tenocytes were isolated from patients with rotator cuff tears and pre-incubated with TNF-α (10 ng/ml), DEXA (1 µM), or both of them for 10 min, 1 h, and 2 h. Expression of p65, p50, and p52 in the nuclei and cytosol was analyzed by western blotting and immunofluorescence imaging using confocal microscopy. We also evaluated nucleus/cytosol (N/C) ratios of p65, p50, and p52. In our study, the combined treatment with DEXA and TNF-α showed increased N/C ratios of p65, p50, and p52 compared with those in the TNF-α group at all time points. Additionally, in the DEXA group, N/C ratios of p65, p50, and p52 gradually increased from 10 min to 2 h. In conclusion, DEXA promoted the nuclear localization of p65, p50, and p52, but was not effective in inhibiting the inflammatory response of TNF-α-stimulated rotator cuff tear.
Assuntos
Dexametasona/farmacologia , NF-kappa B/metabolismo , Lesões do Manguito Rotador/metabolismo , Transdução de Sinais/efeitos dos fármacos , Tenócitos/efeitos dos fármacos , Fator de Necrose Tumoral alfa/farmacologia , Núcleo Celular/metabolismo , Células Cultivadas , Citosol/metabolismo , Humanos , Lesões do Manguito Rotador/patologia , Tenócitos/metabolismoRESUMO
BACKGROUND: Platelet rich plasma (PRP) is widely used in rotator cuff repairs but its effect on the healing process is unclear. Several cell culture studies on the effect of allogenic PRP have reported promising results but are not transferable to clinical practice. The aim of the present study is to assess the possible effect of autologous PRP on rotator cuff tendon cells. The amount of growth factors involved with tendon-bone healing (PDGF-AB, IGF-1, TGF-ß1, BMP-7 and -12) is quantified. METHODS: Rotator cuff tissue samples were obtained from (n = 24) patients grouped by age (>/< 65 years) and sex into four groups and cells were isolated and characterized. Later, autologous PRP preparations were obtained and the effect was analyzed by means of cell proliferation, collagen I synthesis and expression of collagen I and III. Furthermore, the PRPs were quantified for growth factor content by means of platelet-derived growth factor (PDGF-AB), insulin-like growth factor (IGF-1), transforming growth factor (TGF-ß1), as well as bone morphogenetic protein (BMP) -7 and - 12. RESULTS: Cell proliferation and absolute synthesis of collagen I were positively affected by PRP exposure compared to controls (p < 0.05), but expression and relative synthesis of collagen I (normalized to cell proliferation) were significantly reduced. PRP contained high amounts of IGF-1 and lower levels of TGF-ß1 and PDGF-AB. The amounts of BMP-7 and -12 were below the detection limits. CONCLUSIONS: PRP is a source of growth factors such involved with tendon-bone healing. PRP had an anabolic effect on the human rotator cuff tenocytes of the same individual in vitro by means of cell proliferation and absolute, but not relative collagen I synthesis. These results encourage further studies on clinical outcomes with more comparable standards in terms of preparation and application methods. LEVEL OF EVIDENCE: Controlled laboratory study.
Assuntos
Produtos Biológicos/farmacologia , Plasma Rico em Plaquetas , Lesões do Manguito Rotador/terapia , Manguito Rotador/efeitos dos fármacos , Tenócitos/efeitos dos fármacos , Adulto , Idoso , Artroscopia , Produtos Biológicos/uso terapêutico , Biópsia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Colágeno Tipo I/metabolismo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Cultura Primária de Células , Manguito Rotador/citologia , Manguito Rotador/patologia , Manguito Rotador/cirurgia , Tenócitos/metabolismo , Resultado do Tratamento , Cicatrização/efeitos dos fármacosRESUMO
Tendon rupture induces an inflammatory response characterized by release of pro-inflammatory cytokines and impaired tendon performance. This study sought to investigate the therapeutic effects of simvastatin-loaded porous microspheres (SIM/PMSs) on inflamed tenocytes in vitro and collagenase-induced Achilles tendinitis in vivo. The treatment of SIM/PMSs in lipopolysaccharide (LPS)-treated tenocytes reduced the mRNA expressions of pro-inflammatory cytokines (Matrix metalloproteinase-3 (MMP-3), cyclooxygenase-2 (COX-2), interleukin-6 (IL-6), and tumor necrosis factor-α (TNF-α)). In addition, the local injection of SIM/PMSs into the tendons of collagenase-induced Achilles tendinitis rat models suppressed pro-inflammatory cytokines (MMP-3, COX-2, IL-6, TNF-α, and MMP-13). This local treatment also upregulated anti-inflammatory cytokines (IL-4, IL-10, and IL-13). Furthermore, treatment with SIM/PMSs also improved the alignment of collagen fibrils and effectively prevented collagen disruption in a dose-dependent manner. Therefore, SIM/PMSs treatment resulted in an incremental increase in the collagen content, stiffness, and tensile strength in tendons. This study suggests that SIM/PMSs have great potential for tendon healing and restoration in Achilles tendinitis.
Assuntos
Anti-Inflamatórios/farmacologia , Microesferas , Sinvastatina/farmacologia , Tendinopatia/tratamento farmacológico , Tenócitos/efeitos dos fármacos , Tendão do Calcâneo/patologia , Animais , Anti-Inflamatórios/administração & dosagem , Células Cultivadas , Colágeno/genética , Colágeno/metabolismo , Colagenases/toxicidade , Ciclo-Oxigenase 2/genética , Ciclo-Oxigenase 2/metabolismo , Interleucinas/genética , Interleucinas/metabolismo , Lipopolissacarídeos/toxicidade , Metaloproteinases da Matriz/genética , Metaloproteinases da Matriz/metabolismo , Ratos , Ratos Sprague-Dawley , Sinvastatina/administração & dosagem , Tendinopatia/etiologia , Tenócitos/metabolismo , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismoRESUMO
Diabetes mellitus is associated with damage to tendons, which may result from cellular dysfunction in response to a hyperglycemic environment. Tenocytes express diminished levels of tendon-associated genes under hyperglycemic conditions. In contrast, mechanical stretch enhances tenogenic differentiation. However, whether hyperglycemia increases the non-tenogenic differentiation potential of tenocytes and whether this can be mitigated by mechanical stretch remains elusive. We explored the in vitro effects of high glucose and mechanical stretch on rat primary tenocytes. Specifically, non-tenogenic gene expression, adipogenic potential, cell migration rate, filamentous actin expression, and the activation of signaling pathways were analyzed in tenocytes treated with high glucose, followed by the presence or absence of mechanical stretch. We analyzed tenocyte phenotype in vivo by immunohistochemistry using an STZ (streptozotocin)-induced long-term diabetic mouse model. High glucose-treated tenocytes expressed higher levels of the adipogenic transcription factors PPARγ and C/EBPs. PPARγ was also highly expressed in diabetic tendons. In addition, increased adipogenic differentiation and decreased cell migration induced by high glucose implicated a fibroblast-to-adipocyte phenotypic change. By applying mechanical stretch to tenocytes in high-glucose conditions, adipogenic differentiation was repressed, while cell motility was enhanced, and fibroblastic morphology and gene expression profiles were strengthened. In part, these effects resulted from a stretch-induced activation of ERK (extracellular signal-regulated kinases) and a concomitant inactivation of Akt. Our results show that mechanical stretch alleviates the augmented adipogenic transdifferentiation potential of high glucose-treated tenocytes and helps maintain their fibroblastic characteristics. The alterations induced by high glucose highlight possible pathological mechanisms for diabetic tendinopathy. Furthermore, the beneficial effects of mechanical stretch on tenocytes suggest that an appropriate physical load possesses therapeutic potential for diabetic tendinopathy.
Assuntos
Adipócitos/efeitos dos fármacos , Diabetes Mellitus Experimental/terapia , Glucose/farmacologia , Mecanotransdução Celular/genética , Estresse Mecânico , Tenócitos/efeitos dos fármacos , Adipócitos/metabolismo , Adipócitos/patologia , Adipogenia/efeitos dos fármacos , Adipogenia/genética , Animais , Fenômenos Biomecânicos , Proteína beta Intensificadora de Ligação a CCAAT/genética , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Transdiferenciação Celular/efeitos dos fármacos , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/genética , Diabetes Mellitus Experimental/patologia , Regulação da Expressão Gênica , Masculino , Camundongos , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , PPAR gama/genética , PPAR gama/metabolismo , Cultura Primária de Células , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Estreptozocina , Tendões/efeitos dos fármacos , Tendões/metabolismo , Tendões/patologia , Tenócitos/metabolismo , Tenócitos/patologiaRESUMO
BACKGROUND: Local injections of anesthetics, NSAIDs, and corticosteroids for tendinopathies are empirically used. They are believed to have some cytotoxicity toward tenocytes. The maximal efficacy dosages of local injections should be determined. A commercial 2D microfluidic xCELLigence system had been developed to detect real-time cellular proliferation and their responses to different stimuli and had been used in several biomedical applications. The purpose of this study is to determine if human tenocytes can successfully proliferate inside xCELLigence system and the result has high correlation with conventional cell culture methods in the same condition. METHODS: First passage of human tenocytes was seeded in xCELLigence and conventional 24-well plates. Ketorolac tromethamine, bupivacaine, methylprednisolone, and betamethasone with different concentrations (100, 50, and 10% diluted of clinical usage) were exposed in both systems. Gene expression of type I collagen, type III collagen, tenascin-C, decorin, and scleraxis were compared between two systems. RESULTS: Human tenocytes could proliferate both in xCELLigence and conventional cell culture systems. Cytotoxicity of each drug revealed dose-dependency when exposed to tenocytes in both systems. Significance was found between groups. All the four drugs had comparable cytotoxicity in their 100% concentration. When 50% concentration was used, betamethasone had a relatively decreased cytotoxicity among them in xCELLigence but not in conventional culture. When 10% concentration was used, betamethasone had the least cytotoxicity. Strong and positive correlation was found between cell index of xCELLigence and result of WST-1 assay (Pearson's correlation [r] = 0.914). Positive correlation of gene expression between tenocytes in xCELLigence and conventional culture was also observed. Type I collagen: [r] = 0.823; type III collagen: [r] = 0.899; tenascin-C: [r] = 0.917; decorin: [r] = 0.874; and scleraxis: [r] = 0.965. CONCLUSIONS: Human tenocytes could proliferate inside xCELLigence system. These responses varied when tenocytes were exposed to different concentrations of ketorolac tromethamine, bupivacaine, methylprednisolone, and betamethasone. The result of cell proliferation and gene expression of tenocytes in both xCELLigence and conventional culture system is strongly correlated. CLINICAL RELEVANCE: xCELLigence culture system may replace conventional cell culture, which made real-time tenocyte proliferation monitoring possible.
Assuntos
Técnicas Biossensoriais/métodos , Técnicas de Cultura de Células/métodos , Proliferação de Células/efeitos dos fármacos , Sistemas Computacionais , Técnicas Analíticas Microfluídicas/métodos , Tenócitos/efeitos dos fármacos , Corticosteroides/toxicidade , Anestésicos/toxicidade , Anti-Inflamatórios não Esteroides/toxicidade , Proliferação de Células/fisiologia , Testes Imunológicos de Citotoxicidade/métodos , Avaliação Pré-Clínica de Medicamentos/métodos , Feminino , Humanos , Pessoa de Meia-Idade , Tenócitos/fisiologiaRESUMO
Excessive mechanical loading is a major factor affecting heterotopic ossification (HO), which is a major pathological alteration in calcific tendinopathy. However, physical therapies with mechanical loading as the functional element have exhibited promising results in the treatment of calcific tendinopathy. The dual effects that mechanical loading may have on the pathogenesis and rehabilitation of calcified tendinopathy remain unclear. The present study was designed to investigate the effects of mechanical loading on HO in calcific tendinopathy. In the present study, a tendon cell in vitro stretch model and an Achilles tenotomy rat model were used to simulate different elongation mechanical loading scenarios in order to investigate the effects of mechanical loading on HO of the tendon. In addition, rapamycin, a selective mammalian target of rapamycin complex1 (mTORC1) signaling pathway inhibitor, was employed to determine whether mechanical loading modulates heterotopic ossification in calcific tendinopathy through the mTORC1 signaling pathway. The data indicate that mechanical loading modulated HO of the tendon through the mTORC1 signaling pathway, and that low elongation mechanical loading attenuated HO, while high elongation mechanical loading accelerated HO in vivo. This study may improve the understanding of the effect of physical therapies used to treat calcific tendinopathy, so as to guide clinical treatment more effectively. Furthermore, rapamycin may be a potential drug for the treatment of calcific tendinopathy.
Assuntos
Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Mecanotransdução Celular , Ossificação Heterotópica/metabolismo , Tendinopatia/metabolismo , Suporte de Carga/fisiologia , Tendão do Calcâneo/cirurgia , Animais , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/genética , Ossificação Heterotópica/genética , Ossificação Heterotópica/patologia , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Sirolimo/farmacologia , Tendinopatia/genética , Tendinopatia/patologia , Tenócitos/citologia , Tenócitos/efeitos dos fármacos , Tenócitos/metabolismo , Tenotomia/métodos , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismoRESUMO
One of the objectives of rotator cuff repairs is to achieve biological healing and recovery in the tendon-bone zone. Some clinical evaluations reported the feasibility of tendon healing based on the stimulations of electric field and platelet-rich plasma (PRP). However, because of lack of appropriate tool for in vitro primary culture under complicated conditions, the efficacy and standard protocol of these healing approaches are still controversial among clinical experts. In this study, a novel co-culture device was developed for the study of tenocytes proliferation under single and combined stimulations of electric field and PRP. The device was a culture well divided into three sub-chambers separated by a barrier and embedded with a pair of parallel plate electrodes. Tenocytes and PRP gel could be respectively loaded into the sub-chambers and cultured with interlinked medium. Hence, tenocytes could concurrently receive a uniform electric field and platelet-derived growth factors by diffusion. Results revealed that the proliferation of tenocytes could be significantly enhanced by these stimulations. The device provides a precise and practical approach for the in vitro study of tendon healing, especially for PRP study. Moreover, optimization of the conditions of electric field and PRP could be determined by in vitro screening procedure before surgery to provide a personalized therapy.
Assuntos
Técnicas de Cocultura/instrumentação , Estimulação Elétrica , Plasma Rico em Plaquetas/metabolismo , Tenócitos/citologia , Proliferação de Células/efeitos dos fármacos , Desenho de Equipamento , Humanos , Tenócitos/efeitos dos fármacosRESUMO
INTRODUCTION: The purpose of this study was to evaluate the effect of allogenic leukocyte-reduced platelet-rich plasma on human tenocytes after treatment with prednisolone and to develop a standardization of its application for clinical practice. METHODS: A leukocyte-reduced PRP was produced using the Arthrex Double Syringe (Arthrex, Inc., Naples, FL, USA), in a modified single-spin separation method. Human tenocytes were isolated from discarded rotator cuff segments. Tenocytes were cultured in the presence of PRP and prednisolone, both alone and in combination. Control samples were treated in media containing 2% FCS for 72 h. After 72 h of incubation, cell cycle kinetics of tenocytes were analyzed to assess proliferation. RESULTS: Incubation of the tenocytes with PRP alone for 48 h led to high proliferation rate (10% PRP, 28.0 ± 10.5%; 20% PRP, 40.9 ± 3.3%). Incubation in the presence of prednisolone led to a significant decrease of the proliferation rate (5.2 ± 3.1%; p < 0.05). Treatment with PRP for 48 h significantly increased the proliferation of tenocytes in a dose-dependent manner (10% PRP, 28.0 ± 10.5%; 20% PRP, 40.9 ± 3.3%; p < 0.05). The presence of prednisolone resulted in a decreased tenocyte proliferation (5.2 ± 3.1%; p < 0.05), whereas addition of PRP for 24 and 48 h after prednisolone exposure did not show any compensating effect independent of PRPs concentration (10% PRP, 3.7 ± 3.0%; 20% PRP, 2.5 ± 2.5%). However, a significantly increased cell proliferation of tenocytes was evident when PRP was applied 24 h after prednisolone incubation for 48 h (31.0 ± 3.4 and 34.3 ± 4.7%). CONCLUSION: The use of leukocyte-reduced PRP stimulates the proliferation of tenocytes and antagonizes the negative effect of prednisolone 24 h after treatment. Addition of PRP 48 h after treatment with prednisolone has no positive effect on the proliferation rate of tenocytes.
Assuntos
Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Plasma Rico em Plaquetas , Prednisolona/farmacologia , Tenócitos , Células Cultivadas , Humanos , Manguito Rotador/citologia , Tenócitos/citologia , Tenócitos/efeitos dos fármacosRESUMO
PURPOSE: To analyze the ability of ropivacaine, bupivacaine, and triamcinolone to induce apoptosis and necrosis in fibroblasts, tenocytes, and human mesenchymal stem cells. METHODS: Human dermal fibroblasts, adipose-derived human mesenchymal stem cells (hMSCs), and tenocytes gained from the rotator cuff tendon were seeded with a cell density of 0.5 × 104/cm2. One specimen of ropivacaine, bupivacaine, and triamcinolone was tested separately on the cells with separate concentrations of 0.5%, 0.25%, and 0.125% for each specimen. The negative control received no agent, only a change of medium. The incubation period for each agent was 30 minutes. After a change of medium and 1 hour, 24 hours, and 7 days of incubation, 104 cells were harvested and analyzed via fluorescence-activated cell sorting with double-staining with annexin V and propidium iodide. Statistical analysis to determine significant difference (P < .05) between the groups with SPSS statistics 23 through one-way analysis of variance with a univariate general linear model was performed. RESULTS: Bupivacaine showed necrosis-inducing effects on fibroblasts and tenocytes, with the necrotic effect peaking at 0.5% and 0.25%. Ropivacaine and triamcinolone caused no significant necrosis. Compared with fibroblasts and tenocytes, hMSCs did not show significant necrotic or apoptotic effects after exposure to bupivacaine. Overall, no significant differences in apoptosis were detected between different cell lines, varying concentrations, or time measurements. CONCLUSIONS: Bupivacaine 0.5% and 0.25% have the most necrosis-inducing effects on fibroblasts and tenocytes. Ropivacaine caused less necrosis than bupivaine. Compared with fibroblasts and tenocytes, hMSCs were not affected by necrosis using any of the tested agents. A significant apoptosis-inducing effect could not be detected for the different cell lines. CLINICAL RELEVANCE: Possible cell toxicity raises questions of concern for intra-articular injections using local anesthetics and corticosteroids. The present study demonstrates the necrotic and apoptotic effects of ropivacaine, bupivacaine, and triamcinolone and may give recommendations for intra-articular use of local anesthetics and corticosteroids.
Assuntos
Amidas/toxicidade , Bupivacaína/toxicidade , Fibroblastos/efeitos dos fármacos , Células-Tronco Mesenquimais/efeitos dos fármacos , Tenócitos/efeitos dos fármacos , Triancinolona/toxicidade , Adulto , Amidas/administração & dosagem , Anestésicos Locais/farmacologia , Apoptose/efeitos dos fármacos , Bupivacaína/administração & dosagem , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Fibroblastos/patologia , Citometria de Fluxo , Glucocorticoides/administração & dosagem , Glucocorticoides/toxicidade , Humanos , Células-Tronco Mesenquimais/patologia , Necrose , Ropivacaina , Manguito Rotador/citologia , Pele/citologia , Tenócitos/patologia , Triancinolona/administração & dosagemRESUMO
Scar formation after filtration surgery of glaucoma is mainly caused by excessive synthesis of new extracellular matrix (ECM) and contraction of subconjunctival tissue mediated by human Tenon fibroblasts (HTFs) and the transforming growth factor (TGF-ß1). Montelukast, a potent and specific cysteinyl leukotriene receptor 1 (cysLT1R) antagonist, is a licensed drug clinically used for the treatment of bronchial asthma. In this study, we investigated the effects of montelukast on the contractility of HTFs cultured in a three-dimensional collagen gel. We found that cysLT1R was expressed in HTFs. Interestingly, the expression of cysLT1R was increased in response to TGF-ß1 in a dose dependent manner, suggesting its potential role in TGF-ß1 induced fibrosis. Importantly, we found that montelukast inhibited TGF-ß1-induced collagen gel contraction mediated by HTFs in a concentration- and time-dependent manner. In addition, TGF-ß1-induced expression of MMP-1 and MMP-3, generation of fibronectin and type I collagen production, focal adhesion kinase (FAK) and paxillin phosphorylation in HTFs were also ameliorated by montelukast in a dose dependent manner. These results suggested that montelukast might provide therapeutic possibilities for inhibition of scar formation after such surgery.
Assuntos
Acetatos/administração & dosagem , Proteínas da Matriz Extracelular/metabolismo , Matriz Extracelular/metabolismo , Matriz Extracelular/patologia , Antagonistas de Leucotrienos/administração & dosagem , Quinolinas/administração & dosagem , Receptores de Leucotrienos/efeitos dos fármacos , Tenócitos/metabolismo , Células Cultivadas , Ciclopropanos , Relação Dose-Resposta a Droga , Matriz Extracelular/efeitos dos fármacos , Humanos , Receptores de Leucotrienos/metabolismo , Sulfetos , Tenócitos/efeitos dos fármacos , Cicatrização/efeitos dos fármacos , Cicatrização/fisiologiaRESUMO
BACKGROUND: Although there are many studies discussing the etiological and pathological factors leading to both, acute and chronic tendon injuries, the pathophysiology of tendon injuries is still not clearly understood. Although most lesions are uncomplicated, treatment is long and unsatisfactory due to the poor vascularity of tendon tissue. Platelet mediator concentrate (PMC) contains many growth factors derived from platelets, which can promote wound healing. In this study we investigate the effects of PMC on tenocyte proliferation and differentiation in order to provide an experimental basis for tissue regeneration strategies and to develop new treatment concepts. METHODS: Using enzyme linked immunosorbent assay (ELISA) we were able to quantify the several growth factors and cytokines found in PMC. Tenocytes were isolated both from human and from mouse Achilles tendons and stimulated with PMC. CyQuant® and Cell Titer Blue® assays were carried out to analyze tendon growth and viability at different concentrations of PMC. Real time RT-PCR was used to analyze tenocyte gene expression with or without PMC treatment. Immunohistochemistry was carried out to detect the tenocyte-specific antibody tenomodulin (TNMD) and scleraxis (SCX). RESULTS: We were able to detect numerous mediators such as platelet derived growth factor BB (PDGF-BB), interleukin 6 (IL-6), vascular endothelial growth factor (VEGF), tumor necrosis factor (TNF-α), transforming growth factor beta 1 (TGF-ß1), and bone morphogenetic proteins 2, 4 and 7 (BMP-4, BMP-2, BMP-7) in PMC. It was possible to show a positive effect of PMC on human tendon cell growth and viability in a dose-dependent manner. Furthermore, PMC treatment led to induction of gene expression of scleraxis (SCX), type I collagen A 1 (Col1A1) and TNMD by tenocytes. CONCLUSIONS: We suggest that the use of autologous PMC may be a suitable addition to conventional tendon therapy that is capable of increasing and optimizing tendon healing and reducing the risk of recurrence.