Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
1.
Heliyon ; 10(14): e34192, 2024 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-39100446

RESUMEN

Objective: To explore the mechanism and efficacy of gel in the treatment of posttraumatic osteoarthritis (PTOA), combined with hyaluronic acid (HA) and bone marrow mesenchymal stem cell exosomes (BMSC-EXOs), and to explain its role in alleviating oxidative stress damage induced by mitochondrial reactive oxygen species (ROS). Methods: How is the therapeutic potential of toa influenced by bone marrow mesenchymal stem cells-EXO to be evaluated both in vitro and in vivo. In vitro, BMSC-EXOs were extracted and characterized from rat specimens and labeled with Dil. Rat primary chondrocytes were then isolated to create a cellular PTOA model. BMSC-EXOs + HA group, BMSC-EXOs + HA + 740Y-P group, model group, BMSC-EXOs group, HA group, and control group were included in the cell group, and the function of cartilage matrix and the level of oxidative stress could be evaluated. Cartilage matrix integrity and oxidative stress can be assessed by grouping rats. At the same time, a rat model of ptosis can be established by excision of the anterior cruciate ligament, and joint rehabilitation, with pro-inflammatory and Enzyme-linked immunosorbent assay (ELISA) can be used to determine anti-inflammatory markers. Result: sThe combined use of BMSC-EXOs and HA gel was found to significantly reduce oxidative stress in chondrocytes and PTOA rat models, improving cartilage mechanical properties more effectively than BMSC-EXOs alone. Conclusion: BMSC-EXOs combined with HA gel offer a promising treatment for PTOA by modulating damage caused by mitochondrial ROS-induced oxidative stress.

2.
Injury ; 55(10): 111766, 2024 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-39106534

RESUMEN

BACKGROUND: The sustentaculum tali is a biomechanically important stabilizer of the hindfoot and contributes to articular congruency of the subtalar joint. Sustentaculum injury associated with a talus fracture has been described infrequently and treatment of this combined injury varies. The purpose of this study was to describe and evaluate the outcomes of the combined talus and sustentaculum fracture. METHODS: Retrospective chart and radiographic review was performed on all talus fractures (n = 436) requiring operative fixation over a 21-year period at a single Level-1 trauma center. All talus fractures with sustentaculum fractures were included. Statistical analysis was performed using Chi-squared and Fishers exact tests where appropriate. RESULTS: Sustentaculum fractures occurred in 6.2 % (n = 27) of patients with talus fractures. Average follow-up was 14 months; 18.5 % were open fractures, 88.8 % were from high-energy mechanisms, and 44.4 % were polytraumas. Diagnosis of the sustentaculum fracture was missed on presenting radiographs in 69.2 % (n = 18). The most common associated talus fracture was a talar neck fracture (40.7 %) and the majority (73.7 %) were Hawkins II. Overall, 40.7 % (n = 11) of the sustentaculum fractures were treated with independent fixation and 7.4 % (n = 2) were treated with acute subtalar arthrodesis. Subtalar post-traumatic osteoarthritis (PTOA) at final follow-up was seen in 23.1 % of combined injuries. Independent sustentaculum fixation did not influence the rate of PTOA or re-operation (p = 0.92, p = 0.91, respectively). CONCLUSION: Talar fractures have an associated sustentaculum fracture in approximately 6 % of cases, especially with Hawkins II fracture-dislocations. Over two-thirds of the associated sustentaculum fractures were missed on presenting radiographs, reiterating the need for heightened awareness and consideration of advanced imaging for all talus fractures. The rate of PTOA following these combined injuries at mean follow-up of 24 months does not exceed established rates after isolated talus fractures. Further research is required to determine the optimal management of the sustentaculum in these combined injuries. LEVEL OF EVIDENCE: IV.

4.
Orthop Clin North Am ; 55(4): 461-469, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39216951

RESUMEN

Post-traumatic osteoarthritis (PTOA) occurs following injury to joints. It accounts for approximately 12% of osteoarthritis and has far-reaching effects on individual patients and social/health care systems. Present work focuses on evaluating the role of the post-traumatic inflammatory response in the development and progression of the disease. As there is minimal evidence to suggest the capacity of cartilage to undergo self-healing, most of this work focuses strictly on the avoidance or prevention of PTOA as opposed to treatment solutions following its onset. Ongoing and future investigations may reveal therapeutic targets for possible intervention and ultimately the prophylaxis of PTOA development.


Asunto(s)
Traumatismos del Tobillo , Traumatismos de la Rodilla , Osteoartritis de la Rodilla , Humanos , Traumatismos de la Rodilla/complicaciones , Traumatismos del Tobillo/complicaciones , Traumatismos del Tobillo/terapia , Osteoartritis de la Rodilla/etiología , Osteoartritis/etiología
5.
Bioact Mater ; 37: 378-392, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38689658

RESUMEN

Posttraumatic osteoarthritis (PTOA) patients are often diagnosed by X-ray imaging at a middle-late stage when drug interventions are less effective. Early PTOA is characterized by overexpressed matrix metalloprotease 13 (MMP13). Herein, we constructed an integrated diagnosis and treatment micelle modified with MMP13 enzyme-detachable, cyanine 5 (Cy5)-containing PEG, black hole quencher-3 (BHQ3), and cRGD ligands and loaded with siRNA silencing MMP13 (siM13), namely ERMs@siM13. ERMs@siM13 could be cleaved by MMP13 in the diseased cartilage tissues to detach the PEG shell, causing cRGD exposure. Accordingly, the ligand exposure promoted micelle uptake by the diseased chondrocytes by binding to cell surface αvß3 integrin, increasing intracellular siM13 delivery for on-demand MMP13 downregulation. Meanwhile, the Cy5 fluorescence was restored by detaching from the BHQ3-containing micelle, precisely reflecting the diseased cartilage state. In particular, the intensity of Cy5 fluorescence generated by ERMs@siM13 that hinged on the MMP13 levels could reflect the PTOA severity, enabling the physicians to adjust the therapeutic regimen. Finally, in the murine PTOA model, ERMs@siM13 could diagnose the early-stage PTOA, perform timely interventions, and monitor the OA progression level during treatment through a real-time detection of MMP13. Therefore, ERMs@siM13 represents an appealing approach for early-stage PTOA theranostics.

6.
Osteoarthritis Cartilage ; 32(6): 680-689, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38432607

RESUMEN

OBJECTIVE: Phlpp1 inhibition is a potential therapeutic strategy for cartilage regeneration and prevention of post-traumatic osteoarthritis (PTOA). To understand how Phlpp1 loss affects cartilage structure, cartilage elastic modulus was measured with atomic force microscopy (AFM) in male and female mice after injury. METHODS: Osteoarthritis was induced in male and female Wildtype (WT) and Phlpp1-/- mice by destabilization of the medial meniscus (DMM). At various timepoints post-injury, activity was measured, and knee joints examined with AFM and histology. In another cohort of WT mice, the PHLPP inhibitor NSC117079 was intra-articularly injected 4 weeks after injury. RESULTS: Male WT mice showed decreased activity and histological signs of cartilage damage at 12 but not 6-weeks post-DMM. Female mice showed a less severe response to DMM by comparison, with no histological changes seen at any time point. In both sexes the elastic modulus of medial condylar cartilage was decreased in WT mice but not Phlpp1-/- mice after DMM as measured by AFM. By 6-weeks, cartilage modulus had decreased from 2 MPa to 1 MPa in WT mice. Phlpp1-/- mice showed no change in modulus at 6-weeks and only a 25% decrease at 12-weeks. The PHLPP inhibitor NSC117079 protected cartilage structure and prevented signs of OA 6-weeks post-injury. CONCLUSIONS: AFM is a sensitive method for detecting early changes in articular cartilage post-injury. Phlpp1 suppression, either through genetic deletion or pharmacological inhibition, protects cartilage degradation in a model of PTOA, validating Phlpp1 as a therapeutic target for PTOA.


Asunto(s)
Cartílago Articular , Fosfoproteínas Fosfatasas , Animales , Cartílago Articular/patología , Cartílago Articular/efectos de los fármacos , Fosfoproteínas Fosfatasas/antagonistas & inhibidores , Fosfoproteínas Fosfatasas/genética , Masculino , Femenino , Ratones , Modelos Animales de Enfermedad , Proteínas Nucleares/genética , Proteínas Nucleares/antagonistas & inhibidores , Ratones Noqueados , Microscopía de Fuerza Atómica , Osteoartritis/patología , Módulo de Elasticidad , Osteoartritis de la Rodilla/etiología , Osteoartritis de la Rodilla/patología , Lesiones de Menisco Tibial/complicaciones
7.
J Hand Surg Am ; 49(8): 757-765, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38180412

RESUMEN

PURPOSE: The aim of this prospective, randomized, controlled, double-blinded pilot study was to determine the rates of post-traumatic osteoarthritis and assess joint space width in the presence or absence of a single intra-articular injection of corticosteroid after an acute, intra-articular distal radius fracture (DRF). METHODS: Forty patients received a single, intra-articular, radiocarpal joint injection of 4 mg of dexamethasone (DEX) (n = 19) or normal saline placebo (n = 21) within 2 weeks of a surgically or nonsurgically treated intra-articular DRF. The primary outcome measure was minimum radiocarpal joint space width (mJSW) on noncontrast computed tomography scans at 2 years postinjection. Secondary outcomes were obtained at 3 months, 6 months, 1 year, and 2 years postinjection and included Disabilities of the Arm, Shoulder, and Hand; Michigan Hand Questionnaire; Patient-Rated Wrist Evaluation; wrist range of motion; and grip strength. RESULTS: At 2-year follow-up, there was no difference in mean mJSW between the DEX group (2.2 mm; standard deviation, 0.6; range, 1.4-3.2) and the placebo group (2.3 mm; standard deviation, 0.7; range, 0.9-3.9). Further, there were no differences in any secondary outcome measures at any postinjection follow-up interval. CONCLUSIONS: Radiocarpal joint injection of corticosteroid within 2 weeks of an intra-articular DRF does not appear to affect the development of post-traumatic osteoarthritis within 2 years follow-up in a small pilot cohort. TYPE OF STUDY/LEVEL OF EVIDENCE: Therapeutic II.


Asunto(s)
Dexametasona , Glucocorticoides , Osteoartritis , Fracturas del Radio , Fracturas de la Muñeca , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Dexametasona/administración & dosificación , Dexametasona/uso terapéutico , Método Doble Ciego , Glucocorticoides/administración & dosificación , Fuerza de la Mano , Inyecciones Intraarticulares , Fracturas Intraarticulares/complicaciones , Fracturas Intraarticulares/diagnóstico por imagen , Fracturas Intraarticulares/tratamiento farmacológico , Osteoartritis/etiología , Osteoartritis/prevención & control , Proyectos Piloto , Estudios Prospectivos , Fracturas del Radio/complicaciones , Fracturas del Radio/diagnóstico por imagen , Fracturas del Radio/tratamiento farmacológico , Rango del Movimiento Articular , Tomografía Computarizada por Rayos X , Resultado del Tratamiento , Fracturas de la Muñeca/complicaciones , Fracturas de la Muñeca/diagnóstico por imagen , Fracturas de la Muñeca/tratamiento farmacológico , Articulación de la Muñeca
8.
J Athl Train ; 2024 Jan 20.
Artículo en Inglés | MEDLINE | ID: mdl-38243733

RESUMEN

CONTEXT: Early identification of knee osteoarthritis (OA) symptoms after anterior cruciate ligament reconstruction (ACLR) could enable timely interventions to improve long-term outcomes. However, little is known about the change in early OA symptoms from 6 to 12 months following ACLR. OBJECTIVE: To evaluate the change over time in meeting classification criteria for early knee OA symptoms from 6 to 12 months following ACLR. DESIGN: Prospective Cohort Study. SETTING: Research laboratory. PATIENTS OR OTHER PARTICIPANTS: 82 participants aged 13-35 years who underwent unilateral primary ACLR. On average, participants' 1st and 2nd visits were 6.2 and 12.1 months post-ACLR. MAIN OUTCOME MEASURES: Early OA symptoms were classified using generic (Luyten Original) and patient population-specific (Luyten PASS) thresholds on Knee Injury and Osteoarthritis Outcome Score (KOOS) subscales. Changes in meeting early OA criteria were compared between an initial and follow-up visit at an average of 6 and 12 months post-ACLR, respectively. RESULTS: Twenty-two percent of participants exhibited persistent early OA symptoms across both visits using both the Luyten Original and PASS criteria. From initial to follow-up visit, 18-27% had resolution of early OA symptoms while 4-9% developed incident symptoms. In total, 48-51% had no early OA symptoms at either visit. There were no differences between change in early OA status between adults and adolescents. CONCLUSIONS: Nearly one quarter of participants exhibited persistent early knee OA symptoms based on KOOS thresholds from 6 to 12 months post-ACLR. Determining if this symptom persistence predicts worse long-term outcomes could inform the need for timely interventions after ACLR. Future research should examine if resolving persistent symptoms in this critical window improves later outcomes. Tracking early OA symptoms over time may identify high-risk patients who could benefit from early treatment.

9.
Osteoarthritis Cartilage ; 31(12): 1554-1566, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37742942

RESUMEN

OBJECTIVE: There is no disease-modifying treatment for posttraumatic osteoarthritis (PTOA). This may be partly due to an incomplete understanding of synovitis, which has been causally linked to PTOA progression. The microscopic and transcriptomic changes in synovium seen in early- to mid-stage PTOA were evaluated to better characterize this knowledge gap. METHODS: Seventy-two Yucatan minipigs underwent transection of the anterior cruciate ligament (ACL). Subjects were randomized to no further intervention, ligament reconstruction, or ligament repair, followed by microscopic synovium evaluation and RNA-sequencing at 1, 4, and 52 weeks. Six additional subjects received no ligament transection and served as 1- and 4-week controls and 12 contralateral knees served as 52-week controls. RESULTS: Synovial lining thickness, stromal cellularity, and overall microscopic synovitis reached their highest levels in the first few weeks following injury. Inflammatory infiltration continued to increase over the course of a year. Leaving the ACL transected, reconstructing the ligament, or repairing the ligament did not modulate synovitis development at 1, 4, or 52 weeks. Differential gene expression analysis of PTOA-affected synovium compared to control synovium revealed increased cell proliferation, angiogenesis, collagen breakdown, and diminished lipid metabolism at 1 and 4 weeks, and increased axonogenesis and focal adhesion with reduced immune activation at 52 weeks. CONCLUSIONS: Synovitis was present one year after ACL injury and was not alleviated by surgical intervention. Gene expression in early synovitis was characterized by cell proliferation, angiogenesis, proteolysis, and reduced lipolysis, which was followed by nerve growth and cellular adhesion with less immune activation at 52 weeks.


Asunto(s)
Lesiones del Ligamento Cruzado Anterior , Osteoartritis , Sinovitis , Animales , Ligamento Cruzado Anterior/cirugía , Lesiones del Ligamento Cruzado Anterior/cirugía , Perfilación de la Expresión Génica , Osteoartritis/metabolismo , Porcinos , Porcinos Enanos/genética , Membrana Sinovial/metabolismo , Sinovitis/metabolismo , Transcriptoma
10.
Bioengineering (Basel) ; 10(9)2023 Sep 06.
Artículo en Inglés | MEDLINE | ID: mdl-37760151

RESUMEN

In regenerative medicine, experimental animal models are commonly used to study potential effects of human cells as therapeutic candidates. Although some studies describe certain cells, such as mesenchymal stromal cells (MSC) or human primary cells, as hypoimmunogenic and therefore unable to trigger strong inflammatory host responses, other studies report antibody formation and immune rejection following xenotransplantation. Accordingly, the goal of our study was to test the cellular retention and survival of human-induced pluripotent stem cell (iPSCs)-derived MSCs (iMSCs) and primary nucleus pulposus cells (NPCs) following their xenotransplantation into immune-privileged knee joints (14 days) and intervertebral discs (IVD; 7 days) of immunocompromised Nude and immunocompetent Sprague Dawley (SD) rats. At the end of both experiments, we could demonstrate that both rat types revealed comparably low levels of systemic IL-6 and IgM inflammation markers, as assessed via ELISA. Furthermore, the number of recovered cells was with no significant difference between both rat types. Conclusively, our results show that xenogeneic injection of human iMSC and NPC into immunoprivileged knee and IVD sites did not lead to an elevated inflammatory response in immunocompetent rats when compared to immunocompromised rats. Hence, immunocompetent rats represent suitable animals for xenotransplantation studies targeting immunoprivileged sites.

11.
Life (Basel) ; 13(7)2023 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-37511942

RESUMEN

BACKGROUND: Senescence, a characteristic of cellular aging and inflammation, has been linked to the acceleration of osteoarthritis. The purpose of this study is to prospectively identify, measure, and compare senescent profiles in synovial fluid and peripheral blood in patients with an acute knee injury within 48 h. METHODS: Seven subjects, aged 18-60 years, with an acute ACL tear with effusion were prospectively enrolled. Synovial fluid and peripheral blood samples were collected and analyzed by flow cytometry, using senescent markers C12FDG and CD87. The senescent versus pro-regenerative phenotype was probed at a gene and protein level using qRT-PCR and multiplex immunoassays. RESULTS: C12FDG and CD87 positive senescent cells were detected in the synovial fluid and peripheral blood of all patients. Pro-inflammatory IL-1ß gene expression measured in synovial fluid was significantly higher (p = 0.0156) than systemic/blood expression. Senescent-associated factor MMP-3 and regenerative factor TIMP-2 were significantly higher in synovial fluid compared to blood serum. Senescent-associated factor MMP-9 and regenerative factor TGFß-2 were significantly elevated in serum compared to synovial fluid. Correlation analysis revealed that C12FDG++/CD87++ senescent cells in synovial fluid positively correlated with age-related growth-regulated-oncogene (ρ = 1.00, p < 0.001), IFNγ (ρ = 1.00, p < 0.001), IL-8 (ρ = 0.90, p = 0.0374), and gene marker p16 (ρ = 0.83, p = 0.0416). CONCLUSIONS: There is an abundance of senescent cells locally and systemically after an acute ACL tear without a significant difference between those present in peripheral blood compared to synovial fluid. This preliminary data may have a role in identifying strategies to modify the acute environment within the synovial fluid, either at the time of acute ligament injury or reconstruction surgery.

12.
Foot Ankle Int ; 44(9): 922-930, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37329280

RESUMEN

BACKGROUND: The first stage of fracture healing consists of hematoma formation with recruitment of proinflammatory cytokines and matrix metalloproteinases. Unfortunately, when there is an intra-articular fracture, these inflammatory mediators are not retained at the fracture site, but instead, envelop the healthy cartilage of the entire joint via the synovial fluid fracture hematoma (SFFH). These inflammatory cytokines and matrix metalloproteinases are known factors in the progression of osteoarthritis and rheumatoid arthritis. Despite the known inflammatory contents of the SFFH, little research has been done on the effects of the SFFH on healthy cartilage with regard to cell death and alteration in gene expression that could lead to posttraumatic osteoarthritis (PTOA). METHODS: SFFH was collected from 12 patients with intraarticular ankle fracture at the time of surgery. Separately, C20A4 immortalized human chondrocytes were 3-dimensionally cultured to create scaffold-free cartilage tissue analogs (CTAs) to simulate healthy cartilage. Experimental CTAs (n = 12) were exposed to 100% SFFH for 3 days, washed, and transferred to complete media for 3 days. Control CTAs (n = 12) were simultaneously cultured in complete medium without exposure to SFFH. Subsequently, CTAs were harvested and underwent biochemical, histological, and gene expression analysis. RESULTS: Exposure of CTAs to ankle SFFH for 3 days significantly decreased chondrocyte viability by 34% (P = .027). Gene expression of both COL2A1 and SOX9 were significantly decreased after exposure to SFFH (P = .012 and P = .0013 respectively), while there was no difference in COL1A1, RUNX2, and MMP13 gene expression. Quantitative analysis of Picrosirius red staining demonstrated increased collagen I deposition with poor ultrastructural organization in SFFH-exposed CTAs. CONCLUSION: Exposure of an organoid model of healthy cartilage tissue to SFFH after intraarticular ankle fracture resulted in decreased chondrocyte viability, decreased expression of genes regulating normal chondrocyte phenotype, and altered matrix ultrastructure indicating differentiation toward an osteoarthritis phenotype. CLINICAL RELEVANCE: The majority of ankle fracture open reduction and internal fixation does not occur immediately after fracture. In fact, typically these fractures are treated several days to weeks later in order to let the swelling subside. This means that the healthy innocent bystander cartilage not involved in the fracture is exposed to SFFH during this time. In this study, the SFFH caused decreased chondrocyte viability and specific altered gene expression that might have the potential to induce osteoarthritis. These data suggest that early intervention after intraarticular ankle fracture could possibly mitigate progression toward PTOA.


Asunto(s)
Fracturas de Tobillo , Cartílago Articular , Fracturas Intraarticulares , Osteoartritis , Humanos , Líquido Sinovial/metabolismo , Fracturas de Tobillo/cirugía , Condrocitos , Citocinas/análisis , Osteoartritis/tratamiento farmacológico , Fracturas Intraarticulares/cirugía , Cartílago Articular/patología , Metaloproteinasas de la Matriz/análisis , Expresión Génica
13.
Bioengineering (Basel) ; 10(5)2023 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-37237597

RESUMEN

The relationship between cartilage and synovium is a rapidly growing area of osteoarthritis research. However, to the best of our knowledge, the relationships in gene expression between these two tissues have not been explored in mid-stage disease development. The current study compared the transcriptomes of these two tissues in a large animal model one year following posttraumatic osteoarthritis induction and multiple surgical treatment modalities. Thirty-six Yucatan minipigs underwent transection of the anterior cruciate ligament. Subjects were randomized to no further intervention, ligament reconstruction, or ligament repair augmented with an extracellular matrix (ECM) scaffold, followed by RNA sequencing of the articular cartilage and synovium at 52 weeks after harvest. Twelve intact contralateral knees served as controls. Across all treatment modalities, the primary difference in the transcriptomes was that the articular cartilage had greater upregulation of genes related to immune activation compared to the synovium-once baseline differences between cartilage and synovium were adjusted for. Oppositely, synovium featured greater upregulation of genes related to Wnt signaling compared to articular cartilage. After adjusting for expression differences between cartilage and synovium seen following ligament reconstruction, ligament repair with an ECM scaffold upregulated pathways related to ion homeostasis, tissue remodeling, and collagen catabolism in cartilage relative to synovium. These findings implicate inflammatory pathways within cartilage in the mid-stage development of posttraumatic osteoarthritis, independent of surgical treatment. Moreover, use of an ECM scaffold may exert a chondroprotective effect over gold-standard reconstruction through preferentially activating ion homeostatic and tissue remodeling pathways within cartilage.

14.
Biomolecules ; 13(2)2023 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-36830586

RESUMEN

The terminal complement complex (TCC) has been described as a potential driver in the pathogenesis of posttraumatic osteoarthritis (PTOA). However, sublytic TCC deposition might also play a crucial role in bone development and regeneration. Therefore, we elucidated the effects of TCC on joint-related tissues using a rabbit PTOA model. In brief, a C6-deficient rabbit breed was characterized on genetic, protein, and functional levels. Anterior cruciate ligament transection (ACLT) was performed in C6-deficient (C6-/-) and C6-sufficient (C6+/-) rabbits. After eight weeks, the progression of PTOA was determined histologically. Moreover, the structure of the subchondral bone was evaluated by µCT analysis. C6 deficiency could be attributed to a homozygous 3.6 kb deletion within the C6 gene and subsequent loss of the C5b binding site. Serum from C6-/- animals revealed no hemolytic activity. After ACLT surgery, joints of C6-/- rabbits exhibited significantly lower OA scores, including reduced cartilage damage, hypocellularity, cluster formation, and osteophyte number, as well as lower chondrocyte apoptosis rates and synovial prostaglandin E2 levels. Moreover, ACLT surgery significantly decreased the trabecular number in the subchondral bone of C6-/- rabbits. Overall, the absence of TCC protected from injury-induced OA progression but had minor effects on the micro-structure of the subchondral bone.


Asunto(s)
Cartílago Articular , Osteoartritis , Animales , Conejos , Complejo de Ataque a Membrana del Sistema Complemento/farmacología , Cartílago Articular/patología , Osteoartritis/patología , Ligamento Cruzado Anterior/patología , Ligamento Cruzado Anterior/cirugía , Condrocitos/patología
15.
J Orthop Res ; 41(1): 241-248, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-35451533

RESUMEN

A hallmark of osteoarthritis (OA) is cartilage degeneration, which has been previously correlated with dramatic increases in inflammatory enzymes. Specifically, interleukin-1ß (IL-1ß) and subsequent upregulation of nuclear factor kappa B (NF-κB) is implicated as an important player in the development of posttraumatic osteoarthritis (PTOA). Alpha 2-macroglobulin (A2M) can inhibit this inflammatory pathway, making it a promising therapy for PTOA. Herein, we demonstrate that A2M binds and neutralizes IL-1ß, blocking downstream NF-κB-induced catabolism seen in in vitro. Human chondrocytes (cell line C28) were incubated with A2M protein and then treated with IL-1ß. A2M was labeled with VivoTag™ 680 to localize the protein postincubation. The degree of binding between A2M and IL-1ß was evaluated through immunoprecipitation (IP). Catabolic proteins, including IL-1ß and NF-kB, were detected by Western blot. Pro-inflammatory and chondrocyte-related gene expression was examined by qRT-PCR. VivoTag™ 680-labeled A2M was observed in the cytoplasm of C28 human chondrocytes by fluorescence microscopy. IP experiments demonstrated that A2M could bind IL-1ß. Additionally, western blot analysis revealed that A2M neutralized IL-1ß and NF-κB in a dose-dependent manner. Moreover, A2M decreased levels of MMPs and TNF-α and increased the expression of cartilage protective genes Col2, Type2, Smad4, and aggrecan. Mostly importantly, A2M was shown to directly neutralize IL-1ß to downregulate the pro-inflammatory responses mediated by the NF-kB pathway. These results demonstrate a mechanism by which A2M reduces inflammatory catabolic activity and protects cartilage after joint injury. Further in vivo studies are needed to fully understand the potential of A2M as a novel PTOA therapy.


Asunto(s)
FN-kappa B , alfa 2-Macroglobulinas Asociadas al Embarazo , Humanos , Embarazo , Femenino , Interleucina-1beta , Mediadores de Inflamación , alfa-Macroglobulinas
16.
Clin Biomech (Bristol, Avon) ; 101: 105854, 2023 01.
Artículo en Inglés | MEDLINE | ID: mdl-36542910

RESUMEN

BACKGROUND: Large animal fracture models that allow for anatomic fracture fixation are currently lacking. It was hypothesized that a compressed air impaction system can generate a reproducible tibial plafond fracture and be adjustable to create fractures consistent with high and low energy fractures seen in humans. METHODS: Pilot testing of the impaction system was done by impacting polyurethane foam blocks at varying compressed air pressures. A guillotine impaction test was performed on the same foam blocks to create an energy conversion. A total of 12 porcine hindlimb hindlimbs were subjected to low-energy (42.2 J) and high-energy (73.9 J) impact to create tibial plafond fractures. FINDINGS: Guillotine impaction test demonstrated strong correlations between potential energy and foam block impaction depth (R2 = 0.99). Compressed air impaction system test strongly correlated with foam block impaction depth (R2 = 0.99). All six porcine hindlimbs in the low-energy group developed simple coronal split tibial plafond fractures. All six porcine hindlimbs in the high-energy group developed complex, multi-fragmentary tibial plafond fractures. INTERPRETATION: This porcine fracture model created tibial plafond fracture patterns with similar fracture morphology as human patients without violation of the soft tissue structures or adjacent joints. This model would allow for anatomic fixation, the study of post-traumatic osteoarthritis, or the delivery of locally targeted therapeutics to the ankle joint.


Asunto(s)
Fracturas de Tobillo , Fracturas de la Tibia , Humanos , Animales , Porcinos , Fracturas de la Tibia/cirugía , Fracturas de Tobillo/cirugía , Fijación de Fractura , Fijación Interna de Fracturas , Estudios Retrospectivos
17.
Osteoarthr Cartil Open ; 4(1): 100227, 2022 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-36474470

RESUMEN

Objective: The objective of this study is to understand the role of altered in vivo mechanical environments in knee joints post anterior cruciate ligament (ACL)-injury in chondrocyte vulnerability against mechanical stimuli and in the progression of post-traumatic osteoarthritis (PT-OA). Methods: Differential in vivo mechanical environments were induced by unilateral ACL-injury (uni-ACL-I) and bilateral ACL-injury (bi-ACL-I) in 8-week-old female C57BL/6 mice. The gait parameters, the mechano-vulnerability of in situ chondrocytes, Young's moduli of cartilage extracellular matrix (ECM), and the histological assessment of OA severity (OARSI score) were compared between control and experimental groups at 0∼8-weeks post-ACL-injury. Results: We found that bi-ACL-I mice experience higher joint-loading on their both injured limbs, but uni-ACL-I mice balance their joint-loading between injured and uninjured hind limbs resulting in a reduced joint-loading during gait. We also found that at 4- and 8-week post-injury the higher weight-bearing hind limbs (i.e., bi-ACL-I) had the increased area of chondrocyte death induced by impact loading and higher OARSI score than the lower weight-bearing limbs (uni-ACL-I). Additionally, we found that at 8-weeks post-injury the ECM became stiffer in bi-ACL-I joints and softer in uni-ACL-I joints. Conclusions: Our results show that ACL-injured limbs with lower in vivo joint-loading develops PT-OA significantly slower than injured limbs with higher joint-loading during gait. Our data also indicate that articular chondrocytes in severe PT-OA are more fragile from mechanical impacts than chondrocytes in healthy or mild PT-OA. Thus, preserving physiologic joint-loads on injured joints will reduce chondrocyte death post-injury and may delay PT-OA progression.

18.
Osteoarthr Cartil Open ; 4(2): 100266, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-36475289

RESUMEN

Objective: During articular fracture reconstruction, orthopedic surgeons are frequently faced with the dilemma of retaining small articular fragments versus discarding these fragments. The purpose of this study was to compare post-traumatic osteoarthritis (PTOA) development between tibial plafond fractures and plafond fractures with a missing articular fragment (MF) in a porcine model. Design: High-energy tibial plafond fractures in skeletally mature Yucatan mini pigs (n â€‹= â€‹12) were created. During surgery, a 3 â€‹× â€‹3 mm section of the articular surface was removed in six animals (MF group). Ankle synovial fluid was analyzed for IL-1ß, IL-1Ra, IL-6, IL-8, and IL-10 concentrations obtained at initial surgery and 12 weeks post-surgery. Plafond and talus sections were evaluated for subchondral bone porosity and stained with Sanderson's Rapid Bone Stain and blindly evaluated to determine the Osteoarthritis Research Society International (OARSI) grade and vascular invasion. Results: Fractured ankles had greater concentrations of IL-1ß, IL-1Ra, IL-6, IL-8, and IL-10 compared to control ankles. There was no difference in cytokine concentrations between fractured and fractured â€‹+ â€‹MF ankles. Fractured ankles had significantly greater bone porosity, vascular invasion, and OARSI grade as compared to the control group. In comparing tibial plafonds, the MF group had significantly more bone porosity, more vascular invasion, and a higher average OARSI grade than the anatomically reconstructed group. In comparing the talus, the MF group had higher average OARSI grade and similar bone porosity. Conclusions: Articular fractures with a MF had worse PTOA development as measured by bone porosity, vascular invasion, and OARSI grade than the anatomically reconstructed fractures.

19.
Acta Biomater ; 153: 243-259, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36374749

RESUMEN

There are no drugs or treatment methods known to prevent the development of post-traumatic osteoarthritis (PTOA), a type of osteoarthritis (OA) that is triggered by traumatic joint injuries and accounts for ∼12% of the nearly 600 million OA cases worldwide. Lack of effective drug delivery techniques remains a major challenge in developing clinically effective treatments, but cationic delivery carriers can help overcome this challenge. Scaling up treatments that are effective in in vitro models to achieve success in preclinical in vivo models and clinical trials is also a challenging problem in the field. Here we use a cationic green fluorescent protein (GFP) as a carrier to deliver Insulin-Like Growth Factor 1 (IGF-1), a drug considered as a potential therapeutic for PTOA. GFP-IGF-1 conjugates were first synthesized as fusion proteins with different polypeptide linkers, and their transport properties were characterized in human cartilage explants. In vitro experimental data were used to develop a predictive mathematical transport model that was validated using an independent in vitro experimental data set. The model was used to predict the transport of these fusion proteins upon intra-articular injection into human knee joints. The predictions included results for the rate and extent of fusion protein penetration into cartilage, and the maximum levels of fusion proteins that would escape into systemic circulation through the joint capsule. Together, our transport measurements and model set the stage for translation of such explant culture studies to in vivo preclinical studies and potentially clinical application. STATEMENT OF SIGNIFICANCE: The lack of blood supply in cartilage and rapid clearance of drugs injected into human knees presents a major challenge in developing clinically effective treatments for osteoarthritis. Cationic delivery carriers can target negatively charged cartilage and help overcome this problem. Scaling up treatments that are effective in vitro to achieve success in vivo is also challenging. Here, we use a cationic green fluorescent protein (GFP) to deliver Insulin-Like Growth Factor-1 (IGF-1) into cartilage. Experiments measuring transport of GFP-IGF-1 fusion proteins in human cartilage explants were used to develop and validate a mathematical model to predict fusion protein transport upon injection into human knee joints. This work translates such explant culture studies to in vivo preclinical studies and potentially clinical application.


Asunto(s)
Cartílago Articular , Factor I del Crecimiento Similar a la Insulina , Osteoartritis , Humanos , Cartílago Articular/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Factor I del Crecimiento Similar a la Insulina/farmacología , Articulación de la Rodilla , Osteoartritis/tratamiento farmacológico , Proteínas Recombinantes de Fusión/farmacología , Sistemas de Liberación de Medicamentos
20.
Front Bioeng Biotechnol ; 10: 970235, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36312551

RESUMEN

Meniscal tearing in the knee increases the risk of post-traumatic osteoarthritis (OA) in patients. The therapeutic application of tissue-specific mesenchymal progenitor cells is currently being investigated as an emerging biologic strategy to help improve healing of musculoskeletal tissues like meniscal fibrocartilage and articular hyaline cartilage. However, many of these approaches involve isolating cells from healthy tissues, and the low yield of rare progenitor populations (< 1% of total cells residing in tissues) can make finding a readily available cell source for therapeutic use a significant logistical challenge. In the present study, we investigated the therapeutic efficacy of using expanded cartilage-derived and bone marrow-derived progenitor cell lines, which were stabilized using retroviral SV40, for repair of meniscus injury in a rodent model. Our findings indicate that these cell lines express the same cell surface marker phenotype of primary cells (CD54+, CD90+, CD105+, CD166+), and that they exhibit improved proliferative capacity that is suitable for extensive expansion. Skeletally mature male athymic rats treated with 3.2 million cartilage-derived progenitor cell line exhibited approximately 79% greater meniscal tear reintegration/healing, compared to injured animals that left untreated, and 76% greater compared to animals treated with the same number of marrow-derived stromal cells. Histological analysis of articular surfaces also showed that cartilage-derived progenitor cell line treated animals exhibited reduced post-traumatic OA associated articular cartilage degeneration. Stable cell line treatment did not cause tumor formation or off-target engraftment in animals. Taken together, we present a proof-of-concept study demonstrating, for the first time, that intra-articular injection of a stable human cartilage-derived progenitor cell line stimulates meniscus tear healing and provide chondroprotection in an animal model. These outcomes suggest that the use of stable cell lines may help overcome cell source limitations for cell-based medicine.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...