Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.361
Filter
1.
Crit Rev Biomed Eng ; 52(4): 17-28, 2024.
Article in English | MEDLINE | ID: mdl-38780103

ABSTRACT

In this study, we examine the behavior of articular cartilage equilibrated in a salt (NaCl) solution during non-Newtonian fluid flow that follows an Ostwald-de Waele model. A linearly elastic and isotropic rectangular strip of cartilage is considered for analysis. A continuum theory of mixtures has been employed to develop a coupled system of partial differential equations for the solid displacement and the fluid pressure by considering the important factor of the ion concentration by assuming the cartilage as a deformable porous media. The coupled system of partial differential equations is solved using the numerical method named method of lines. In most cases, shear-thinning fluid is compared to the shear-thickening fluid to magnify the difference. Graphical results show that shear-thickening fluids bring more solid deformation and shows less fluid pressure in comparison to the shear-thinning fluids.


Subject(s)
Cartilage, Articular , Pressure , Cartilage, Articular/physiology , Models, Biological , Humans , Ions , Animals , Rheology/methods , Elasticity , Sodium Chloride/chemistry , Viscosity , Porosity
2.
Langmuir ; 40(20): 10648-10662, 2024 May 21.
Article in English | MEDLINE | ID: mdl-38712915

ABSTRACT

This study presents new insights into the potential role of polyelectrolyte interfaces in regulating low friction and interstitial fluid pressurization of cartilage. Polymer brushes composed of hydrophilic 3-sulfopropyl methacrylate potassium salt (SPMK) tethered to a PEEK substrate (SPMK-g-PEEK) are a compelling biomimetic solution for interfacing with cartilage, inspired by the natural lubricating biopolyelectrolyte constituents of synovial fluid. These SPMK-g-PEEK surfaces exhibit a hydrated compliant layer approximately 5 µm thick, demonstrating the ability to maintain low friction coefficients (µ ∼ 0.01) across a wide speed range (0.1-200 mm/s) under physiological loads (0.75-1.2 MPa). A novel polyelectrolyte-enhanced tribological rehydration mechanism is elucidated, capable of recovering up to ∼12% cartilage strain and subsequently facilitating cartilage interstitial fluid recovery, under loads ranging from 0.25 to 2.21 MPa. This is attributed to the combined effects of fluid confinement within the contact gap and the enhanced elastohydrodynamic behavior of polymer brushes. Contrary to conventional theories that emphasize interstitial fluid pressurization in regulating cartilage lubrication, this work demonstrates that SPMK-g-PEEK's frictional behavior with cartilage is independent of these factors and provides unabating aqueous lubrication. Polyelectrolyte-enhanced tribological rehydration can occur within a static contact area and operates independently of known mechanisms of cartilage interstitial fluid recovery established for converging or migrating cartilage contacts. These findings challenge existing paradigms, proposing a novel polyelectrolyte-cartilage tribological mechanism not exclusively reliant on interstitial fluid pressurization or cartilage contact geometry. The implications of this research extend to a broader understanding of synovial joint lubrication, offering insights into the development of joint replacement materials that more accurately replicate the natural functionality of cartilage.


Subject(s)
Lubrication , Polymers , Polymers/chemistry , Animals , Polyelectrolytes/chemistry , Polyethylene Glycols/chemistry , Cartilage/chemistry , Cartilage/drug effects , Surface Properties , Benzophenones/chemistry , Cartilage, Articular/chemistry , Cartilage, Articular/physiology , Ketones/chemistry
3.
J Biomech ; 169: 112135, 2024 May.
Article in English | MEDLINE | ID: mdl-38744145

ABSTRACT

Articular cartilage exhibits site-specific biomechanical properties. However, no study has comprehensively characterized site-specific cartilage properties from the same knee joints at different stages of osteoarthritis (OA). Cylindrical osteochondral explants (n = 381) were harvested from donor-matched lateral and medial tibia, lateral and medial femur, patella, and trochlea of cadaveric knees (N = 17). Indentation test was used to measure the elastic and viscoelastic mechanical properties of the samples, and Osteoarthritis Research Society International (OARSI) grading system was used to categorize the samples into normal (OARSI 0-1), early OA (OARSI 2-3), and advanced OA (OARSI 4-5) groups. OA-related changes in cartilage mechanical properties were site-specific. In the lateral and medial tibia and trochlea sites, equilibrium, instantaneous and dynamic moduli were higher (p < 0.001) in normal tissue than in early and advanced OA tissue. In lateral and medial femur, equilibrium, instantaneous and dynamic moduli were smaller in advanced OA, but not in early OA, than in normal tissue. The phase difference (0.1-0.25 Hz) between stress and strain was significantly smaller (p < 0.05) in advanced OA than in normal tissue across all sites except medial tibia. Our results indicated that in contrast to femoral and patellar cartilage, equilibrium, instantaneous and dynamic moduli of the tibia and trochlear cartilage decreased in early OA. These may suggest that the tibia and trochlear cartilage degrades faster than the femoral and patellar cartilage. The information is relevant for developing site-specific computational models and engineered cartilage constructs.


Subject(s)
Cartilage, Articular , Knee Joint , Osteoarthritis, Knee , Humans , Cartilage, Articular/physiopathology , Cartilage, Articular/physiology , Cartilage, Articular/pathology , Knee Joint/physiopathology , Aged , Osteoarthritis, Knee/physiopathology , Male , Female , Middle Aged , Biomechanical Phenomena , Elasticity , Viscosity , Tibia/physiopathology , Femur/physiopathology , Femur/physiology , Aged, 80 and over , Adult , Stress, Mechanical
4.
J Biomech ; 169: 112133, 2024 May.
Article in English | MEDLINE | ID: mdl-38744146

ABSTRACT

Abnormal loading is thought to play a key role in the disease progression of cartilage, but our understanding of how cartilage compositional measurements respond to acute compressive loading in-vivo is limited. Ten healthy subjects were scanned at two timepoints (7 ± 3 days apart) with a 3 T magnetic resonance imaging (MRI) scanner. Scanning sessions included T1ρ and T2* acquisitions of each knee in two conditions: unloaded (traditional MRI setup) and loaded in compression at 40 % bodyweight as applied by an MRI-compatible loading device. T1ρ and T2* parameters were quantified for contacting cartilage (tibial and femoral) and non-contacting cartilage (posterior femoral condyle) regions. Significant effects of load were found in contacting regions for both T1ρ and T2*. The effect of load (loaded minus unloaded) in femoral contacting regions ranged from 4.1 to 6.9 ms for T1ρ, and 3.5 to 13.7 ms for T2*, whereas tibial contacting regions ranged from -5.6 to -1.7 ms for T1ρ, and -2.1 to 0.7 ms for T2*. Notably, the responses to load in the femoral and tibial cartilage revealed opposite effects. No significant differences were found in response to load between the two visits. This is the first study that analyzed the effects of acute loading on T1ρ and T2* measurements in human femoral and tibial cartilage separately. The results suggest the effect of acute compressive loading on T1ρ and T2* was: 1) opposite in the femoral and tibial cartilage; 2) larger in contacting regions than in non-contacting regions of the femoral cartilage; and 3) not different visit-to-visit.


Subject(s)
Cartilage, Articular , Femur , Magnetic Resonance Imaging , Tibia , Weight-Bearing , Humans , Cartilage, Articular/physiology , Cartilage, Articular/diagnostic imaging , Femur/diagnostic imaging , Femur/physiology , Male , Adult , Female , Magnetic Resonance Imaging/methods , Tibia/diagnostic imaging , Tibia/physiology , Weight-Bearing/physiology , Knee Joint/physiology , Knee Joint/diagnostic imaging , Compressive Strength/physiology
5.
Med Eng Phys ; 126: 104130, 2024 04.
Article in English | MEDLINE | ID: mdl-38621832

ABSTRACT

Biphasic models have been widely used to simulate the time-dependent biomechanical response of soft tissues. Modelling techniques of joints with biphasic weight-bearing soft tissues have been markedly improved over the last decade, enhancing our understanding of the function, degenerative mechanism and outcomes of interventions of joints. This paper reviews the recent advances, challenges and opportunities in computational models of joints with biphasic weight-bearing soft tissues. The review begins with an introduction of the function and degeneration of joints from a biomechanical aspect. Different constitutive models of articular cartilage, in particular biphasic materials, are illustrated in the context of the study of contact mechanics in joints. Approaches, advances and major findings of biphasic models of the hip and knee are presented, followed by a discussion of the challenges awaiting to be addressed, including the convergence issue, high computational cost and inadequate validation. Finally, opportunities and clinical insights in the areas of subject-specific modeling and tissue engineering are provided and discussed.


Subject(s)
Cartilage, Articular , Models, Biological , Humans , Biomechanical Phenomena , Joints/physiology , Cartilage, Articular/physiology , Computer Simulation , Knee Joint/physiology , Finite Element Analysis
6.
Biomater Adv ; 160: 213857, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38657287

ABSTRACT

Articular cartilage injury impairs joint function and necessitates orthopedic intervention to restore the structure and function of the cartilage. Extracellular matrix (ECM) scaffolds derived from bone marrow mesenchymal stem cells (BMSCs) can effectively promote cell adhesion, proliferation, and chondrogenesis. However, pre-shaped ECM scaffolds have limited applicability due to their poor fit with the irregular surface of most articular cartilage defects. In this study, we fabricated an injectable active ECM hydrogel from autologous BMSCs-derived ECM by freeze-drying, liquid nitrogen milling, and enzymatic digestion. Moreover, our in vitro and in vivo results demonstrated that the prepared hydrogel enhanced chondrocyte adhesion and proliferation, chondrogenesis, cartilage regeneration, and integration with host tissue, respectively. These findings indicate that active ECM components can provide trophic support for cell proliferation and differentiation, restoring the structure and function of damaged cartilage.


Subject(s)
Cartilage, Articular , Chondrocytes , Chondrogenesis , Extracellular Matrix , Hydrogels , Mesenchymal Stem Cells , Regeneration , Tissue Engineering , Tissue Scaffolds , Extracellular Matrix/chemistry , Extracellular Matrix/metabolism , Animals , Mesenchymal Stem Cells/cytology , Cartilage, Articular/physiology , Cartilage, Articular/injuries , Hydrogels/chemistry , Tissue Scaffolds/chemistry , Chondrocytes/transplantation , Tissue Engineering/methods , Cell Proliferation , Cell Differentiation , Rabbits , Cell Adhesion , Humans , Injections
7.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 41(2): 328-334, 2024 Apr 25.
Article in Chinese | MEDLINE | ID: mdl-38686414

ABSTRACT

Superficial cartilage defect is an important factor that causes osteoarthritis. Therefore, it is very important to investigate the influence of superficial cartilage defects on its surface morphology and mechanical properties. In this study, the knee joint cartilage samples of adult pig were prepared, which were treated by enzymolysis with chymotrypsin and physical removal with electric friction pen, respectively. Normal cartilage and surface treated cartilage were divided into five groups: control group (normal cartilage group), chymotrypsin immersion group, chymotrypsin wiping group, removal 10% group with electric friction pen, and removal 20% group with electric friction pen. The surface morphology and structure of five groups of samples were characterized by laser spectrum confocal microscopy and environmental field scanning electron microscopy, and the mechanical properties of each group of samples were evaluated by tensile tests. The results show that the surface arithmetic mean height and fracture strength of the control group were the smallest, and the fracture strain was the largest. The surface arithmetic mean height and fracture strength of the removal 20% group with electric friction pen were the largest, and the fracture strain was the smallest. The surface arithmetic mean height, fracture strength and fracture strain values of the other three groups were all between the above two groups, but the surface arithmetic mean height and fracture strength of the removal 10% group with electric friction pen, the chymotrypsin wiping group and the chymotrypsin soaking group decreased successively, and the fracture strain increased successively. In addition, we carried out a study on the elastic modulus of different groups, and the results showed that the elastic modulus of the control group was the smallest, and the elastic modulus of the removal 20% group with electric friction pen was the largest. The above study revealed that the defect of the superficial area of cartilage changed its surface morphology and structure, and reduced its mechanical properties. The research results are of great significance for the prevention and repair of cartilage injury.


Subject(s)
Cartilage, Articular , Animals , Swine , Cartilage, Articular/physiology , Surface Properties , Biomechanical Phenomena , Knee Joint/physiology , Stress, Mechanical , Tensile Strength , Chymotrypsin/metabolism , Microscopy, Electron, Scanning
8.
Foot Ankle Clin ; 29(2): 281-290, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38679439

ABSTRACT

Bone Marrow Stimulation of osteochondral lesions of the talus has been shown to be a successful way to treat cartilage injuries. Newer data suggest that Bone Marrow Stimulation is best reserved for osteochondral lesions of the talus Sizes Less Than 107.4 mm2 in area. Additionally, newer smaller and deeper techniques to perform bone marrow stimulation have resulted in less subchondral bone damage, less cancellous compaction, and superior bone marrow access with multiple trabecular access channels. Biologic adjuvants such as platelet-rich plasma (PRP), hyaluronic acid (HA), and bone marrow aspirate concentrate (BMAC) may lead to better functional outcomes when used concomitant to bone marrow stimulation.


Subject(s)
Talus , Humans , Talus/injuries , Talus/surgery , Cartilage, Articular/injuries , Cartilage, Articular/surgery , Cartilage, Articular/physiology , Platelet-Rich Plasma , Bone Marrow , Bone Regeneration/physiology
9.
Foot Ankle Clin ; 29(2): 291-305, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38679440

ABSTRACT

Osteochondral lesion of the talus (OLT) is a commune cause of chronic ankle pain. Symptomatic lesions require surgical treatment. Currently, lesions with diameter less than 107.4 mm2 are treated with bone marrow stimulating technique with notable success rate. However, more extensive lesions show less predictable surgical results. Autologous matrix-induced chondrogenesis has proven to provide satisfactory medium and long-term results on OLTs. In the current review, we describe an all-arthroscopic technique and the Milan-Tel Aviv lesion assessment protocol.


Subject(s)
Arthroscopy , Talus , Humans , Talus/surgery , Arthroscopy/methods , Cartilage, Articular/surgery , Cartilage, Articular/physiology , Chondrogenesis/physiology
10.
J Biomech Eng ; 146(8)2024 Aug 01.
Article in English | MEDLINE | ID: mdl-38530647

ABSTRACT

Articular cartilage (AC) is a load-bearing tissue that covers long bones in synovial joints. The biphasic/poroelastic mechanical properties of AC help it to protect joints by distributing loads, absorbing impact forces, and reducing friction. Unfortunately, alterations in these mechanical properties adversely impact cartilage function and precede joint degeneration in the form of osteoarthritis (OA). Thus, understanding what factors regulate the poroelastic mechanical properties of cartilage is of great scientific and clinical interest. Transgenic mouse models provide a valuable platform to delineate how specific genes contribute to cartilage mechanical properties. However, the poroelastic mechanical properties of murine articular cartilage are challenging to measure due to its small size (thickness ∼ 50 microns). In the current study, our objective was to test whether the poroelastic mechanical properties of murine articular cartilage can be determined based solely on time-dependent cell death measurements under constant loading conditions. We hypothesized that in murine articular cartilage subjected to constant, sub-impact loading from an incongruent surface, cell death area and tissue strain are closely correlated. We further hypothesized that the relationship between cell death area and tissue strain can be used-in combination with inverse finite element modeling-to compute poroelastic mechanical properties. To test these hypotheses, murine cartilage-on-bone explants from different anatomical locations were subjected to constant loading conditions by an incongruent surface in a custom device. Cell death area increased over time and scaled linearly with strain, which rose in magnitude over time due to poroelastic creep. Thus, we were able to infer tissue strain from cell death area measurements. Moreover, using tissue strain values inferred from cell death area measurements, we applied an inverse finite element modeling procedure to compute poroelastic material properties and acquired data consistent with previous studies. Collectively, our findings demonstrate in the key role poroelastic creep plays in mediating cell survival in mechanically loaded cartilage and verify that cell death area can be used as a surrogate measure of tissue strain that enables determination of murine cartilage mechanical properties.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Mice , Chondrocytes/physiology , Stress, Mechanical , Cartilage, Articular/physiology , Cell Death
11.
Cell Tissue Bank ; 25(2): 633-648, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38319426

ABSTRACT

Osteochondral allograft (OCA) transplantation involves grafting of natural hyaline cartilage and supporting subchondral bone into the cartilage defect area to restore its biomechanical and tissue structure. However, differences in biomechanical properties and donor-host matching may impair the integration of articular cartilage (AC). This study analyzed the biomechanical properties of the AC in different regions of different sites of the knee joint and provided a novel approach to OCA transplantation. Intact stifle joints from skeletally mature pigs were collected from a local abattoir less than 8 h after slaughter. OCAs were collected from different regions of the joints. The patella and the tibial plateau were divided into medial and lateral regions, while the trochlea and femoral condyle were divided into six regions. The OCAs were analyzed and compared for Young's modulus, the compressive modulus, and cartilage thickness. Young's modulus, cartilage thickness, and compressive modulus of OCA were significantly different in different regions of the joints. A negative correlation was observed between Young's modulus and the proportion of the subchondral bone (r = - 0.4241, P < 0.0001). Cartilage thickness was positively correlated with Young's modulus (r = 0.4473, P < 0.0001) and the compressive modulus (r = 0.3678, P < 0.0001). During OCA transplantation, OCAs should be transplanted in the same regions, or at the closest possible regions to maintain consistency of the biomechanical properties and cartilage thickness of the donor and recipient, to ensure smooth integration with the surrounding tissue. A 7 mm depth achieved a higher Young's modulus, and may represent the ideal length.


Subject(s)
Allografts , Cartilage, Articular , Knee Joint , Animals , Cartilage, Articular/physiology , Knee Joint/physiology , Knee Joint/surgery , Biomechanical Phenomena , Swine , Elastic Modulus , Bone Transplantation/methods
12.
Med Sci Sports Exerc ; 56(6): 1026-1035, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38233979

ABSTRACT

PURPOSE: The purpose of this study was to determine whether regular running distance and biomechanics are related to medial central femur cartilage (MCFC) structure. METHODS: The cross-sectional study sample consisted of 1164 runners and nonrunners aged 18-65 yr. Participants completed questionnaires on physical activity and their running history. We performed quantitative magnetic resonance imaging of knee cartilage-T2 relaxation time (T2) mapping (high T2 indicates cartilage degeneration)-and a running biomechanical analysis using a three-dimensional motion capture system. A 14-d monitoring of the physical activity was conducted. RESULTS: Those aged 35-49 yr were at 84% higher odds of having MCFC T2 in the highest level (85th percentile, P < 0.05) compared with youngest adults indicating that MCFC structures may be altered with aging. Being male was associated with 34% lower odds of having T2 at the highest level ( P < 0.05) compared with females. Nonrunners and runners with the highest weekly running distance were more likely to have a high T2 compared with runners with running distance of 6-20 km·wk -1 ( P < 0.05). In addition, the maximal knee internal adduction moment was associated with a 19% lower odds of having T2 at the highest level ( P < 0.05). CONCLUSIONS: Females compared with males and a middle-aged cohort compared with the younger cohort seemed to be associated with the degeneration of MCFC structures. Runners who ran 6-20 km·wk -1 were associated with a higher quality of their MCFC compared with highly active individuals and nonrunners. Knee frontal plane biomechanics was related to MCFC structure indicating a possibility of modifying the medial knee collagen fibril network through regular running.


Subject(s)
Cartilage, Articular , Knee Joint , Magnetic Resonance Imaging , Running , Humans , Running/physiology , Male , Female , Cross-Sectional Studies , Adult , Knee Joint/physiology , Knee Joint/diagnostic imaging , Middle Aged , Cartilage, Articular/physiology , Cartilage, Articular/anatomy & histology , Cartilage, Articular/diagnostic imaging , Biomechanical Phenomena , Young Adult , Adolescent , Aged , Age Factors , Sex Factors , Femur/physiology , Femur/anatomy & histology , Femur/diagnostic imaging
13.
Adv Biol (Weinh) ; 8(1): e2300268, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37688354

ABSTRACT

One of the main components of articular cartilage is the chondrocyte's pericellular matrix (PCM), which is critical for regulating mechanotransduction, biochemical cues, and healthy cartilage development. Here, individual primary human chondrocytes (PHC) are encapsulated and cultured in 50 µm diameter alginate microgels using drop-based microfluidics. This unique culturing method enables PCM formation and manipulation of individual cells. Over ten days, matrix formation is observed using autofluorescence imaging, and the elastic moduli of isolated cells are measured using AFM. Matrix production and elastic modulus increase are observed for the chondrons cultured in microgels. Furthermore, the elastic modulus of cells grown in microgels increases ≈ten-fold over ten days, nearly reaching the elastic modulus of in vivo PCM. The AFM data is further analyzed using a Gaussian mixture model and shows that the population of PHCs grown in microgels exhibit two distinct populations with elastic moduli averaging 9.0 and 38.0 kPa. Overall, this work shows that microgels provide an excellent culture platform for the growth and isolation of PHCs, enabling PCM formation that is mechanically similar to native PCM. The microgel culture platform presented here has the potential to revolutionize cartilage regeneration procedures through the inclusion of in vitro developed PCM.


Subject(s)
Cartilage, Articular , Microgels , Humans , Chondrocytes/physiology , Microscopy, Atomic Force , Extracellular Matrix/physiology , Mechanotransduction, Cellular , Cartilage, Articular/physiology
14.
J Orthop Res ; 42(4): 745-752, 2024 Apr.
Article in English | MEDLINE | ID: mdl-37975270

ABSTRACT

The association of knee joint osteoarthritis and altered frictional properties of the degenerated cartilage remains ambiguous, because previous in vitro studies did not consider the characteristic loads and velocities during gait. Therefore, the aim of this study was to quantify the friction behavior of degenerated human cartilage under characteristic stance and swing phase conditions. A dynamic pin-on-plate tribometer was used to test the tribological systems of cartilage against cartilage and cartilage against glass, both with synthetic synovial fluid as lubricant. Using the International Cartilage Repair Society classification, the cartilage samples were assigned to a mildly or a severely degenerated group before testing. Friction coefficients were calculated under stance and swing phase conditions at the beginning of the test and after 600 s of testing. The most important finding of this study is that cartilage against glass couplings displayed significantly higher friction for the severely degenerated samples compared to the mildly degenerated ones, whereas cartilage against cartilage couplings only indicated slight tendencies under the observed test conditions. Consequently, care should be taken when transferring in vitro findings from cartilage against cartilage couplings to predict the friction behavior in vivo. Therefore, we recommend in vitro tribological testing methods which account for gait-like loading conditions and to replicate physiological material pairings, particularly in preclinical medical device validation studies.


Subject(s)
Cartilage, Articular , Osteoarthritis, Knee , Humans , Friction , Cartilage, Articular/physiology , Knee Joint , Synovial Fluid/physiology , Stress, Mechanical
15.
Small ; 20(11): e2304088, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37939310

ABSTRACT

The use of natural cartilage extracellular matrix (ECM) has gained widespread attention in the field of cartilage tissue engineering. However, current approaches for delivering functional scaffolds for osteoarthritis (OA) therapy rely on knee surgery, which is limited by the narrow and complex structure of the articular cavity and carries the risk of injuring surrounding tissues. This work introduces a novel cell microcarrier, magnetized cartilage ECM-derived scaffolds (M-CEDSs), which are derived from decellularized natural porcine cartilage ECM. Human bone marrow mesenchymal stem cells are selected for their therapeutic potential in OA treatments. Owing to their natural composition, M-CEDSs have a biomechanical environment similar to that of human cartilage and can efficiently load functional cells while maintaining high mobility. The cells are released spontaneously at a target location for at least 20 days. Furthermore, cell-seeded M-CEDSs show better knee joint function recovery than control groups 3 weeks after surgery in preclinical experiments, and ex vivo experiments reveal that M-CEDSs can rapidly aggregate inside tissue samples. This work demonstrates the use of decellularized microrobots for cell delivery and their in vivo therapeutic effects in preclinical tests.


Subject(s)
Cartilage, Articular , Mesenchymal Stem Cells , Osteoarthritis , Animals , Swine , Humans , Cartilage, Articular/physiology , Tissue Engineering , Extracellular Matrix/chemistry , Magnetic Phenomena , Tissue Scaffolds/chemistry
16.
BMC Musculoskelet Disord ; 24(1): 964, 2023 Dec 11.
Article in English | MEDLINE | ID: mdl-38082264

ABSTRACT

BACKGROUND: The patellofemoral joint is a challenging environment for treating chondral defects. Among the surgical options for the treatment of chondral defects, the single-stage Autologous Matrix-Induced Chondrogenesis (AMIC) procedure uses a porcine collagen I/III membrane to enhance bone-marrow stimulation. However, longer term outcomes data are rare for this specific indication. In order to provide real-world information, an ongoing registry has been established to record patient data and outcomes when AMIC is used to treat chondral and osteochondral lesions. METHODS: Patient data were retrieved from an ongoing, prospective, multisite registry of patients who had undergone AMIC treatment of chondral defects. We identified 64 patients who had undergone AMIC for patellofemoral chondral defects and for whom pre-operative and at least 1 post-operative score were available were included in this retrospective data analysis. Outcomes were assessed via the KOOS, VAS pain, and the Lysholm scores. Outcomes at the post-operative time-points were analysed using a factorial ANOVA with post-hoc testing while linear regression was used to assess associations between the change in the Lysholm score and lesion size. RESULTS: There was a significant improvement in Lysholm, VAS pain, and KOOS scores from pre-operative to the 1st year post-operative (p < 0.001), and this was maintained during the follow-up. CONCLUSIONS: The forces exerted on the patellofemoral joint make this a challenging scenario for chondral repair. Our data demonstrates that the AMIC procedure with a collagen I/III membrane is an effective treatment for retropatellar cartilage lesions, and provides reliable results, with decreased pain and improved function. Importantly, these improvements were maintained through the follow-up period.


Subject(s)
Cartilage Diseases , Cartilage, Articular , Humans , Animals , Swine , Cartilage, Articular/surgery , Cartilage, Articular/physiology , Retrospective Studies , Chondrogenesis , Prospective Studies , Cartilage Diseases/surgery , Treatment Outcome , Collagen Type I , Transplantation, Autologous , Registries , Pain
17.
Biomater Adv ; 153: 213567, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37540940

ABSTRACT

Tissue-engineered articular cartilage constructs are currently not able to equal native tissues in terms of mechanical and biological properties. A major cause lies in the deficiency in engineering the biomechanical microenvironment (BMME) of articular chondrocytes. In this work, to engineer the BMME of articular chondrocytes, heterogeneous hydrogel structures of gelatin methacrylated (GelMA) containing differential-stiffness domains were first fabricated, and then periodic dynamic mechanical stimulations were applied to the hydrogel structures. The chondrocyte phenotype of ATDC5 cells was enhanced as the spatial differentiation in stiffness was increased in the hydrogel structures and was further strengthened by dynamic mechanical stimulation. It was speculated that the mechanical signals generated by the engineered BMME were sensed by the cells through the integrin ß1-FAK signaling pathway. This study revealed the key role of the combined effects of differential and dynamic BMME on the chondrocyte phenotype, which could provide theoretical guidance for highly active tissue-engineered articular cartilage.


Subject(s)
Cartilage, Articular , Chondrocytes , Chondrocytes/metabolism , Hydrogels/analysis , Gelatin , Cartilage, Articular/physiology , Tissue Engineering
18.
Acta Biomater ; 168: 372-387, 2023 09 15.
Article in English | MEDLINE | ID: mdl-37481194

ABSTRACT

Osteoarthritis (OA) is a widespread clinical disease characterized by cartilage degeneration in middle-aged and elderly people. Currently, there is no effective treatment for OA apart from total joint replacement in advanced stages. Mesenchymal stem cells (MSCs) are a type of adult stem cell with diverse differentiation capabilities and immunomodulatory potentials. MSCs are known to effectively regulate the cartilage microenvironment, promote cartilage regeneration, and alleviate OA symptoms. As a result, they are promising sources of cells for OA therapy. Recent studies have revealed the presence of resident MSCs in synovial fluid, synovial membrane, and articular cartilage, which can be collected as knee joint-derived MSCs (KJD-MSC). Several preclinical and clinical studies have demonstrated that KJD-MSCs have great potential for OA treatment, whether applied alone, in combination with biomaterials, or as exocrine MSCs. In this article, we will review the characteristics of MSCs in the joints, including their cytological characteristics, such as proliferation, cartilage differentiation, and immunomodulatory abilities, as well as the biological function of MSC exosomes. We will also discuss the use of tissue engineering in OA treatment and introduce the concept of a new generation of stem cell-based tissue engineering therapy, including the use of engineering, gene therapy, and gene editing techniques to create KJD-MSCs or KJD-MSC derivative exosomes with improved functionality and targeted delivery. These advances aim to maximize the efficiency of cartilage tissue engineering and provide new strategies to overcome the bottleneck of OA therapy. STATEMENT OF SIGNIFICANCE: This research will provide new insights into the medicinal benefit of Joint resident Mesenchymal Stem Cells (MSCs), specifically on its cartilage tissue engineering ability. Through this review, the community will further realize promoting joint resident mesenchymal stem cells, especially cartilage progenitor/MSC-like progenitor cells (CPSC), as a preventive measure against osteoarthritis and cartilage injury. People and medical institutions may also consider cartilage derived MSC as an alternative approach against cartilage degeneration. Moreover, the discussion presented in this study will convey valuable information for future research that will explore the medicinal benefits of cartilage derived MSC.


Subject(s)
Cartilage, Articular , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Osteoarthritis , Aged , Adult , Middle Aged , Humans , Tissue Engineering/methods , Cartilage, Articular/physiology , Osteoarthritis/metabolism , Knee Joint , Mesenchymal Stem Cell Transplantation/methods
19.
Adv Sci (Weinh) ; 10(27): e2207715, 2023 09.
Article in English | MEDLINE | ID: mdl-37518822

ABSTRACT

The microenvironment and stem cell fate guidance of post-traumatic articular cartilage regeneration is primarily the focus of cartilage tissue engineering. In articular cartilage, stem cells are characterized by overlapping lineages and uneven effectiveness. Within the first 12 weeks after trauma, the articular inflammatory microenvironment (AIME) plays a decisive role in determining the fate of stem cells and cartilage. The development of fibrocartilage and osteophyte hyperplasia is an adverse outcome of chronic inflammation, which results from an imbalance in the AIME during the cartilage tissue repair process. In this review, the sources for the different types of stem cells and their fate are summarized. The main pathophysiological events that occur within the AIME as well as their protagonists are also discussed. Additionally, regulatory strategies that may guide the fate of stem cells within the AIME are proposed. Finally, strategies that provide insight into AIME pathophysiology are discussed and the design of new materials that match the post-traumatic progress of AIME pathophysiology in a spatial and temporal manner is guided. Thus, by regulating an appropriately modified inflammatory microenvironment, efficient stem cell-mediated tissue repair may be achieved.


Subject(s)
Arthritis , Cartilage, Articular , Humans , Regeneration/physiology , Tissue Engineering/methods , Stem Cells , Cartilage, Articular/injuries , Cartilage, Articular/physiology , Wound Healing
20.
Proc Inst Mech Eng H ; 237(7): 879-889, 2023 Jul.
Article in English | MEDLINE | ID: mdl-37345411

ABSTRACT

The purpose of this study was to create a preliminary set of experimentally validated Finite Element Analysis (FEA) models, in order to predict the dynamic mechanical behaviour of human articular cartilage (AC). Current models consider static loading with limited independent experimental validation, while the models for this study assess dynamic loading of AC, with direct comparison and validation to physical testing. Three different FEA models of AC were constructed, which considered both linear elastic and hyperelastic models; Neo-Hookean and Ogden. Models were validated using the data collected from compression testing of human femoral heads across 0-1.7 MPa (quasi-static tests and dynamic mechanical analysis). The linear elastic model was inadequate, with a 10-fold over prediction of the displacement dynamic amplitude. The Neo-Hookean model accurately predicted the dynamic amplitude but failed to predict the initial compression of the cartilage, with a 10 times overprediction. The Ogden model provided the best results, with both the initial compression lying within one standard deviation of that observed in the validation data set, and the dynamic amplitude of the same order of magnitude. In conclusion, this study has found that the fast dynamic response of human AC is best represented by a third order Ogden model.


Subject(s)
Cartilage, Articular , Humans , Cartilage, Articular/physiology , Stress, Mechanical , Pressure , Finite Element Analysis , Models, Biological , Elasticity , Biomechanical Phenomena
SELECTION OF CITATIONS
SEARCH DETAIL
...