Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34.932
Filter
1.
Spectrochim Acta A Mol Biomol Spectrosc ; 324: 125000, 2025 Jan 05.
Article in English | MEDLINE | ID: mdl-39180968

ABSTRACT

Fourier transform infrared spectroscopy (FTIRS) can provide rich information on the composition and content of samples, enabling the detection of subtle changes in tissue composition and structure. This study represents the first application of FTIRS to investigate cartilage under microgravity. Simulated microgravity cartilage model was firstly established by tail-suspension (TS) for 7, 14 and 21 days, which would be compared to control samples. A self-developed hollow optical fiber attenuated total reflection (HOF-ATR) probe coupled with a FTIR spectrometer was used for the spectral acquisition of cartilage samples in situ, and one-way analysis of variance (ANOVA) was employed to analyze the changes in the contents of cartilage matrix at different stages. The results indicate that cartilage degenerates in microgravity, the collagen content gradually decreases with the TS time, and the structure of collagen fibers changes. The trends of proteoglycan content and collagen integrity show an initial decrease followed by an increase, ultimately significantly decreasing. The findings provide the basis for the cartilage degeneration in microgravity with TS time, which must be of real significance for space science and health detection.


Subject(s)
Cartilage, Articular , Collagen , Weightlessness Simulation , Spectroscopy, Fourier Transform Infrared/methods , Cartilage, Articular/pathology , Cartilage, Articular/chemistry , Cartilage, Articular/metabolism , Collagen/analysis , Collagen/metabolism , Collagen/chemistry , Animals , Proteoglycans/analysis , Male
2.
Biomaterials ; 312: 122720, 2025 Jan.
Article in English | MEDLINE | ID: mdl-39084098

ABSTRACT

Mesenchymal stem cells (MSCs) are expected to be useful therapeutics in osteoarthritis (OA), the most common joint disorder characterized by cartilage degradation. However, evidence is limited with regard to cartilage repair in clinical trials because of the uncontrolled differentiation and weak cartilage-targeting ability of MSCs after injection. To overcome these drawbacks, here we synthesized CuO@MSN nanoparticles (NPs) to deliver Sox9 plasmid DNA (favoring chondrogenesis) and recombinant protein Bmp7 (inhibiting hypertrophy). After taking up CuO@MSN/Sox9/Bmp7 (CSB NPs), the expressions of chondrogenic markers were enhanced while hypertrophic markers were decreased in response to these CSB-engineered MSCs. Moreover, a cartilage-targeted peptide (designated as peptide W) was conjugated onto the surface of MSCs via a click chemistry reaction, thereby prolonging the residence time of MSCs in both the knee joint cavity of mice and human-derived cartilage. In a surgery-induced OA mouse model, the NP and peptide dual-modified W-CSB-MSCs showed an enhancing therapeutic effect on cartilage repair in knee joints compared with other engineered MSCs after intra-articular injection. Most importantly, W-CSB-MSCs accelerated cartilage regeneration in damaged cartilage explants derived from OA patients. Thus, this new peptide and NPs dual engineering strategy shows potential for clinical applications to boost cartilage repair in OA using MSC therapy.


Subject(s)
Cell Differentiation , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells , Nanoparticles , Osteoarthritis , Peptides , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Animals , Osteoarthritis/therapy , Osteoarthritis/pathology , Nanoparticles/chemistry , Humans , Cell Differentiation/drug effects , Peptides/chemistry , Mesenchymal Stem Cell Transplantation/methods , Chondrogenesis/drug effects , Mice , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Cartilage, Articular/pathology , Cartilage, Articular/drug effects , Bone Morphogenetic Protein 7/chemistry , Bone Morphogenetic Protein 7/pharmacology , Tissue Engineering/methods , Regeneration/drug effects
4.
Sci Adv ; 10(37): eadp4408, 2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39259800

ABSTRACT

The rapid advancement of cell therapies underscores the importance of understanding fundamental cellular attributes. Among these, cell fitness-how transplanted cells adapt to new microenvironments and maintain functional stability in vivo-is crucial. This study identifies a chemical compound, FPH2, that enhances the fitness of human chondrocytes and the repair of articular cartilage, which is typically nonregenerative. Through drug screening, FPH2 was shown to broadly improve cell performance, especially in maintaining chondrocyte phenotype and enhancing migration. Single-cell transcriptomics indicated that FPH2 induced a super-fit cell state. The mechanism primarily involves the inhibition of carnitine palmitoyl transferase I and the optimization of metabolic homeostasis. In animal models, FPH2-treated human chondrocytes substantially improved cartilage regeneration, demonstrating well-integrated tissue interfaces in rats. In addition, an acellular FPH2-loaded hydrogel proved effective in preventing the onset of osteoarthritis. This research provides a viable and safe method to enhance chondrocyte fitness, offering insights into the self-regulatory mechanisms of cell fitness.


Subject(s)
Cartilage, Articular , Chondrocytes , Regeneration , Chondrocytes/metabolism , Chondrocytes/cytology , Chondrocytes/drug effects , Animals , Humans , Cartilage, Articular/metabolism , Rats , Osteoarthritis/metabolism , Osteoarthritis/therapy , Hydrogels/chemistry , Cell Movement/drug effects
5.
J Nanobiotechnology ; 22(1): 555, 2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39261846

ABSTRACT

BACKGROUND: The pathogenesis of osteoarthritis (OA) involves the progressive degradation of articular cartilage. Exosomes derived from mesenchymal stem cells (MSC-EXOs) have been shown to mitigate joint pathological injury by attenuating cartilage destruction. Optimization the yield and therapeutic efficacy of exosomes derived from MSCs is crucial for promoting their clinical translation. The preconditioning of MSCs enhances the therapeutic potential of engineered exosomes, offering promising prospects for application by enabling controlled and quantifiable external stimulation. This study aims to address these issues by employing pro-inflammatory preconditioning of MSCs to enhance exosome production and augment their therapeutic efficacy for OA. METHODS: The exosomes were isolated from the supernatant of infrapatellar fat pad (IPFP)-MSCs preconditioned with a pro-inflammatory factor, TNF-α, and their production was subsequently quantified. The exosome secretion-related pathways in IPFP-MSCs were evaluated through high-throughput transcriptome sequencing analysis, q-PCR and western blot analysis before and after TNF-α preconditioning. Furthermore, exosomes derived from TNF-α preconditioned IPFP-MSCs (IPFP-MSC-EXOsTNF-α) were administered intra-articularly in an OA mouse model, and subsequent evaluations were conducted to assess joint pathology and gait alterations. The expression of proteins involved in the maintenance of cartilage homeostasis within the exosomes was determined through proteomic analysis. RESULTS: The preconditioning with TNF-α significantly enhanced the exosome secretion of IPFP-MSCs compared to unpreconditioned MSCs. The potential mechanism involved the activation of the PI3K/AKT signaling pathway in IPFP-MSCs by TNF-α precondition, leading to an up-regulation of autophagy-related protein 16 like 1(ATG16L1) levels, which subsequently facilitated exosome secretion. The intra-articular administration of IPFP-MSC-EXOsTNF-α demonstrated superior efficacy in ameliorating pathological changes in the joints of OA mice. The preconditioning of TNF-α enhanced the up-regulation of low-density lipoprotein receptor-related protein 1 (LRP1) levels in IPFP-MSC-EXOsTNF-α, thereby exerting chondroprotective effects. CONCLUSION: TNF-α preconditioning constitutes an effective and promising method for optimizing the therapeutic effects of IPFP-MSCs derived exosomes in the treatment of OA.


Subject(s)
Exosomes , Mesenchymal Stem Cells , Osteoarthritis , Tumor Necrosis Factor-alpha , Exosomes/metabolism , Animals , Mesenchymal Stem Cells/metabolism , Tumor Necrosis Factor-alpha/metabolism , Mice , Osteoarthritis/therapy , Osteoarthritis/metabolism , Adipose Tissue/cytology , Mice, Inbred C57BL , Male , Disease Models, Animal , Cartilage, Articular/metabolism , Mesenchymal Stem Cell Transplantation/methods , Cells, Cultured , Humans
6.
J Orthop Surg Res ; 19(1): 550, 2024 Sep 09.
Article in English | MEDLINE | ID: mdl-39252111

ABSTRACT

Osteoarthritis (OA) is a chronic joint disease characterized by the degeneration, destruction, and excessive ossification of articular cartilage. The prevalence of OA is rising annually, concomitant with the aging global population and increasing rates of obesity. This condition imposes a substantial and escalating burden on individual health, healthcare systems, and broader social and economic frameworks. The etiology of OA is multifaceted and not fully understood. Current research suggests that the death of chondrocytes, encompassing mechanisms such as cellular apoptosis, pyroptosis, autophagy, ferroptosis and cuproptosis, contributes to both the initiation and progression of the disease. These cell death pathways not only diminish the population of chondrocytes but also exacerbate joint damage through the induction of inflammation and other deleterious processes. This paper delineates the morphological characteristics associated with various modes of cell death and summarizes current research results on the molecular mechanisms of different cell death patterns in OA. The objective is to review the advancements in understanding chondrocyte cell death in OA, thereby offering novel insights for potential clinical interventions.


Subject(s)
Cell Death , Chondrocytes , Disease Progression , Osteoarthritis , Chondrocytes/pathology , Humans , Osteoarthritis/pathology , Osteoarthritis/therapy , Cell Death/physiology , Apoptosis/physiology , Cartilage, Articular/pathology , Autophagy/physiology , Animals , Pyroptosis/physiology , Ferroptosis/physiology
7.
Int J Rheum Dis ; 27(9): e15323, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39221886

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a prevalent degenerative disease. We explored the role and regulatory mechanisms of lncRNA-FAS-AS1 in OA progression. METHODS: We exposed human immortalized chondrocytes to IL-1ß for 24 h to induce an OA cell model. The target molecule levels were assessed using western blot and quantitative real-time PCR (RT-qPCR). Cell viability and apoptosis were measured using CCK-8 and flow cytometry. The m6A modification of FAS-AS1 was determined using MeRIP. We examined the binding relationships between FAS-AS1, Fragile X mental retardation 1 (FMR1), and A disintegrin and metalloproteinase 8 (ADAM8) using RIP and RNA pull-down. The OA animal model was established by separating the medial collateral ligament and medial meniscus. Safranin-O staining and Mankin's scale were employed to evaluate pathological changes within the cartilage. RESULTS: FAS-AS1, METTL14, and ADAM8 were upregulated, and the JAK/STAT3 signaling pathway was activated in OA mice and IL-1ß-induced chondrocytes. FAS-AS1 knockdown inhibited extracellular matrix degradation in IL-1ß-induced chondrocytes; however, ADAM8 overexpression reversed this effect. FAS-AS1 maintained the stability of ADAM8 mRNA by recruiting FMR1. METTL14 knockdown repressed FAS-AS1 expression in an m6A-dependent manner. FAS-AS1 overexpression reversed the inhibitory effects of METTL14 knockdown on JAK/STAT3 signaling and cartilage damage in the OA model both in vitro and in vivo. CONCLUSION: METTL14-mediated FAS-AS1 promotes OA progression through the FMR1/ADAM8/JAK/STAT3 axis.


Subject(s)
ADAM Proteins , Chondrocytes , Disease Progression , Membrane Proteins , RNA, Long Noncoding , STAT3 Transcription Factor , Signal Transduction , Up-Regulation , Animals , Humans , Male , Mice , ADAM Proteins/metabolism , ADAM Proteins/genetics , Adenosine/analogs & derivatives , Apoptosis , Arthritis, Experimental/metabolism , Arthritis, Experimental/genetics , Arthritis, Experimental/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Cell Line , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Interleukin-1beta/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Methyltransferases/metabolism , Methyltransferases/genetics , Mice, Inbred C57BL , Osteoarthritis/metabolism , Osteoarthritis/genetics , Osteoarthritis/pathology , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/genetics , Osteoarthritis, Knee/pathology , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/genetics
8.
BMC Vet Res ; 20(1): 394, 2024 Sep 06.
Article in English | MEDLINE | ID: mdl-39242548

ABSTRACT

BACKGROUND: The dromedary camel (Camelus dromedarius) carpal joint presents multiple joints and constitutes several bones and soft tissues. Radiography and/or ultrasonography of the carpus are challenging due to structural superimposition. High-field magnetic resonance imaging (MRI) technique precludes superimposed tissues and offers high soft tissue contrast in multiple sequences and planes. Hence, understanding the normal MRI anatomy is crucial during clinical investigations. Magnetic resonance imaging is highly sensitive for investigation of soft tissues and articular cartilage; therefore, it is extensively used for outlining joint anatomy and evaluation of a wide range of musculoskeletal conditions. MRI images of a specific anatomical region acquired by using multiple sequences in various planes are necessary for a complete MRI examination. Given the dearth of information on the MRI features of the dromedary camel carpus, the current study demonstrates the MRI appearance of the clinically significant structures in the camel carpus in various sequences and planes using a high-field 1.5 Tesla superconducting magnet. For this purpose, twelve cadaveric forelimbs, obtained from 6 clinically sound lameness free adult dromedary camels, were examined. RESULTS: The cortex and medulla of the radius, carpal bones and metacarpus were evaluated. Articular cartilage of the carpal joints was depicted and showed intermediate intensity. Carpal tendons expressed lower signal intensity in all pulse sequences. The collateral and inter-carpal ligaments showed mixed signal intensity. CONCLUSIONS: The obtained data outlines the validation of MRI for investigation of the camel carpus and could set as a reference for interpretation in clinical patients.


Subject(s)
Camelus , Magnetic Resonance Imaging , Animals , Camelus/anatomy & histology , Magnetic Resonance Imaging/veterinary , Magnetic Resonance Imaging/methods , Carpus, Animal/diagnostic imaging , Carpus, Animal/anatomy & histology , Female , Male , Cartilage, Articular/diagnostic imaging , Cartilage, Articular/anatomy & histology
9.
Physiol Res ; 73(4): 633-642, 2024 Aug 31.
Article in English | MEDLINE | ID: mdl-39264083

ABSTRACT

The study aims to elucidate the therapeutic mechanism of Baicalin (BAI) in alleviating cartilage injury in osteoarthritic (OA) rat models, concentrating on its regulation of the miR-766-3p/AIFM1 axis. An OA rat model was developed with unilateral anterior cruciate ligament transection (ACLT). Interventions comprised of BAI treatment and intra-articular administration of miR-766-3p inhibitor. For evaluation, histopathological staining was conducted to investigate the pathological severity of knee cartilage injury. The levels of oxidative stress (OS) indicators including MDA, SOD, and GSH-Px, were quantified using colorimetric assays. Inflammatory factors (IFs; TNF-?, IL-1?, and IL-6) in knee joint lavage fluids were assessed using ELISA, while RT-PCR was employed to quantify miR-766-3p expression. TUNEL apoptosis staining was utilized to detect chondrocyte apoptosis, and western blotting examined autophagy-related markers (LC3, Beclin, p62), extracellular matrix (ECM) synthesis-associated indices (COL2A, ACAN, MMP13), and apoptosis-inducing factor mitochondrion-associated 1 (AIFM1). Histological examination revealed a marked amelioration of cartilage injury in the BAI-treated OA rat models compared to controls. BAI treatment significantly reduced inflammation and OS of knee joint fluid, activated autophagy, and decreased chondrocyte apoptosis and ECM degradation. Interestingly, the inhibitory effects of BAI on these pathological markers were significantly decreased by the miR-766-3p inhibitor. Further assessment revealed that BAI efficiently promoted miR-766-3p expression while inhibiting AIFM1 protein expression. BAI potentially mitigates articular cartilage injury in OA rats, likely through modulation of miR-766-3p/AIFM1 axis. Keywords: Baicalin, microRNA, AIFM1, Osteoarthritisv, Rat.


Subject(s)
Flavonoids , MicroRNAs , Rats, Sprague-Dawley , Animals , Flavonoids/pharmacology , Flavonoids/therapeutic use , MicroRNAs/metabolism , MicroRNAs/genetics , MicroRNAs/biosynthesis , Rats , Male , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Apoptosis Inducing Factor/metabolism , Oxidative Stress/drug effects , Apoptosis/drug effects
10.
Nutrients ; 16(17)2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39275306

ABSTRACT

The extracellular matrix of cartilage primarily constitutes of collagen and aggrecan. Cartilage degradation starts with aggrecan loss in osteoarthritis (OA). Vitamin D (VD) plays an essential role in several inflammation-related diseases and can protect the collagen in cartilage during OA. The present study focused on the role of VD in aggrecan turnover of human articular chondrocytes treated with tumor necrosis factor α (TNF-α) and the possible mechanism. Treatment with different doses of VD and different periods of intervention with TNF-α and TGF-ß1 receptor (TGFßR1) inhibitor SB525334 were investigated. The viability of human chondrocytes and extracellular secretion of TGF-ß1 were measured. The expression of intracellular TGFßR1 and VD receptor was examined. Transcriptional and translational levels of aggrecan and the related metabolic factors were analyzed. The results showed that TNF-α markedly reduced the viability, TGFßR1 expressions and aggrecan levels of human chondrocytes, and increased disintegrin and metalloproteinase with thrombospondin motifs. The alterations were partially inhibited by VD treatment. Furthermore, the effects of VD were blocked by the TGFßR1 inhibitor SB525334 in TNF-α-treated cells. VD may prevent proteoglycan loss due to TNF-α via TGF-ß1 signaling in human chondrocytes.


Subject(s)
Aggrecans , Cartilage, Articular , Chondrocytes , Proteoglycans , Signal Transduction , Transforming Growth Factor beta1 , Tumor Necrosis Factor-alpha , Vitamin D , Humans , Chondrocytes/metabolism , Chondrocytes/drug effects , Signal Transduction/drug effects , Transforming Growth Factor beta1/metabolism , Aggrecans/metabolism , Tumor Necrosis Factor-alpha/metabolism , Vitamin D/pharmacology , Proteoglycans/metabolism , Proteoglycans/pharmacology , Cartilage, Articular/metabolism , Cartilage, Articular/drug effects , Cells, Cultured , Cell Survival/drug effects , Osteoarthritis/metabolism , Receptor, Transforming Growth Factor-beta Type I/metabolism , Receptors, Calcitriol/metabolism
11.
J Cell Mol Med ; 28(18): e70095, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39289794

ABSTRACT

EphrinB2, a member of the Ephrin family, has been linked to several orthopaedic conditions. Nevertheless, the correlation between ephrinB2 and post-traumatic arthritis (PTOA) remains unclear. Human PTOA cartilage from human and mouse knee joints was systematically analysed to investigate the relationship between EphrinB2 and PTOA using SO-FG and toluidine blue staining, micro-CT, histomorphometry, immunohistochemistry, immunofluorescence, lentiviral articular injection and in situ end labeling (TUNEL) assays. EphrinB2 expression was significantly downregulated in PTOA chondrocytes. Blocking EphrinB2 increased the breakdown of cartilage matrix in mice with PTOA via reducing the process of chondrocyte autophagy. The presence of severe cartilage damage was evident, as indicated by a considerable decrease in both cartilage thickness and area, accompanied by an increase in chondrocyte death. Altogether, EphrinB2 is required for the maintenance of cartilage homeostasis in post-traumatic arthritis, and EphrinB2 ablation is associated with accelerated chondrocyte matrix degeneration, finally causing damage to the articular cartilage.


Subject(s)
Autophagy , Cartilage, Articular , Chondrocytes , Ephrin-B2 , Homeostasis , Chondrocytes/metabolism , Chondrocytes/pathology , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Ephrin-B2/metabolism , Ephrin-B2/genetics , Humans , Mice , Male , Mice, Inbred C57BL , Female
12.
Int J Mol Sci ; 25(18)2024 Sep 13.
Article in English | MEDLINE | ID: mdl-39337384

ABSTRACT

Hemophilia, which is a rare disease, results from congenital deficiencies of coagulation factors VIII and IX, respectively, leading to spontaneous bleeding into joints, resulting in hemophilic arthropathy (HA). HA involves complex processes, including synovial proliferation, angiogenesis, and tissue remodeling. Despite ongoing research, factors contributing to HA progression, especially in adults with severe HA experiencing joint pain, remain unclear. Blood markers, particularly collagen-related ones, have been explored to assess joint health in hemophilia. For example, markers like CTX-I and CTX-II reflect bone and cartilage turnover, respectively. Studies indicate elevated levels of certain markers post-bleeding episodes, suggesting joint health changes. However, longitudinal studies on collagen turnover and basement membrane or endothelial cell markers in relation to joint outcomes, particularly during painful episodes, are scarce. Given the role of the CX3CL1/CX3XR1 axis in arthritis, other studies investigate its involvement in HA. The importance of different inflammatory and bone damage biomarkers should be assessed, alongside articular cartilage and synovial membrane morphology, aiming to enhance understanding of hemophilic arthropathy progression.


Subject(s)
Biomarkers , Hemophilia A , Humans , Hemophilia A/complications , Hemophilia A/metabolism , Hemophilia A/pathology , Hemarthrosis/metabolism , Hemarthrosis/etiology , Hemarthrosis/pathology , Joint Diseases/metabolism , Joint Diseases/pathology , Joint Diseases/etiology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Synovial Membrane/metabolism , Synovial Membrane/pathology
13.
Int J Mol Sci ; 25(18)2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39337473

ABSTRACT

Cartilage repair remains a major challenge in human orthopedic medicine, necessitating the application of innovative strategies to overcome existing technical and clinical limitations. Adhesive hydrogels have emerged as promising candidates for cartilage repair promotion and tissue engineering, offering key advantages such as enhanced tissue integration and therapeutic potential. This comprehensive review navigates the landscape of adhesive hydrogels in cartilage repair, discussing identified challenges, shortcomings of current treatment options, and unique advantages of adhesive hydrogel products and scaffolds. While emphasizing the critical need for in situ lateral integration with surrounding tissues, we dissect current limitations and outline future perspectives for hydrogel scaffolds in cartilage repair. Moreover, we examine the clinical translation pathway and regulatory considerations specific to adhesive hydrogels. Overall, this review synthesizes the existing insights and knowledge gaps and highlights directions for future research regarding adhesive hydrogel-based devices in advancing cartilage tissue engineering.


Subject(s)
Hydrogels , Tissue Engineering , Tissue Scaffolds , Hydrogels/chemistry , Humans , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Cartilage, Articular , Cartilage/metabolism , Tissue Adhesives/chemistry , Tissue Adhesives/pharmacology
14.
Nutrients ; 16(18)2024 Sep 14.
Article in English | MEDLINE | ID: mdl-39339707

ABSTRACT

BACKGROUND: Hesperetin, a flavonoid derived from citrus fruits, exhibits potent antioxidant and anti-inflammatory activities and has been implicated in cartilage protection. However, its effectiveness against T-2 toxin-induced knee cartilage damage remains unclear. METHODS: In this study, high-throughput sequencing analysis was employed to identify the key signaling pathways involved in T-2 toxin-induced articular cartilage damage in rats. Animal models were divided into the following groups: control, low-dose T-2 toxin, high-dose T-2 toxin, T-2 toxin + hesperetin, hesperetin, and vehicle. Pathological staining and immunohistochemistry were used to assess pathological changes, as well as the expression levels of the cartilage matrix-related proteins MMP13 and collagen II, along with the activation of the p38 MAPK signaling pathway. Additionally, primary rat chondrocytes were cultured to establish an in vitro model for investigating the underlying mechanism. RESULTS: High-throughput sequencing analysis revealed the involvement of the MAPK signaling pathway in T-2 toxin-induced articular cartilage damage in rats. Hesperetin intervention in T-2 toxin-exposed rats attenuated pathological cartilage damage. Immunohistochemistry results demonstrated a significant reduction in collagen II protein expression in the high-dose T-2 toxin group (p < 0.01), accompanied by a significant increase in MMP13 protein expression (p < 0.01). In both the articular cartilage and the epiphyseal plate, the T-2 toxin + hesperetin group exhibited significantly higher collagen II protein expression than the high-dose T-2 toxin group (p < 0.05), along with significantly lower MMP13 protein expression (p < 0.05). Hesperetin inhibited the over-activation of the p38/MEF2C signaling axis induced by T-2 toxin in primary rat chondrocytes. Compared to the T-2 toxin group, the T-2 toxin + hesperetin group showed significantly reduced phosphorylation levels of p38 and protein expression levels of MEF2C (p < 0.001 or p < 0.05). Moreover, the T-2 toxin + hesperetin group exhibited a significant decrease in MMP13 protein expression (p < 0.05) and a significant increase in collagen II protein expression (p < 0.01) compared to the T-2 toxin group. CONCLUSIONS: T-2 toxin activates the p38 MAPK signaling pathway, causing knee cartilage damage in rats. Treatment with hesperetin inhibits the p38/MEF2C signaling axis, regulates collagen II and MMP13 protein expression, and reduces cartilage injury significantly.


Subject(s)
Cartilage, Articular , Chondrocytes , Hesperidin , T-2 Toxin , p38 Mitogen-Activated Protein Kinases , Animals , Male , Rats , Cartilage, Articular/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/metabolism , Cells, Cultured , Chondrocytes/drug effects , Chondrocytes/metabolism , Collagen Type II/metabolism , Disease Models, Animal , Hesperidin/pharmacology , MAP Kinase Signaling System/drug effects , Matrix Metalloproteinase 13/metabolism , Matrix Metalloproteinase 13/genetics , p38 Mitogen-Activated Protein Kinases/metabolism , Rats, Sprague-Dawley , Signal Transduction/drug effects , T-2 Toxin/toxicity
15.
Stem Cell Res Ther ; 15(1): 308, 2024 Sep 16.
Article in English | MEDLINE | ID: mdl-39285485

ABSTRACT

BACKGROUND: Articular cartilage degeneration can result from injury, age, or arthritis, causing significant joint pain and disability without surgical intervention. Currently, the only FDA cell-based therapy for articular cartilage injury is Autologous Chondrocyte Implantation (ACI); however, this procedure is costly, time-intensive, and requires multiple treatments. Mesenchymal stromal cells (MSCs) are an attractive alternative autologous therapy due to their availability and ability to robustly differentiate into chondrocytes for transplantation with good safety profiles. However, treatment outcomes are variable due to donor-to-donor variability as well as intrapopulation heterogeneity and unstandardized MSC manufacturing protocols. Process improvements that reduce cell heterogeneity while increasing donor cell numbers with improved chondrogenic potential during expansion culture are needed to realize the full potential of MSC therapy. METHODS: In this study, we investigated the potential of MSC metabolic modulation during expansion to enhance their chondrogenic commitment by varying the nutrient composition, including glucose, pyruvate, glutamine, and ascorbic acid in culture media. We tested the effect of metabolic modulation in short-term (one passage) and long-term (up to seven passages). We measured metabolic state, cell size, population doubling time, and senescence and employed novel tools including micro-magnetic resonance relaxometry (µMRR) relaxation time (T2) to characterize the effects of AA on improved MSC expansion and chondrogenic potential. RESULTS: Our data show that the addition of 1 mM L-ascorbic acid-2-phosphate (AA) to cultures for one passage during MSC expansion prior to initiation of differentiation improves chondrogenic differentiation. We further demonstrate that AA treatment reduced the proportion of senescent cells and cell heterogeneity also allowing for long-term expansion that led to a > 300-fold increase in yield of MSCs with enhanced chondrogenic potential compared to untreated cells. AA-treated MSCs with improved chondrogenic potential showed a robust shift in metabolic profile to OXPHOS and higher µMRR T2 values, identifying critical quality attributes that could be implemented in MSC manufacturing for articular cartilage repair. CONCLUSIONS: Our results suggest an improved MSC manufacturing process that can enhance chondrogenic potential by targeting MSC metabolism and integrating process analytic tools during expansion.


Subject(s)
Cartilage, Articular , Chondrocytes , Mesenchymal Stem Cells , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Cartilage, Articular/metabolism , Humans , Chondrocytes/metabolism , Chondrocytes/cytology , Chondrogenesis/drug effects , Cell Differentiation , Cells, Cultured , Cell Proliferation , Mesenchymal Stem Cell Transplantation/methods , Animals
16.
Am J Sports Med ; 52(11): 2782-2791, 2024 09.
Article in English | MEDLINE | ID: mdl-39276119

ABSTRACT

BACKGROUND: Matrix-induced autologous chondrocyte implantation (MACI), the third-generation of the technique, is an established procedure for the treatment of focal cartilage defects in the knee. However, the literature lacks long-term results of MACI with good statistical power. PURPOSE: To determine long-term survival and patient-reported outcomes (PROs) in a representative cohort and to identify patient- and surgery-related parameters that may influence long-term clinical outcomes. STUDY DESIGN: Case series; Level of evidence, 4. METHODS: A total of 103 patients were clinically evaluated at the current follow-up of 8.1 years (range, 5-11.9 years). PRO measures (PROMs) included the Knee injury and Osteoarthritis Outcome Score (KOOS), EQ-5D, visual analog scale for pain, and Tegner Activity Scale. Magnetic resonance imaging results were evaluated by using the AMADEUS (area measurement and depth and underlying structures) and MOCART (magnetic resonance observation of cartilage repair tissue) 2.0 knee score classification systems. Potential factors influencing PROs were first identified univariately and investigated in a multivariate regression model. RESULTS: The defects had a mean size of 4.8 cm2 (range, 1.2-12 cm2) and were predominantly femorotibial (66%). The mean Kaplan-Meier survival rate of revision for any reason was 97.2% ± 1.6% at 10 years. In comparison to preoperative values, all PROMs were significantly improved at the current follow-up (P < .05). The MOCART 2.0 score peaked at 12 months (mean, 80.2 ± 15.3 months) and showed no significant change at 96 months (mean, 76.1 ± 19.5 months; P = .142). The linear multivariate regression model identified an association of body mass index (BMI), MOCART 2.0 score, and number of previous knee surgeries with KOOS (R2 = 0.41; f2 = 0.69). Further analysis of the individual determinants revealed an optimal BMI range of 20 to 29 for favorable PROs at 96 months. Significant correlations of MOCART subscores with the overall KOOS were found for graft surface and structure, bony reaction, and subchondral detectable changes. Only 30% of patients with 2 previous surgeries and 20% of patients with 3 previous surgeries achieved a Patient Acceptable Symptom State (χ2 = 10.93; P = .012). CONCLUSION: The present study shows consistently good long-term clinical outcomes after MACI with a low revision rate and high patient satisfaction. BMI and number of previous knee surgeries may influence clinical outcomes and should be considered in patient selection and education. There is a correlation between graft structure, subchondral bone changes on magnetic resonance imaging, and long-term PROMs.


Subject(s)
Chondrocytes , Patient Reported Outcome Measures , Transplantation, Autologous , Humans , Chondrocytes/transplantation , Female , Male , Adult , Middle Aged , Young Adult , Cartilage, Articular/surgery , Cartilage, Articular/injuries , Knee Injuries/surgery , Follow-Up Studies , Magnetic Resonance Imaging , Adolescent , Treatment Outcome , Knee Joint/surgery
17.
Medicine (Baltimore) ; 103(38): e39586, 2024 Sep 20.
Article in English | MEDLINE | ID: mdl-39312372

ABSTRACT

Chronic ankle pain significantly impairs daily activities and athletic performance with osteochondral lesions of the talus (OLT) in Hepple stages IV and V, which are often causative factors. This study aimed to assess the efficacy and safety of autologous osteochondral transplantation (AOT) for the treatment of these conditions. This retrospective study was conducted from May 2020 to May 2023 at Cangzhou Traditional Chinese and Western Medicine Combined Hospital, including patients with a diagnosis of Hepple stage IV or V OLT confirmed by magnetic resonance imaging (MRI) and arthroscopy. Surgical interventions involved arthroscopic debridement, followed by AOT or limited arthrotomy based on the location and size of the lesion. Preoperative and postoperative evaluations used the Visual Analog Scale, American Orthopedic Foot and Ankle Society Ankle-Hindfoot Scale, MRI-Based Cartilage Repair Tissue Scoring, and the International Knee Documentation Committee Knee Evaluation Form. Statistical analysis was conducted using paired-sample t tests to compare the preoperative and postoperative data. Twenty patients were included, revealing significant postoperative improvements in Visual Analog Scale, American Orthopedic Foot and Ankle Society, and MRI-based cartilage repair tissue scores (P < .05). The radiographic findings suggested effective cartilage regeneration. No adverse effects were observed in the donor knee sites, as confirmed by the stable pre- and postoperative International Knee Documentation Committee Knee Evaluation Form scores. Recovery of physical abilities was achieved on average within 7.3 weeks for daily activities and 13.4 weeks for sports activities. AOT effectively treats Hepple stage IV-V OLT, improves ankle function, promotes cartilage regrowth, and allows quick resumption of daily and athletic activities without compromising donor-site integrity.


Subject(s)
Bone Transplantation , Chondrocytes , Ilium , Transplantation, Autologous , Humans , Retrospective Studies , Female , Male , Adult , Bone Transplantation/methods , Transplantation, Autologous/methods , Ilium/transplantation , Chondrocytes/transplantation , Periosteum/transplantation , Talus/surgery , Middle Aged , Cartilage, Articular/surgery , Arthroplasty, Subchondral/methods , Arthroscopy/methods , Magnetic Resonance Imaging , Debridement/methods , Treatment Outcome , Young Adult , Ankle Joint/surgery , Ankle Joint/diagnostic imaging
18.
Biomolecules ; 14(9)2024 Aug 26.
Article in English | MEDLINE | ID: mdl-39334835

ABSTRACT

Introduction: Osteoarthritis is a degenerative condition of the cartilage, often common among the population and occurs frequently with aging. Many factors are decisive for the development of its pathogenesis such as age, obesity, trauma, mechanical load, and modification of synovial biology. The main features of osteoarthritis are chondrocytes and cartilage matrix loss, which lead to pain, loss of function of the whole joint, and disability, representing a relevant health problem. Recently, a new therapeutic approach based on cell therapy has been studying the regenerative ability of mesenchymal stem cells for osteoarthritic chondrocytes. Aim: This in vitro study clarifies the regenerative effects of multipotent adipose-derived stem cells and the pluripotent amniotic epithelial stem cells on arthrosis chondrocytes by performing co-culture experiments. Methods: We studied the regenerative potential of secretome (soluble factors and extracellular vesicles), mesenchymal stem cells, and the adipose stromal vascular fraction. The regenerative effects were evaluated by gene and protein expression analysis of articular cartilage-specific genes and proteins like col2a1, acan, and sox9. Results: Mesenchymal stem cells, secretome, and adipose stromal vascular fractions influenced the cartilage genes and protein expression. Conclusions: The results indicate that the treatment with mesenchymal stem cells could be the best biological approach for cartilage regenerative medicine.


Subject(s)
Cartilage, Articular , Chondrocytes , Mesenchymal Stem Cells , Osteoarthritis , Secretome , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Humans , Secretome/metabolism , Chondrocytes/metabolism , Osteoarthritis/therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , SOX9 Transcription Factor/metabolism , SOX9 Transcription Factor/genetics , Coculture Techniques , Collagen Type II/metabolism , Collagen Type II/genetics , Aggrecans/metabolism , Aggrecans/genetics , Cells, Cultured
19.
Int J Mol Sci ; 25(18)2024 Sep 11.
Article in English | MEDLINE | ID: mdl-39337321

ABSTRACT

The existing in vitro and in vivo models for studying osteoarthritis have significant limitations in replicating the complexity of joint tissues. This research aims to validate a Tissue-On-a-Chip system for osteoarthritis research. Osteochondral tissues obtained from knee replacement surgeries of patients with osteoarthritis were cultured in an Organ-On-a-Chip system. This system was designed to supply oxygen and glucose to the cartilage from the bone. The distribution of oxygen and glucose was evaluated by fluorescence using Image-iT Green Hypoxia and 2-NBDG, respectively. Cytotoxicity was measured using lactate dehydrogenase (LDH) levels in chip cultures compared to plate cultures (12 tissues per method). Glycosaminoglycans (GAGs), alkaline phosphatase (ALP), Coll2-1, and procollagen type II N-terminal propeptide (PIINP) were measured in the perfused medium of the Tissue-On-a-Chip over a period of 70 days. Fluorescence of Image-iT Green Hypoxia was observed only in the cartilage area, while 2-NBDG was distributed throughout the tissue. An increase in LDH levels was noted in the plate cultures on day 24 and in the Tissue-On-a-Chip cultures on day 63. Compared to the start of the culture, GAG content increased on day 52, while ALP showed variations. A notable increase in GAG, ALP, and Coll2-1 levels was observed on day 59. PIINP levels remained stable throughout the experiment. The validated osteochondral Tissue-On-a-Chip system can replicate the joint microenvironment, with hypoxic conditions in cartilage and normoxic conditions in bone. Tissue survival and component stability were maintained for approximately two months. This platform is a useful tool for evaluating new drugs and represents a viable alternative to animal models.


Subject(s)
Lab-On-A-Chip Devices , Osteoarthritis , Humans , Osteoarthritis/metabolism , Osteoarthritis/pathology , Glycosaminoglycans/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Oxygen/metabolism , Glucose/metabolism , L-Lactate Dehydrogenase/metabolism , Alkaline Phosphatase/metabolism
20.
Nat Microbiol ; 9(10): 2538-2552, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39261580

ABSTRACT

Joint pain and osteoarthritis can occur as coronavirus disease 2019 (COVID-19) sequelae after infection. However, little is known about the damage to articular cartilage. Here we illustrate knee joint damage after wild-type, Delta and Omicron variants of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection in vivo. Rapid joint injury with cystic lesions at the osteochondral junction was observed in two patients with post-COVID osteoarthritis and recapitulated in a golden Syrian hamster model. SARS-CoV-2-activated endothelin-1 signalling increased vascular permeability and caused viral spike proteins leakage into the subchondral bone. Osteoclast activation, chondrocyte dropout and cyst formation were confirmed histologically. The US Food and Drug Administration-approved endothelin receptor antagonist, macitentan, mitigated cystic lesions and preserved chondrocyte number in the acute phase of viral infection in hamsters. Delayed macitentan treatment at post-acute infection phase alleviated chondrocyte senescence and restored subchondral bone loss. It is worth noting that it could also attenuate viral spike-induced joint pain. Our work suggests endothelin receptor blockade as a novel therapeutic strategy for post-COVID arthritis.


Subject(s)
COVID-19 , Disease Models, Animal , Endothelin Receptor Antagonists , Mesocricetus , Osteoarthritis , Pyrimidines , SARS-CoV-2 , Animals , SARS-CoV-2/drug effects , SARS-CoV-2/pathogenicity , Humans , COVID-19/virology , COVID-19/complications , COVID-19/metabolism , COVID-19/pathology , Osteoarthritis/drug therapy , Osteoarthritis/virology , Osteoarthritis/pathology , Osteoarthritis/metabolism , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Endothelin Receptor Antagonists/pharmacology , Endothelin Receptor Antagonists/therapeutic use , Sulfonamides/pharmacology , Cricetinae , Male , COVID-19 Drug Treatment , Chondrocytes/virology , Chondrocytes/metabolism , Chondrocytes/drug effects , Cartilage, Articular/pathology , Cartilage, Articular/virology , Cartilage, Articular/metabolism , Cartilage, Articular/drug effects , Receptors, Endothelin/metabolism , Endothelin-1/metabolism , Female , Spike Glycoprotein, Coronavirus/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL