Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 264
Filtrar
1.
Life Sci ; 346: 122630, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38614296

RESUMEN

Bone remodeling is the balance between osteoblasts and osteoclasts. Bone diseases such as osteoporosis and osteoarthritis are associated with imbalanced bone remodeling. Skeletal injury leads to limited motor function and pain. Neurophilin was initially identified in axons, and its various ligands and roles in bone remodeling, angiogenesis, neuropathic pain and immune regulation were later discovered. Neurophilin promotes osteoblast mineralization and inhibits osteoclast differentiation and its function. Neuropolin-1 provides channels for immune cell chemotaxis and cytokine diffusion and leads to pain. Neuropolin-1 regulates the proportion of T helper type 17 (Th17) and regulatory T cells (Treg cells), and affects bone immunity. Vascular endothelial growth factors (VEGF) combine with neuropilin and promote angiogenesis. Class 3 semaphorins (Sema3a) compete with VEGF to bind neuropilin, which reduces angiogenesis and rejects sympathetic nerves. This review elaborates on the structure and general physiological functions of neuropilin and summarizes the role of neuropilin and its ligands in bone and cartilage diseases. Finally, treatment strategies and future research directions based on neuropilin are proposed.


Asunto(s)
Enfermedades Óseas , Neuropilinas , Humanos , Animales , Enfermedades Óseas/metabolismo , Enfermedades Óseas/fisiopatología , Neuropilinas/metabolismo , Neuropilinas/fisiología , Enfermedades de los Cartílagos/metabolismo , Enfermedades de los Cartílagos/fisiopatología , Remodelación Ósea/fisiología
2.
J Cell Mol Med ; 28(7): e18242, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38509736

RESUMEN

Articular cartilage defect is challenged by insufficient regenerative ability of cartilage. Catalpol (CA), the primary active component of Rehmanniae Radix, could exert protective effects against various diseases. However, the impact of CA on the treatment of articular cartilage injuries is still unclear. In this study, full-thickness articular cartilage defect was induced in a mouse model via surgery. The animals were intraperitoneally injected with CA for 4 or 8 weeks. According to the results of macroscopic observation, micro-computed tomography CT (µCT), histological and immunohistochemistry staining, CA treatment could promote mouse cartilage repair, resulting in cartilage regeneration, bone structure improvement and matrix anabolism. Specifically, an increase in the expression of CD90, the marker of mesenchymal stem cells (MSCs), in the cartilage was observed. In addition, we evaluated the migratory and chondrogenic effects of CA on MSCs. Different concentration of CA was added to C3H10 T1/2 cells. The results showed that CA enhanced cell migration and chondrogenesis without affecting proliferation. Collectively, our findings indicate that CA may be effective for the treatment of cartilage defects via stimulation of endogenous MSCs.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Glucósidos Iridoides , Trasplante de Células Madre Mesenquimatosas , Células Madre Mesenquimatosas , Animales , Ratones , Cartílago Articular/patología , Microtomografía por Rayos X , Diferenciación Celular , Enfermedades de los Cartílagos/metabolismo , Trasplante de Células Madre Mesenquimatosas/métodos , Condrogénesis
3.
Arthritis Res Ther ; 26(1): 66, 2024 Mar 11.
Artículo en Inglés | MEDLINE | ID: mdl-38468277

RESUMEN

BACKGROUND: CRISPR-Cas9-based genome engineering represents a powerful therapeutic tool for cartilage tissue engineering and for understanding molecular pathways driving cartilage diseases. However, primary chondrocytes are difficult to transfect and rapidly dedifferentiate during monolayer (2D) cell culture, making the lengthy expansion of a single-cell-derived edited clonal population not feasible. For this reason, functional genetics studies focused on cartilage and rheumatic diseases have long been carried out in cellular models that poorly recapitulate the native molecular properties of human cartilaginous tissue (e.g., cell lines, induced pluripotent stem cells). Here, we set out to develop a non-viral CRISPR-Cas9, bulk-gene editing method suitable for chondrocyte populations from different cartilaginous sources. METHODS: We screened electroporation and lipid nanoparticles for ribonucleoprotein (RNP) delivery in primary polydactyly chondrocytes, and optimized RNP reagents assembly. We knocked out RELA (also known as p65), a subunit of the nuclear factor kappa B (NF-κB), in polydactyly chondrocytes and further characterized knockout (KO) cells with RT-qPCR and Western Blot. We tested RELA KO in chondrocytes from diverse cartilaginous sources and characterized their phenotype with RT-qPCR. We examined the chondrogenic potential of wild-type (WT) and KO cell pellets in presence and absence of interleukin-1ß (IL-1ß). RESULTS: We established electroporation as the optimal transfection technique for chondrocytes enhancing transfection and editing efficiency, while preserving high cell viability. We knocked out RELA with an unprecedented efficiency of ~90%, confirming lower inflammatory pathways activation upon IL-1ß stimulation compared to unedited cells. Our protocol could be easily transferred to primary human chondrocytes harvested from osteoarthritis (OA) patients, human FE002 chondroprogenitor cells, bovine chondrocytes, and a human chondrocyte cell line, achieving comparable mean RELA KO editing levels using the same protocol. All KO pellets from primary human chondrocytes retained chondrogenic ability equivalent to WT cells, and additionally displayed enhanced matrix retention under inflamed conditions. CONCLUSIONS: We showcased the applicability of our bulk gene editing method to develop effective autologous and allogeneic off-the-shelf gene therapies strategies and to enable functional genetics studies in human chondrocytes to unravel molecular mechanisms of cartilage diseases.


Asunto(s)
Enfermedades de los Cartílagos , Polidactilia , Humanos , Animales , Bovinos , Condrocitos/metabolismo , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Interleucina-1beta/metabolismo , Enfermedades de los Cartílagos/metabolismo , Polidactilia/metabolismo
4.
Int J Mol Sci ; 24(23)2023 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-38069268

RESUMEN

The effective and long-term treatment of cartilage defects is an unmet need among patients worldwide. In the past, several synthetic and natural biomaterials have been designed to support functional articular cartilage formation. However, they have mostly failed to enhance the terminal stage of chondrogenic differentiation, leading to scar tissue formation after the operation. Growth factors substantially regulate cartilage regeneration by acting on receptors to trigger intracellular signaling and cell recruitment for tissue regeneration. In this study, we investigated the effect of recombinant insulin-like growth factor 1 (rIGF-1), loaded in fibrin microbeads (FibIGF1), on cartilage regeneration. rIGF-1-loaded fibrin microbeads were injected into full-thickness cartilage defects in the knees of goats. The stability, integration, and quality of tissue repair were evaluated at 1 and 6 months by gross morphology, histology, and collagen type II staining. The in vivo results showed that compared to plain fibrin samples, particularly at 6 months, FibIGF1 improved the functional cartilage formation, confirmed through gross morphology, histology, and collagen type II immunostaining. FibIGF1 could be a promising candidate for cartilage repair in the clinic.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Humanos , Animales , Colágeno Tipo II/metabolismo , Fibrina/metabolismo , Cabras , Cartílago Articular/metabolismo , Enfermedades de los Cartílagos/metabolismo , Condrocitos
5.
Int J Mol Sci ; 24(19)2023 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-37834131

RESUMEN

Osteoarthritis (OA) is a worldwide joint disease. However, the precise mechanism causing OA remains unclear. Our primary aim was to identify vital biomarkers associated with the mechano-inflammatory aspect of OA, providing potential diagnostic and therapeutic targets for OA. Thirty OA patients who underwent total knee arthroplasty were recruited, and cartilage samples were obtained from both the lateral tibial plateau (LTP) and medial tibial plateau (MTP). GO and KEGG enrichment analyses were performed, and the protein-protein interaction (PPI) assessment was conducted for hub genes. The effect of PSD95 inhibition on cartilage degeneration was also conducted and analyzed. A total of 1247 upregulated and 244 downregulated DEGs were identified. Significant differences were observed between MTP and LTP in mechanical stress-related genes and activated sensory neurons based on a self-contrast model of human knee OA. Cluster analysis identified DLG4 as the hub gene. Cyclic loading stress increased PSD95 (encoded by DLG4) expression in LTP cartilage, and PSD95 inhibitors could alleviate OA progression. This study suggests that inhibiting PSD95 could be a potential therapeutic strategy for preventing articular cartilage degradation.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Osteoartritis de la Rodilla , Humanos , Osteoartritis de la Rodilla/genética , Osteoartritis de la Rodilla/metabolismo , Articulación de la Rodilla/metabolismo , Enfermedades de los Cartílagos/metabolismo , Tibia , Factores de Transcripción/metabolismo
6.
Int J Mol Sci ; 24(20)2023 Oct 13.
Artículo en Inglés | MEDLINE | ID: mdl-37894837

RESUMEN

This study performs an analysis that will enable the evaluation of the quality, durability, and structure of repaired cartilaginous extracellular matrix tissue using an autologous-based particulated autograft cartilage and platelet-rich plasma treatment (PACI + PRP). A single-blind controlled experiment was conducted on 28 sheep to evaluate the efficacy of the PACI + PRP treatment for cartilage defects. Full-thickness 8 mm diameter defects were created in the weight-bearing area of both knees. The right knees received PACI + PRP. The left knees were treated with Ringer's lactate solution (RLS) or hyaluronic acid (HA) injections. Sheep were euthanized at 9- or 18-months post-surgery. An extensive immunohistochemical analysis was performed to assess collagen types (I, II, III, V, VI, IX, X, XI) and aggrecan positivity. A semiquantitative scoring system provided a detailed evaluation of immunostaining. Collagens and aggrecan scores in the PACI + PRP groups were similar to healthy cartilage. Significant differences were found in collagens associated with matrix maturity (II and V), degradation (IX), structure and mechanics (VI), and hypertrophy (X) between healthy cartilage and RLS- or HA-repaired cartilage. The PACI + PRP treatment advanced the repair cartilage process in chondral defects with mature hyaline cartilage and enhanced the structural and mechanical qualities with better consistent cartilage, less susceptible to degradation and without hypertrophic formation over time.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Plasma Rico en Plaquetas , Animales , Ovinos , Cartílago Articular/cirugía , Agrecanos/genética , Agrecanos/metabolismo , Método Simple Ciego , Enfermedades de los Cartílagos/metabolismo , Plasma Rico en Plaquetas/metabolismo , Ácido Hialurónico/metabolismo , Colágeno/metabolismo
7.
Int J Mol Sci ; 24(20)2023 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-37894859

RESUMEN

Mesenchymal stem cell (MSC)-based exosomes have garnered attention as a viable therapeutic for post-traumatic cartilage injury and osteoarthritis of the knee; however, efforts for application have been limited due to issues with variable dosing and rapid clearance in vivo. Scaffolds laden with MSC-based exosomes have recently been investigated as a solution to these issues. Here, we review in vivo studies and highlight key strengths and potential clinical uses of exosome-scaffold therapeutics for treatment of post-traumatic cartilage injury and osteoarthritis. In vivo animal studies were gathered using keywords related to the topic, revealing 466 studies after removal of duplicate papers. Inclusion and exclusion criteria were applied for abstract screening and full-text review. Thirteen relevant studies were identified for analysis and extraction. Three predominant scaffold subtypes were identified: hydrogels, acellular extracellular matrices, and hyaluronic acid. Each scaffold-exosome design showcased unique properties with relation to gross findings, tissue histology, biomechanics, and gene expression. All designs demonstrated a reduction in inflammation and induction of tissue regeneration. The results of our review show that current exosome-scaffold therapeutics demonstrate the capability to halt and even reverse the course of post-traumatic cartilage injury and osteoarthritis. While this treatment modality shows incredible promise, future research should aim to characterize long-term biocompatibility and optimize scaffold designs for human treatment.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Exosomas , Osteoartritis de la Rodilla , Animales , Humanos , Osteoartritis de la Rodilla/patología , Exosomas/metabolismo , Enfermedades de los Cartílagos/metabolismo , Articulación de la Rodilla/patología , Cartílago/patología , Cartílago Articular/patología , Andamios del Tejido
8.
Int J Mol Sci ; 24(14)2023 Jul 18.
Artículo en Inglés | MEDLINE | ID: mdl-37511339

RESUMEN

The treatment of chondral and osteochondral defects is challenging. These types of lesions are painful and progress to osteoarthritis over time. Tissue engineering offers tools to address this unmet medical need. The use of an autologous cartilage construct consisting of hyaline cartilage chips embedded in plasma rich in growth factors (PRGF) has been proposed as a therapeutic alternative. The purpose of this study was to dig into the potential mechanisms behind the in vitro remodelling process that might explain the clinical success of this technique and facilitate its optimisation. Chondrocyte viability and cellular behaviour over eight weeks of in vitro culture, type II collagen synthesis, the dual delivery of growth factors by hyaline cartilage and PRGF matrix, and the ultrastructure of the construct and its remodelling were characterised. The main finding of this research is that the cartilage fragments embedded in the three-dimensional PRGF scaffold contain viable chondrocytes that are able to migrate into the fibrin network, proliferate and synthesise extracellular matrix after the second week of in vitro culture. The characterization of this three-dimensional matrix is key to unravelling the molecular kinetics responsible for its efficacy.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Humanos , Cartílago Hialino/metabolismo , Condrocitos/metabolismo , Enfermedades de los Cartílagos/metabolismo , Plasma , Ingeniería de Tejidos/métodos
9.
Int J Mol Sci ; 24(13)2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37445948

RESUMEN

Osteoarthritis (OA) is the most common form of arthritis and joint disorder worldwide. Metabolic reprogramming of osteoarthritic chondrocytes from oxidative phosphorylation to glycolysis results in the accumulation of lactate from glycolytic metabolite pyruvate by lactate dehydrogenase A (LDHA), leading to cartilage degeneration. In the present study, we investigated the protective effects of the intra-articular administration of oxamate (LDHA inhibitor) against OA development and glycolysis-related protein expression in experimental OA rats. The animals were randomly allocated into four groups: Sham, anterior cruciate ligament transection (ACLT), ACLT + oxamate (0.25 and 2.5 mg/kg). Oxamate-treated groups received an intra-articular injection of oxamate once a week for 5 weeks. Intra-articular oxamate significantly reduced the weight-bearing defects and knee width in ACLT rats. Histopathological analyses showed that oxamate caused significantly less cartilage degeneration in the ACLT rats. Oxamate exerts hypertrophic effects in articular cartilage chondrocytes by inhibiting glucose transporter 1, glucose transporter 3, hexokinase II, pyruvate kinase M2, pyruvate dehydrogenase kinases 1 and 2, pyruvate dehydrogenase kinase 2, and LHDA. Further analysis revealed that oxamate significantly reduced chondrocyte apoptosis in articular cartilage. Oxamate attenuates nociception, inflammation, cartilage degradation, and chondrocyte apoptosis and possibly attenuates glycolysis-related protein expression in ACLT-induced OA rats. The present findings will facilitate future research on LDHA inhibitors in prevention strategies for OA progression.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Osteoartritis , Ratas , Animales , Lactato Deshidrogenasa 5/metabolismo , Nocicepción , Osteoartritis/metabolismo , Condrocitos/metabolismo , Cartílago Articular/metabolismo , Enfermedades de los Cartílagos/metabolismo , Modelos Animales de Enfermedad
10.
Int J Mol Sci ; 24(13)2023 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-37446002

RESUMEN

Proteoglycans are vital components of the extracellular matrix in articular cartilage, providing biomechanical properties crucial for its proper functioning. They are key players in chondral diseases, specifically in the degradation of the extracellular matrix. Evaluating proteoglycan molecules can serve as a biomarker for joint degradation in osteoarthritis patients, as well as assessing the quality of repaired tissue following different treatment strategies for chondral injuries. Despite ongoing research, understanding osteoarthritis and cartilage repair remains unclear, making the identification of key molecules essential for early diagnosis and effective treatment. This review offers an overview of proteoglycans as primary molecules in articular cartilage. It describes the various types of proteoglycans present in both healthy and damaged cartilage, highlighting their roles. Additionally, the review emphasizes the importance of assessing proteoglycans to evaluate the quality of repaired articular tissue. It concludes by providing a visual and narrative description of aggrecan distribution and presence in healthy cartilage. Proteoglycans, such as aggrecan, biglycan, decorin, perlecan, and versican, significantly contribute to maintaining the health of articular cartilage and the cartilage repair process. Therefore, studying these proteoglycans is vital for early diagnosis, evaluating the quality of repaired cartilage, and assessing treatment effectiveness.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Osteoartritis , Humanos , Agrecanos/metabolismo , Cartílago Articular/metabolismo , Decorina/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Biglicano/metabolismo , Osteoartritis/diagnóstico , Osteoartritis/metabolismo , Enfermedades de los Cartílagos/metabolismo , Lectinas Tipo C/metabolismo
11.
Life Sci ; 326: 121804, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37245839

RESUMEN

Fibroblast growth factor 7 (FGF7), also known as keratinocyte growth factor (KGF), shows a crucial biological significance in tissue development, wound repair, tumorigenesis, and immune reconstruction. In the skeletal system, FGF7 directs the cellular synaptic extension of individual cells and facilities functional gap junction intercellular communication of a collective of cells. Moreover, it promotes the osteogenic differentiation of stem cells via a cytoplasmic signaling network. For cartilage, reports have indicated the potential role of FGF7 on the regulation of key molecules Cx43 in cartilage and Runx2 in hypertrophic cartilage. However, the molecular mechanism of FGF7 in chondrocyte behaviors and cartilage pathological process remains largely unknown. In this review, we systematically summarize the recent biological function of FGF7 and its regulatory role on chondrocytes and cartilage diseases, especially through the hot focus of two key molecules, Runx2 and Cx43. The current knowledge of FGF7 on the physiological and pathological processes of chondrocytes and cartilage provides us new cues for wound repair of cartilage defect and therapy of cartilage diseases.


Asunto(s)
Enfermedades de los Cartílagos , Factor 7 de Crecimiento de Fibroblastos , Humanos , Factor 7 de Crecimiento de Fibroblastos/metabolismo , Conexina 43/metabolismo , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Osteogénesis , Cartílago/metabolismo , Diferenciación Celular , Condrocitos/metabolismo , Enfermedades de los Cartílagos/metabolismo , Enfermedades de los Cartílagos/patología
12.
Biomater Adv ; 137: 212845, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35929274

RESUMEN

BACKGROUND: Tissue engineering using adipose-derived mesenchymal stem cells (ADSCs) promotes the regeneration of articular cartilage. However, insulin-like growth factor 1 (IGF-1), which is used to induce the differentiation of ADSCs into chondrocytes during treatment, is prone to instability and short tissue retention. METHODS: Nap-FFG-GYGSSSRRAPQT was used as an IGF-1 mimicking molecule. MTT and CCK-8 assays were performed to evaluate the proliferation ability of ADSCs. QRT-PCR and Western blot assays were used to assess the expression of cartilage-related genes. International Cartilage Regeneration and Joint Preservation Society (ICRS) scoring was used for the evaluation of cartilage repair. Repaired tissues were analyzed by hematoxylin-eosin, Safranin-O and immunohistochemical staining. RESULTS: Nap-FFG-GYGSSRRAPQT stimulated the proliferation and migration of ADSCs through the activation of IGF-1 receptor. Gel Nap-FFG-GYGSSRRAPQT treatment upregulated the expression of cartilage-related genes in ADSCs. ADSCs/Gel Nap-FFG-GYGSSRRAPQT treatment significantly promoted the regeneration of cartilages. CONCLUSION: Self-assembled IGF-1 peptide, Nap-FFG-GYGSSRRAPQT, can induce ADSC differentiation and proliferation to repair cartilage injury.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Tejido Adiposo , Enfermedades de los Cartílagos/metabolismo , Cartílago Articular/fisiología , Diferenciación Celular , Humanos , Factor I del Crecimiento Similar a la Insulina/genética
13.
Arthritis Res Ther ; 24(1): 174, 2022 07 22.
Artículo en Inglés | MEDLINE | ID: mdl-35869508

RESUMEN

Osteoarthritis (OA) is an age-related cartilage degenerative disease, and chondrocyte senescence has been extensively studied in recent years. Increased numbers of senescent chondrocytes are found in OA cartilage. Selective clearance of senescent chondrocytes in a post-traumatic osteoarthritis (PTOA) mouse model ameliorated OA development, while intraarticular injection of senescent cells induced mouse OA. However, the means and extent to which senescence affects OA remain unclear. Here, we review the latent mechanism of senescence in OA and propose potential therapeutic methods to target OA-related senescence, with an emphasis on immunotherapies. Natural killer (NK) cells participate in the elimination of senescent cells in multiple organs. A relatively comprehensive discussion is presented in that section. Risk factors for OA are ageing, obesity, metabolic disorders and mechanical overload. Determining the relationship between known risk factors and senescence will help elucidate OA pathogenesis and identify optimal treatments.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Osteoartritis , Animales , Enfermedades de los Cartílagos/metabolismo , Cartílago Articular/patología , Senescencia Celular , Condrocitos/metabolismo , Ratones , Osteoartritis/metabolismo
14.
Osteoarthritis Cartilage ; 30(8): 1116-1129, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35569800

RESUMEN

OBJECTIVE: Hip osteoarthritis (OA) affects all components of the osteochondral unit, leading to bone marrow (BM) lesions, and unknown consequences on BM cell functionality. We analyzed the cellular composition in OA-affected acetabula compared to proximal femur shafts obtained of hip OA patients to reveal yet not explored immune and stem cell compartments. DESIGN: Combining flow cytometry, cellular assays and transcription analyses, we performed extensive ex vivo phenotyping of acetabular BM cells from 18 hip OA patients, comparing them with their counterparts from patient-matched femoral shaft BM samples. Findings were related to differences in skeletal sites and age. RESULTS: Acetabular BM had a greater frequency of T-lymphocytes, non-hematopoietic cells and colony-forming units fibroblastic potential than femoral BM. The incidence of acetabular CD45+CD3+ T-lymphocytes increased (95% CI: 0.1770 to 0.0.8416), while clonogenic hematopoietic progenitors declined (95% CI: -0.9023 to -0.2399) with age of patients. On the other side, in femoral BM, we observed higher B-lymphocyte, myeloid and erythroid cell frequencies. Acetabular mesenchymal stromal cells (MSCs) showed a senescent profile associated with the expression of survival and inflammation-related genes. Efficient osteogenic and chondrogenic differentiation was detected in acetabular MSCs, while adipogenesis was more pronounced in their femoral counterparts. CONCLUSION: Our results suggest that distinctions in BM cellular compartments and MSCs may be due to the influence of the OA-stressed microenvironment, but also acetabular vs femoral shaft-specific peculiarities cannot be excluded. These results bring new knowledge on acetabular BM cell populations and may be addressed as novel pathogenic mechanisms and therapeutic targets in OA.


Asunto(s)
Enfermedades de los Cartílagos , Osteoartritis de la Cadera , Acetábulo , Médula Ósea , Células de la Médula Ósea , Enfermedades de los Cartílagos/metabolismo , Diferenciación Celular , Humanos , Osteoartritis de la Cadera/metabolismo , Células Madre
15.
J Bone Miner Res ; 37(3): 397-410, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35124831

RESUMEN

Induced pluripotent stem cell (iPSC) technology allows pathomechanistic and therapeutic investigation of human heritable disorders affecting tissue types whose collection from patients is difficult or even impossible. Among them are cartilage diseases. Over the past decade, iPSC-chondrocyte disease models have been shown to exhibit several key aspects of known disease mechanisms. Concurrently, an increasing number of protocols to differentiate iPSCs into chondrocytes have been published, each with its respective (dis)advantages. In this review we provide a comprehensive overview of the different differentiation approaches, the hitherto described iPSC-chondrocyte disease models and mechanistic and/or therapeutic insights that have been derived from their investigation, and the current model limitations. Key lessons are that the most appropriate differentiation approach is dependent upon the cartilage disease under investigation and that further optimization is still required to recapitulate the in vivo cartilage. © 2022 American Society for Bone and Mineral Research (ASBMR).


Asunto(s)
Enfermedades de los Cartílagos , Células Madre Pluripotentes Inducidas , Cartílago/metabolismo , Enfermedades de los Cartílagos/metabolismo , Diferenciación Celular , Condrocitos/metabolismo , Descubrimiento de Drogas , Humanos
16.
Rheumatology (Oxford) ; 61(2): 856-864, 2022 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33989379

RESUMEN

OBJECTIVES: OA is a complex genetic disease with different risk factors contributing to its development. One of the genes, TNFRSF11B, previously identified with gain-of-function mutation in a family with early-onset OA with chondrocalcinosis, is among the highest upregulated genes in lesioned OA cartilage (RAAK-study). Here, we determined the role of TNFRSF11B overexpression in development of OA. METHODS: Human primary articular chondrocytes (9 donors RAAK study) were transduced using lentiviral particles with or without TNFRSF11B. Cells were cultured for 1 week in a 3 D in-vitro chondrogenic model. TNFRSF11B overexpression was confirmed by RT-qPCR, immunohistochemistry and ELISA. Effects of TNFRSF11B overexpression on cartilage matrix deposition, matrix mineralization, and genes highly correlated to TNFRSF11B in RNA-sequencing dataset (r >0.75) were determined by RT-qPCR. Additionally, glycosaminoglycans and collagen deposition were visualized with Alcian blue staining and immunohistochemistry (COL1 and COL2). RESULTS: Overexpression of TNFRSF11B resulted in strong upregulation of MMP13, COL2A1 and COL1A1. Likewise, mineralization and osteoblast characteristic markers RUNX2, ASPN and OGN showed a consistent increase. Among 30 genes highly correlated to TNFRSF11B, expression of only eight changed significantly, with BMP6 showing the highest increase (9-fold) while expression of RANK and RANKL remained unchanged indicating previously unknown downstream pathways of TNFRSF11B in cartilage. CONCLUSION: Results of our 3D in vitro chondrogenesis model indicate that upregulation of TNFRSF11B in lesioned OA cartilage may act as a direct driving factor for chondrocyte to osteoblast transition observed in OA pathophysiology. This transition does not appear to act via the OPG/RANK/RANKL triad common in bone remodeling.


Asunto(s)
Enfermedades de los Cartílagos/etiología , Osteoartritis/etiología , Osteoprotegerina/metabolismo , Anciano , Cartílago/metabolismo , Enfermedades de los Cartílagos/metabolismo , Células Cultivadas , Condrocitos/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Humanos , Osteoartritis/metabolismo , Reacción en Cadena de la Polimerasa
17.
Acta Biomater ; 140: 23-42, 2022 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-34896634

RESUMEN

The ability of articular cartilage to repair itself is limited because it lacks blood vessels, nerves, and lymph tissue. Once damaged, it can lead to joint swelling and pain, accelerating the progression of osteoarthritis. To date, complete regeneration of hyaline cartilage exhibiting mechanical properties remains an elusive goal, despite the many available technologies. The inflammatory milieu created by cartilage damage is critical for chondrocyte death and hypertrophy, extracellular matrix breakdown, ectopic bone formation, and progression of cartilage injury to osteoarthritis. In the inflammatory microenvironment, mesenchymal stem cells (MSCs) undergo aberrant differentiation, and chondrocytes begin to convert or dedifferentiate into cells with a fibroblast phenotype, thereby resulting in fibrocartilage with poor mechanical qualities. All these factors suggest that inflammatory problems may be a major stumbling block to cartilage repair. To produce a milieu conducive to cartilage repair, multi-dimensional management of the joint inflammatory microenvironment in place and time is required. Therefore, this calls for elucidation of the immune microenvironment of cartilage repair after injury. This review provides a brief overview of: (1) the pathogenesis of cartilage injury; (2) immune cells in cartilage injury and repair; (3) effects of inflammatory cytokines on cartilage repair; (4) clinical strategies for treating cartilage defects; and (5) strategies for targeted immunoregulation in cartilage repair. STATEMENT OF SIGNIFICANCE: Immune response is increasingly considered the key factor affecting cartilage repair. It has both negative and positive regulatory effects on the process of regeneration and repair. Proinflammatory factors are secreted in large numbers, and necrotic cartilage is removed. During the repair period, immune cells can secrete anti-inflammatory factors and chondrogenic cytokines, which can inhibit inflammation and promote cartilage repair. However, inflammatory factors persist, which accelerate the degradation of the cartilage matrix. Furthermore, in an inflammatory microenvironment, MSCs undergo abnormal differentiation, and chondrocytes begin to transform or dedifferentiate into fibroblast-like cells, forming fibrocartilage with poor mechanical properties. Consequently, cartilage regeneration requires multi-dimensional regulation of the joint inflammatory microenvironment in space and time to make it conducive to cartilage regeneration.


Asunto(s)
Enfermedades de los Cartílagos , Cartílago Articular , Osteoartritis , Enfermedades de los Cartílagos/metabolismo , Diferenciación Celular/genética , Condrocitos , Condrogénesis , Humanos , Osteoartritis/patología , Ingeniería de Tejidos
18.
Int J Mol Sci ; 22(24)2021 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-34947976

RESUMEN

Adjuvant therapy in autologous chondrocyte implantation (ACI) can control the post-traumatic environment and guide graft maturation to support cartilage repair. To investigate both aspects, we examined potential chondro-regenerative effects of lysed platelet concentrate (PC) and supplementary interleukin 10 (IL-10) on mechanically injured cartilage and on clinically used ACI scaffolds. ACI remnants and human cartilage explants, which were applied to an uniaxial unconfined compression as injury model, were treated with human IL-10 and/or PC from thrombocyte concentrates. We analyzed nuclear blebbing/TUNEL, sGAG content, immunohistochemistry, and the expression of COL1A1, COL2A1, COL10A1, SOX9, and ACAN. Post-injuriously, PC was associated with less cell death, increased COL2A1 expression, and decreased COL10A1 expression and, interestingly, the combination with Il-10 or Il-10 alone had no additional effects, except on COL10A1, which was most effectively decreased by the combination of PC and Il-10. The expression of COL2A1 or SOX9 was statistically not modulated by these substances. In contrast, in chondrocytes in ACI grafts the combination of PC and IL-10 had the most pronounced effects on all parameters except ACAN. Thus, using adjuvants such as PC and IL-10, preferably in combination, is a promising strategy for enhancing repair and graft maturation of autologous transplanted chondrocytes after cartilage injury.


Asunto(s)
Factores Biológicos/farmacología , Plaquetas/química , Enfermedades de los Cartílagos/terapia , Condrocitos/trasplante , Interleucina-10/farmacología , Agrecanos/metabolismo , Enfermedades de los Cartílagos/etiología , Enfermedades de los Cartílagos/metabolismo , Células Cultivadas , Condrocitos/citología , Colágeno/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , Factor de Transcripción SOX9/metabolismo , Estrés Mecánico , Trasplante Autólogo
19.
Int J Mol Sci ; 22(24)2021 Dec 11.
Artículo en Inglés | MEDLINE | ID: mdl-34948124

RESUMEN

Several collagen subtypes have been identified in hyaline articular cartilage. The main and most abundant collagens are type II, IX and XI collagens. The minor and less abundant collagens are type III, IV, V, VI, X, XII, XIV, XVI, XXII, and XXVII collagens. All these collagens have been found to play a key role in healthy cartilage, regardless of whether they are more or less abundant. Additionally, an exhaustive evaluation of collagen fibrils in a repaired cartilage tissue after a chondral lesion is necessary to determine the quality of the repaired tissue and even whether or not this repaired tissue is considered hyaline cartilage. Therefore, this review aims to describe in depth all the collagen types found in the normal articular cartilage structure, and based on this, establish the parameters that allow one to consider a repaired cartilage tissue as a hyaline cartilage.


Asunto(s)
Enfermedades de los Cartílagos/metabolismo , Cartílago Articular/metabolismo , Colágeno/metabolismo , Cartílago Hialino/metabolismo , Animales , Enfermedades de los Cartílagos/patología , Cartílago Articular/patología , Humanos , Cartílago Hialino/patología
20.
Oxid Med Cell Longev ; 2021: 4190098, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34777686

RESUMEN

Osteoarthritis (OA), a highly prevalent chronic joint disease, involves a complex network of inflammatory mediators that not only triggers pain and cartilage degeneration but also accelerates disease progression. Traditional Chinese medicinal shenjinhuoxue mixture (SHM) shows anti-inflammatory and analgesic effects against OA with remarkable clinical efficacy. This study explored the mechanism underlying anti-OA properties of SHM and evaluated its efficacy and safety via in vivo experiments. Through network pharmacology and published literature, we identified the key active phytochemicals in SHM, including ß-sitosterol, oleanolic acid, licochalcone A, quercetin, isorhamnetin, kaempferol, morusin, lupeol, and pinocembrin; the pivotal targets of which are TLR-4 and NF-κB, eliciting anti-OA activity. These phytochemicals can enter the active pockets of TLR-4 and NF-κB with docking score ≤ -3.86 kcal/mol, as shown in molecular docking models. By using surface plasmon resonance assay, licochalcone A and oleanolic acid were found to have good TLR-4-binding affinity. In OA rats, oral SHM at mid and high doses (8.72 g/kg and 26.2 g/kg) over 6 weeks significantly alleviated mechanical and thermal hyperalgesia (P < 0.0001). Accordingly, the expression of inflammatory mediators (TLR-4, interleukin (IL-) 1 receptor-associated kinase 1 (IRAK1), NF-κB-p65, tumor necrosis factor (TNF-) α, IL-6, and IL-1ß), receptor activator of the NF-κB ligand (RANKL), and transient receptor potential vanilloid 1 (TRPV1) in the synovial and cartilage tissue of OA rats was significantly decreased (P < 0.05). Moreover, pathological observation illustrated amelioration of cartilage degeneration and joint injury. In chronic toxicity experiment of rats, SHM at 60 mg/kg demonstrated the safety. SHM had an anti-inflammatory effect through a synergistic combination of active phytochemicals to attenuate pain and cartilage degeneration by inhibiting TLR-4 and NF-κB activation. This study provided the experimental foundation for the development of SHM into a more effective dosage form or new drugs for OA treatment.


Asunto(s)
Enfermedades de los Cartílagos/prevención & control , Inflamación/prevención & control , FN-kappa B/antagonistas & inhibidores , Osteoartritis/complicaciones , Dolor/prevención & control , Fitoquímicos/farmacología , Receptor Toll-Like 4/antagonistas & inhibidores , Animales , Antiinflamatorios/farmacología , Enfermedades de los Cartílagos/etiología , Enfermedades de los Cartílagos/metabolismo , Enfermedades de los Cartílagos/patología , Modelos Animales de Enfermedad , Inflamación/etiología , Inflamación/metabolismo , Inflamación/patología , Masculino , Medicina Tradicional China , Ratones , Ratones Endogámicos C57BL , Dolor/etiología , Dolor/metabolismo , Dolor/patología , Extractos Vegetales/farmacología , Sustancias Protectoras/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...