Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 4.094
Filter
1.
Int J Mol Sci ; 25(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38891793

ABSTRACT

Joint-resident chondrogenic precursor cells have become a significant therapeutic option due to the lack of regenerative capacity in articular cartilage. Progenitor cells are located in the superficial zone of the articular cartilage, producing lubricin/Prg4 to decrease friction of cartilage surfaces during joint movement. Prg4-positive progenitors are crucial in maintaining the joint's structure and functionality. The disappearance of progenitor cells leads to changes in articular hyaline cartilage over time, subchondral bone abnormalities, and the formation of ectopic ossification. Genetic labeling cell technology has been the main tool used to characterize Prg4-expressing progenitor cells of articular cartilage in vivo through drug injection at different time points. This technology allows for the determination of the origin of progenitor cells and the tracking of their progeny during joint development and cartilage damage. We endeavored to highlight the currently known information about the Prg4-producing cell population in the joint to underline the significance of the role of these cells in the development of articular cartilage and its homeostasis. This review focuses on superficial progenitors in the joint, how they contribute to postnatal articular cartilage formation, their capacity for regeneration, and the consequences of Prg4 deficiency in these cells. We have accumulated information about the Prg4+ cell population of articular cartilage obtained through various elegantly designed experiments using transgenic technologies to identify potential opportunities for further research.


Subject(s)
Cartilage, Articular , Proteoglycans , Stem Cells , Cartilage, Articular/metabolism , Cartilage, Articular/cytology , Animals , Humans , Stem Cells/metabolism , Stem Cells/cytology , Proteoglycans/metabolism , Chondrogenesis , Chondrocytes/metabolism , Chondrocytes/cytology , Cell Differentiation , Regeneration
2.
Food Chem Toxicol ; 189: 114724, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38734200

ABSTRACT

Notch signaling regulates cartilage formation and homeostasis. Kashin-Beck Disease (KBD), an endemic osteochondropathy, is characterized by severe cartilage degradation. The etiology of KBD is related to the exposure of HT-2 toxin, a mycotoxin and primary metabolite of T-2 toxin. This study aims to explore the role of HT-2 toxin in the Notch signaling regulation and extracellular matrix (ECM) metabolism of hiPSCs-Chondrocytes. Immunohistochemistry and qRT-PCR were employed to investigate the expression of Notch pathway molecules in KBD articular cartilage and primary chondrocytes. hiPSCs-Chondrocytes, derived from hiPSCs, were treated with 100 ng/mL HT-2 toxin and the γ-secretase inhibitor (DAPT) for 48h, respectively. The markers related to the Notch signaling pathway and ECM were assessed using qRT-PCR and Western blot. Notch pathway dysregulation was prominent in KBD cartilage. HT-2 toxin exposure caused cytotoxicity in hiPSCs-Chondrocytes, and activated Notch signaling by increasing the mRNA and protein levels of NOTCH1 and HES1. HT-2 toxin also upregulated ECM catabolic enzymes and downregulated ECM components (COL2A1 and ACAN), indicating ECM degradation. DAPT-mediated Notch signaling inhibition suppressed the mRNA and protein level of ADAMTS5 expression while enhancing ECM component expression in hiPSCs-Chondrocytes. This study suggests that HT-2 toxin may induce ECM degradation in hiPSCs-Chondrocytes through activating Notch signaling.


Subject(s)
Chondrocytes , Extracellular Matrix , Induced Pluripotent Stem Cells , Receptors, Notch , Signal Transduction , T-2 Toxin , Humans , Signal Transduction/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Extracellular Matrix/metabolism , Extracellular Matrix/drug effects , T-2 Toxin/toxicity , Induced Pluripotent Stem Cells/metabolism , Induced Pluripotent Stem Cells/drug effects , Receptors, Notch/metabolism , Receptors, Notch/genetics , Kashin-Beck Disease/metabolism , Cartilage, Articular/metabolism , Cartilage, Articular/cytology , Cartilage, Articular/drug effects , Transcription Factor HES-1/metabolism , Transcription Factor HES-1/genetics , Cells, Cultured
3.
Biofabrication ; 16(3)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38768586

ABSTRACT

Toward the translation of allogeneic cell therapy products, cell banks are needed not only to manufacture the final human product but also during the preclinical evaluation of an animal-based analogous cellular product (ACP). These cell banks need to be established at both the master cell bank (MCB) level and the working cell bank (WCB) level. Inasmuch as most of the development of cell therapy products is at academic centers, it is imperative that academic researchers understand how to establish MCBs and WCBs within an academic environment. To illustrate this process, using articular cartilage as the model, a cell bank for an ACP was developed (MCBs at passage 2, WCBs at passage 5) to produce self-assembled neocartilage for preclinical evaluation (constructs at passage 7). The cell bank system is estimated to be able to produce between 160 000 and 400 000 constructs for each of the six MCBs. Overall, the ACP cell bank yielded constructs that are analogous to the intended human product, which is critical toward conducting preclinical evaluations of the ACP for inclusion in an Investigational New Drug application to the FDA.


Subject(s)
Cell- and Tissue-Based Therapy , Humans , Animals , Cartilage, Articular/cytology , Tissue Engineering , Tissue Banks
4.
PLoS One ; 19(5): e0297947, 2024.
Article in English | MEDLINE | ID: mdl-38768116

ABSTRACT

In various biological systems, analyzing how cell behaviors are coordinated over time would enable a deeper understanding of tissue-scale response to physiologic or superphysiologic stimuli. Such data is necessary for establishing both normal tissue function and the sequence of events after injury that lead to chronic disease. However, collecting and analyzing these large datasets presents a challenge-such systems are time-consuming to process, and the overwhelming scale of data makes it difficult to parse overall behaviors. This problem calls for an analysis technique that can quickly provide an overview of the groups present in the entire system and also produce meaningful categorization of cell behaviors. Here, we demonstrate the application of an unsupervised method-the Variational Autoencoder (VAE)-to learn the features of cells in cartilage tissue after impact-induced injury and identify meaningful clusters of chondrocyte behavior. This technique quickly generated new insights into the spatial distribution of specific cell behavior phenotypes and connected specific peracute calcium signaling timeseries with long term cellular outcomes, demonstrating the value of the VAE technique.


Subject(s)
Cartilage, Articular , Chondrocytes , Cartilage, Articular/cytology , Chondrocytes/cytology , Animals , Cluster Analysis , Calcium Signaling
5.
Sci Rep ; 14(1): 11765, 2024 05 23.
Article in English | MEDLINE | ID: mdl-38782958

ABSTRACT

In vitro use of articular cartilage on an organ-on-a-chip (OOAC) via microfluidics is challenging owing to the dense extracellular matrix (ECM) composed of numerous protein moieties and few chondrocytes, which has limited proliferation potential and microscale translation. Hence, this study proposes a novel approach for using a combination of biopolymers and decellularised ECM (dECM) as a bioink additive in the development of scalable OOAC using a microfluidic platform. The bioink was tested with native chondrocytes and mesenchymal stem cell-induced chondrocytes using biopolymers of alginate and chitosan composite hydrogels. Two-dimensional (2D) and three-dimensional (3D) biomimetic tissue construction approaches have been used to characterise the morphology and cellular marker expression (by histology and confocal laser scanning microscopy), viability (cell viability dye using flow cytometry), and genotypic expression of ECM-specific markers (by quantitative PCR). The results demonstrated that the bioink had a significant impact on the increase in phenotypic and genotypic expression, with a statistical significance level of p < 0.05 according to Student's t-test. The use of a cell-laden biopolymer as a bioink optimised the niche conditions for obtaining hyaline-type cartilage under culture conditions, paving the way for testing mechano-responsive properties and translating these findings to a cartilage-on-a-chip microfluidics system.


Subject(s)
Alginates , Cartilage, Articular , Chitosan , Chondrocytes , Extracellular Matrix , Tissue Engineering , Chitosan/chemistry , Alginates/chemistry , Cartilage, Articular/metabolism , Cartilage, Articular/cytology , Animals , Extracellular Matrix/metabolism , Chondrocytes/metabolism , Chondrocytes/cytology , Tissue Engineering/methods , Biopolymers/chemistry , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Tissue Scaffolds/chemistry , Lab-On-A-Chip Devices , Hydrogels/chemistry , Cells, Cultured , Cell Survival , Microphysiological Systems
6.
J Forensic Sci ; 69(3): 1094-1101, 2024 May.
Article in English | MEDLINE | ID: mdl-38491758

ABSTRACT

The aim of the present study was to investigate the effects of time, temperature, and burial in a natural environment on the viability of chondrocytes in porcine femoral condyles using confocal laser scanning microscopy. Hind trotters from 10 pigs were buried or left unburied. Samples were collected daily and stained with a combination of vital dyes (calcein-AM and ethidium homodimer-1). The chondrocytes showed an intense staining corresponding to their vitality. In the first 3 days, viability decreased slowly and showed no statistical difference between buried and unburied samples. After the first 3 days, it decreased rapidly, with the viability of the buried samples being 66% on day 4, decreasing to 25% on day 8 and to 16% on day 10, while in the unburied samples it decreased to 43% on day 4, 13% on day 8 and 5% on day 10. Our results indicate a time, temperature, and burial dependent decrease in chondrocyte viability and suggest the use of chondrocyte viability as a marker for estimating PMI in both the natural environment and in animals, as well as its potential use in humans.


Subject(s)
Burial , Cartilage, Articular , Cell Survival , Chondrocytes , Microscopy, Confocal , Postmortem Changes , Temperature , Animals , Chondrocytes/cytology , Cartilage, Articular/cytology , Swine , Time Factors , Seasons , Forensic Pathology , Fluorescent Dyes , Femur/cytology
7.
Nature ; 622(7984): 834-841, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37794190

ABSTRACT

Although haemoglobin is a known carrier of oxygen in erythrocytes that functions to transport oxygen over a long range, its physiological roles outside erythrocytes are largely elusive1,2. Here we found that chondrocytes produced massive amounts of haemoglobin to form eosin-positive bodies in their cytoplasm. The haemoglobin body (Hedy) is a membraneless condensate characterized by phase separation. Production of haemoglobin in chondrocytes is controlled by hypoxia and is dependent on KLF1 rather than the HIF1/2α pathway. Deletion of haemoglobin in chondrocytes leads to Hedy loss along with severe hypoxia, enhanced glycolysis and extensive cell death in the centre of cartilaginous tissue, which is attributed to the loss of the Hedy-controlled oxygen supply under hypoxic conditions. These results demonstrate an extra-erythrocyte role of haemoglobin in chondrocytes, and uncover a heretofore unrecognized mechanism in which chondrocytes survive a hypoxic environment through Hedy.


Subject(s)
Adaptation, Physiological , Cell Hypoxia , Chondrocytes , Hemoglobins , Humans , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cell Death , Cell Hypoxia/physiology , Chondrocytes/metabolism , Cytoplasm/metabolism , Eosine Yellowish-(YS)/metabolism , Erythrocytes/metabolism , Glycolysis , Hemoglobins/deficiency , Hemoglobins/genetics , Hemoglobins/metabolism , Oxygen/metabolism
8.
Elife ; 122023 02 15.
Article in English | MEDLINE | ID: mdl-36790146

ABSTRACT

The origin and differentiation mechanism of articular chondrocytes remain poorly understood. Broadly, the difference in developmental mechanisms of articular and growth-plate cartilage is still less elucidated. Here, we identified that the nuclear factor of activated T-cells cytoplasmic 1 (NFATc1) is a crucial regulator of articular, but not growth-plate, chondrocyte differentiation during development. At the early stage of mouse knee development (embryonic day 13.5), NFATc1-expressing cells were mainly located in the flanking region of the joint interzone. With development, NFATc1-expressing cells generated almost all articular chondrocytes but not chondrocytes in limb growth-plate primordium. NFATc1-expressing cells displayed prominent capacities for colony formation and multipotent differentiation. Transcriptome analyses revealed a set of characteristic genes in NFATc1-enriched articular cartilage progenitors. Strikingly, the expression of NFATc1 was diminished with articular chondrocyte differentiation, and suppressing NFATc1 expression in articular cartilage progenitors was sufficient to induce spontaneous chondrogenesis while overexpressing NFATc1 suppresses chondrogenesis. Mechanistically, NFATc1 negatively regulated the transcriptional activity of the Col2a1 gene. Thus, our results reveal that NFATc1 characterizes articular, but not growth-plate, cartilage progenitors during development and negatively determines articular chondrocyte differentiation at least partly through regulating COL2A1 gene transcription.


Within the body are about 300 joints connecting bones together. Many factors ­ including trauma, inflammation, aging, and genetic changes ­ can affect the cushion tissue covering the end of the bones in these joints known as articular cartilage. This can lead to diseases such as osteoarthritis which cause chronic pain, and in some cases disability. To treat such conditions, it is essential to know how cells in the articular cartilage are formed during development. In the embryo, most cells come from groups of progenitor cells that are programmed to produce specific types of tissue. But which progenitor cells are responsible for producing the main cells in articular cartilage, chondrocytes, and the mechanisms that govern this transformation are poorly understood. In 2016, a group of researchers found that the gene for the protein NFATc1, which is important for building bone, is also expressed in a group of progenitor cells at the site where ligaments insert into bone in mice. Inactivation of NFATc1 in these progenitor cells has also been shown to cause abnormal cartilage to form, a condition termed osteochondromas. Building on this work, Zhang, Wang et al. ­ including some of the researchers involved in the 2016 study ­ set out to find whether NFATc1 is also involved in the normal development of articular chondrocytes. To investigate, the team used genetically modified mice in which any cells with NFATc1 also had a green fluorescent protein, and tracked these cells and their progeny over the course of joint development. This led them to discover a group of NFATc1-containing progenitor cells that gave rise to almost all articular chondrocytes in the knee joint. Further experiments revealed that when NFATc1 was removed, this made the progenitors become articular chondrocytes very quickly. In contrast, when the cells had excess amounts of the protein, the formation of articular chondrocytes was significantly reduced. This suggests that the level of NFATc1 governs when progenitors develop into articular chondrocytes. These findings have provided a way to track the progenitors of articular chondrocytes throughout development and study how articular cartilage is formed. In the future, this work could help researchers develop treatment strategies for osteoarthritis and other cartilage-based diseases. However, before this can happen, further work is needed to confirm that the effects observed in this study also relate to humans.


Subject(s)
Cartilage, Articular , Chondrocytes , NFATC Transcription Factors , Animals , Mice , Cartilage, Articular/cytology , Chondrocytes/cytology , NFATC Transcription Factors/metabolism , Gene Expression Profiling , Cell Differentiation , Embryo, Mammalian/cytology
9.
Development ; 149(11)2022 06 01.
Article in English | MEDLINE | ID: mdl-35451016

ABSTRACT

It has been established in the mouse model that during embryogenesis joint cartilage is generated from a specialized progenitor cell type, distinct from that responsible for the formation of growth plate cartilage. We recently found that mesodermal progeny of human pluripotent stem cells gave rise to two types of chondrogenic mesenchymal cells in culture: SOX9+ and GDF5+ cells. The fast-growing SOX9+ cells formed in vitro cartilage that expressed chondrocyte hypertrophy markers and readily underwent mineralization after ectopic transplantation. In contrast, the slowly growing GDF5+ cells derived from SOX9+ cells formed cartilage that tended to express low to undetectable levels of chondrocyte hypertrophy markers, but expressed PRG4, a marker of embryonic articular chondrocytes. The GDF5+-derived cartilage remained largely unmineralized in vivo. Interestingly, chondrocytes derived from the GDF5+ cells seemed to elicit these activities via non-cell-autonomous mechanisms. Genome-wide transcriptomic analyses suggested that GDF5+ cells might contain a teno/ligamento-genic potential, whereas SOX9+ cells resembled neural crest-like progeny-derived chondroprogenitors. Thus, human pluripotent stem cell-derived GDF5+ cells specified to generate permanent-like cartilage seem to emerge coincidentally with the commitment of the SOX9+ progeny to the tendon/ligament lineage.


Subject(s)
Cartilage, Articular , Chondrocytes , Pluripotent Stem Cells , Animals , Cartilage, Articular/cytology , Cartilage, Articular/metabolism , Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrocytes/pathology , Chondrogenesis , Growth Differentiation Factor 5/metabolism , Humans , Hypertrophy , Mice , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism
10.
FASEB J ; 36(4): e22225, 2022 04.
Article in English | MEDLINE | ID: mdl-35224777

ABSTRACT

Neocartilage tissue engineering aims to address the shortcomings of current clinical treatments for articular cartilage indications. However, advancement is required toward neocartilage functionality (mechanical and biochemical properties) and translatability (construct size, gross morphology, passage number, cell source, and cell type). Using fluid-induced shear (FIS) stress, a potent mechanical stimulus, over four phases, this work investigates FIS stress' efficacy toward creating large neocartilage derived from highly passaged minipig costal chondrocytes, a species relevant to the preclinical regulatory process. In Phase I, FIS stress application timing was investigated in bovine articular chondrocytes and found to improve the aggregate modulus of neocartilage by 151% over unstimulated controls when stimulated during the maturation stage. In Phase II, FIS stress stimulation was translated from bovine articular chondrocytes to expanded minipig costal chondrocytes, yielding a 46% improvement in aggregate modulus over nonstimulated controls. In Phase III, bioactive factors were combined with FIS stress to improve the shear modulus by 115% over bioactive factor-only controls. The translatability of neocartilage was improved in Phase IV by utilizing highly passaged cells to form constructs more than 9-times larger in the area (11 × 17 mm), yielding an improved aggregate modulus by 134% and a flat morphology compared to free-floating, bioactive factor-only controls. Overall, this study represents a significant step toward generating mechanically robust, large constructs necessary for animal studies, and eventually, human clinical studies.


Subject(s)
Cartilage, Articular/physiology , Chondrocytes/physiology , Hydrodynamics , Mechanotransduction, Cellular , Stress, Mechanical , Tissue Engineering/methods , Animals , Cartilage, Articular/cytology , Cattle , Cell Culture Techniques , Cell Proliferation , Cells, Cultured , Chondrocytes/cytology , Swine , Swine, Miniature
11.
J Cell Physiol ; 237(4): 2258-2270, 2022 04.
Article in English | MEDLINE | ID: mdl-35147979

ABSTRACT

Immune regulation of osteochondral defect regeneration has not yet been rigorously characterized. Although macrophages have been demonstrated to regulate the regeneration process in various tissues, their direct contribution to cartilage regeneration remains to be investigated, particularly the functions of polarized macrophage subpopulations. In this study, we investigated the origins and functions of macrophages during healing of osteochondral injury in the murine model. Upon osteochondral injury, joint macrophages are predominantly derived from circulating monocytes. Macrophages are essential for spontaneous cartilage regeneration in juvenile C57BL/6 mice, by modulating proliferation and apoptosis around the injury site. Exogeneous macrophages also exhibit therapeutic potential in promoting cartilage regeneration in adult mice with poor regenerative capacity, possibly via regulation of PDGFRα+  stem cells, with this process being influenced by initial phenotype and administration timing. Only M2c macrophages are able to promote regeneration of both cartilage tissues and subchondral bone. Overall, we reveal the direct link between macrophages and osteochondral regeneration and highlight the key roles of relevant immunological niches in successful regeneration.


Subject(s)
Cartilage, Articular , Macrophages/physiology , Wound Healing , Animals , Cartilage, Articular/cytology , Cartilage, Articular/injuries , Cartilage, Articular/physiology , Mice , Mice, Inbred C57BL
12.
Oxid Med Cell Longev ; 2022: 5635797, 2022.
Article in English | MEDLINE | ID: mdl-35154568

ABSTRACT

Osteoarthritis (OA) is a chronic degenerative disease featured by cartilage erosion and inflammation. Luteolin, a member of the flavonoid family, has been shown to exert anti-inflammatory and antioxidative activities. However, the potential biological effects and underlying mechanism of luteolin on chondrocytes and OA progression remain largely elusive. In this study, the potential effect and mechanism of luteolin on OA were investigated in vitro and in vivo. Our data revealed that luteolin inhibited H2O2-induced cell death, apoptosis, oxidative stress, programmed necrosis, and inflammatory mediator production in primary murine chondrocytes. In addition, luteolin could activate the AMPK and Nrf2 pathways, and AMPK serves as a positive upstream regulator of Nrf2. In vivo results demonstrated the therapeutic effects of luteolin on OA in the DMM mouse model. Collectively, our findings showed that luteolin might serve as a novel and effective treatment for OA and provided a new research direction for clinical OA therapies.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Antioxidants/administration & dosage , Biological Products/administration & dosage , Chondrocytes/drug effects , Chondrocytes/metabolism , Disease Progression , Hydrogen Peroxide/adverse effects , Luteolin/administration & dosage , MAP Kinase Signaling System/drug effects , NF-E2-Related Factor 2/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Oxidative Stress/drug effects , AMP-Activated Protein Kinases/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cartilage, Articular/cytology , Cells, Cultured , Disease Models, Animal , Gene Knockdown Techniques/methods , MAP Kinase Signaling System/genetics , Male , Mice , Mice, Inbred C57BL , NF-E2-Related Factor 2/genetics , Osteoarthritis/pathology , Oxidative Stress/genetics , Transduction, Genetic/methods , Treatment Outcome
13.
Int J Mol Sci ; 23(2)2022 Jan 15.
Article in English | MEDLINE | ID: mdl-35055112

ABSTRACT

In 3D bioprinting for cartilage regeneration, bioinks that support chondrogenic development are of key importance. Growth factors covalently bound in non-printable hydrogels have been shown to effectively promote chondrogenesis. However, studies that investigate the functionality of tethered growth factors within 3D printable bioinks are still lacking. Therefore, in this study, we established a dual-stage crosslinked hyaluronic acid-based bioink that enabled covalent tethering of transforming growth factor-beta 1 (TGF-ß1). Bone marrow-derived mesenchymal stromal cells (MSCs) were cultured over three weeks in vitro, and chondrogenic differentiation of MSCs within bioink constructs with tethered TGF-ß1 was markedly enhanced, as compared to constructs with non-covalently incorporated TGF-ß1. This was substantiated with regard to early TGF-ß1 signaling, chondrogenic gene expression, qualitative and quantitative ECM deposition and distribution, and resulting construct stiffness. Furthermore, it was successfully demonstrated, in a comparative analysis of cast and printed bioinks, that covalently tethered TGF-ß1 maintained its functionality after 3D printing. Taken together, the presented ink composition enabled the generation of high-quality cartilaginous tissues without the need for continuous exogenous growth factor supply and, thus, bears great potential for future investigation towards cartilage regeneration. Furthermore, growth factor tethering within bioinks, potentially leading to superior tissue development, may also be explored for other biofabrication applications.


Subject(s)
Bioprinting/methods , Cartilage, Articular/cytology , Hyaluronic Acid/chemistry , Mesenchymal Stem Cells/cytology , Transforming Growth Factor beta1/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cell Differentiation , Cells, Cultured , Extracellular Matrix/metabolism , Humans , Hydrogels , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds , Transforming Growth Factor beta1/chemistry
14.
J Nanobiotechnology ; 20(1): 25, 2022 Jan 06.
Article in English | MEDLINE | ID: mdl-34991615

ABSTRACT

BACKGROUND: The regeneration and repair of articular cartilage remains a major challenge for clinicians and scientists due to the poor intrinsic healing of this tissue. Since cartilage injuries are often clinically irregular, tissue-engineered scaffolds that can be easily molded to fill cartilage defects of any shape that fit tightly into the host cartilage are needed. METHOD: In this study, bone marrow mesenchymal stem cell (BMSC) affinity peptide sequence PFSSTKT (PFS)-modified chondrocyte extracellular matrix (ECM) particles combined with GelMA hydrogel were constructed. RESULTS: In vitro experiments showed that the pore size and porosity of the solid-supported composite scaffolds were appropriate and that the scaffolds provided a three-dimensional microenvironment supporting cell adhesion, proliferation and chondrogenic differentiation. In vitro experiments also showed that GelMA/ECM-PFS could regulate the migration of rabbit BMSCs. Two weeks after implantation in vivo, the GelMA/ECM-PFS functional scaffold system promoted the recruitment of endogenous mesenchymal stem cells from the defect site. GelMA/ECM-PFS achieved successful hyaline cartilage repair in rabbits in vivo, while the control treatment mostly resulted in fibrous tissue repair. CONCLUSION: This combination of endogenous cell recruitment and chondrogenesis is an ideal strategy for repairing irregular cartilage defects.


Subject(s)
Chondrogenesis/drug effects , Decellularized Extracellular Matrix , Hydrogels , Oligopeptides , Tissue Scaffolds/chemistry , Animals , Cartilage, Articular/cytology , Decellularized Extracellular Matrix/chemistry , Decellularized Extracellular Matrix/pharmacology , Hydrogels/chemistry , Hydrogels/pharmacology , Male , Mesenchymal Stem Cells/drug effects , Oligopeptides/chemistry , Oligopeptides/pharmacology , Rabbits , Tissue Engineering/methods
15.
Cells ; 10(12)2021 12 14.
Article in English | MEDLINE | ID: mdl-34944042

ABSTRACT

Cartilage stem/progenitor cells (CSPCs) are cartilage-specific, multipotent progenitor cells residing in articular cartilage. In this study, we investigated the characteristics and potential of human CSPCs combined with poly(lactic-co-glycolic acid) (PLGA) scaffolds to induce osteochondral regeneration in rabbit knees. We isolated CSPCs from human adult articular cartilage undergoing total knee replacement (TKR) surgery. We characterized CSPCs and compared them with infrapatellar fat pad-derived stem cells (IFPs) in a colony formation assay and by multilineage differentiation analysis in vitro. We further evaluated the osteochondral regeneration of the CSPC-loaded PLGA scaffold during osteochondral defect repair in rabbits. The characteristics of CSPCs were similar to those of mesenchymal stem cells (MSCs) and exhibited chondrogenic and osteogenic phenotypes without chemical induction. For in vivo analysis, CSPC-loaded PLGA scaffolds produced a hyaline-like cartilaginous tissue, which showed good integration with the host tissue and subchondral bone. Furthermore, CSPCs migrated in response to injury to promote subchondral bone regeneration. Overall, we demonstrated that CSPCs can promote osteochondral regeneration. A monophasic approach of using diseased CSPCs combined with a PLGA scaffold may be beneficial for repairing complex tissues, such as osteochondral tissue.


Subject(s)
Cartilage, Articular/cytology , Cell Differentiation , Chondrogenesis , Stem Cells/cytology , Tissue Scaffolds/chemistry , Adult , Aged , Aged, 80 and over , Animals , Bone Regeneration , Cell Lineage , Cell Shape , Cells, Cultured , Colony-Forming Units Assay , Humans , Immunophenotyping , Male , Middle Aged , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Porosity , Rabbits , X-Ray Microtomography
16.
Int J Mol Sci ; 22(24)2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34948394

ABSTRACT

The composition and organisation of the extracellular matrix (ECM), particularly the pericellular matrix (PCM), in articular cartilage is critical to its biomechanical functionality; the presence of proteoglycans such as aggrecan, entrapped within a type II collagen fibrillar network, confers mechanical resilience underweight-bearing. Furthermore, components of the PCM including type VI collagen, perlecan, small leucine-rich proteoglycans-decorin and biglycan-and fibronectin facilitate the transduction of both biomechanical and biochemical signals to the residing chondrocytes, thereby regulating the process of mechanotransduction in cartilage. In this review, we summarise the literature reporting on the bidirectional reciprocity of the ECM in chondrocyte mechano-signalling and articular cartilage homeostasis. Specifically, we discuss studies that have characterised the response of articular cartilage to mechanical perturbations in the local tissue environment and how the magnitude or type of loading applied elicits cellular behaviours to effect change. In vivo, including transgenic approaches, and in vitro studies have illustrated how physiological loading maintains a homeostatic balance of anabolic and catabolic activities, involving the direct engagement of many PCM molecules in orchestrating this slow but consistent turnover of the cartilage matrix. Furthermore, we document studies characterising how abnormal, non-physiological loading including excessive loading or joint trauma negatively impacts matrix molecule biosynthesis and/or organisation, affecting PCM mechanical properties and reducing the tissue's ability to withstand load. We present compelling evidence showing that reciprocal engagement of the cells with this altered ECM environment can thus impact tissue homeostasis and, if sustained, can result in cartilage degradation and onset of osteoarthritis pathology. Enhanced dysregulation of PCM/ECM turnover is partially driven by mechanically mediated proteolytic degradation of cartilage ECM components. This generates bioactive breakdown fragments such as fibronectin, biglycan and lumican fragments, which can subsequently activate or inhibit additional signalling pathways including those involved in inflammation. Finally, we discuss how bidirectionality within the ECM is critically important in enabling the chondrocytes to synthesise and release PCM/ECM molecules, growth factors, pro-inflammatory cytokines and proteolytic enzymes, under a specified load, to influence PCM/ECM composition and mechanical properties in cartilage health and disease.


Subject(s)
Cartilage, Articular/metabolism , Extracellular Matrix/metabolism , Mechanotransduction, Cellular , Animals , Cartilage, Articular/cytology , Cartilage, Articular/pathology , Humans , Osteoarthritis/metabolism , Osteoarthritis/pathology , Signal Transduction
17.
Cell Mol Life Sci ; 78(24): 8127-8155, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34783870

ABSTRACT

Articular cartilage damage caused by sports injury or osteoarthritis (OA) has gained increased attention as a worldwide health burden. Pharmaceutical treatments are considered cost-effective means of promoting cartilage regeneration, but are limited by their inability to generate sufficient functional chondrocytes and modify disease progression. Small molecular chemical compounds are an abundant source of new pharmaceutical therapeutics for cartilage regeneration, as they have advantages in design, fabrication, and application, and, when used in combination, act as powerful tools for manipulating cellular fate. In this review, we present current achievements in the development of small molecular drugs for cartilage regeneration, particularly in the fields of chondrocyte generation and reversion of chondrocyte degenerative phenotypes. Several clinically or preclinically available small molecules, which have been shown to facilitate chondrogenesis, chondrocyte dedifferentiation, and cellular reprogramming, and subsequently ameliorate cartilage degeneration by targeting inflammation, matrix degradation, metabolism, and epigenetics, are summarized. Notably, this review introduces essential parameters for high-throughput screening strategies, including models of different chondrogenic cell sources, phenotype readout methodologies, and transferable advanced systems from other fields. Overall, this review provides new insights into future pharmaceutical therapies for cartilage regeneration.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/cytology , Chondrogenesis , Osteoarthritis/therapy , Pharmaceutical Preparations/administration & dosage , Regeneration , Small Molecule Libraries/pharmacology , Animals , Cartilage, Articular/drug effects , Cell Differentiation , Chondrocytes/drug effects , High-Throughput Screening Assays , Humans , Osteoarthritis/pathology , Tissue Engineering
18.
Int J Mol Sci ; 22(21)2021 Oct 29.
Article in English | MEDLINE | ID: mdl-34769199

ABSTRACT

Resorbable polyglycolic acid (PGA) chondrocyte grafts are clinically established for human articular cartilage defects. Long-term implant performance was addressed in a standardized in vitro model. PGA implants (+/- bovine chondrocytes) were placed inside cartilage rings punched out of bovine femoral trochleas (outer Ø 6 mm; inner defect Ø 2 mm) and cultured for 84 days (12 weeks). Cartilage/PGA hybrids were subsequently analyzed by histology (hematoxylin/eosin; safranin O), immunohistochemistry (aggrecan, collagens 1 and 2), protein assays, quantitative real-time polymerase chain reactions, and implant push-out force measurements. Cartilage/PGA hybrids remained vital with intact matrix until 12 weeks, limited loss of proteoglycans from "host" cartilage or cartilage-PGA interface, and progressively diminishing release of proteoglycans into the supernatant. By contrast, the collagen 2 content in cartilage and cartilage-PGA interface remained approximately constant during culture (with only little collagen 1). Both implants (+/- cells) displayed implant colonization and progressively increased aggrecan and collagen 2 mRNA, but significantly decreased push-out forces over time. Cell-loaded PGA showed significantly accelerated cell colonization and significantly extended deposition of aggrecan. Augmented chondrogenic differentiation in PGA and cartilage/PGA-interface for up to 84 days suggests initial cartilage regeneration. Due to the PGA resorbability, however, the model exhibits limitations in assessing the "lateral implant bonding".


Subject(s)
Cartilage, Articular/physiology , Chondrocytes/cytology , Polyglycolic Acid/chemistry , Regeneration , Tissue Scaffolds/chemistry , Absorbable Implants , Animals , Cartilage, Articular/cytology , Cartilage, Articular/injuries , Cattle , Cells, Cultured , Chondrocytes/metabolism , Chondrogenesis , Disease Models, Animal , Tissue Engineering
19.
Molecules ; 26(19)2021 Sep 22.
Article in English | MEDLINE | ID: mdl-34641274

ABSTRACT

Articular cartilage (AC) damage is quite common, but due to AC's poor self-healing ability, the damage can easily develop into osteoarthritis (OA). To solve this problem, we developed a microsphere/hydrogel system that provides two growth factors that promote cartilage repair: transforming growth factor-ß3 (TGF-ß3) to enhance cartilage tissue formation and ghrelin synergy TGF-ß to significantly enhance the chondrogenic differentiation. The hydrogel and microspheres were characterized in vitro, and the biocompatibility of the system was verified. Double emulsion solvent extraction technology (w/o/w) is used to encapsulate TGF-ß3 and ghrelin into microspheres, and these microspheres are encapsulated in a hydrogel to continuously release TGF-ß3 and ghrelin. According to the chondrogenic differentiation ability of mesenchymal stem cells (MSCs) in vitro, the concentrations of the two growth factors were optimized to promote cartilage regeneration.


Subject(s)
Cartilage, Articular/cytology , Ghrelin/pharmacology , Mesenchymal Stem Cells/cytology , Transforming Growth Factor beta3/pharmacology , Cartilage, Articular/drug effects , Cartilage, Articular/metabolism , Cell Culture Techniques , Cells, Cultured , Chondrogenesis/drug effects , Culture Media/chemistry , Glycosaminoglycans/metabolism , Humans , Hydrogels , Materials Testing , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Microspheres , Regenerative Medicine
20.
Int J Mol Sci ; 22(19)2021 Sep 24.
Article in English | MEDLINE | ID: mdl-34638629

ABSTRACT

Tissue and organ failure has induced immense economic and healthcare concerns across the world. Tissue engineering is an interdisciplinary biomedical approach which aims to address the issues intrinsic to organ donation by providing an alternative strategy to tissue and organ transplantation. This review is specifically focused on cartilage tissue. Cartilage defects cannot readily regenerate, and thus research into tissue engineering approaches is relevant as a potential treatment option. Cells, scaffolds, and growth factors are three components that can be utilized to regenerate new tissue, and in particular recent advances in microparticle technology have excellent potential to revolutionize cartilage tissue regeneration. First, microspheres can be used for drug delivery by injecting them into the cartilage tissue or joint space to reduce pain and stimulate regeneration. They can also be used as controlled release systems within tissue engineering constructs. Additionally, microcarriers can act as a surface for stem cells or chondrocytes to adhere to and expand, generating large amounts of cells, which are necessary for clinically relevant cell therapies. Finally, a newer application of microparticles is to form them together into granular hydrogels to act as scaffolds for tissue engineering or to use in bioprinting. Tissue engineering has the potential to revolutionize the space of cartilage regeneration, but additional research is needed to allow for clinical translation. Microparticles are a key enabling technology in this regard.


Subject(s)
Cartilage, Articular/cytology , Tissue Engineering/methods , Animals , Cartilage Diseases/therapy , Cell Differentiation/physiology , Chondrocytes/cytology , Humans , Microspheres
SELECTION OF CITATIONS
SEARCH DETAIL
...