Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 3.960
2.
ACS Biomater Sci Eng ; 10(5): 2983-2994, 2024 May 13.
Article En | MEDLINE | ID: mdl-38634615

Calcified cartilage digested by chondroclasts provides an excellent scaffold to initiate bone formation. We analyzed bioactive proteins and microarchitecture of calcified cartilage either separately or in combination and evaluated biomimetic osteogenic culture conditions of surface-coated micropatterning. To do so, we prepared a crude extract from porcine femoral growth plates, which enhanced in vitro mineralization when coated on flat-bottom culture dishes, and identified four candidate proteins by fractionation and mass spectrometry. Murine homologues of two candidates, desmoglein 4 (DSG4) and peroxiredoxin 6 (PRDX6), significantly promoted osteogenic activity based on in vitro mineralization and osteoblast differentiation. Moreover, we observed DSG4 and PRDX6 protein expression in mouse femur. In addition, we designed circular, triangular, and honeycomb micropatterns with 30 or 50 µm units, either isolated or connected, to mimic hypertrophic chondrocyte-sized compartments. Isolated, larger honeycomb patterns particularly enhanced osteogenesis in vitro. Mineralization on micropatterns was positively correlated with the reduction of osteoblast migration distance in live cell imaging. Finally, we evaluated possible combinatorial effects of coat proteins and micropatterns and observed an additive effect of DSG4 or PRDX6 coating with micropatterns. These data suggest that combining a bioactive surface coating with osteogenic micropatterns may recapitulate initiation of bone formation during endochondral ossification.


Osteogenesis , Animals , Osteogenesis/drug effects , Mice , Swine , Osteoblasts/metabolism , Osteoblasts/cytology , Osteoblasts/drug effects , Cell Differentiation/drug effects , Cartilage/metabolism , Cartilage/cytology , Peroxiredoxin VI/metabolism , Calcification, Physiologic/drug effects
3.
Biosci Rep ; 44(5)2024 May 29.
Article En | MEDLINE | ID: mdl-38563479

Osteoarthritis (OA) is a long-term, persistent joint disorder characterized by bone and cartilage degradation, resulting in tightness, pain, and restricted movement. Current attempts in cartilage regeneration are cell-based therapies using stem cells. Multipotent stem cells, such as mesenchymal stem cells (MSCs), and pluripotent stem cells, such as embryonic stem cells (ESCs), have been used to regenerate cartilage. However, since the discovery of human-induced pluripotent stem cells (hiPSCs) in 2007, it was seen as a potential source for regenerative chondrogenic therapy as it overcomes the ethical issues surrounding the use of ESCs and the immunological and differentiation limitations of MSCs. This literature review focuses on chondrogenic differentiation and 3D bioprinting technologies using hiPSCS, suggesting them as a viable source for successful tissue engineering. METHODS: A literature search was conducted using scientific search engines, PubMed, MEDLINE, and Google Scholar databases with the terms 'Cartilage tissue engineering' and 'stem cells' to retrieve published literature on chondrogenic differentiation and tissue engineering using MSCs, ESCs, and hiPSCs. RESULTS: hiPSCs may provide an effective and autologous treatment for focal chondral lesions, though further research is needed to explore the potential of such technologies. CONCLUSIONS: This review has provided a comprehensive overview of these technologies and the potential applications for hiPSCs in regenerative medicine.


Cell Differentiation , Chondrogenesis , Induced Pluripotent Stem Cells , Tissue Engineering , Humans , Tissue Engineering/methods , Induced Pluripotent Stem Cells/cytology , Induced Pluripotent Stem Cells/metabolism , Animals , Osteoarthritis/therapy , Osteoarthritis/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Regenerative Medicine/methods , Cartilage/metabolism , Cartilage/cytology , Bioprinting/methods , Printing, Three-Dimensional , Regeneration
4.
BMC Biotechnol ; 24(1): 25, 2024 Apr 30.
Article En | MEDLINE | ID: mdl-38689309

The reconstruction of a stable, nipple-shaped cartilage graft that precisely matches the natural nipple in shape and size on the contralateral side is a clinical challenge. While 3D printing technology can efficiently and accurately manufacture customized complex structures, it faces limitations due to inadequate blood supply, which hampers the stability of nipple-shaped cartilage grafts produced using this technology. To address this issue, we employed a biodegradable biomaterial, Poly(lactic-co-glycolic acid) (PLGA), loaded with Cell-Free Fat Extract (Ceffe). Ceffe has demonstrated the ability to promote angiogenesis and cell proliferation, making it an ideal bio-ink for bioprinting precise nipple-shaped cartilage grafts. We utilized the Ceffe/PLGA scaffold to create a porous structure with a precise nipple shape. This scaffold exhibited favorable porosity and pore size, ensuring stable shape maintenance and satisfactory biomechanical properties. Importantly, it could release Ceffe in a sustained manner. Our in vitro results confirmed the scaffold's good biocompatibility and its ability to promote angiogenesis, as evidenced by supporting chondrocyte proliferation and endothelial cell migration and tube formation. Furthermore, after 8 weeks of in vivo culture, the Ceffe/PLGA scaffold seeded with chondrocytes regenerated into a cartilage support structure with a precise nipple shape. Compared to the pure PLGA group, the Ceffe/PLGA scaffold showed remarkable vascular formation, highlighting the beneficial effects of Ceffe. These findings suggest that our designed Ceffe/PLGA scaffold with a nipple shape represents a promising strategy for precise nipple-shaped cartilage regeneration, laying a foundation for subsequent nipple reconstruction.


Cartilage , Chondrocytes , Polylactic Acid-Polyglycolic Acid Copolymer , Printing, Three-Dimensional , Tissue Engineering , Tissue Scaffolds , Tissue Scaffolds/chemistry , Animals , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Tissue Engineering/methods , Chondrocytes/cytology , Cartilage/cytology , Cartilage/growth & development , Cell Proliferation/drug effects , Biocompatible Materials/chemistry , Rabbits , Porosity , Polyglycolic Acid/chemistry , Neovascularization, Physiologic/drug effects
5.
J Biol Chem ; 299(6): 104805, 2023 06.
Article En | MEDLINE | ID: mdl-37172728

Bone development starts with condensations of undifferentiated mesenchymal cells that set a framework for future bones within the primordium. In the endochondral pathway, mesenchymal cells inside the condensation differentiate into chondrocytes and perichondrial cells in a SOX9-dependent mechanism. However, the identity of mesenchymal cells outside the condensation and how they participate in developing bones remain undefined. Here we show that mesenchymal cells surrounding the condensation contribute to both cartilage and perichondrium, robustly generating chondrocytes, osteoblasts, and marrow stromal cells in developing bones. Single-cell RNA-seq analysis of Prrx1-cre-marked limb bud mesenchymal cells at E11.5 reveals that Notch effector Hes1 is expressed in a mutually exclusive manner with Sox9 that is expressed in pre-cartilaginous condensations. Analysis of a Notch signaling reporter CBF1:H2B-Venus reveals that peri-condensation mesenchymal cells are active for Notch signaling. In vivo lineage-tracing analysis using Hes1-creER identifies that Hes1+ early mesenchymal cells surrounding the SOX9+ condensation at E10.5 contribute to both cartilage and perichondrium at E13.5, subsequently becoming growth plate chondrocytes, osteoblasts of trabecular and cortical bones, and marrow stromal cells in postnatal bones. In contrast, Hes1+ cells in the perichondrium at E12.5 or E14.5 do not generate chondrocytes within cartilage, contributing to osteoblasts and marrow stromal cells only through the perichondrial route. Therefore, Hes1+ peri-condensation mesenchymal cells give rise to cells of the skeletal lineage through cartilage-dependent and independent pathways, supporting the theory that early mesenchymal cells outside the condensation also play important roles in early bone development.


Bone Development , Bone and Bones , Cartilage , Cell Differentiation , Cell Lineage , Chondrocytes , Mesenchymal Stem Cells , Transcription Factor HES-1 , Animals , Mice , Bone and Bones/cytology , Cartilage/cytology , Cartilage/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Osteoblasts/cytology , Osteoblasts/metabolism , Transcription Factor HES-1/metabolism , Stromal Cells/cytology , Stromal Cells/metabolism , Receptors, Notch/metabolism
6.
Nature ; 612(7940): 546-554, 2022 12.
Article En | MEDLINE | ID: mdl-36477541

Insufficient intracellular anabolism is a crucial factor involved in many pathological processes in the body1,2. The anabolism of intracellular substances requires the consumption of sufficient intracellular energy and the production of reducing equivalents. ATP acts as an 'energy currency' for biological processes in cells3,4, and the reduced form of NADPH is a key electron donor that provides reducing power for anabolism5. Under pathological conditions, it is difficult to correct impaired anabolism and to increase insufficient levels of ATP and NADPH to optimum concentrations1,4,6-8. Here we develop an independent and controllable nanosized plant-derived photosynthetic system based on nanothylakoid units (NTUs). To enable cross-species applications, we use a specific mature cell membrane (the chondrocyte membrane (CM)) for camouflage encapsulation. As proof of concept, we demonstrate that these CM-NTUs enter chondrocytes through membrane fusion, avoid lysosome degradation and achieve rapid penetration. Moreover, the CM-NTUs increase intracellular ATP and NADPH levels in situ following exposure to light and improve anabolism in degenerated chondrocytes. They can also systemically correct energy imbalance and restore cellular metabolism to improve cartilage homeostasis and protect against pathological progression of osteoarthritis. Our therapeutic strategy for degenerative diseases is based on a natural photosynthetic system that can controllably enhance cell anabolism by independently providing key energy and metabolic carriers. This study also provides an enhanced understanding of the preparation and application of bioorganisms and composite biomaterials for the treatment of disease.


Chondrocytes , Osteoarthritis , Photosynthesis , Plants , Humans , Adenosine Triphosphate/metabolism , Chondrocytes/metabolism , NADP/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Osteoarthritis/therapy , Plants/metabolism , Cartilage/cytology , Cartilage/metabolism , Homeostasis , Energy Metabolism , Membrane Fusion
7.
Nat Commun ; 13(1): 3960, 2022 07 08.
Article En | MEDLINE | ID: mdl-35803931

Mesenchymal stromal cells (MSCs) differentiation into different lineages is precisely controlled by signaling pathways. Given that protein kinases play a crucial role in signal transduction, here we show that Microtubule Associated Serine/Threonine Kinase Family Member 4 (Mast4) serves as an important mediator of TGF-ß and Wnt signal transduction in regulating chondro-osteogenic differentiation of MSCs. Suppression of Mast4 by TGF-ß1 led to increased Sox9 stability by blocking Mast4-induced Sox9 serine 494 phosphorylation and subsequent proteasomal degradation, ultimately enhancing chondrogenesis of MSCs. On the other hand, Mast4 protein, which stability was enhanced by Wnt-mediated inhibition of GSK-3ß and subsequent Smurf1 recruitment, promoted ß-catenin nuclear localization and Runx2 activity, increasing osteogenesis of MSCs. Consistently, Mast4-/- mice demonstrated excessive cartilage synthesis, while exhibiting osteoporotic phenotype. Interestingly, Mast4 depletion in MSCs facilitated cartilage formation and regeneration in vivo. Altogether, our findings uncover essential roles of Mast4 in determining the fate of MSC development into cartilage or bone.


Bone and Bones , Cartilage , Mesenchymal Stem Cells , Microtubule-Associated Proteins , Protein Serine-Threonine Kinases , Animals , Female , Mice , Bone and Bones/cytology , Bone and Bones/metabolism , Cartilage/cytology , Cartilage/metabolism , Cell Differentiation/genetics , Chondrogenesis/genetics , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microtubule-Associated Proteins/genetics , Osteogenesis/genetics , Protein Serine-Threonine Kinases/genetics , Transforming Growth Factor beta/metabolism , Wnt Signaling Pathway
8.
Development ; 149(4)2022 02 15.
Article En | MEDLINE | ID: mdl-35132438

Cranial neural crest cell (NCC)-derived chondrocyte precursors undergo a dynamic differentiation and maturation process to establish a scaffold for subsequent bone formation, alterations in which contribute to congenital birth defects. Here, we demonstrate that transcription factor and histone methyltransferase proteins Prdm3 and Prdm16 control the differentiation switch of cranial NCCs to craniofacial cartilage. Loss of either paralog results in hypoplastic and disorganized chondrocytes due to impaired cellular orientation and polarity. We show that these proteins regulate cartilage differentiation by controlling the timing of Wnt/ß-catenin activity in strikingly different ways: Prdm3 represses whereas Prdm16 activates global gene expression, although both act by regulating Wnt enhanceosome activity and chromatin accessibility. Finally, we show that manipulating Wnt/ß-catenin signaling pharmacologically or generating prdm3-/-;prdm16-/- double mutants rescues craniofacial cartilage defects. Our findings reveal upstream regulatory roles for Prdm3 and Prdm16 in cranial NCCs to control Wnt/ß-catenin transcriptional activity during chondrocyte differentiation to ensure proper development of the craniofacial skeleton.


Cell Differentiation , MDS1 and EVI1 Complex Locus Protein/metabolism , Wnt Signaling Pathway/genetics , Zebrafish Proteins/metabolism , Animals , Cartilage/cytology , Cartilage/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis , Chromatin/metabolism , Chromatin Assembly and Disassembly , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation, Developmental , MDS1 and EVI1 Complex Locus Protein/deficiency , MDS1 and EVI1 Complex Locus Protein/genetics , Mice , Mice, Knockout , Neural Crest/cytology , Neural Crest/metabolism , Regulatory Sequences, Nucleic Acid , Skull/cytology , Skull/metabolism , Wnt Proteins/metabolism , Zebrafish , Zebrafish Proteins/deficiency , Zebrafish Proteins/genetics , beta Catenin/metabolism
9.
Nat Commun ; 13(1): 571, 2022 01 28.
Article En | MEDLINE | ID: mdl-35091558

Developmental osteogenesis, physiological bone remodelling and fracture healing require removal of matrix and cellular debris. Osteoclasts generated by the fusion of circulating monocytes degrade bone, whereas the identity of the cells responsible for cartilage resorption is a long-standing and controversial question. Here we show that matrix degradation and chondrocyte phagocytosis are mediated by fatty acid binding protein 5-expressing cells representing septoclasts, which have a mesenchymal origin and are not derived from haematopoietic cells. The Notch ligand Delta-like 4, provided by endothelial cells, is necessary for septoclast specification and developmental bone growth. Consistent with the termination of growth, septoclasts disappear in adult and ageing bone, but re-emerge in association with growing vessels during fracture healing. We propose that cartilage degradation is mediated by rare, specialized cells distinct from osteoclasts. Our findings have implications for fracture healing, which is frequently impaired in aging humans.


Cartilage/metabolism , Fracture Healing/physiology , Mesenchymal Stem Cells/metabolism , Osteoclasts/metabolism , Osteogenesis/physiology , Animals , Bone and Bones/cytology , Bone and Bones/metabolism , Bone and Bones/ultrastructure , Cartilage/cytology , Cells, Cultured , Chondrocytes/cytology , Chondrocytes/metabolism , Fatty Acid-Binding Proteins/genetics , Fatty Acid-Binding Proteins/metabolism , Female , Fracture Healing/genetics , Humans , Male , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Microscopy, Immunoelectron , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Osteoclasts/cytology , Osteogenesis/genetics , RNA-Seq/methods
10.
BMC Complement Med Ther ; 22(1): 25, 2022 Jan 27.
Article En | MEDLINE | ID: mdl-35086536

BACKGROUND: Osteoarthritis (OA) treatment aims to improve inflammation and delay cartilage degeneration. However, there is no effective strategy presently available. Ononin, a representative isoflavone glycoside component extracted from natural Chinese herbs, exerts anti-inflammatory and proliferative effects. However, the therapeutic effect of ononin on chondrocyte inflammation remains unclear. METHODS: In this study, we explored the therapeutic effect and potential mechanism of ononin in OA by establishing an interleukin-1 beta (IL-1ß)-induced chondrocyte inflammation model. RESULTS: Our results verified that ononin alleviated the IL-1ß-induced decrease in chondrocyte viability, attenuated the overexpression of the inflammatory factors tumour necrosis factor α (TNF-α) and interleukin 6 (IL-6), and simultaneously inhibited the expression of cartilage extracellular matrix (ECM)-degrading enzymes such as matrix metalloproteinase-13 (MMP-13). Furthermore, the decomposition of Collagen II protein could be alleviated in the OA model by ononin. Finally, ononin improved chondrocyte inflammation by downregulating the mitogen-activated protein kinase (MAPK) and nuclear factor kappa-B (NF-κB) signalling pathways. CONCLUSION: Our findings suggested that ononin could inhibit the IL-1ß-induced proinflammatory response and ECM degradation in chondrocytes by interfering with the abnormal activation of the MAPK and NF-κB pathways, indicating its protective effect against OA.


Cartilage/drug effects , Glucosides/pharmacology , Inflammation/metabolism , Interleukin-1beta/metabolism , Isoflavones/pharmacology , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoarthritis , Animals , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Cartilage/cytology , Cartilage/metabolism , Cartilage/pathology , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrocytes/pathology , Down-Regulation , Glucosides/therapeutic use , Inflammation/drug therapy , Isoflavones/therapeutic use , MAP Kinase Signaling System , Male , Matrix Metalloproteinase 13/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Osteoarthritis/pathology , Phytotherapy , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats, Sprague-Dawley , Signal Transduction , Tumor Necrosis Factor-alpha/metabolism
11.
Int J Mol Sci ; 22(21)2021 Oct 21.
Article En | MEDLINE | ID: mdl-34768781

The study was aimed at the applicability of a bioink based on 4% collagen and chondrocytes for de novo cartilage formation. Extrusion-based bioprinting was used for the biofabrication. The printing parameters were tuned to obtain stable material flow. In vivo data proved the ability of the tested bioink to form a cartilage within five to six weeks after the subcutaneous scaffold implantation. Certain areas of cartilage formation were detected as early as in one week. The resulting cartilage tissue had a distinctive structure with groups of isogenic cells as well as a high content of glycosaminoglycans and type II collagen.


Bioprinting/methods , Cartilage/cytology , Tissue Engineering/methods , Animals , Cartilage/metabolism , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrogenesis , Collagen/metabolism , Printing, Three-Dimensional/instrumentation , Rats , Tissue Scaffolds/chemistry
12.
J Extracell Vesicles ; 10(13): e12160, 2021 11.
Article En | MEDLINE | ID: mdl-34724347

Mesenchymal stem cell-derived small extracellular vesicles (MSC-sEVs) possess a great therapeutical potential for osteoarthritis (OA) treatment. However, the steric and electrostatic hindrance of cartilage matrix leads to very limited distribution of MSC-sEVs in cartilage and low bioavailability of MSC-sEVs after intra-articular injection. To overcome this, a strategy to reverse the surface charge of MSC-sEVs by modifying the MSC-sEVs with a novel cationic amphiphilic macromolecule namely ε-polylysine-polyethylene-distearyl phosphatidylethanolamine (PPD) was developed in this study. Through incubation with 100 µg/ml PPD, positively charged MSC-sEVs (PPD-sEVs) were obtained, and the modification process showed nearly no disturbance to the integrity and contents of sEVs and exhibited good stability under the interference of anionic macromolecules. A more effective cellular uptake and homeostasis modulation ability of PPD-sEVs than unmodified MSC-sEVs to chondrocytes was demonstrated. More importantly, PPD-sEVs demonstrated significantly enhanced cartilage uptake, cartilage penetration, and joint retention capacity as compared to MSC-sEVs. Intra-articular injection of PPD-sEVs into a mouse OA model showed significantly improved bioavailability than MSC-sEVs, which resulted in enhanced therapeutic efficacy with reduced injection frequency. In general, this study provides a facile and effective strategy to improve the intra-articular bioavailability of MSC-sEVs and has a great potential to accelerate the clinical practice of MSC-sEVs based OA therapy.


Cell- and Tissue-Based Therapy/methods , Extracellular Vesicles/drug effects , Mesenchymal Stem Cells/cytology , Osteoarthritis/therapy , Phosphatidylethanolamines/chemistry , Phosphatidylethanolamines/pharmacology , Polyethylene Glycols/chemistry , Polyethylene Glycols/pharmacology , Polylysine/chemistry , Polylysine/pharmacology , Adolescent , Animals , Cartilage/cytology , Cells, Cultured , Chondrocytes/metabolism , Disease Models, Animal , Humans , Induced Pluripotent Stem Cells/cytology , Injections, Intra-Articular , Male , Mice , Mice, Inbred C57BL , Swine , Treatment Outcome
13.
Bioengineered ; 12(1): 8622-8634, 2021 12.
Article En | MEDLINE | ID: mdl-34629037

Osteoarthritis (OA) is a common degenerative disease in middle-aged and elderly people. Our previous study has proved that microRNA-7 (miR-7) exacerbated the OA process. This study was aimed to explore the downstream genes and mechanism regulated by miR-7 to affect OA. Multiple EGF-like-domains 9 (MEGF9) was the predicted target of miR-7 by databases. Luciferase report experiment results confirmed that MEGF9 could bind to miR-7. Among the 10 collected pairs of OA and healthy samples, the expression levels of miR-7 and MEGF9 were both up-regulated when compared with healthy subjects by qRT-PCR and immunohistochemistry (IHC). The increased MEGF9 levels were due to the interaction with epidermal growth factor receptor (EGFR) by co-immunoprecipitation. Evaluations found that upregulation of miR-7 or MEGF9 can increase the expression of EGFR, matrix metalloproteinase-13 (MMP-13) and a disintegrin like and metallopeptidase with thrombospondin type 1 motif 5 (ADAMTS-5), so as to aggravate cartilage degradation. In addition, this effect induced by miR-7/EGFR/MEGF9 axis was by activation of PI3K/AKT signaling. The IHC and western blot assay results on OA model mice also demonstrated that miR-7/EGFR/MEGF9 axis regulated cartilage degradation in vivo. In summary, miR-7/EGFR/MEGF9 axis may perform a crucial function in the regulation of OA, providing potential for OA treatment.


Cartilage , Membrane Proteins/genetics , MicroRNAs/genetics , Nerve Tissue Proteins/genetics , Osteoarthritis , Animals , Cartilage/cytology , Cartilage/metabolism , Cartilage/pathology , Cells, Cultured , Chondrocytes/metabolism , ErbB Receptors/genetics , ErbB Receptors/metabolism , Humans , Male , Membrane Proteins/metabolism , Mice , MicroRNAs/metabolism , Nerve Tissue Proteins/metabolism , Osteoarthritis/metabolism , Osteoarthritis/pathology , Phosphatidylinositol 3-Kinases/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
14.
Int J Mol Sci ; 22(19)2021 Oct 08.
Article En | MEDLINE | ID: mdl-34639200

To date, pharmacological strategies designed to accelerate bone fracture healing are lacking. We subjected 8-week-old C57BL/6 male mice to closed, transverse, mid-diaphyseal tibial fractures and treated them with intraperitoneal injection of a vehicle or r-irisin (100 µg/kg/weekly) immediately following fracture for 10 days or 28 days. Histological analysis of the cartilaginous callus at 10 days showed a threefold increase in Collagen Type X (p = 0.0012) and a reduced content of proteoglycans (40%; p = 0.0018). Osteoclast count within the callus showed a 2.4-fold increase compared with untreated mice (p = 0.026), indicating a more advanced stage of endochondral ossification of the callus during the early stage of fracture repair. Further evidence that irisin induced the transition of cartilage callus into bony callus was provided by a twofold reduction in the expression of SOX9 (p = 0.0058) and a 2.2-fold increase in RUNX2 (p = 0.0137). Twenty-eight days post-fracture, microCT analyses showed that total callus volume and bone volume were increased by 68% (p = 0.0003) and 67% (p = 0.0093), respectively, and bone mineral content was 74% higher (p = 0.0012) in irisin-treated mice than in controls. Our findings suggest that irisin promotes bone formation in the bony callus and accelerates the fracture repair process, suggesting a possible use as a novel pharmacologic modulator of fracture healing.


Cartilage/cytology , Fibronectins/administration & dosage , Fracture Healing , Fractures, Bone/therapy , Osteoclasts/cytology , Osteogenesis , Recombinant Proteins/administration & dosage , Animals , Cartilage/metabolism , Male , Mice , Mice, Inbred C57BL , Osteoclasts/metabolism
15.
Commun Biol ; 4(1): 1125, 2021 09 24.
Article En | MEDLINE | ID: mdl-34561538

Previous findings on dinosaur cartilage material from the Late Cretaceous of Montana suggested that cartilage is a vertebrate tissue with unique characteristics that favor nuclear preservation. Here, we analyze additional dinosaur cartilage in Caudipteryx (STM4-3) from the Early Cretaceous Jehol biota of Northeast China. The cartilage fragment is highly diagenetically altered when observed in ground-sections but shows exquisite preservation after demineralization. It reveals transparent, alumino-silicified chondrocytes and brown, ironized chondrocytes. The histochemical stain Hematoxylin and Eosin (that stains the nucleus and cytoplasm in extant cells) was applied to both the demineralized cartilage of Caudipteryx and that of a chicken. The two specimens reacted identically, and one dinosaur chondrocyte revealed a nucleus with fossilized threads of chromatin. This is the second example of fossilized chromatin threads in a vertebrate material. These data show that some of the original nuclear biochemistry is preserved in this dinosaur cartilage material and further support the hypothesis that cartilage is very prone to nuclear fossilization and a perfect candidate to further understand DNA preservation in deep time.


Cartilage/cytology , Cell Nucleus/chemistry , Dinosaurs , Fossils , Animals , China
16.
Biomed Mater ; 16(6)2021 09 28.
Article En | MEDLINE | ID: mdl-34507313

Cartilage damage is one of the main causes of disability, and 3D bioprinting technology can produce complex structures that are particularly suitable for constructing a customized and irregular tissue engineering scaffold for cartilage repair. Alginate is an attractive biomaterial for bioinks because of its good biological safety profile and fast ionic gelation. However, ionically crosslinked alginate hydrogels are recognized as lacking enough mechanical property and long-term stability due to ion exchange. Here, we developed a double crosslinked alginate (DC-Alg) hydrogel for 3D bioprinting, and human umbilical cord mesenchymal stem cells (huMSCs) could differentiate into chondrocytes on its printed 3D scaffold after 4 weeks' culture. We performed sequential modification of alginate with L-cysteine and 5-norbornene-2-methylamine, and the DC-Alg hydrogels were obtained in the presence of CaCl2and ultraviolet light with stronger mechanical properties than those of the single ionic crosslinked alginate hydrogels, which was similar to natural cartilage. They also had better stability and could be maintained in DMEM medium for over 1 month, as well good viability for huMSCs. Moreover, the DC-Alg as 3D printing inks demonstrated a better printing accuracy (∼200 µm). After 4 weeks culture of huMSCs in the 3D printed DC-Alg scaffolds, the expressions of chondrogenic genes such asaggrecan (agg), collagen II (col II), and SRY-box transcription factor9(sox-9) were obviously observed, indicating the differentiation of huMSCs into cartilage. Immumohistochemical staining analysis further exhibited cartilage tissue developed well in the 3D printed scaffolds. Our study is the first demonstration of DC-Alg in 3D printing for MSC differentiation into cartilage, which shows a potential application in cartilage defect repair.


Alginates , Bioprinting , Cartilage , Printing, Three-Dimensional , Tissue Engineering/methods , Alginates/chemistry , Alginates/pharmacology , Cartilage/cytology , Cartilage/metabolism , Cell Differentiation/drug effects , Chondrocytes/drug effects , Chondrocytes/metabolism , Chondrogenesis/drug effects , Female , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism
17.
Carbohydr Polym ; 270: 118330, 2021 Oct 15.
Article En | MEDLINE | ID: mdl-34364592

Based on the gradient distribution of structure and composition in biological cartilage tissue, we designed a gradient hydrogel scaffold by the moving photomask, using chondroitin sulfate and poly (γ-glutamic acid) as crude materials. The hydrogel scaffold had a gradient distribution of cross-linking density, which can be verified from the results of SEM and swelling behavior. Besides, the hydrogel exhibited great viscoelastic, toughness (70% strain), and strength properties (600 kPa). Additionally, the gradient hydrogel's superior cell compatibility was proved through the MTT, live/dead staining assays, and 3D cell culture experiments. Remarkably, the results of in vitro stem cell differentiation experiments showed that the duration of light directly affected the differentiation extent of stem cells, demonstrating that the gradient hydrogel scaffold can better simulate the function of natural cartilage than the homogeneous one. Due to these outstanding characteristics, this gradient hydrogel is a potential scaffold for cartilage tissue engineering.


Cartilage/cytology , Chondroitin Sulfates/chemistry , Hydrogels/chemistry , Polyglutamic Acid/analogs & derivatives , Stem Cells/metabolism , Tissue Engineering/methods , Animals , Cartilage/metabolism , Cell Differentiation , Cell Line , Chondrocytes/metabolism , Mice , Microscopy, Electron, Scanning/methods , Polyglutamic Acid/chemistry , Rheology/methods , Thermogravimetry/methods , Tissue Scaffolds/chemistry
18.
J Struct Biol ; 213(4): 107781, 2021 12.
Article En | MEDLINE | ID: mdl-34411695

The interphase region at the base of the growth plate includes blood vessels, cells and mineralized tissues. In this region, cartilage is mineralized and replaced with bone. Blood vessel extremities permeate this space providing nutrients, oxygen and signaling factors. All these different components form a complex intertwined 3D structure. Here we use cryo-FIB SEM to elaborate this 3D structure without removing the water. As it is challenging to image mineralized and unmineralized tissues in a hydrated state, we provide technical details of the parameters used. We obtained two FIB SEM image stacks that show that the blood vessels are in intimate contact not only with cells, but in some locations also with mineralized tissues. There are abundant red blood cells at the extremities of the vessels. We also documented large multinucleated cells in contact with mineralized cartilage and possibly also with bone. We observed membrane bound mineralized particles in these cells, as well as in blood serum, but not in the hypertrophic chondrocytes. We confirm that there is an open pathway from the blood vessel extremities to the mineralizing cartilage. Based on the sparsity of the mineralized particles, we conclude that mainly ions in solution are used for mineralizing cartilage and bone, but these are augmented by the supply of mineralized particles.


Cartilage/ultrastructure , Cryoelectron Microscopy/methods , Growth Plate/ultrastructure , Imaging, Three-Dimensional/methods , Microscopy, Electron, Scanning/methods , Tibia/ultrastructure , Animals , Basement Membrane/ultrastructure , Blood Vessels/cytology , Blood Vessels/ultrastructure , Bone Development , Calcification, Physiologic , Cartilage/cytology , Cartilage/growth & development , Cell Differentiation , Chondrocytes/cytology , Chondrocytes/metabolism , Chondrocytes/ultrastructure , Extracellular Matrix/metabolism , Extracellular Matrix/ultrastructure , Female , Growth Plate/cytology , Growth Plate/growth & development , Mice, Inbred BALB C , Morphogenesis , Tibia/cytology , Tibia/growth & development
19.
Int J Mol Sci ; 22(14)2021 Jul 14.
Article En | MEDLINE | ID: mdl-34299147

The mandibular and hyoid arches collectively make up the facial skeleton, also known as the viscerocranium. Although all three germ layers come together to assemble the pharyngeal arches, the majority of tissue within viscerocranial skeletal components differentiates from the neural crest. Since nearly one third of all birth defects in humans affect the craniofacial region, it is important to understand how signalling pathways and transcription factors govern the embryogenesis and skeletogenesis of the viscerocranium. This review focuses on mouse and zebrafish models of craniofacial development. We highlight gene regulatory networks directing the patterning and osteochondrogenesis of the mandibular and hyoid arches that are actually conserved among all gnathostomes. The first part of this review describes the anatomy and development of mandibular and hyoid arches in both species. The second part analyses cell signalling and transcription factors that ensure the specificity of individual structures along the anatomical axes. The third part discusses the genes and molecules that control the formation of bone and cartilage within mandibular and hyoid arches and how dysregulation of molecular signalling influences the development of skeletal components of the viscerocranium. In conclusion, we notice that mandibular malformations in humans and mice often co-occur with hyoid malformations and pinpoint the similar molecular machinery controlling the development of mandibular and hyoid arches.


Body Patterning , Cartilage/embryology , Hyoid Bone/embryology , Mandible/embryology , Animals , Cartilage/cytology
20.
Cell Tissue Res ; 386(2): 309-320, 2021 Nov.
Article En | MEDLINE | ID: mdl-34241697

Cartilage has little intrinsic capacity for repair, so transplantation of exogenous cartilage cells is considered a realistic option for cartilage regeneration. We explored whether human-induced pluripotent stem cells (hiPSCs) could represent such unlimited cell sources for neo-cartilage comparable to human primary articular chondrocytes (hPACs) or human bone marrow-derived mesenchymal stromal cells (hBMSCs). For this, chondroprogenitor cells (hiCPCs) and hiPSC-derived mesenchymal stromal cells (hiMSCs) were generated from two independent hiPSC lines and characterized by morphology, flow cytometry, and differentiation potential. Chondrogenesis was compared to hBMSCs and hPACs by histology, immunohistochemistry, and RT-qPCR, while similarities were estimated based on Pearson correlations using a panel of 20 relevant genes. Our data show successful differentiations of hiPSC into hiMSCs and hiCPCs. Characteristic hBMSC markers were shared between hBMSCs and hiMSCs, with the exception of CD146 and CD45. However, neo-cartilage generated from hiMSCs showed low resemblances when compared to hBMSCs (53%) and hPACs (39%) characterized by lower collagen type 2 and higher collagen type 1 expression. Contrarily, hiCPC neo-cartilage generated neo-cartilage more similar to hPACs (65%), with stronger expression of matrix deposition markers. Our study shows that taking a stepwise approach to generate neo-cartilage from hiPSCs via chondroprogenitor cells results in strong similarities to neo-cartilage of hPACs within 3 weeks following chondrogenesis, making them a potential candidate for regenerative therapies. Contrarily, neo-cartilage deposited by hiMSCs seems more prone to hypertrophic characteristics compared to hPACs. We therefore compared chondrocytes derived from hiMSCs and hiCPCs with hPACs and hBMSCs to outline similarities and differences between their neo-cartilage and establish their potential suitability for regenerative medicine and disease modelling.


Cartilage/cytology , Chondrocytes/cytology , Induced Pluripotent Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Cartilage/metabolism , Cell Differentiation , Cell Line , Chondrocytes/metabolism , Chondrogenesis , Humans , Induced Pluripotent Stem Cells/metabolism , Mesenchymal Stem Cells/metabolism , Transcriptome
...