Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Biochem Biophys Res Commun ; 727: 150277, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38936225

ABSTRACT

With the aging of the global demographic, the prevention and treatment of osteoporosis are becoming crucial issues. The gradual loss of self-renewal and osteogenic differentiation capabilities in bone marrow stromal cells (BMSCs) is one of the key factors contributing to osteoporosis. To explore the regulatory mechanisms of BMSCs differentiation, we collected bone marrow cells of femoral heads from patients undergoing total hip arthroplasty for single-cell RNA sequencing analysis. Single-cell RNA sequencing revealed significantly reduced CRIP1 (Cysteine-Rich Intestinal Protein 1) expression and osteogenic capacity in the BMSCs of osteoporosis patients compared to non-osteoporosis group. CRIP1 is a gene that encodes a member of the LIM/double zinc finger protein family, which is involved in the regulation of various cellular processes including cell growth, development, and differentiation. CRIP1 knockdown resulted in decreased alkaline phosphatase activity, mineralization and expression of osteogenic markers, indicating impaired osteogenic differentiation. Conversely, CRIP1 overexpression, both in vitro and in vivo, enhanced osteogenic differentiation and rescued bone mass reduction in ovariectomy-induced osteoporosis mice model. The study further established CRIP1's modulation of osteogenesis through the Wnt signaling pathway, suggesting that targeting CRIP1 could offer a novel approach for osteoporosis treatment by promoting bone formation and preventing bone loss.

2.
Cell Mol Life Sci ; 81(1): 260, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38878096

ABSTRACT

The pathological advancement of osteoporosis is caused by the uneven development of bone marrow-derived mesenchymal stem cells (BMSCs) in terms of osteogenesis and adipogenesis. While the role of EEF1B2 in intellectual disability and tumorigenesis is well established, its function in the bone-fat switch of BMSCs is still largely unexplored. During the process of osteogenic differentiation, we observed an increase in the expression of EEF1B2, while a decrease in its expression was noted during adipogenesis. Suppression of EEF1B2 hindered the process of osteogenic differentiation and mineralization while promoting adipogenic differentiation. On the contrary, overexpression of EEF1B2 enhanced osteogenesis and strongly inhibited adipogenesis. Furthermore, the excessive expression of EEF1B2 in the tibias has the potential to mitigate bone loss and decrease marrow adiposity in mice with osteoporosis. In terms of mechanism, the suppression of ß-catenin activity occurred when EEF1B2 function was suppressed during osteogenesis. Our collective findings indicate that EEF1B2 functions as a regulator, influencing the differentiation of BMSCs and maintaining a balance between bone and fat. Our finding highlights its potential as a therapeutic target for diseases related to bone metabolism.


Subject(s)
Adipogenesis , Cell Differentiation , Mesenchymal Stem Cells , Osteogenesis , Osteoporosis , Wnt Signaling Pathway , beta Catenin , Animals , Male , Mice , Adipogenesis/genetics , beta Catenin/metabolism , Bone Marrow Cells/metabolism , Bone Marrow Cells/cytology , Cells, Cultured , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Mice, Inbred C57BL , Osteogenesis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Peptide Elongation Factor 1/metabolism , Guanine Nucleotide Exchange Factors/metabolism
3.
FASEB J ; 38(9): e23657, 2024 May 15.
Article in English | MEDLINE | ID: mdl-38713087

ABSTRACT

The pathogenesis of osteoporosis (OP) is closely associated with the disrupted balance between osteogenesis and adipogenesis in bone marrow-derived mesenchymal stem cells (BMSCs). We analyzed published single-cell RNA sequencing (scRNA-seq) data to dissect the transcriptomic profiles of bone marrow-derived cells in OP, reviewing 56 377 cells across eight scRNA-seq datasets from femoral heads (osteoporosis or osteopenia n = 5, osteoarthritis n = 3). Seventeen genes, including carboxypeptidase M (CPM), were identified as key osteogenesis-adipogenesis regulators through comprehensive gene set enrichment, differential expression, regulon activity, and pseudotime analyses. In vitro, CPM knockdown reduced osteogenesis and promoted adipogenesis in BMSCs, while adenovirus-mediated CPM overexpression had the reverse effects. In vivo, intraosseous injection of CPM-overexpressing BMSCs mitigated bone loss in ovariectomized mice. Integrated scRNA-seq and bulk RNA sequencing analyses provided insight into the MAPK/ERK pathway's role in the CPM-mediated regulation of BMSC osteogenesis and adipogenesis; specifically, CPM overexpression enhanced MAPK/ERK signaling and osteogenesis. In contrast, the ERK1/2 inhibitor binimetinib negated the effects of CPM overexpression. Overall, our findings identify CPM as a pivotal regulator of BMSC differentiation, which provides new clues for the mechanistic study of OP.


Subject(s)
Adipogenesis , Carboxypeptidases , MAP Kinase Signaling System , Mesenchymal Stem Cells , Osteogenesis , Single-Cell Analysis , Animals , Female , Humans , Mice , Carboxypeptidases/metabolism , Carboxypeptidases/genetics , Cell Differentiation , GPI-Linked Proteins , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Metalloendopeptidases , Mice, Inbred C57BL , Osteogenesis/physiology , Osteogenesis/genetics , Osteoporosis/genetics , Osteoporosis/metabolism , Osteoporosis/pathology , Transcriptome
4.
Res Sq ; 2024 May 03.
Article in English | MEDLINE | ID: mdl-38234849

ABSTRACT

The IncRNA Malat1 was initially believed to be dispensable for physiology due to the lack of observable phenotypes in Malat1 knockout (KO) mice. However, our study challenges this conclusion. We found that both Malat1 KO and conditional KO mice in the osteoblast lineage exhibit significant osteoporosis. Mechanistically, Malat1 acts as an intrinsic regulator in osteoblasts to promote osteogenesis. Interestingly, Malat1 does not directly affect osteoclastogenesis but inhibits osteoclastogenesis in a non-autonomous manner in vivo via integrating crosstalk between multiple cell types, including osteoblasts, osteoclasts and chondrocytes. Our findings substantiate the existence of a novel remodeling network in which Malat1 serves as a central regulator by binding to ß-catenin and functioning through the ß-catenin-OPG/Jagged1 pathway in osteoblasts and chondrocytes. In pathological conditions, Malat1 significantly promotes bone regeneration in fracture healing. Bone homeostasis and regeneration are crucial to well-being. Our discoveries establish a previous unrecognized paradigm model of Malat1 function in the skeletal system, providing novel mechanistic insights into how a lncRNA integrates cellular crosstalk and molecular networks to fine tune tissue homeostasis, remodeling and repair.

5.
J Orthop Surg Res ; 18(1): 751, 2023 Oct 04.
Article in English | MEDLINE | ID: mdl-37794464

ABSTRACT

BACKGROUND: GEM (GTP-binding protein overexpressed in skeletal muscle) is one of the atypical small GTPase subfamily members recently identified as a regulator of cell differentiation. Abnormal chondrogenesis coupled with an imbalance in the turnover of cartilaginous matrix formation is highly relevant to the onset and progression of osteoarthritis (OA). However, how GEM regulates chondrogenic differentiation remains unexplored. METHODS: Cartilage tissues were obtained from OA patients and graded according to the ORASI and ICRS grading systems. The expression alteration of GEM was detected in the Grade 4 cartilage compared to Grade 0 and verified in OA mimic culture systems. Next, to investigate the specific function of GEM during these processes, we generated a Gem knockdown (Gem-Kd) system by transfecting siRNA targeting Gem into ATDC5 cells. Acan, Col2a1, Sox9, and Wnt target genes of Gem-Kd ATDC5 cells were detected during induction. The transcriptomic sequencing analysis was performed to investigate the mechanism of GEM regulation. Wnt signaling pathways were verified by real-time PCR and immunoblot analysis. Finally, a rescue model generated by treating Gem-KD ATDC5 cells with a Wnt signaling agonist was established to validate the mechanism identified by RNA sequencing analysis. RESULTS: A decreased expression of GEM in OA patients' cartilage tissues and OA mimic chondrocytes was observed. While during chondrogenesis differentiation and cartilage matrix formation, the expression of GEM was increased. Gem silencing suppressed chondrogenic differentiation and the expressions of Acan, Col2a1, and Sox9. RNA sequencing analysis revealed that Wnt signaling was downregulated in Gem-Kd cells. Decreased expression of Wnt signaling associated genes and the total ß-CATENIN in the nucleus and cytoplasm were observed. The exogenous Wnt activation exhibited reversed effect on Gem loss-of-function cells. CONCLUSION: These findings collectively validated that GEM functions as a novel regulator mediating chondrogenic differentiation and cartilage matrix formation through Wnt/ß-catenin signaling.


Subject(s)
Cartilage, Articular , Osteoarthritis , Humans , Wnt Signaling Pathway/genetics , beta Catenin/genetics , beta Catenin/metabolism , Chondrogenesis/genetics , Cartilage/metabolism , Chondrocytes/metabolism , Cell Differentiation/genetics , Osteoarthritis/genetics , Osteoarthritis/metabolism , Cartilage, Articular/metabolism , Cells, Cultured
6.
iScience ; 26(8): 107200, 2023 Aug 18.
Article in English | MEDLINE | ID: mdl-37554462

ABSTRACT

Human epiphyseal development has been mainly investigated through radiological and histological approaches, uncovering few details of cellular temporal genetic alternations. Using single-cell RNA sequencing, we investigated the dynamic transcriptome changes during post-conception weeks (PCWs) 15-25 of human distal femoral epiphysis cells. We find epiphyseal cells contain multiple subtypes distinguished by specific markers, gene signatures, Gene Ontology (GO) enrichment analysis, and gene set variation analysis (GSVA). We identify the populations committed to cartilage or ossification at this time, although the secondary ossification centers (SOCs) have not formed. We describe the temporal alternation in transcriptional expression utilizing trajectories, transcriptional regulatory networks, and intercellular communication analyses. Moreover, we find the emergence of the ossification-committed population is correlated with the COL2A1-(ITGA2/11+ITGB1) signaling. NOTCH signaling may contribute to the formation of cartilage canals and ossification via NOTCH signaling. Our findings will advance the understanding of single-cell genetic changes underlying fetal epiphysis development.

7.
Commun Biol ; 5(1): 324, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35388143

ABSTRACT

Alcohol-induced osteonecrosis of the femoral head (ONFH) is a disabling disease with a high incidence and elusive pathogenesis. Here, we used single-cell RNA sequencing to explore the transcriptomic landscape of mid- and advanced-stage alcohol-induced ONFH. Cells derived from age-matched hip osteoarthritis and femoral neck fracture samples were used as control. Our bioinformatics analysis revealed the disorder of osteogenic-adipogenic differentiation of stromal cells in ONFH and altered regulons such as MEF2C and JUND. In addition, we reported that one of the endothelial cell clusters with ACKR1 expression exhibited strong chemotaxis and a weak angiogenic ability and expanded with disease progression. Furthermore, ligand-receptor-based cell-cell interaction analysis indicated that ACKR1+ endothelial cells might specifically communicate with stromal cells through the VISFATIN and SELE pathways, thus influencing stromal cell differentiation in ONFH. Overall, our data revealed single cell transcriptome characteristics in alcohol-induced ONFH, which may contribute to the further investigation of ONFH pathogenesis.


Subject(s)
Osteonecrosis , Transcriptome , Endothelial Cells/pathology , Ethanol , Femur Head/pathology , Gene Expression Profiling , Humans , Osteonecrosis/pathology , Stromal Cells
8.
Bioengineered ; 12(1): 997-1007, 2021 12.
Article in English | MEDLINE | ID: mdl-33749514

ABSTRACT

Osteoarthritis (OA) occurs mostly in the knees, hips, finger interphalangeal joints, and spinal facet joints, and is characterized by cartilage degeneration. The existing bulk RNA sequencing (bulk RNA-seq) and single-cell sequencing (scRNA-seq) data for chondrocytes in the osteoarthritic knee joint provide the expression profiles of entire cell populations and individual cells, respectively. Here, we aimed to analyze these two types of sequencing data in order to obtain a more comprehensive understanding of OA. We compared the analysis results of bulk RNA-seq and scRNA-seq from the dataset GSE114007 and the dataset GSE104782, respectively, and identified the differentially expressed genes (DEGs). Then, we tried to find the key The transcription factor is a more fomal term (TFs) and long non-coding RNA (lncRNA) regulation. We highlighted 271 genes that were simultaneously suggested by these two types of data and provided their possible expression pattern in OA. Among the 271 genes, we identified 14 TFs, and TWIST2, MYBL2, RELA, JUN, KLF4, and PTTG1 could be the key TFs for the 271 genes. We also found that 8 lncRNAs among the 271 genes and the lncRNA regulation between CYTOR and NRP1 could contribute to the pain and vascularization of cartilage in the osteoarthritic knee. In short, our research combined the analysis results of bulk RNA-seq and scRNA-seq data for OA chondrocytes, which will contribute to further elucidation of the molecular mechanisms of OA pathogenesis.[Figure: see text].


Subject(s)
Chondrocytes/metabolism , Gene Expression Regulation , Knee Joint/pathology , Osteoarthritis/genetics , Osteoarthritis/pathology , Sequence Analysis, RNA , Single-Cell Analysis , Gene Expression Profiling , Gene Regulatory Networks , Humans , Kruppel-Like Factor 4 , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
9.
Elife ; 92020 12 29.
Article in English | MEDLINE | ID: mdl-33373293

ABSTRACT

Bone remodeling involves a balance between bone resorption and formation. The mechanisms underlying bone remodeling are not well understood. DEF6 is recently identified as a novel loci associated with bone mineral density. However, it is unclear how Def6 impacts bone remodeling. We identify Def6 as a novel osteoblastic regulator that suppresses osteoblastogenesis and bone formation. Def6 deficiency enhances both bone resorption and osteogenesis. The enhanced bone resorption in Def6-/- mice dominates, leading to osteoporosis. Mechanistically, Def6 inhibits the differentiation of both osteoclasts and osteoblasts via a common mechanism through endogenous type-I IFN-mediated feedback inhibition. RNAseq analysis shows expression of a group of IFN stimulated genes (ISGs) during osteoblastogenesis. Furthermore, we found that Def6 is a key upstream regulator of IFNß and ISG expression in osteoblasts. Collectively, our results identify a novel immunoregulatory function of Def6 in bone remodeling, and shed insights into the interaction between immune system and bone.


Subject(s)
Interferon-gamma/physiology , Osteoblasts/physiology , Osteogenesis/physiology , Animals , Gene Expression Regulation , Interferon Regulatory Factors/physiology , Interferon-gamma/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout
10.
Nan Fang Yi Ke Da Xue Xue Bao ; 40(11): 1656-1661, 2020 Nov 30.
Article in Chinese | MEDLINE | ID: mdl-33243734

ABSTRACT

OBJECTIVE: To assess the value of Ploton silver staining and phalloidin-iFlour 488 staining in observation of the morphology of osteocyte dendrites of mice at different developmental stages. METHODS: The humerus and femurs were harvested from mice at 0 (P0), 5 (P5), 15 (P15), 21 (P21), 28 (P28), and 35 days (P35) after birth to prepare cryo-sections and paraffin sections. HE staining of P35 mouse femur sections served as a reference for observing osteocytes in the trabecular bone and cortical bone. The humeral sections at different developmental stages were stained with Ploton silver staining to observe the morphology of osteocytes and canaliculi, and the canalicular lengths in the cortical and trabecular bones of the humerus of the mice in each developmental stage were recorded. The cryo-sections of the humerus from P10 and P15 mice were stained with phalloidin iFlour-488 to observe the morphology of osteocytes and measurement of the length of osteocyte dendrites in the cortical bone. RESULTS: In the trabecular bone of the humerus of P0-P15 mice, Ploton silver staining only visualized the outline of the osteocytes, and the morphology of the canaliculi was poorly defined. In P21 or older mice, Ploton silver staining revealed the morphology of the trabecular bone osteocytes and the canaliculi, which were neatly arranged and whose lengths increased significantly with age (P21 vs P28, P < 0.05; P21 vs P35, P < 0.05). In the humeral cortical bone of P15 mice, the morphology of the osteocytes and canalicular could be observed with Ploton silver staining, and the length of the regularly arranged canaliculi of the osteocytes increased significantly with age (P15 vs P21, P < 0.005; P15 vs P28, P < 0.0001; P15 vs P35, P < 0.0001). Phalloidin iFlour-488 staining was capable of visualizing the complete morphology of the osteocytes at P10 and P15; the osteocyte dendrites elongated progressively with age (P10 vs P15, P < 0.01) to form connections with the surrounding osteocytes. CONCLUSIONS: Mouse osteocyte dendrites elongate progressively and their arrangement gradually becomes regular with age. Ploton silver staining can clearly visualize the morphology of the osteocytes and the canaliculi in adult mice but not in mice in early stages of development. Phalloidin iFlour-488 staining for labeling the cytoskeleton can be applied for mouse osteocytes at all developmental stages and allows morphological observation of mouse osteocytes in early developmental stages.


Subject(s)
Bone and Bones , Osteocytes , Animals , Dendrites , Mice , Phalloidine , Silver Staining
11.
Nan Fang Yi Ke Da Xue Xue Bao ; 39(9): 1038-1044, 2019 Sep 30.
Article in Chinese | MEDLINE | ID: mdl-31640956

ABSTRACT

OBJECTIVE: To optimize the method for embedding multiple undecalcified mouse tibias in plastic blocks, improve the efficiency and stability of plastic embedding and reduce the detachment rate of plastic slides. METHODS: Thirty undecalcified tibias from 15 B6 mice were used for plastic embedding after calcein labeling, fixation, dehydration and infiltration. The tibias were embedded in cylindrical plastic blocks with a diameter of 4 mm. For each bone, the 1/4 proximal tibia was cut off, and the remaining 3/4 was used for re-embedding. Five bones were embedded in a single block with each bone standing closely on the surface of a flat plate. The samples were randomized into control and experimental groups in all the processes of embedding, sectioning and staining. In the 3 groups with modified embedment, flowing CO2 was added into the embedding solution, embedding solution was applied to the section surface, and the slides were heated at 95 ℃ for 15 min. The polymerization time, slide detachment rate, bone formation and osteoblast parameters were analyzed. RESULTS: We prepared 6 plastic blocks, each containing 5 tibias, whose cross sections were on the same plane. The blocks were completely polymerized and suitable for sectioning. Flowing CO2 into the embedding solution reduced the polymerization time and increased the rate of complete polymerization. Application of the embedding solution on the section surface significantly reduced the detachment rate of the sections (P < 0.05) without affecting bone formation analysis (P > 0.05). Heating the slides significantly lowered the detachment rate of the sections (P < 0.05) without affecting osteoblast analysis (P > 0.05). CONCLUSIONS: The optimized method allows effective embedding of multiple undecalcified mice tibias in the same block and can be an ideal method for histological analysis of undecalcified bones.


Subject(s)
Plastics , Tibia , Tissue Embedding/methods , Animals , Mice , Staining and Labeling
12.
Sci Rep ; 9(1): 4236, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30862896

ABSTRACT

Excessive osteoclastic bone erosion disrupts normal bone remodeling and leads to bone loss in many skeletal diseases, including inflammatory arthritis, such as rheumatoid arthritis (RA) and psoriatic arthritis, periodontitis and peri-prosthetic loosening. Functional control of osteoclasts is critical for the maintenance of bone homeostasis. However, the mechanisms that restrain osteoclast resorptive function are not fully understood. In this study, we identify a previously unrecognized role for G-protein Gα13 in inhibition of osteoclast adhesion, fusion and bone resorptive function. Gα13 is highly expressed in mature multinucleated osteoclasts, but not during early differentiation. Deficiency of Gα13 in myeloid osteoclast lineage (Gα13ΔM/ΔM mice) leads to super spread morphology of multinucleated giant osteoclasts with elevated bone resorptive capacity, corroborated with an osteoporotic bone phenotype in the Gα13ΔM/ΔM mice. Mechanistically, Gα13 functions as a brake that restrains the c-Src, Pyk2, RhoA-Rock2 mediated signaling pathways and related gene expressions to control the ability of osteoclasts in fusion, adhesion, actin cytoskeletal remodeling and resorption. Genome wide analysis reveals cytoskeleton related genes that are suppressed by Gα13, identifying Gα13 as a critical cytoskeletal regulator in osteoclasts. We also identify a genome wide regulation of genes responsible for mitochondrial biogenesis and function by Gα13 in osteoclasts. Furthermore, the significant correlation between Gα13 expression levels, TNF activity and RA disease activity in RA patients suggests that the Gα13 mediated mechanisms represent attractive therapeutic targets for diseases associated with excessive bone resorption.


Subject(s)
Cytoskeleton/metabolism , GTP-Binding Protein alpha Subunits, G12-G13/metabolism , Mitochondria/metabolism , Osteoclasts/metabolism , Animals , Bone Resorption/genetics , Bone Resorption/metabolism , Cell Adhesion , Cell Fusion , Cytoskeleton/genetics , GTP-Binding Protein alpha Subunits, G12-G13/genetics , Genome-Wide Association Study , Mice , Mice, Transgenic , Mitochondria/genetics
13.
Nat Commun ; 9(1): 4108, 2018 10 05.
Article in English | MEDLINE | ID: mdl-30291236

ABSTRACT

Targeting microRNAs recently shows significant therapeutic promise; however, such progress is underdeveloped in treatment of skeletal diseases with osteolysis, such as osteoporosis and rheumatoid arthritis (RA). Here, we identified miR-182 as a key osteoclastogenic regulator in bone homeostasis and diseases. Myeloid-specific deletion of miR-182 protects mice against excessive osteoclastogenesis and bone resorption in disease models of ovariectomy-induced osteoporosis and inflammatory arthritis. Pharmacological treatment of these diseases with miR-182 inhibitors completely suppresses pathologic bone erosion. Mechanistically, we identify protein kinase double-stranded RNA-dependent (PKR) as a new and essential miR-182 target that is a novel inhibitor of osteoclastogenesis via regulation of the endogenous interferon (IFN)-ß-mediated autocrine feedback loop. The expression levels of miR-182, PKR, and IFN-ß are altered in RA and are significantly correlated with the osteoclastogenic capacity of RA monocytes. Our findings reveal a previously unrecognized regulatory network mediated by miR-182-PKR-IFN-ß axis in osteoclastogenesis, and highlight the therapeutic implications of miR-182 inhibition in osteoprotection.


Subject(s)
Bone Resorption/prevention & control , Interferon-beta/metabolism , MicroRNAs/metabolism , Osteogenesis , eIF-2 Kinase/metabolism , Animals , Arthritis, Rheumatoid/complications , Arthritis, Rheumatoid/metabolism , Autocrine Communication , Bone Resorption/etiology , Female , Homeostasis , Male , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/antagonists & inhibitors , Monocytes/physiology
14.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(6): 716-722, 2018 Jun 20.
Article in Chinese | MEDLINE | ID: mdl-29997095

ABSTRACT

OBJECTIVE: To evaluate the effect of a novel biomaterial in repairing large cranial defects in rats. METHODS: Eighteen SD rats were used to establish rat modes of large cranial defect (8 mm in diameter). The rat models were randomized into 3 groups and the cranial defects were repaired using different scaffold materials, namely CPC paste prepared with distilled water (CPC control group), CPC paste mixed with 10% chitosan (CPC/CN group), or CPC paste with 10% chitosan and 300 mg adenosine (CPC/CN/AD group). The defects were examined 12 weeks after the surgery with X-ray, CT, HE staining and quantitative assessments. RESULTS: X-ray showed that the defect was repaired in all the groups. The fracture line became obscure and the defects were almost fully repaired by regenerated bone tissues in CPC/CN/AD group, which was consistent with CT findings. In all the 3 groups, HE staining revealed the presence of new bones in the defects and new vessels in and around the new bones without inflammatory cells. The new bone area was significantly greater in CPC/CN/AD group than in CPC/CN group and CPC control group (P<0.05). The new vessel density was the highest in CPC/CN/AD group (P>0.05) but similar between CPC/CN group and CPC control group (P>0.05). CONCLUSION: This novel calcium phosphate cement pre-loaded with chitosan and small molecule adenosine can better promote bone regeneration than calcium phosphate cement for repairing large bone defects to serve as a good replacement material for bone regeneration.


Subject(s)
Adenosine/administration & dosage , Bone Cements/therapeutic use , Bone Regeneration/drug effects , Calcium Phosphates/therapeutic use , Chitosan/administration & dosage , Skull Fractures/therapy , Adenosine/chemistry , Animals , Calcium Phosphates/administration & dosage , Random Allocation , Rats , Rats, Sprague-Dawley
15.
Nan Fang Yi Ke Da Xue Xue Bao ; 38(1): 62-68, 2018 Jan 30.
Article in Chinese | MEDLINE | ID: mdl-33177019

ABSTRACT

OBJECTIVE: To assess the effect of ascorbic acid/ferric chloride (AA/FeCl3) in attenuating cartilage damage in rats with osteoarthritis. METHODS: Thirty adult male Wistar rats with surgically induced osteoarthritis were randomized into 2 groups for treatment with intra-articular injection of saline (control group) or AA/FeCl3 mixture (AA group) once a week starting from the third week after the operation. At 6, 9, and 12 weeks after the operation, 5 rats from each group were sacrificed for observing subchondral bone changes on X-ray films and evaluation of cartilage degeneration in the right knee joints using safranin-O/Fast green staining and a modified OARSI scoring system. The degradation of the cartilage matrix was observed by immunohistochemical staining for type Ⅱ collagen. RESULTS: X-ray examination in saline control group revealed the presence of osteophytes and narrowing of the joint space at 9 weeks, and the joint line disappeared at 12 weeks after the surgery; only slight irregularity of the articular surface was observed in the AA group at 9 and 12 weeks. OARSI scores were significantly lower in AA group than in the control group at 9 weeks (18.67±0.67 vs 12.17±2.75; P < 0.05) and 12 weeks (20.11±1.84 vs 13.77± 0.40; P < 0.05) but not at 6 weeks after the surgery. The content of type 2 collagen in AA group was significantly higher than that in the control group at 6 weeks (0.36±0.039 vs 0.49±0.029; P < 0.05) and 9 weeks after the surgery (0.25±0.041 vs 0.38±0.040; P < 0.05). CONCLUSIONS: Early intra-articular injection of AA/FeCl3 can effectively delay the progression of post-traumatic osteoarthritis in rats.

SELECTION OF CITATIONS
SEARCH DETAIL
...