Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
J Clin Invest ; 134(10)2024 Mar 21.
Article in English | MEDLINE | ID: mdl-38512413

ABSTRACT

Elevated bone resorption and diminished bone formation have been recognized as the primary features of glucocorticoid-associated skeletal disorders. However, the direct effects of excess glucocorticoids on bone turnover remain unclear. Here, we explored the outcomes of exogenous glucocorticoid treatment on bone loss and delayed fracture healing in mice and found that reduced bone turnover was a dominant feature, resulting in a net loss of bone mass. The primary effect of glucocorticoids on osteogenic differentiation was not inhibitory; instead, they cooperated with macrophages to facilitate osteogenesis. Impaired local nutrient status - notably, obstructed fatty acid transportation - was a key factor contributing to glucocorticoid-induced impairment of bone turnover in vivo. Furthermore, fatty acid oxidation in macrophages fueled the ability of glucocorticoid-liganded receptors to enter the nucleus and then promoted the expression of BMP2, a key cytokine that facilitates osteogenesis. Metabolic reprogramming by localized fatty acid delivery partly rescued glucocorticoid-induced pathology by restoring a healthier immune-metabolic milieu. These data provide insights into the multifactorial metabolic mechanisms by which glucocorticoids generate skeletal disorders, thus suggesting possible therapeutic avenues.


Subject(s)
Bone Remodeling , Glucocorticoids , Osteogenesis , Animals , Mice , Glucocorticoids/pharmacology , Osteogenesis/drug effects , Bone Remodeling/drug effects , Macrophages/metabolism , Macrophages/immunology , Macrophages/drug effects , Bone Morphogenetic Protein 2/metabolism , Bone Morphogenetic Protein 2/genetics , Fatty Acids/metabolism , Bone and Bones/metabolism , Bone and Bones/drug effects , Bone and Bones/immunology , Cellular Microenvironment/drug effects
2.
Adv Sci (Weinh) ; 10(22): e2300897, 2023 08.
Article in English | MEDLINE | ID: mdl-37218542

ABSTRACT

The knowledge of osteoarthritis (OA) has nowadays been extended from a focalized cartilage disorder to a multifactorial disease. Although recent investigations have reported that infrapatellar fat pad (IPFP) can trigger inflammation in the knee joint, the mechanisms behind the role of IPFP on knee OA progression remain to be defined. Here, dysregulated osteopontin (OPN) and integrin ß3 signaling are found in the OA specimens of both human and mice. It is further demonstrated that IPFP-derived OPN participates in OA progression, including activated matrix metallopeptidase 9 in chondrocyte hypertrophy and integrin ß3 in IPFP fibrosis. Motivated by these findings, an injectable nanogel is fabricated to provide sustained release of siRNA Cd61 (RGD- Nanogel/siRNA Cd61) that targets integrins. The RGD- Nanogel possesses excellent biocompatibility and desired targeting abilities both in vitro and in vivo. Local injection of RGD- Nanogel/siRNA Cd61 robustly alleviates the cartilage degeneration, suppresses the advancement of tidemark, and reduces the subchondral trabecular bone mass in OA mice. Taken together, this study provides an avenue for developing RGD- Nanogel/siRNA Cd61 therapy to mitigate OA progression via blocking OPN-integrin ß3 signaling in IPFP.


Subject(s)
Cartilage, Articular , Osteoarthritis, Knee , Humans , Mice , Animals , Integrin beta3 , Nanogels , Osteopontin , Knee Joint , Adipose Tissue , RNA, Small Interfering/genetics , Oligopeptides
3.
ACS Nano ; 16(11): 18071-18089, 2022 11 22.
Article in English | MEDLINE | ID: mdl-36108267

ABSTRACT

Bone grafting is frequently conducted to treat bone defects caused by trauma and tumor removal, yet with significant medical and socioeconomic burdens. Space-occupying bone substitutes remain challenging in the control of osteointegration, and meanwhile activation of endogenous periosteal cells by using non-space-occupying implants to promote new bone formation becomes another therapeutic strategy. Here, we fabricated a magnesium-based artificial bandage with optimal micropatterns for activating periosteum-associated biomineralization. Collagen was self-assembled on the surface of magnesium oxide nanoparticles embedded electrospun fibrous membranes as a hierarchical bandage structure to facilitate the integration with periosteum in situ. After the implantation on the surface of cortical bone in vivo, magnesium ions were released to generate a pro-osteogenic immune microenvironment by activating the endogenous periosteal macrophages into M2 phenotype and, meanwhile, promote blood vessel formation and neurite outgrowth. In a cortical bone defect model, magnesium-based artificial bandage guided the surrounding newly formed bone tissue to cover the defected area. Taken together, our study suggests that the strategy of stimulating bone formation can be achieved with magnesium delivery to periosteum in situ and the proposed periosteal bandages act as a bioactive media for accelerating bone healing.


Subject(s)
Nanoparticles , Osteogenesis , Magnesium Oxide/pharmacology , Bone Regeneration , Magnesium/pharmacology , Periosteum/physiology , Periosteum/transplantation , Cortical Bone , Bandages
4.
Adv Healthc Mater ; 11(18): e2201679, 2022 09.
Article in English | MEDLINE | ID: mdl-36026579

ABSTRACT

Guided bone regeneration (GBR) therapy demonstrates a prominent curative effect on the management of craniomaxillofacial (CMF) bone defects. In this study, a GBR membrane consisting of a microporous layer and a struvite-nanowire-doped fibrous layer is constructed via non-solvent induced phase separation, followed by an electrospinning procedure to treat critical-sized calvarial defects. The microporous layer shows selective permeability for excluding the rapid-growing non-osteogenic tissues and potential wound stabilization. The nanowire-like struvite is synthesized as the deliverable therapeutic agent within the fibrous layer to facilitate bone regeneration. Such a membrane displays a well-developed heterogeneous architecture, satisfactory mechanical performance, and long-lasting characteristics. The in vitro biological evaluation reveals that apart from being a strong barrier, the bilayer struvite-laden membrane can actively promote cellular adhesion, proliferation, and osteogenic differentiation. Consequently, the multifunctional struvite-doped membranes are applied to treat 5 mm-sized bilateral calvarial defects in rats, resulting in overall improved healing outcomes compared with the untreated or the struvite-free membrane-treated group, which is characterized by enhanced osteogenesis and significantly increased new bone formation. The encouraging preclinical results reveal the great potential of the bilayer struvite-doped membrane as a clinical GBR device for augmenting large-area CMF bone reconstruction.


Subject(s)
Nanowires , Osteogenesis , Animals , Biocompatible Materials/pharmacology , Bone Regeneration , Membranes, Artificial , Rats , Struvite/pharmacology
5.
ACS Appl Mater Interfaces ; 14(35): 39830-39842, 2022 Sep 07.
Article in English | MEDLINE | ID: mdl-36026585

ABSTRACT

Guided bone regeneration (GBR) technique using a barrier membrane holds great potential to allow the single-stage reconstruction of critical-sized bone defects. Here, bioactive nanoneedle-like magnesium oxychloride ceramics (MOCs) are synthesized and recruited as an osteoinductive factor within a polycaprolactone-gelatin A (PCL-GelA) membranous matrix to generate a periosteum-mimicking biphasic GBR membrane (PCL-GelA/MOC) to accelerate calvarial defect repair. The PCL-GelA/MOC membrane acts as a shield for defect areas and a reservoir of osteoinductive molecules, which provides a favorable microenvironment for supporting cell proliferation, infiltration, and differentiation. This membrane leads to accelerated osteogenesis and angiogenesis, effectual defect bridging, and significantly enhanced bone regeneration when applied to a 5 mm sized rat calvarial defect. This makes this innovative and multifunctional GBR membrane a suitable candidate for clinical applications with promising curative efficacy.


Subject(s)
Bone Regeneration , Magnesium , Animals , Ceramics/pharmacology , Osteogenesis , Periosteum , Rats
6.
Nat Commun ; 13(1): 427, 2022 01 20.
Article in English | MEDLINE | ID: mdl-35058428

ABSTRACT

Epididymal white adipose tissue (eWAT) secretes an array of cytokines to regulate the metabolism of organs and tissues in high-fat diet (HFD)-induced obesity, but its effects on bone metabolism are not well understood. Here, we report that macrophages in eWAT are a main source of osteopontin, which selectively circulates to the bone marrow and promotes the degradation of the bone matrix by activating osteoclasts, as well as modulating bone marrow-derived macrophages (BMDMs) to engulf the lipid droplets released from adipocytes in the bone marrow of mice. However, the lactate accumulation induced by osteopontin regulation blocks both lipolysis and osteoclastogenesis in BMDMs by limiting the energy regeneration by ATP6V0d2 in lysosomes. Both surgical removal of eWAT and local injection of either clodronate liposomes (for depleting macrophages) or osteopontin-neutralizing antibody show comparable amelioration of HFD-induced bone loss in mice. These results provide an avenue for developing therapeutic strategies to mitigate obesity-related bone disorders.


Subject(s)
Adipose Tissue/metabolism , Bone and Bones/metabolism , Epididymis/metabolism , Homeostasis , Macrophages/metabolism , Osteopontin/metabolism , Adipose Tissue/diagnostic imaging , Adipose Tissue, White/diagnostic imaging , Adipose Tissue, White/metabolism , Animals , Body Weight , Bone Resorption/pathology , Bone and Bones/diagnostic imaging , CD11b Antigen/metabolism , Cancellous Bone/diagnostic imaging , Cancellous Bone/metabolism , Diet, High-Fat , Inflammation/pathology , Lipid Metabolism , Lysosomes/metabolism , Male , Mice, Inbred C57BL , Models, Biological , Organ Size , Protein Subunits/metabolism , Vacuolar Proton-Translocating ATPases/metabolism , X-Ray Microtomography
7.
Adv Sci (Weinh) ; 9(1): e2103005, 2022 01.
Article in English | MEDLINE | ID: mdl-34708571

ABSTRACT

The neuronal engagement of the peripheral nerve system plays a crucial role in regulating fracture healing, but how to modulate the neuronal activity to enhance fracture healing remains unexploited. Here it is shown that electrical stimulation (ES) directly promotes the biosynthesis and release of calcitonin gene-related peptide (CGRP) by activating Ca2+ /CaMKII/CREB signaling pathway and action potential, respectively. To accelerate rat femoral osteoporotic fracture healing which presents with decline of CGRP, soft electrodes are engineered and they are implanted at L3 and L4 dorsal root ganglions (DRGs). ES delivered at DRGs for the first two weeks after fracture increases CGRP expression in both DRGs and fracture callus. It is also identified that CGRP is indispensable for type-H vessel formation, a biological event coupling angiogenesis and osteogenesis, contributing to ES-enhanced osteoporotic fracture healing. This proof-of-concept study shows for the first time that ES at lumbar DRGs can effectively promote femoral fracture healing, offering an innovative strategy using bioelectronic device to enhance bone regeneration.


Subject(s)
Bone Regeneration/physiology , Calcitonin Gene-Related Peptide/metabolism , Electric Stimulation/instrumentation , Electric Stimulation/methods , Fracture Healing/physiology , Ganglia, Spinal/metabolism , Osteoporotic Fractures/therapy , Animals , Disease Models, Animal , Osteoporotic Fractures/metabolism , Rats
8.
Front Pharmacol ; 12: 730587, 2021.
Article in English | MEDLINE | ID: mdl-34497524

ABSTRACT

Background: Osteoarthritis (OA) is one of the main causes of disability in the elderly population, accompanied by a series of underlying pathologic changes, such as cartilage degradation, synovitis, subchondral bone sclerosis, and meniscus injury. The present study aimed to identify key genes, signaling pathways, and miRNAs in knee OA associated with the entire joint components, and to explain the potential mechanisms using computational analysis. Methods: The differentially expressed genes (DEGs) in cartilage, synovium, subchondral bone, and meniscus were identified using the Gene Expression Omnibus 2R (GEO2R) analysis based on dataset from GSE43923, GSE12021, GSE98918, and GSE51588, respectively and visualized in Volcano Plot. Venn diagram analyses were performed to identify the overlapping DEGs (overlapping DEGs) that expressed in at least two types of tissues mentioned above. Gene Ontology (GO) enrichment analysis, Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, protein-protein interaction (PPI) analysis, and module analysis were conducted. Furthermore, qRT-PCR was performed to validate above results using our clinical specimens. Results: As a result, a total of 236 overlapping DEGs were identified, of which 160 were upregulated and 76 were downregulated. Through enrichment analysis and constructing the PPI network and miRNA-mRNA network, knee OA-related key genes, such as HEY1, AHR, VEGFA, MYC, and CXCL12 were identified. Clinical validation by qRT-PCR experiments further supported above computational results. In addition, knee OA-related key miRNAs such as miR-101, miR-181a, miR-29, miR-9, and miR-221, and pathways such as Wnt signaling, HIF-1 signaling, PI3K-Akt signaling, and axon guidance pathways were also identified. Among above identified knee OA-related key genes, pathways and miRNAs, genes such as AHR, HEY1, MYC, GAP43, and PTN, pathways like axon guidance, and miRNAs such as miR-17, miR-21, miR-155, miR-185, and miR-1 are lack of research and worthy for future investigation. Conclusion: The present informatic study for the first time provides insight to the potential therapeutic targets of knee OA by comprehensively analyzing the overlapping genes differentially expressed in multiple joint components and their relevant signaling pathways and interactive miRNAs.

9.
Nanomicro Lett ; 13(1): 149, 2021 Jun 23.
Article in English | MEDLINE | ID: mdl-34160733

ABSTRACT

Osteoarthritis is the most prevalent chronic and debilitating joint disease, resulting in huge medical and socioeconomic burdens. Intra-articular administration of agents is clinically used for pain management. However, the effectiveness is inapparent caused by the rapid clearance of agents. To overcome this issue, nanoparticles as delivery systems hold considerable promise for local control of the pharmacokinetics of therapeutic agents. Given the therapeutic programs are inseparable from pathological progress of osteoarthritis, an ideal delivery system should allow the release of therapeutic agents upon specific features of disorders. In this review, we firstly introduce the pathological features of osteoarthritis and the design concept for accurate localization within cartilage for sustained drug release. Then, we review the interactions of nanoparticles with cartilage microenvironment and the rational design. Furthermore, we highlight advances in the therapeutic schemes according to the pathology signals. Finally, armed with an updated understanding of the pathological mechanisms, we place an emphasis on the development of "smart" bioresponsive and multiple modality nanoparticles on the near horizon to interact with the pathological signals. We anticipate that the exploration of nanoparticles by balancing the efficacy, safety, and complexity will lay down a solid foundation tangible for clinical translation.

10.
Biomaterials ; 275: 120984, 2021 08.
Article in English | MEDLINE | ID: mdl-34186235

ABSTRACT

Critical size bone defects are frequently caused by accidental trauma, oncologic surgery, and infection. Distraction osteogenesis (DO) is a useful technique to promote the repair of critical size bone defects. However, DO is usually a lengthy treatment, therefore accompanied with increased risks of complications such as infections and delayed union. Here, we demonstrated that magnesium (Mg) nail implantation into the marrow cavity degraded gradually accompanied with about 4-fold increase of new bone formation and over 5-fold of new vessel formation as compared with DO alone group in the 5 mm femoral segmental defect rat model at 2 weeks after distraction. Mg nail upregulated the expression of calcitonin gene-related peptide (CGRP) in the new bone as compared with the DO alone group. We further revealed that blockade of the sensory nerve by overdose capsaicin blunted Mg nail enhanced critical size bone defect repair during the DO process. CGRP concentration-dependently promoted endothelial cell migration and tube formation. Meanwhile, CGRP promoted the phosphorylation of focal adhesion kinase (FAK) at Y397 site and elevated the expression of vascular endothelial growth factor A (VEGFA). Moreover, inhibitor/antagonist of CGRP receptor, FAK, and VEGF receptor blocked the Mg nail stimulated vessel and bone formation. We revealed, for the first time, a CGRP-FAK-VEGF signaling axis linking sensory nerve and endothelial cells, which may be the main mechanism underlying Mg-enhanced critical size bone defect repair when combined with DO, suggesting a great potential of Mg implants in reducing DO treatment time for clinical applications.


Subject(s)
Calcitonin Gene-Related Peptide , Osteogenesis, Distraction , Animals , Bone Regeneration , Calcitonin , Endothelial Cells , Focal Adhesion Protein-Tyrosine Kinases , Magnesium , Osteogenesis , Rats , Vascular Endothelial Growth Factor A
11.
Bioact Mater ; 6(8): 2511-2522, 2021 Aug.
Article in English | MEDLINE | ID: mdl-33665494

ABSTRACT

INTRODUCTION: Magnesium (Mg) has a prophylactic potential against the onset of hyperlipidemia. Similar to statin, Mg is recommended as lipid-lowering medication for hypercholesterolemia and concomitantly exhibits an association with increased bone mass. The combination of statin with Mg ions (Mg2+) may be able to alleviate the high-fat diet (HFD)-induced bone loss and reduce the side-effects of statin. This study aimed to explore the feasibility of combined Mg2+ with simvastatin (SIM) for treating HFD-induced bone loss in mice and the involving mechanisms. MATERIALS AND METHODS: C57BL/6 male mice were fed with a HFD or a normal-fat diet (NFD). Mice were intraperitoneally injected SIM and/or orally received water with additional Mg2+ until sacrificed. Enzyme-linked immunosorbent assay was performed to measure cytokines and cholesterol in serum and liver lysates. Bone mineral density (BMD) and microarchitecture were assessed by micro-computed tomography (µCT) in different groups. The adipogenesis in palmitate pre-treated HepG2 cells was performed under various treatments. RESULTS: µCT analysis showed that the trabecular bone mass was significantly lower in the HFD-fed group than that in NFD-fed group since week 8. The cortical thickness in HFD-fed group had a significant decrease at week 24, as compared with NFD-fed group. The combination of Mg2+ and SIM significantly attenuated the trabecular bone loss in HFD-fed mice via arresting the osteoclast formation and bone resorption. Besides, such combination also reduced the hepatocytic synthesis of cholesterol and inhibited matrix metallopeptidase 13 (Mmp13) mRNA expression in pre-osteoclasts. CONCLUSIONS: The combination of Mg2+ and SIM shows a synergistic effect on attenuating the HFD-induced bone loss. Our current formulation may be a cost-effective alternative treatment to be indicated for obesity-related bone loss.

12.
Tissue Eng Part A ; 27(1-2): 87-102, 2021 01.
Article in English | MEDLINE | ID: mdl-32375579

ABSTRACT

Distraction osteogenesis (DO) is a well-established surgical technique for treating bone defect and limb lengthening. The major drawback of DO is the long treatment period as the external fixator has to be kept in place until consolidation is completed. Calcitonin gene-related peptide (CGRP) has been reported to promote angiogenesis by affecting endothelial progenitor cells (EPCs) in limb ischemia and wound healing. Thus, the goal of this study was to evaluate the angiogenic effect of exogenous CGRP on bone regeneration in a rat DO model. Exogenous CGRP was directly injected into the bone defect after each cycle of distraction in vivo. Microcomputed tomography, biomechanical test, and histological analysis were performed to assess the new bone formation. Angiography and immunofluorescence were performed to assess the formation of blood vessels. CD31+CD144+ EPCs in the bone defect were quantified with flow cytometry. In in vitro study, bone marrow stem cells (BMSCs) were used to investigate the effect of CGRP on EPCs production during endothelial differentiation. Our results showed that CGRP significantly promoted bone regeneration and vessel formation after consolidation. CGRP significantly increased the fraction of CD31+CD144+EPCs and the capillary density in the bone defect at the end of distraction phase. CGRP increased EPC population in the endothelial differentiation of BMSCs in vitro by activating PI3K/AKT signaling pathway. Furthermore, differentiated EPCs rapidly assembled into tube-like structures and promoted osteogenic differentiation of BMSCs. In conclusion, CGRP increased EPC population and promoted blood vessel formation and bone regeneration at the defect region in a DO model. Impact statement Distraction osteogenesis (DO) is a well-established surgical technique for limb lengthening and bone defect. The disadvantage of this technique is that external fixator is needed to be kept in place for about 12 months. This may result in increased risk of infection, financial burden, and negative psychological impacts. In this study, we have injected calcitonin gene-related peptide (CGRP) into the defect region after distraction and found that CGRP enhanced vessel formation and bone regeneration in a rat DO model. This suggests that a controlled delivery system for CGRP could be developed and applied clinically for accelerating bone regeneration in patients with DO.


Subject(s)
Osteogenesis, Distraction , Osteogenesis , Animals , Bone Regeneration , Calcitonin , Calcitonin Gene-Related Peptide , Phosphatidylinositol 3-Kinases , Rats , X-Ray Microtomography
13.
Bioact Mater ; 6(5): 1341-1352, 2021 May.
Article in English | MEDLINE | ID: mdl-33210027

ABSTRACT

INTRODUCTION: We previously demonstrated that magnesium ions (Mg2+) was a novel therapeutic alternative for osteoarthritis (OA) through promoting the hypoxia inducible factor-1α (HIF-1α)-mediated cartilage matrix synthesis. However, oxidative stress can inhibit the expression of HIF-1α, amplify the inflammation that potentially impairs the therapeutic efficacy of Mg2+ in OA. Vitamin (VC), a potent antioxidant, may enhance the efficacy of Mg2+ in OA treatment. This study aims to investigate the efficacy of combination of Mg2+ and VC on alleviating joint destruction and pain in OA. MATERIAL AND METHODS: Anterior cruciate ligament transection with partial medial meniscectomy induced mice OA model were randomly received intra-articular injection of either saline, MgCl2 (0.5 mol/L), VC (3 mg/ml) or MgCl2 (0.5 mol/L) plus VC (3 mg/ml) at week 2 post-operation, twice weekly, for 2 weeks. Joint pain and pathological changes were assessed by gait analysis, histology, western blotting and micro-CT. RESULTS: Mg2+ and VC showed additive effects to significantly alleviate the joint destruction and pain. The efficacy of this combined therapy could sustain for 3 months after the last injection. We demonstrated that VC enhanced the promotive effect of Mg2+ on HIF-1α expression in cartilage. Additionally, combination of Mg2+ and VC markedly promoted the M2 polarization of macrophages in synovium. Furthermore, combination of Mg2+ and VC inhibited osteophyte formation and expressions of pain-related neuropeptides. CONCLUSIONS: Intra-articular administration of Mg2+ and VC additively alleviates joint destruction and pain in OA. Our current formulation may be a cost-effective alternative treatment for OA.

14.
Ageing Res Rev ; 62: 101098, 2020 09.
Article in English | MEDLINE | ID: mdl-32535273

ABSTRACT

Osteoporosis is a highly prevalent disorder characterized by the loss of bone mass and microarchitecture deterioration of bone tissue, attributed to various factors, including menopause (primary), aging (primary) and adverse effects of relevant medications (secondary). In recent decades, knowledge regarding the etiological mechanisms underpinning osteoporosis emphasizes that bone cellular homeostasis, including the maintenance of cell functions, differentiation, and the response to stress, is tightly regulated by autophagy, which is a cell survival mechanism for eliminating and recycling damaged proteins and organelles. With the important roles in the maintenance of cellular homeostasis and organ function, autophagy has emerged as a potential target for the prevention and treatment of osteoporosis. In this review, we update and discuss the pathophysiology of autophagy in normal bone cell life cycle and metabolism. Then, the alternations of autophagy in primary and secondary osteoporosis, and the accompanied pathological process are discussed. Finally, we discuss current strategies, limitations, and challenges involved in targeting relevant pathways and propose strategies by which such hurdles may be circumvented in the future for their translation into clinical validations and applications for the prevention and treatment of osteoporosis.


Subject(s)
Autophagy , Osteoporosis , Aging , Bone and Bones , Female , Homeostasis , Humans , Osteoporosis/drug therapy
15.
J Biomed Mater Res A ; 106(11): 2944-2954, 2018 11.
Article in English | MEDLINE | ID: mdl-30329209

ABSTRACT

In this study, we designed a polyvinyl alcohol (PVA)-alginate based hydrogel and evaluated its cytocompatibility and printability. The samples were fabricated by 3D printing using a freeze-thaw process. The scanning electron microscope, material testing machine, rheometer, and cell counting kit-8 assay were used to examine the morphology, mechanical properties, rheological properties, and cytocompatiblity of the scaffolds, respectively. The mechanical strength, cytocompatiblity, crosslinking time, and printability were remarkably improved with the use of PVA. To sum up, our data suggest that hybrid bio-ink is more appropriate for precise 3D bioprinting due to its rapid prototyping capability and better cytocompatibility. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 106A: 2944-2954, 2018.


Subject(s)
Alginates/chemistry , Biocompatible Materials/chemistry , Bioprinting/methods , Polyvinyl Alcohol/chemistry , Printing, Three-Dimensional , Tissue Scaffolds/chemistry , Animals , Cell Adhesion , Cell Line , Cell Survival , Materials Testing , Mice , Osteoblasts/cytology , Tensile Strength , Tissue Engineering
16.
Medicine (Baltimore) ; 97(21): e10884, 2018 May.
Article in English | MEDLINE | ID: mdl-29794796

ABSTRACT

It suggests that a high leptin level may increase the risk of venous thromboembolism (VTE) in animal studies. However, clinical studies in this field are still largely unexplored. Our objective was to evaluate the relationship between the preoperative serum leptin levels and postoperative VTE incidence in osteoarthritis (OA) patients who underwent total knee arthroplasty (TKA) at our institute.We conducted a prospective and cross-sectional study in these OA patients from March 2014 to March 2016. Preoperative leptin levels were analyzed by Luminex assays. VTE was assessed preoperatively and on postoperative day 5 and 7. The potential risk factors for VTE were also documented.We enrolled 203 OA patients. No PE was detected and DVT was diagnosed in 34 patients postoperatively. There were significant differences between the median leptin levels in DVT group and non-DVT group [25.13 ng/mL (interquartile range, 14.51-44.31) vs 18.71 ng/mL (8.26-28.99), P = .007]. The relative risk of DVT significantly increased with natural logarithm (ln) leptin (per SD increase) (OR 2.37, 95% confidence interval (95% CI), 1.29-4.33, P = .005). Multivariate analyses adjusted for potential confounders showed ln leptin (per SD increase) was significantly associated with the relative risk of DVT (OR 2.17, 95% CI, 1.01-4.64, P = .046). When patients were subdivided into tertiles according to their leptin values, the OR for DVT increased with increasing tertiles of serum leptin (OR 1.03, 95% CI, 1.01-1.06, P for trend = .023).In the present study, our results indicate that a high preoperative leptin level may be an independent risk factor for postoperative DVT.


Subject(s)
Arthroplasty, Replacement, Knee/adverse effects , Leptin/blood , Osteoarthritis, Knee/surgery , Venous Thrombosis/metabolism , Aged , Cross-Sectional Studies , Female , Humans , Incidence , Male , Middle Aged , Postoperative Complications/epidemiology , Preoperative Period , Prospective Studies , Risk Factors , Ultrasonography, Doppler, Duplex/methods , Venous Thromboembolism/diagnostic imaging , Venous Thromboembolism/epidemiology , Venous Thromboembolism/metabolism , Venous Thrombosis/complications , Venous Thrombosis/diagnostic imaging , Venous Thrombosis/etiology
17.
J Bone Miner Metab ; 36(2): 133-147, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28357594

ABSTRACT

Bone fracture healing is achieved through the proliferation and differentiation of stem cells, while bone marrow stem cells (BMSCs) contribute to endochondral ossification. During fracture healing, mesenchymal progenitor cells first form a cartilaginous blastema that becomes vascularized to recruit precursor cells of osteoblasts through the bone morphogenetic protein 2 (Bmp2)/Smad-dependent Runx2 pathway. Statins deplete geranylgeranyl diphosphate (GGPP), which participates in the regulation of BMSCs differentiation, through the inhibition of 3-hydroxy-3-methylglutaryl (HMG)-CoA reductase, leading to impaired protein geranylgeranylation, which strongly impacts the bone synthesis induced by Bmp2. Accordingly, we would like to investigate the role of geranylgeranyl diphosphate synthase 1 (Ggps1) in bone fracture via endochondral ossification in mice. We used a Cre-loxP system, namely the tamoxifen-inducible Collagen 2-CreERT2 Ggps1 fl/fl, to eliminate specifically the Ggps1 activity in chondrocytes of 8-10-week-old mice. We found that the endochondral bone formation, calcification and vasculogenesis of the bony callus were accelerated in fractures in Ggps1-/-mice. Together, the results of this study confirm that the specific deletion of Ggps1, using the Collagen 2-CreERT2 mice, will accelerate the fracture healing process by activating the Bmp2/Smad-dependent Runx2 pathway. In addition, we managed to improve the fracture healing process by inhibiting the Ggps1 activity and its related products with statin drugs.


Subject(s)
Chondrocytes/metabolism , Farnesyltranstransferase/metabolism , Fracture Healing , Gene Knockdown Techniques , Multienzyme Complexes/metabolism , Osteogenesis , Animals , Biomarkers/metabolism , Biomechanical Phenomena , Bone Morphogenetic Protein 2/metabolism , Bony Callus/metabolism , Bony Callus/pathology , Core Binding Factor Alpha 1 Subunit/metabolism , Farnesyltranstransferase/deficiency , Femur/blood supply , Femur/diagnostic imaging , Femur/pathology , Femur/physiopathology , Fracture Healing/genetics , Gene Expression Regulation , Ki-67 Antigen/metabolism , Male , Mice, Knockout , Multienzyme Complexes/deficiency , Neovascularization, Physiologic , Osteogenesis/genetics , Signal Transduction , Smad Proteins/metabolism , Transforming Growth Factor beta/metabolism
19.
Medicine (Baltimore) ; 96(29): e7604, 2017 Jul.
Article in English | MEDLINE | ID: mdl-28723807

ABSTRACT

BACKGROUND: The effectiveness of preoperative thromboprophylaxis remains obscure in patients with femoral neck fracture. The purpose of the current study was to investigate whether these patients benefit from preoperative thromboprophylaxis. METHODS: In this prospective, randomized controlled trial, a total of 80 patients with femoral neck fracture were randomly assigned to receive either rivaroxaban or conservative treatment before surgery. For all patients, color Doppler ultrasound of both lower extremities was performed immediately after admission. The primary efficacy outcome was venous thromboembolism (VTE) defined as deep vein thrombosis (DVT) or pulmonary embolism (PE). The primary safety outcome was major bleeding. RESULT: Compared with conservative treatment, rivaroxaban could significantly reduce the incidence of DVT from 19.5% (8/41) to 2.6% (1/39) (P = .016). Preoperatively, there were a total of 9 occurrences of DVT including 8 DVT in the conservative treatment group and 1 in the oral rivaroxaban group. All cases of DVT were asymptomatic, with 8 of them diagnosed as isolated muscular calf vein thromboses. There were no differences between the 2 groups in terms of the overall incidence of major bleeding. CONCLUSION: Thromboprophylaxis with rivaroxaban prior to surgery can effectively reduce the risk of preoperative DVT for patients with femoral neck fracture without increasing the risk of bleeding.


Subject(s)
Factor Xa Inhibitors/administration & dosage , Femoral Neck Fractures/drug therapy , Femoral Neck Fractures/surgery , Pulmonary Embolism/prevention & control , Rivaroxaban/administration & dosage , Venous Thrombosis/prevention & control , Administration, Oral , Aged , Aged, 80 and over , Conservative Treatment , Factor Xa Inhibitors/adverse effects , Female , Femoral Neck Fractures/diagnostic imaging , Hemorrhage/etiology , Humans , Incidence , Male , Middle Aged , Postoperative Complications/prevention & control , Preoperative Care , Pulmonary Embolism/etiology , Rivaroxaban/adverse effects , Treatment Outcome , Ultrasonography, Doppler, Color , Venous Thrombosis/etiology
20.
Biomed Res Int ; 2017: 8260487, 2017.
Article in English | MEDLINE | ID: mdl-28612028

ABSTRACT

BACKGROUND: Deep vein thrombosis (DVT) is one of the major complications of total joint arthroplasty (TJA). Chronic kidney dysfunction (CKD) has proven to promote a proinflammatory and prothrombotic state and is prevalent among patients undergoing TJA. The purpose of this study is to identify whether CKD increase the risk of DVT following TJA. METHODS: In a retrospective study, 1274 patients who underwent primary TJA were studied. CKD is graded in 5 stages. Univariate and multivariate analysis were used to identify the association of CKD and its severity with postoperative DVT. RESULTS: There were 1139 (89.4%) participants with normal kidney function, 103 (8.1%) with mildly decreased kidney function, and 32 (2.5%) with stage 3 and 4 CKD. A total of 244 patients (19.2%) were diagnosed with DVT. Sixty-four patients (5.0%) developed symptomatic DVT. Advanced age, female gender, malignancy, and eGFR showed significant association with total DVT. BMI, thrombosis history, malignancy, and eGFR were associated with symptomatic DVT. After adjusting for age, gender, BMI, and malignancy, eGFR was found to be related to both total and symptomatic DVT. CONCLUSIONS: CKD is an important risk factor for both total and symptomatic DVT following TJA. Postoperative prophylaxis should be made a priority in this population.


Subject(s)
Arthroplasty, Replacement, Hip/adverse effects , Arthroplasty, Replacement, Knee/adverse effects , Postoperative Complications/epidemiology , Renal Insufficiency, Chronic , Venous Thrombosis , Adolescent , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Renal Insufficiency, Chronic/epidemiology , Renal Insufficiency, Chronic/etiology , Retrospective Studies , Risk Factors , Venous Thrombosis/epidemiology , Venous Thrombosis/etiology
SELECTION OF CITATIONS
SEARCH DETAIL
...