Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Front Mol Biosci ; 11: 1268019, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38903180

RESUMO

Skeletal diseases impose a considerable burden on society. The clinical and tissue-engineering therapies applied to alleviate such diseases frequently result in complications and are inadequately effective. Research has shifted from conventional therapies based on mesenchymal stem cells (MSCs) to exosomes derived from MSCs. Exosomes are natural nanocarriers of endogenous DNA, RNA, proteins, and lipids and have a low immune clearance rate and good barrier penetration and allow targeted delivery of therapeutics. MSC-derived exosomes (MSC-exosomes) have the characteristics of both MSCs and exosomes, and so they can have both immunosuppressive and tissue-regenerative effects. Despite advances in our knowledge of MSC-exosomes, their regulatory mechanisms and functionalities are unclear. Here we review the therapeutic potential of MSC-exosomes for skeletal diseases.

2.
Arch Pharm (Weinheim) ; : e2400066, 2024 May 29.
Artigo em Inglês | MEDLINE | ID: mdl-38809025

RESUMO

Oncogenic overexpression or activation of C-terminal Src kinase (CSK) has been shown to play an important role in triple-negative breast cancer (TNBC) progression, including tumor initiation, growth, metastasis, drug resistance. This revelation has pivoted the focus toward CSK as a potential target for novel treatments. However, until now, there are few inhibitors designed to target the CSK protein. Responding to this, our research has implemented a comprehensive virtual screening protocol. By integrating energy-based screening methods with AI-driven scoring functions, such as Attentive FP, and employing rigorous rescoring methods like Glide docking and molecular mechanics generalized Born surface area (MM/GBSA), we have systematically sought out inhibitors of CSK. This approach led to the discovery of a compound with a potent CSK inhibitory activity, reflected by an IC50 value of 1.6 nM under a homogeneous time-resolved fluorescence (HTRF) bioassay. Subsequently, molecule 2 exhibits strong growth inhibition of MD anderson - metastatic breast (MDA-MB) -231, Hs578T, and SUM159 cells, showing a level of growth inhibition comparable to that observed with dasatinib. Treatment with molecule 2 also induced significant G1 phase accumulation and cell apoptosis. Furthermore, we have explored the explicit binding interactions of the compound with CSK using molecular dynamics simulations, providing valuable insights into its mechanism of action.

3.
Stem Cell Rev Rep ; 19(4): 968-982, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36609902

RESUMO

BACKGROUND: Epidermal growth factor-like domain protein 7 (EGFL7) is a secreted protein that is differentially expressed in the bone microenvironment; however, the effect of EGFL7 on the osteogenesis of human bone marrow mesenchymal stem cells (hBMSCs) is largely unknown. METHODS: EGFL7 expression in the fracture microenvironment was analyzed based on the Gene Expression Omnibus (GEO) database. Knockdown of EGFL7 by small interfering RNA (siRNA) and in vitro stimulation with recombinant human EGFL7 (rhEGFL7) protein were used to assess alterations in downstream signaling and changes in the osteogenic differentiation and proliferation of hBMSCs. A γ-secretase inhibitor was used to further explore whether inhibition of Notch signaling rescued the osteogenic-inhibitory effect of EGFL7 knockdown in hBMSCs. A femoral defect model was established to verify the effect of recombinant mouse EGFL7 on bone healing in vivo. RESULTS: EGFL7 expression increased during hBMSC osteogenesis. Knockdown of EGFL7 impaired hBMSC osteogenesis and activated Notch1/NICD/Hes1 signaling. rhEGFL7 promoted hBMSC osteogenesis and downregulated Notch1 signaling. The osteoblast-inhibitory effect of EGFL7 knockdown was rescued by Notch1 signaling inhibition. Recombinant EGFL7 led to enhanced bone healing in mice with femoral defects. CONCLUSIONS: EGFL7 promotes osteogenesis of hBMSCs partly via downregulation of Notch1 signaling.


Assuntos
Células-Tronco Mesenquimais , Osteogênese , Humanos , Camundongos , Animais , Osteogênese/genética , Regulação para Baixo/genética , Células Cultivadas , Transdução de Sinais , Diferenciação Celular/genética , Peptídeos e Proteínas de Sinalização Intercelular/metabolismo , Osteoblastos/metabolismo , RNA Interferente Pequeno/metabolismo , Fatores de Transcrição/metabolismo , Fatores de Transcrição HES-1/genética , Fatores de Transcrição HES-1/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo
4.
Bioact Mater ; 19: 429-443, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-35574058

RESUMO

The imbalance of bone homeostasis is the root cause of osteoporosis. However current therapeutic approaches mainly focus on either anabolic or catabolic pathways, which often fail to turn the imbalanced bone metabolism around. Herein we reported that a SIRT-1 agonist mediated molecular therapeutic strategy to reverse the imbalance in bone homeostasis by simultaneously regulating osteogenesis and osteoclastogenesis via locally sustained release of SRT2104 from mineral coated acellular matrix microparticles. Immobilization of SRT2104 on mineral coating (MAM/SRT) harnessing their electrostatic interactions resulted in sustained release of SIRT-1 agonist for over 30 days. MAM/SRT not only enhanced osteogenic differentiation and mineralization, but also attenuated the formation and function of excessive osteoclasts via integrating multiple vital upstream signals (ß-catenin, FoxOs, Runx2, NFATc1, etc.) in vitro. Osteoporosis animal model also validated that it accelerated osteoporotic bone healing and improved osseointegration of the surrounding bone. Overall, our work proposes a promising strategy to treat osteoporotic bone defects by reversing the imbalance in bone homeostasis using designated small molecule drug delivery systems.

5.
J Vis Exp ; (202)2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38163268

RESUMO

Unilateral biportal endoscopy (UBE) is a minimally invasive spinal surgery technique increasingly employed in treating degenerative lumbar diseases, such as lumbar disc herniation, lumbar spinal stenosis, and spondylolisthesis. In UBE, two independent yet interconnected surgical channels are established-one for the endoscope and the other for surgical instruments-providing a broad and clear surgical field of view. UBE offers several advantages over traditional open surgery, including reduced tissue damage, shorter hospital stays, and faster recovery times. Additionally, it combines the benefits of microscopic surgery and interlaminar endoscopy, enhancing flexibility, accuracy, and reliability during the procedure. The learning curve for UBE is shorter than that for transforaminal endoscopy, as the surgical processes closely resemble those of conventional open surgery. Despite its favorable clinical outcomes, such as reduced blood loss and shorter hospitalization, UBE carries potential complications, including epidural hematoma, dural injury, and compression of the outlet nerve root. To mitigate these risks, it is crucial to ensure appropriate patient selection, apply the correct surgical technique, and engage in careful postoperative monitoring. This article provides a detailed summary of the step-by-step surgical techniques used in UBE for treating lumbar disc herniation. It serves as a comprehensive guide to enhance practitioners' understanding of UBE. The presentation also underscores the importance of rigorous training and expertise to ensure optimal patient outcomes.


Assuntos
Deslocamento do Disco Intervertebral , Humanos , Deslocamento do Disco Intervertebral/diagnóstico por imagem , Deslocamento do Disco Intervertebral/cirurgia , Reprodutibilidade dos Testes , Vértebras Lombares/diagnóstico por imagem , Vértebras Lombares/cirurgia , Endoscopia/métodos , Procedimentos Cirúrgicos Minimamente Invasivos , Endoscopia Gastrointestinal , Resultado do Tratamento , Estudos Retrospectivos
6.
Biomolecules ; 12(11)2022 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-36358952

RESUMO

Osteoporosis and osteoporotic fractures comprise a substantial health and socioeconomic burden. The leading cause of osteoporosis is an imbalance in bone formation and bone resorption caused by hyperactive osteoclasts. Therefore, a new strategy to suppress osteoclastogenesis is needed. Parkin is likely closely associated with bone metabolism, although its role in osteoclastogenesis is unclear. In this study, the Parkin protein inhibited the receptor activator of nuclear factor-κB ligand (RANKL)-induced osteoclast formation, osteoclast-specific gene expression, F-actin ring formation, and bone resorption pit formation in vitro. Moreover, depletion of Parkin enhanced RANKL-induced osteoclast formation, osteoclast-specific gene expression, F-actin ring formation, and bone resorption pit formation. Reactive oxygen species (ROS) activity was suppressed, while autophagy was upregulated with the presence of the Parkin protein. ROS activity was upregulated and autophagy was decreased due to Parkin knockdown. In addition, intravenous administration of Parkin rescued ovariectomy-induced bone loss and reduced osteoclastogenesis in vivo. Collectively, Parkin has therapeutic potential for diseases associated with overactive osteoclasts.


Assuntos
Reabsorção Óssea , Osteoporose , Humanos , Feminino , Animais , Camundongos , Ligante RANK/farmacologia , Osteogênese , Espécies Reativas de Oxigênio/metabolismo , Actinas/metabolismo , Reabsorção Óssea/tratamento farmacológico , Reabsorção Óssea/genética , Ovariectomia/efeitos adversos , Ubiquitina-Proteína Ligases/genética , Osteoporose/tratamento farmacológico , Osteoporose/etiologia , Diferenciação Celular , NF-kappa B/metabolismo , Camundongos Endogâmicos C57BL
7.
Adv Healthc Mater ; 11(19): e2200787, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35851764

RESUMO

Osteoporosis (OP) is the most common orthopedic disease in the elderly and the main cause of age-related mortality and disability. However, no satisfactory intervention is currently available in clinical practice. Thus, an effective therapy to prevent or delay the development of OP should be devised. Osteoclastogenesis overactivation and excessive bone resorption are the main characteristics of OP. Accordingly, a paradigm for nanozyme-mediated normalization of the disease microenvironment to regulate osteoclast differentiation and delay OP is proposed. Hollow Prussian blue nanozymes (HPBZs) are prepared via template-free hydrothermal synthesis and selected as representative nanozymes. The intrinsic osteoclast activity-remodeling bioactivities of the HPBZs are explored in vitro and in vivo, focusing on their impact on osteogenesis and specific molecular mechanisms using an OP murine model. The HPBZs significantly normalize the OP microenvironment, thereby inhibiting osteoclast formation and osteoclast resorption, possibly owing to the suppression of intracellular reactive oxygen species generation, the mitogen-activated protein kinase, and nuclear factor κB signaling pathways. Consistently, in an ovariectomy-induced OP murine model, HPBZ treatment significantly attenuates osteoporotic bone loss in vivo. The findings confirm the HPBZ-mediated normalization of the disease microenvironment for the treatment of OP and suggest its application to other inflammation-related diseases.


Assuntos
Reabsorção Óssea , Osteoporose , Idoso , Animais , Reabsorção Óssea/metabolismo , Diferenciação Celular , Modelos Animais de Doenças , Feminino , Ferrocianetos , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Osteoclastos/metabolismo , Osteogênese , Osteoporose/tratamento farmacológico , Osteoporose/etiologia , Ovariectomia/efeitos adversos , Ligante RANK/metabolismo , Espécies Reativas de Oxigênio/metabolismo
8.
Acta Biomater ; 128: 150-162, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33894346

RESUMO

Osteochondral regeneration is an orchestrated process of inflammatory immunity, host cell response, and implant degradation in tissue engineering. Here, the effects of a platelet-rich plasma (PRP)-gelatin methacryloyl (GelMA) hydrogel scaffold fabricated using the digital micro-mirror device (DMD) technique for osteochondral repair were investigated in a rabbit model. GelMA hydrogels with different PRP concentrations were fabricated, and their roles in bone marrow mesenchymal stem cells (BMSCs) and macrophage polarization in vitro were investigated. The incorporation of 20% PRP into the hydrogel showed optimal effects on the proliferation, migration, and osteogenic and chondrogenic differentiation of BMSCs. The 20% PRP-GelMA (v/v) hydrogel also promoted M2 polarization with high expression of Arg1 and CD206. Compared to the 20% PRP group, the 50% PRP group showed similar biological roles in BMSCs but less extent of osteogenesis. In the vivo study, the 20% PRP-GelMA composite was used for osteochondral reconstruction and showed more cartilage and subchondral bone regeneration than that observed using the pure GelMA hydrogel. The PRP-GelMA group exhibited more M2 macrophage infiltration and less M1 macrophage presentation at three time points as compared to the nontreatment group. The expression of Arg1 in the PRP-GelMA group increased significantly at 6 weeks but decreased to a lower level at 12 weeks, while CD163 showed sustained high expression until 18 weeks. Our findings demonstrated that the 3D-printed PRP-GelMA composite could promote osteochondral repair through immune regulation by M2 polarization and could be a potential candidate for osteochondral tissue engineering. STATEMENT OF SIGNIFICANCE: PRP-GelMA hydrogels promoted the migration and osteogenic and chondrogenic differentiation of BMSCs. PRP-GelMA hydrogels participated in immune regulation and M1-to-M2 transition of macrophages. PRP-GelMA hydrogels coordinated and promoted several overlapping osteochondral repair events, including dynamic immune regulation, chemotaxis of MSCs, and osteochondral differentiation. PRP-GelMA hydrogels showed superior cartilage and subchondral bone repair properties.


Assuntos
Gelatina , Plasma Rico em Plaquetas , Animais , Gelatina/farmacologia , Hidrogéis/farmacologia , Macrófagos , Impressão Tridimensional , Coelhos , Engenharia Tecidual , Alicerces Teciduais
9.
Bioact Mater ; 6(8): 2439-2451, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33553826

RESUMO

Osteoarthritis (OA) is the main cause of disability in the elderly. Effective intervention in the early and middle stage of osteoarthritis can greatly prevent or slow down the development of the disease, and reduce the probability of joint replacement. However, there is to date no effective intervention for early and middle-stage OA. OA microenvironment mainly destroys the balance of oxidative stress, extracellular matrix synthesis and degradation of chondrocytes under the joint action of biological and mechanical factors. Herein, hollow Prussian blue nanozymes (HPBzymes) were designed via a modified hydrothermal template-free method. The aim of this study was to investigate the effects of HPBzymes on chondrocytes and the progression of OA. The intrinsic bioactivities of HPBzymes were excavated in vitro and in vivo, remodeling microenvironment for significantly protecting chondrocytes and delaying the progression of traumatic OA by inhibiting reactive oxygen species (ROS) and Rac1/nuclear factor kappa-B (NF-κB) signaling in a rat model. HPBzyme significantly diminished interleukin (IL)-1ß-stimulated inflammation, extracellular matrix degradation, and apoptosis of human chondrocytes. HPBzyme attenuated the expression of Rac1 and the ROS levels and prevented the release and nuclear translocation of NF-κB. Deeply digging the intrinsic bioactivities of nanozyme with single component to remodel microenvironment is an effective strategy for ROS-associated chronic diseases. This study reveals that excavating the bioactivities of nanomedicine deserves attention for diagnosis and treatment of severe diseases.

10.
Postgrad Med J ; 97(1145): 150-155, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32114493

RESUMO

OBJECTIVES: The aim of this study was to evaluate the effect of antiosteoporotic drugs on preventing periprosthetic bone loss in calcar 6 and 12 months after total hip arthroplasty. METHODS: The network meta-analysis was conducted guided by the Preferred Reporting Items for Systematic Reviews and Meta-analysis (PRISMA) guideline. A systematic literature search was conducted and 21 studies that enrolled a total of 955 patients with 9 antiosteoporotic drugs met the inclusion criteria. Network meta-analysis and conventional meta-analysis were carried out for calculating standard mean difference (SMD) and the surface under cumulative ranking curve (SUCRA) of the bone mineral density (BMD) in calcar (Gruen zone 7) as well as bone turnover markers (BTMs) including bone-specific alkaline phosphatase (BSAP) and collagen type I cross-linked N telopeptide (NTX) at 6 and 12 months between different antiosteoporotic drugs. RESULTS: At 6 months after total hip arthroplasty, zoledronate (SUCRA=86.4%), risedronate (SUCRA=51.3%) and etidronate (SUCRA=44.5%) were effective in retaining BMD in calcar; zoledronate was significantly more effective than etidronate (SMD=0.65, 95% CI 0.03 to 1.27). Teriparatide (SUCRA=84.5%), denosumab (SUCRA=82.5%), zoledronate (SUCRA=69.2%), alendronate+alfacalcidol (SUCRA=66.2%) and etidronate (SUCRA=51.5%) were the top five drugs in retaining BMD in calcar at 12 months after total hip arthroplasty and the efficacy were comparable. After simultaneously excluding studies in which the prosthesis were cement and the drug dosages as well as treatment durations were inconsistent with those in treating osteoporosis, the above results were robust with the exception that alendronate showed significant efficacy compared with placebo (SMD=1.22, 95% CI 0.46 to 1.99) and was comparable with those effective drugs at 12 months. Long-term residual effect was corroborated only in etidronate, alendronate and zoledronate from previous studies. BTMs were significantly decreased as early as 6 months (SMD of BSAP -0.49, 95% CI -0.84 to -0.13; SMD of NTX -0.93, 95% CI -1.21 to -0.64) and sustained until 12 months (SMD of BSAP -0.27, 95% CI -0.50 to -0.03; SMD of NTX -0.84, 95% CI -1.11 to -0.56) during the prophylaxis. CONCLUSIONS: Antiosteoporotic drugs showed prophylactic efficacy on periprosthetic bone loss after total hip arthroplasty in calcar, the effectiveness varied. Zoledronate was the best recommendation due to its optimal efficacy both within 6 and 12 months as well as its residual effect in the long term. BTMs could be used as indicators for monitoring through the treatment. More head-to-head clinical trials are needed to confirm those findings.


Assuntos
Artroplastia de Quadril , Conservadores da Densidade Óssea/uso terapêutico , Densidade Óssea/efeitos dos fármacos , Reabsorção Óssea/prevenção & controle , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto
11.
Front Cell Dev Biol ; 8: 576104, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33043010

RESUMO

Osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) plays a key role in bone formation. Parkin, an E3 ubiquitin ligase, related to Parkinson's disease and aging. Previous studies have indicated that Parkinson's disease have a higher risk of osteoporotic fracture. To investigate the effects and underlying mechanism of Parkin in the osteogenic differentiation of BMSCs, osteogenic differentiation was analyzed following upregulation or downregulation of Parkin. We found that Parkin was increased during differentiation. Parkin overexpression enhanced osteo-specific markers, and downregulation of Parkin mitigated osteo-specific markers. Moreover, upregulation of Parkin promoted ß-catenin expression and autophagy and vice versa. The upregulation of ß-catenin enhanced autophagy, and the activation of autophagy also increased the expression of ß-catenin in Parkin-downregulated BMSCs. Parkin-overexpressed cell sheets accelerated bone healing in a tibial fracture model. Based on these results, we concluded that Parkin meditates osteoblastic differentiation of BMSCs via ß-catenin and autophagy signaling.

12.
Front Cell Dev Biol ; 8: 703, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32850823

RESUMO

Bergenin is a C-glucoside of 4-O-methyl gallic acid isolated from several medicinal plants and has multiple biological activities. The aim of this study was to assess the potential usefulness of bergenin in hyperuricemia. We found that bergenin reduced serum urate levels in hyperuricemia mice by promoting renal and gut uric acid excretion. Bergenin treatment increased Abcg2 expression both in the kidneys and intestine, while the expression of Slc2a9 was suppressed in the kidney and increased in the intestine. Moreover, bergenin induced ABCG2 expression in HK-2 and Caco-2 cells, as well as SLC2A9 in Caco-2 cells, via the activation of PPARγ. Nevertheless, bergenin suppressed SLC2A9 expression in HK-2 cells by inhibiting the nuclear translocation of p53. Furthermore, bergenin decreased the serum levels of IL-6, IL-1ß, and TNF-α in hyperuricemia mice, and promoted a polarization shift from the M1 to M2 phenotype in RAW264.7 cells. In conclusion, these findings provide evidence supporting the further development of bergenin as a novel therapeutic strategy for hyperuricemia.

13.
J Cell Mol Med ; 24(18): 10444-10457, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32681612

RESUMO

Over-activated osteoclastogenesis, which is initiated by inflammation, has been implicated in osteoporosis. Corilagin, a natural compound extracted from various medicinal herbaceous plants, such as Cinnamomum cassia, has antioxidant and anti-inflammatory activities. We found that Corilagin suppressed osteoclast differentiation in a dose-dependent manner, significantly decreased osteoclast-related gene expression and impaired bone resorption by osteoclasts. Moreover, phosphorylation of members of the nuclear factor-kappaB (NF-κB) and PI3K/AKT signalling pathways was reduced by Corilagin. In a murine model of osteoporosis, Corilagin inhibited osteoclast functions in vivo and restored oestrogen deficiency-induced bone loss. In conclusion, our findings suggested that Corilagin inhibited osteoclastogenesis by down-regulating the NF-κB and PI3K/AKT signalling pathways, thus showing its potential possibility for the treatment of osteoporosis.


Assuntos
Reabsorção Óssea/patologia , Estrogênios/deficiência , Glucosídeos/farmacologia , Taninos Hidrolisáveis/farmacologia , NF-kappa B/metabolismo , Osteogênese/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ligante RANK/farmacologia , Actinas/metabolismo , Animais , Células da Medula Óssea/efeitos dos fármacos , Células da Medula Óssea/metabolismo , Regulação para Baixo/efeitos dos fármacos , Glucosídeos/química , Taninos Hidrolisáveis/química , Camundongos , Camundongos Endogâmicos C57BL , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteoprotegerina/metabolismo , Ovariectomia , Células RAW 264.7 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos
14.
Int Immunopharmacol ; 84: 106550, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-32388216

RESUMO

The metabolicosteopathy known as postmenopausal osteoporosisiscaused by disruption of the balance between bone resorption and osteogenesis, processes that are mediated by osteoclasts and osteoblasts, respectively. The current therapeutic approaches to treating osteoporosis have several limitations. In this study, we demonstrated that the natural chemical compound isoalantolactone (IAL) could inhibit osteoclastogenesis, without affecting osteogenesis. This is the first study reporting a role of IAL in suppressing the receptor activator of nuclear factor-kappa B ligand (RANKL)-induced osteoclast formation in a dose-dependent manner, and downregulating the expression of osteoclast-related marker genes. Furthermore, IAL abrogated the phosphorylation of c-Jun N-terminal kinase (JNK)/p38, NF-κB, and phosphatidylinositol 3-kinase (PI3K)-AKT, and also diminished the expression of osteoclastogenesis-related proteins. In conclusion, our results indicated that IAL has promise for the treatment of osteoporosis and other metabolicbone diseases.


Assuntos
Reabsorção Óssea/tratamento farmacológico , Osteoclastos/efeitos dos fármacos , Osteogênese/efeitos dos fármacos , Sesquiterpenos/uso terapêutico , Actinas/metabolismo , Animais , Catepsina K/genética , Catepsina K/metabolismo , Células Cultivadas , Feminino , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Camundongos Endogâmicos C57BL , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Osteoclastos/metabolismo , Ovariectomia , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-fos/metabolismo , Ligante RANK , Sesquiterpenos/farmacologia , Transdução de Sinais/efeitos dos fármacos
15.
Int J Med Sci ; 17(8): 1083-1094, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32410838

RESUMO

Background: Despite significant advances in the materials and methods development used in surgical repair and postoperative rehabilitation, the adhesion formation remains the most common clinical problem in tendon injuries. Therefore, the development of novel therapies is necessary for targeting at preventing tendon adhesion formation and improving tendon strength. Methods: We used rat fibroblasts for in vitro experiments to determine the optimal concentration of TSA in rats, and then set up negative control group, TSA intervention group, mir-29b interference adenovirus intervention group and TSA and mir-29b interference adenovirus co-intervention group. By comparing cell proliferation and protein expression in different group, we verified the effect and mechanism of drugs on fibroblast function. At the same time, the Sprague-Dawley rat Achilles tendon model in vivo was established in this study, which was divided into sham operation group and operation group. Afterwards in the operation group, mir-29b inhibitor and placebo were injected every 3 days respectively. Then the injection inhibitor group was divided into 5 groups which mean TSA was injected into the marked area at 0, 6, 24 and 72 hours after operation for 1 week, finally all of the rats were died at 3 weeks after operation. Through the observation of general properties, histological observation of Achilles tendon injury, biomechanical test and cell and protein expression in rats' tendon cell, the effect of drugs on tendon adhesion formation was analyzed. Results: We demonstrated that the combination of miR-29b inhibitor and tanshinone IIA(TSA) could prevent tendon adhesion and also enhance tendon strength. Mechanically, the miR-29b inhibitor could activate the TGF-ß/Smad3 pathway to trigger endogenous pathways and induce a high proliferation of fibroblast. Subsequently, we also found adding TSA after 6 hours of miR-29b treatment gave less cell cytotoxicity in our rat model with better outcome of less tendon adhesion and enhanced strength. Conclusion: We conclude that the use of miR-29b inhibitor at the end of the tendon break could initiate endogenous repair mechanism and subsequently use of TSA should be able to inhibit the exogenous repair mechanism. Therefore, the combination of both treatments could prevent tendon adhesion and ensure tendon strength. Our findings suggested that this approach would be a feasible approach for tendon repair.


Assuntos
Abietanos/administração & dosagem , MicroRNAs/antagonistas & inibidores , Complicações Pós-Operatórias/prevenção & controle , Traumatismos dos Tendões/cirurgia , Aderências Teciduais/prevenção & controle , Tendão do Calcâneo/efeitos dos fármacos , Tendão do Calcâneo/lesões , Tendão do Calcâneo/fisiopatologia , Tendão do Calcâneo/cirurgia , Animais , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Células Cultivadas , Modelos Animais de Doenças , Fibroblastos , Humanos , Injeções Intralesionais , MicroRNAs/metabolismo , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/patologia , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Técnicas de Sutura/efeitos adversos , Traumatismos dos Tendões/fisiopatologia , Resistência à Tração/efeitos dos fármacos , Resistência à Tração/fisiologia , Aderências Teciduais/etiologia , Aderências Teciduais/patologia , Cicatrização/efeitos dos fármacos , Cicatrização/genética
16.
FASEB J ; 34(4): 5483-5498, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-32107793

RESUMO

Low back pain (LBP) is one of the most common complains in orthopedic outpatient department and intervertebral disc degeneration (IDD) is one of the most important reasons of LBP. The mechanisms of IDD contain a complex biochemical cascade which includes inflammation, vascular ingrowth, and results in degradation of matrix. In our study, we used both in vitro and in vivo models to investigate the relation between tissue inhibitor of metalloproteinase-3 (TIMP3) expression and IDD. Loss of TIMP3 expression was found in degenerative intervertebral disc (IVD), this change of expression was closely related with the dephosphorylation of smad2/3. Overexpression of TIMP3 significantly inhibited the release of TNF-α and matrix degradation induced by Lipopolysaccharide. Vascular ingrowth was also suppressed by TIMP3 in the in vitro and in vivo models. Further, animal experiments confirmed that the degeneration of IVD was reduced after overexpression of TIMP3 in nucleus pulposus. Taken together, our results indicated TIMP-3 might play an important role in the pathogenesis of IDD and therefore be a potential therapeutic target in the future.


Assuntos
Matriz Extracelular/patologia , Inflamação/patologia , Degeneração do Disco Intervertebral/patologia , Neovascularização Patológica/patologia , Núcleo Pulposo/patologia , Inibidor Tecidual de Metaloproteinase-3/deficiência , Adulto , Idoso , Animais , Células Cultivadas , Matriz Extracelular/metabolismo , Feminino , Humanos , Inflamação/genética , Inflamação/metabolismo , Degeneração do Disco Intervertebral/genética , Degeneração do Disco Intervertebral/metabolismo , Masculino , Pessoa de Meia-Idade , Neovascularização Patológica/genética , Neovascularização Patológica/metabolismo , Núcleo Pulposo/irrigação sanguínea , Núcleo Pulposo/metabolismo , Prognóstico , Ratos Sprague-Dawley , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
17.
Cell Prolif ; 53(2): e12752, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31889368

RESUMO

OBJECTIVES: Insulin-like growth factor-binding protein 7 (IGFBP7) is a low-affinity insulin growth factor (IGF) binder that may play an important role in bone metabolism. We previously reported that IGFBP7 enhanced osteogenic differentiation of bone marrow-derived mesenchymal stem cells (BMSCs) via the Wnt/ß-catenin signalling pathway. In this study, we tried to reveal its function in osteoclast differentiation and osteoporosis. METHODS: We used both in vitro and in vivo studies to investigate the effects of IGFBP7 on RANKL-induced osteoclastogenesis and osteoporosis, together with the underlying molecular mechanisms of these processes. RESULTS: We show that IGFBP7 inhibited receptor activation of nuclear factor-κB (NF-κB) ligand (RANKL)-induced osteoclastogenesis, F-actin ring formation and bone resorption, which was confirmed by using recombinant IGFBP7 protein, lentivirus and siRNA. The NF-κB signalling pathway was inhibited during this process. Moreover, in a mouse ovariectomy-induced osteoporosis model, IGFBP7 treatment attenuated osteoporotic bone loss by inhibiting osteoclast activity. CONCLUSIONS: Taken together, these findings show that IGFBP7 suppressed osteoclastogenesis in vitro and in vivo and suggest that IGFBP7 is a negative regulator of osteoclastogenesis and plays a protective role in osteoporosis. These novel insights into IGFBP7 may facilitate the development of potential treatment strategies for oestrogen deficiency-induced osteoporosis and other osteoclast-related disorders.


Assuntos
Reabsorção Óssea/metabolismo , Estrogênios/metabolismo , Proteínas de Ligação a Fator de Crescimento Semelhante a Insulina/metabolismo , Osteoclastos/metabolismo , Osteogênese/fisiologia , Ligante RANK/metabolismo , Animais , Células da Medula Óssea/metabolismo , Diferenciação Celular/fisiologia , Feminino , Masculino , Células-Tronco Mesenquimais/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Osteoblastos/metabolismo , Osteoporose/metabolismo
18.
Clin Anat ; 33(5): 637-642, 2020 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-31573096

RESUMO

External fixation is a common, efficient technique used for humeral shaft stabilization and elbow fractures. There are reports of radial nerve injuries associated with this procedure. In this study, we investigated the course and variability of the radial nerve along the lateral humerus in relation to the elbow joint to determine a relatively safe zone for lateral pin placement in external fixation. Twenty upper extremities from 10 cadavers were studied. The nerve branches and course of the radial nerve along the lateral humerus were carefully dissected. Straight lines (a, b, and c) were made connecting three landmarks (the acromion, coracoid process, and anterior wall of the axilla) in the proximal upper extremity to the lateral condyle (LC) of the humerus; their intersections with the radial nerve (A, B, and C) were marked. We analyzed whether the intersection positions were correlated with the connecting line lengths. The mean lengths of the connecting lines were (a) 27.24 ± 2.57, (b) 26.18 ± 2.79, and (c) 20.95 ± 1.44 cm; the distance between the intersection points and the LC of the humerus were (Aa) 7.56 ± 1.31, (Bb) 6.90 ± 2.27, and (Cc) 5.01 ± 0.83 cm; and the measured intersection points of the radial nerve in the lateral aspect of the humerus were (A) 18.48%-34.82%, (B) 13.48%-40.00%, and (C) 19.27%-28.05% of the lengths of lines a, b, and c, respectively. Our data provide a more reliable reference to predict the course of the radial nerve on the lateral humerus and define a safe zone for pin placement. Clin. Anat., 33:637-642, 2020. © 2019 Wiley Periodicals, Inc.


Assuntos
Pinos Ortopédicos , Articulação do Cotovelo/inervação , Úmero/inervação , Nervo Radial/anatomia & histologia , Idoso , Idoso de 80 Anos ou mais , Pontos de Referência Anatômicos , Cadáver , Feminino , Fixação de Fratura/métodos , Humanos , Masculino , Pessoa de Meia-Idade
19.
Cell Death Dis ; 10(10): 753, 2019 10 03.
Artigo em Inglês | MEDLINE | ID: mdl-31582734

RESUMO

Interleukin (IL)-37, a pivotal anti-inflammatory cytokine and a fundamental inhibitor of innate immunity, has recently been shown to be abnormally expressed in several autoimmune-related orthopedic diseases, including rheumatoid arthritis, ankylosing spondylitis, and osteoporosis. However, the role of IL-37 during osteogenic differentiation of mesenchymal stem cells (MSCs) remains largely unknown. In this study, extracellular IL-37 significantly increased osteoblast-specific gene expression, the number of mineral deposits, and alkaline phosphatase activity of MSCs. Moreover, a signaling pathway was activated in the presence of IL-37. The enhanced osteogenic differentiation of MSCs due to supplementation of IL-37 was partially rescued by the presence of a PI3K/AKT signaling inhibitor. Using a rat calvarial bone defect model, IL-37 significantly improved bone healing. Collectively, these findings indicate that extracellular IL-37 enhanced osteogenesis of MSCs, at least in part by activation of the PI3K/AKT signaling pathway.


Assuntos
Diferenciação Celular , Espaço Extracelular/metabolismo , Interleucina-1/metabolismo , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Osteogênese , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fosfatase Alcalina/metabolismo , Animais , Cálcio/metabolismo , Morte Celular/genética , Diferenciação Celular/genética , Proliferação de Células/genética , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadeia alfa 1 do Colágeno Tipo I , Subunidade alfa 1 de Fator de Ligação ao Core/genética , Subunidade alfa 1 de Fator de Ligação ao Core/metabolismo , Modelos Animais de Doenças , Regulação da Expressão Gênica , Humanos , Imageamento Tridimensional , Masculino , Osteocalcina/genética , Osteocalcina/metabolismo , Osteogênese/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos Sprague-Dawley , Transdução de Sinais , Crânio/diagnóstico por imagem , Crânio/patologia , Cicatrização
20.
Front Pharmacol ; 10: 618, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31258473

RESUMO

Bone mesenchymal stem cells (BMSCs) are important candidates for bone regeneration. The role of Bergenin, a C-glucoside of 4-O-methyl gallic acid obtained from the species, Bergenia, in BMSC osteogenesis has not yet been elucidated. We therefore investigated the effects of Bergenin on the osteogenesis of BMSCs and found that Bergenin enhanced osteoblast-specific markers and downregulated the adipocyte-specific markers in vitro. Furthermore, using a rat calvarial defect model, we found that Bergenin significantly improved bone healing, as determined by imaging and histological analyses. Moreover, it also upregulated SIRT1 expression. A SIRT1 inhibitor (EX 527) decreased the enhanced bone mineral formation caused by Bergenin. Taken together, these findings show that Bergenin accelerated the osteogenic differentiation of BMSCs, at least partly through the activation of SIRT1.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...