Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Nat Commun ; 13(1): 7139, 2022 11 21.
Artigo em Inglês | MEDLINE | ID: mdl-36414669

RESUMO

Emerging evidence suggests that osteoarthritis is associated with high cholesterol levels in some osteoarthritis patients. However, the specific mechanism under this metabolic osteoarthritis phenotype remains unclear. We find that cholesterol metabolism-related gene, LRP3 (low-density lipoprotein receptor-related protein 3) is significantly reduced in high-cholesterol diet mouse's cartilage. By using Lrp3-/- mice in vivo and LRP3 lentiviral-transduced chondrocytes in vitro, we identify that LRP3 positively regulate chondrocyte extracellular matrix metabolism, and its deficiency aggravate the degeneration of cartilage. Regardless of diet, LRP3 overexpression in cartilage attenuate anterior cruciate ligament transection induced osteoarthritis progression in rats and Lrp3 knockout-induced osteoarthritis progression in mice. LRP3 knockdown upregulate syndecan-4 by activating the Ras signaling pathway. We identify syndecan-4 as a downstream molecular target of LRP3 in osteoarthritis pathogenesis. These findings suggest that cholesterol-LRP3- syndecan-4 axis plays critical roles in osteoarthritis development, and LRP3 gene therapy may provide a therapeutic regimen for osteoarthritis treatment.


Assuntos
Proteínas Relacionadas a Receptor de LDL , Osteoartrite , Sindecana-4 , Animais , Camundongos , Ratos , Cartilagem/metabolismo , Colesterol/metabolismo , Regulação para Baixo , Osteoartrite/metabolismo , Sindecana-4/genética , Sindecana-4/metabolismo , Proteínas Relacionadas a Receptor de LDL/genética , Proteínas Relacionadas a Receptor de LDL/metabolismo
2.
Front Nutr ; 9: 1000947, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36211515

RESUMO

Objective: Rotator cuff injury can be caused by local inflammation and fibrosis of musculotendinous cuff. Hypercholesterolemia can lead to physiological changes of rotator cuff that resemble rotator cuff injury. However, the relationship between lipid metabolism and rotator cuff injury and its potential pathological mechanism remain unclear. Herein, we aimed to investigate the correlation between the plasma lipidome, rotator cuff injury, and successive fatty infiltration pathology, and hoped to identify biomarkers for predicting higher risk or higher severity rotator cuff injury by assessing metabolic perturbations and dyslipidemia using lipidomics. Methods: We quantitatively analyzed 60 lipids species of seven lipids classes and subclasses from 66 subjects using lipidomics. Subjects were divided into four groups: (1) normal rotator cuff with normal clinical routine serum lipid test results (NN group = 13); (2) normal rotator cuff with abnormal clinical routine serum lipid test results (NA group = 10); (3) rotator cuff tear with normal routine serum lipid test results (RN group = 30); (4) rotator cuff tear with abnormal routine serum lipid test results (RA group = 13). Independent-sample t-tests and Kruskal-Wallis tests were used to compare lipid metabolite levels in serum between different groups in patients with rotator cuff tears. The orthogonal partial least squares-discriminant analysis (OPLS-DA) model was used to verify the ability of five lysophosphatidylcholines (LPCs) to distinguish rotator cuff injuries. In the rotator cuff tear group, magnetic resonance imaging (MRI) was used to classify fatty infiltration according to Goutallier's classification. Kruskal-Wallis tests were used to analyze molecular differences between high-grade (grade 3-4) and low-grade (grade 0-2) fatty infiltration groups. Receiver operator characteristic (ROC) curves were drawn for each diagnostic method via different metabolites. The area under the curve (AUC), cutoff, specificity, sensitivity, and accuracy of each diagnostic criterion were calculated. Results: Our results showed that some rotator cuff injury patients yielded unique lipidomic profiles. Based on Kruskal-Wallis tests, our results showed significant differences in three lipid molecules, 17:1 Lyso PI, 18:0-22:6 PE, and 18:3 (Cis) PC, among all four groups independent of clinical blood lipid levels. Also, independent of clinical blood lipid levels, two lipid molecules, 22:0 Lyso PC and 24:0 Lyso PC, were significantly different between the two groups based on Independent sample t-tests. Kruskal-Wallis test results showed that in the rotator cuff tear group, two metabolites (24:0 SM and 16:0 ceramide) differed between high-grade and low-grade fatty infiltration. The AUC values for 22:0 Lyso PC, 24:0 Lyso PC, 18:0-22:6 PE, 24:0 SM, and 16:0 ceramide were 0.6036, 0.6757, 0.6712, 0.8333, and 0.8981, respectively. Conclusion: The results provide insight into how the metabolic mechanisms associated with dyslipidemia impact rotator cuff diseases. Five lipid molecules, 17:1 Lyso PI, 18:0-22:6 PE, 18:3 (Cis) PC, 22:0 Lyso PC, and 24:0 Lyso PC, were closely related to rotator cuff tear based on two statistical analysis methods, independent of clinical routine serum lipid test results, which indicates that lipidomics assays are more sensitive than conventional lipid tests, and more suitable for studying rotator cuff lipid metabolism. In addition, two lipid metabolites, 24:0 SM and 16:0 ceramide, are potentially useful for predicting fatty infiltration severity. Further research with a larger number of samples is needed to verify whether these two metabolites can serve as potential markers of severe fatty infiltration. The findings illuminate how metabolic mechanisms associated with dyslipidemia affect rotator cuff disease.

3.
Orthop J Sports Med ; 10(2): 23259671211051301, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-35187181

RESUMO

BACKGROUND: The accuracy of existing devices for measuring knee laxity is adversely affected by examiner reliability. PURPOSE: To compare the accuracy of a novel automatic knee arthrometer (AKA) to that of the KT-2000 arthrometer for measuring knee laxity after anterior cruciate ligament (ACL) ruptures. STUDY DESIGN: Cohort study; Level of evidence, 2. METHODS: We measured anterior displacement and the anterior displacement difference (ADD) at 134 N of anterior force in 221 healthy volunteers and 200 patients with ACL ruptures. All trials were performed by the same 2 examiners. We first analyzed the effects of examiner, side assessed, and device type using the intraclass correlation coefficient (ICC), t test, and F test. We then used the receiver operating characteristic curve to compare the diagnostic value of the measurements between devices. RESULTS: In repeated measurements for a single healthy volunteer, there were no differences in the variance of the measurements between sides according to the AKA (standard deviation of right vs left knee for examiner A: 0.43 vs 0.58 mm, respectively [P = .39]; for examiner B: 0.49 vs 0.77 mm, respectively [P = .81]), while the KT-2000 measurements showed differences (standard deviation of right vs left knee for examiner A: 1.47 vs 0.80 mm, respectively [P = .02]; for examiner B: 1.78 vs 0.91 mm, respectively [P = .01]). The ADD assessed by the AKA was not significantly different between examiners A and B (0.50 vs 0.75 mm, respectively; P = .27; ICC = 0.83), but the KT-2000 showed a difference (1.07 vs 2.01 mm, respectively; P = .01; ICC = 0.55). The ADD of 20 healthy volunteers assessed by the AKA was less than that by the KT-2000 (0.98 vs 1.41 mm, respectively; P = .04). When comparing the diagnostic value of the 2 devices in the sample of 200 patients with ACL ruptures and 200 healthy controls, the area under the receiver operating characteristic curve for the AKA was larger than that for the KT-2000 (0.93 vs 0.87, respectively; P ≤ .01), and the threshold values were 1.75 and 2.73 mm, respectively. CONCLUSION: The AKA can be used to determine the degree of knee laxity in ACL injuries and to provide indications for treatment.

4.
Acta Biomater ; 131: 262-275, 2021 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-34157451

RESUMO

Decellularized extracellular matrix (dECM) hydrogels are being increasingly investigated for use in bio-inks for three-dimensional cell printing given their good cytocompatibility and biomimetic properties. The osmotic pressure and stiffness of bio-ink are important factors affecting the biological functions of printed cells. However, little attention has been given to the osmotic pressure and stiffness of the dECM bio-inks. Here, we compared three types of commonly used acidic solutions in the bio-fabrication of a tendon derived dECM bio-ink for 3D cell printing (0.5 M acetic acid, 0.1 M hydrochloric acid and 0.02 M hydrochloric acid). We found that low pH value of 0.1 M hydrochloric acid could accelerate the digestion process for dECM powders. This could lead to a much softer dECM hydrogel with storage modulus less than 100 Pa. This soft dECM hydrogel facilitated the spreading and proliferation of stem cells encapsulated within it. It also showed better tendon-inducing ability compared with two others much stiffer dECM hydrogels. However, this over-digested dECM hydrogel was more unstable as it could shrink with the culture time going on. For 0.5 M acetic acid made dECM bio-ink, the hyperosmotic state of the bio-ink led to much lower cellular viability rates. Postprocess (Dilution or dialysis) to tailor the osmotic pressure of hydrogels could be a necessary step before mixed with cells. Thus, kindly choosing the type and concentration of acidic solution is necessary for dECM bio-ink preparation. And a balance should be made between the digestion period, strength of acidic solution, as well as the size and concentration of the dECM powders. STATEMENT OF SIGNIFICANCE: The dECM bio-ink has been widely used in 3D cell printing for tissue engineering and organ modelling. In this study, we found that different types of acid have different digestion and dissolution status for the dECM materials. A much softer tendon derived dECM hydrogel with lower stiffness could facilitate the cellular spreading, proliferation and tendon differentiation. We also demonstrated that the osmotic pressure should be taken care of in the preparation of dECM bio-ink with 0.5 M acetic acid. Thus, kindly choosing the type and concentration of acidic solution is necessary for dECM bio-ink preparation.


Assuntos
Matriz Extracelular , Tinta , Hidrogéis/farmacologia , Impressão Tridimensional , Tendões , Engenharia Tecidual , Tecidos Suporte
5.
Front Cell Dev Biol ; 9: 638382, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33937236

RESUMO

Osteoarthritis (OA) is a highly prevalent and debilitating joint disorder that characterized by progressive destruction of articular cartilage. There is no effective disease-modifying therapy for the condition due to limited understanding of the molecular mechanisms on cartilage maintenance and destruction. Receptor-interacting protein kinase 1 (RIP1)-mediated necroptosis plays a vital role in various diseases, but the involvement of RIP1 in OA pathogenesis remains largely unknown. Here we show that typical necrotic cell morphology is observed within human OA cartilage samples in situ, and that RIP1 is significantly upregulated in cartilage from both OA patients and experimental OA rat models. Intra-articular RIP1 overexpression is sufficient to induce structural and functional defects of cartilage in rats, highlighting the crucial role of RIP1 during OA onset and progression by mediating chondrocyte necroptosis and disrupting extracellular matrix (ECM) metabolism homeostasis. Inhibition of RIP1 activity by its inhibitor necrostatin-1 protects the rats from trauma-induced cartilage degradation as well as limb pain. More importantly, we identify bone morphogenetic protein 7 (BMP7) as a novel downstream target that mediates RIP1-induced chondrocyte necroptosis and OA manifestations, thereby representing a non-canonical regulation mode of necroptosis. Our study supports a model whereby the activation of RIP1-BMP7 functional axis promotes chondrocyte necroptosis and subsequent OA pathogenesis, thus providing a new therapeutic target for OA.

6.
Cancer Genomics Proteomics ; 18(3 Suppl): 461-470, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33994368

RESUMO

BACKGROUND/AIM: Diffuse-type tenosynovial giant cell tumor (TGCT) is a rare benign proliferative synovial neoplasm of uncertain etiology, and the efficacy of surgical resection is not satisfactory. Therefore, there is an urgent need to explore the pathogenesis and identify novel therapeutic targets for TGCT. MATERIALS AND METHODS: Synovial tissues were collected from patients with TGCT and osteoarthritis (OA). Differences of mRNA expression between TGCT and OA were explored using mRNA-seq. In addition, fibroblast-like synoviocytes (FLS) were treated with small interfering RNA (siRNA) or adenovirus in order to knockdown or overexpress ß-arrestin2 (Arrb2), respectively. FLS proliferation and apoptosis were evaluated using the MTT assay and the caspase 3 activity assay, respectively. RESULTS: The expression of Arrb2 in TGCT was significantly higher than that in OA. The overexpression of Arrb2 promoted the proliferation of FLS and inhibited its apoptosis, while knocking down Arrb2 had the opposite effect. Further studies showed that Arrb2 can activate the PI3K-Akt signaling pathway, leading to increased proliferation of TGCT. CONCLUSION: Arrb2 facilitates the proliferation and inhibits the apoptosis of TGCT FLS through activating the PI3K-Akt cell survival pathway, providing new insight into the molecular mechanism of TGCT.


Assuntos
Tumor de Células Gigantes de Bainha Tendinosa/genética , Sinoviócitos/metabolismo , beta-Arrestina 2/metabolismo , Adulto , Apoptose , Proliferação de Células , Feminino , Tumor de Células Gigantes de Bainha Tendinosa/patologia , Humanos , Masculino , Pessoa de Meia-Idade
7.
ACS Biomater Sci Eng ; 7(3): 916-925, 2021 03 08.
Artigo em Inglês | MEDLINE | ID: mdl-33715368

RESUMO

The development of 3D printing techniques has provided a promising platform to study tissue engineering and mechanobiology; however, the pursuit of printability limits the possibility of tailoring scaffolds' mechanical properties. The brittleness of those scaffolds also hinders potential clinical application. To overcome these drawbacks, a double-network ink composed of only natural biomaterials is developed. A shear-thinning hydrogel made of silk fibroin (SF) and methacrylated hyaluronic acid (MAHA) presents a high mechanical modulus with a low concentration of macromers. The physical cross-linking due to protein folding further increases the strength of the scaffolds. The proposed SF/MAHA scaffold exhibits a storage modulus 10 times greater than that of methacrylated gelatin scaffold, along with better flexibility and biodegradation. The synergistic effect between fibroin and hyaluronic allows us to tailor the mechanical strength of scaffolds without compromising their printability. The hierarchy porous structure of the SF/MAHA scaffolds offers a better spatial microenvironment for the migration and proliferation of cells compared to gelatin scaffolds. For the first time, this strategy achieves 3D printing of natural biomaterials with controlled mechanical characteristics by manipulating the cross-linking of peptide chains. The design of such ductile scaffolds with hydrolysis resistance provides a new platform for the mechanobiology research. It also shows promise in the tissue engineering of musculoskeletal system where structural strength is needed.


Assuntos
Fibroínas , Materiais Biocompatíveis , Impressão Tridimensional , Engenharia Tecidual , Tecidos Suporte
8.
Front Cell Dev Biol ; 8: 694, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32903809

RESUMO

Articular cartilage damage remains a tough challenge for clinicians. Stem cells have emerged promising biologics in regenerative medicine. Previous research has widely demonstrated that adipose-derived mesenchymal stem cells (ADSCs) can promote cartilage repair due to their multipotency. However, enzymatic isolation and monolayer expansion of ADSCs decrease their differentiation potential and limit their clinical application. Here, a novel adipose tissue-derived product, extracellular matrix/stromal vascular fraction gel (ECM/SVF-gel), was obtained by simple mechanical shifting and centrifugation to separate the fat oil and concentrate the effective constituents. This study aimed to evaluate the therapeutic effect of this natural biomaterial on the repair of articular cartilage defects. Scanning electron microscopy showed that the fibrous structure in the ECM/SVF-gel was preserved. ADSCs sprouted from the ECM/SVF-gel were characterized by their ability of differentiation into chondrocytes, osteoblasts, and adipocytes. In a rabbit model, critical-sized cartilage defects (diameter, 4 mm; depth, 1.5 mm) were created and treated with microfracture (MF) or a combination of autologous ECM/SVF-gel injection. The knee joints were evaluated at 6 and 12 weeks through magnetic resonance imaging, macroscopic observation, histology, and immunohistochemistry. The International Cartilage Repair Society score and histological score were significantly higher in the ECM/SVF-gel group than those in the MF-treated group. The ECM/SVF-gel distinctly improved cartilage regeneration, integration with surrounding normal cartilage, and the expression of hyaline cartilage marker, type II collagen, in comparison with the MF treatment alone. Overall, the ready-to-use ECM/SVF-gel is a promising therapeutic strategy to facilitate articular cartilage regeneration. Moreover, due to the simple, time-sparing, cost-effective, enzyme-free, and minimally invasive preparation process, this gel provides a valuable alternative to stem cell-based therapy for clinical translation.

9.
Theranostics ; 10(23): 10573-10588, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32929367

RESUMO

Rationale: Pigmented villonodular synovitis (PVNS) is a destructive benign tumor-like hyperplastic disease that occurs in synovial tissue. Fibroblast-like synoviocytes (FLS) are the predominant cell type comprising the structure of the PVNS synovial lining layer. Due to a high recurrence rate, high invasion, migration, and cartilage destruction ability, PVNS causes substantial damage to patients and the efficacy of surgical resection is not satisfactory. Therefore, exploring the pathogenesis and identifying novel therapeutic targets for PVNS are urgently required. Currently, the pathogenesis of PVNS remains unclear, and there is uncertainty and controversy regarding whether PVNS is an inflammatory or a neoplastic disease. Cadherin-11 is a classical molecule that mediates hemophilic cell-to-cell adhesion in FLS and plays an important role in the normal synovium lining layer formation. This study aimed to explore the role of inflammation and cadherin-11 in PVNS pathogenesis and determine the effects of cadherin-11 as a molecular target for PVNS treatment. Methods: FLS were primarily cultured from PVNS patients during arthroscopic synovectomy. The level of cytokines in the PVNS synovial fluid was evaluated using a human antibody array. Cadherin-11 expression of PVNS FLS was detected by qPCR, Western blots, tissue immunohistochemistry, and cell immunofluorescence. Cadherin-11 was down-regulated by siRNA or up-regulated with a plasmid, with or without inflammatory factor stimulation, and PI3K/Akt was inhibited with LY294002. The capacity of migration and invasion of PVNS FLS was tested using Transwell and wound-healing assays. Activation of the nuclear factor-kappaB (NF-κB) and mitogen-activated protein kinase (MAPK) pathways was detected by Western blots. Chondrocyte damage by PVNS FLS was assessed with a co-culture assay. Results: Inflammatory factors (IL-1ß and TNF-α) in the synovial fluid of PVNS patients were significantly up-regulated. Cadherin-11 was highly expressed in the FLS of PVNS patients, and positively correlated with recurrence, extra-articular migration, and cartilage destruction of PVNS. Knocking down of cadherin-11 inhibited the migration and invasion of PVNS FLS. Moreover, inflammatory factors up-regulated the expression of cadherin-11, which activated the NF-κB and MAPK signaling pathways and led to cartilage destruction. Inhibition of cadherin-11 blocked IL-1ß- and TNF-α-induced activation of the above pathways, migration and invasion of PVNS FLS, and damage of chondrocyte. In addition, the elevation of cadherin-11 expression, together with the migration and invasion, of PVNS FLS was down-regulated by the inhibition of the PI3K/Akt signaling pathway. Conclusions: Cadherin-11 plays an important role in the pathogenesis of PVNS and forms a positive feedback loop with inflammatory factors, which further activates the NF-κB and MAPK pathways to trigger an inflammatory cascade. Cadherin-11-mediated inflammation results in PVNS with high recurrence, invasiveness, and strong cartilage destruction ability, and eventually promotes the transformation of PVNS from the initial inflammatory disease to neoplastic disease. Thus, inhibition of cadherin-11 together with its related inflammatory reaction, represents a new therapeutic strategy for PVNS.


Assuntos
Caderinas/metabolismo , Mediadores da Inflamação/metabolismo , Membrana Sinovial/patologia , Sinoviócitos/metabolismo , Sinovite Pigmentada Vilonodular/imunologia , Adenilato Quinase/metabolismo , Adulto , Artroscopia , Caderinas/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/imunologia , Células Cultivadas , Cromonas/farmacologia , Feminino , Técnicas de Silenciamento de Genes , Humanos , Sistema de Sinalização das MAP Quinases/imunologia , Masculino , Pessoa de Meia-Idade , Morfolinas/farmacologia , Fosfatidilinositol 3-Quinases/metabolismo , Fosfatidilinositol 3-Quinases/farmacologia , Inibidores de Fosfoinositídeo-3 Quinase , Cultura Primária de Células , Sinovectomia/métodos , Membrana Sinovial/citologia , Membrana Sinovial/imunologia , Sinoviócitos/imunologia , Sinovite Pigmentada Vilonodular/patologia , Sinovite Pigmentada Vilonodular/cirurgia , Regulação para Cima/efeitos dos fármacos , Regulação para Cima/imunologia
10.
Theranostics ; 10(11): 5090-5106, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32308770

RESUMO

Meniscus deficiency, the most common and refractory disease in human knee joints, often progresses to osteoarthritis (OA) due to abnormal biomechanical distribution and articular cartilage abrasion. However, due to its anisotropic spatial architecture, complex biomechanical microenvironment, and limited vascularity, meniscus repair remains a challenge for clinicians and researchers worldwide. In this study, we developed a 3D printing-based biomimetic and composite tissue-engineered meniscus scaffold consisting of polycaprolactone (PCL)/silk fibroin (SF) with extraordinary biomechanical properties and biocompatibility. We hypothesized that the meticulously tailored composite scaffold could enhance meniscus regeneration and cartilage protection. Methods: The physical property of the scaffold was characterized by scanning electron microscopy (SEM) observation, degradation test, frictional force of interface assessment, biomechanical testing, and fourier transform infrared (FTIR) spectroscopy analysis. To verify the biocompatibility of the scaffold, the viability, morphology, proliferation, differentiation, and extracellular matrix (ECM) production of synovium-derived mesenchymal stem cell (SMSC) on the scaffolds were assessed by LIVE/DEAD staining, alamarBlue assay, ELISA analysis, and qRT-PCR. The recruitment ability of SMSC was tested by dual labeling with CD29 and CD90 by confocal microscope at 1 week after implantation. The functionalized hybrid scaffold was then implanted into the meniscus defects on rabbit knee joint for meniscus regeneration, comparing with the Blank group (no scaffold) and PS group. The regenerated meniscus tissue was evaluated by histological and immunohistochemistry staining, and biomechanical test. Macroscopic and histological scoring was performed to assess the outcome of meniscus regeneration and cartilage protection in vivo. Results: The combination of SF and PCL could greatly balance the biomechanical properties and degradation rate to match the native meniscus. SF sponge, characterized by fine elasticity and low interfacial shear force, enhanced energy absorption capacity of the meniscus and improved chondroprotection. The SMSC-specific affinity peptide (LTHPRWP; L7) was conjugated to the scaffold to further increase the recruitment and retention of endogenous SMSCs. This meticulously tailored scaffold displayed superior biomechanics, structure, and function, creating a favorable microenvironment for SMSC proliferation, differentiation, and extracellular matrix (ECM) production. After 24 weeks of implantation, the histological assessment, biochemical contents, and biomechanical properties demonstrated that the polycaprolactone/silk fibroin-L7 (PS-L7) group was close to the native meniscus group, showing significantly better cartilage protection than the PS group. Conclusion: This tissue engineering scaffold could greatly strengthen meniscus regeneration and chondroprotection. Compared with traditional cell-based therapies, the meniscus tissue engineering approach with advantages of one-step operation and reduced cost has a promising potential for future clinical and translational studies.


Assuntos
Cartilagem Articular/citologia , Fibroínas/química , Menisco/citologia , Células-Tronco Mesenquimais/citologia , Poliésteres/química , Impressão Tridimensional/instrumentação , Engenharia Tecidual/métodos , Tecidos Suporte/química , Animais , Fenômenos Biomecânicos , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Diferenciação Celular , Células Cultivadas , Menisco/efeitos dos fármacos , Menisco/metabolismo , Células-Tronco Mesenquimais/metabolismo , Porosidade , Coelhos
11.
Acta Biomater ; 106: 328-341, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32027991

RESUMO

Mesenchymal stem cells (MSCs)-derived exosomes are being increasingly focused as the new biological pro-regenerative therapeutic agents for various types of tissue injury. Here, we explored the potential of a novel exosome-based therapeutic application combined with a local fibrin delivery strategy for tendon repair. After discovering that bone marrow mesenchymal stem cells-derived exosomes (BMSCs-exos) promoted the proliferation, migration and tenogenic differentiation of tendon stem/progenitor cells (TSPCs) in vitro, we embedded BMSCs-exos in fibrin and injected it into the defect area of rat patellar tendon, and the results showed that the exosomes could be controlled-released from the fibrin, retained within the defect area, and internalized by TSPCs. BMSCs-exos embedded in fibrin significantly improved the histological scores, enhanced the expression of mohawk, tenomodulin, and type I collagen, as well as the mechanical properties of neotendon, and also promoted the proliferation of local TSPCs in vivo. Overall, we demonstrated the beneficial role of BMSCs-exos in tendon regeneration, and that fibrin-exosomes delivery system represents a successful local treatment strategy of exosomes. This study brings prospects in the potential application of exosomes in novel therapies for tendon injury. STATEMENT OF SIGNIFICANCE: Mesenchymal stem cells have been identified as a preferred approach in tissue regeneration. In this study, we reported bone marrow mesenchymal stem cells (BMSCs) promote the proliferation and migration of tendon stem/progenitor cells (TSPCs) via the paracrine signaling effect of the nanoscale exosomes. We also demonstrated that the application of BMSCs-derived exosomes might be a promising approach to activate the regenerative potential of endogenous TSPCs in tendon injured region, and fibrin-exosomes delivery system represents a successful local treatment strategy of exosomes.


Assuntos
Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Exossomos/transplante , Regeneração/fisiologia , Traumatismos dos Tendões/terapia , Tendões/fisiologia , Animais , Diferenciação Celular/fisiologia , Fibrina/química , Hidrogéis/química , Masculino , Células-Tronco Mesenquimais/citologia , Ratos Sprague-Dawley , Tendões/citologia
12.
Am J Transl Res ; 11(10): 6650-6659, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31737215

RESUMO

Cartilage defects are most commonly seen in the knee joint. However, due to the limited self-recovery ability of cartilage, the repair of articular cartilage defects is still a great challenge despite that various approaches have been proposed. We designed a strategy to induce cartilage repair using acellular bone matrix (ABM), thereby creating an appropriate microenvironment for the in-situ cells with an easy surgical application. An in vitro system demonstrated that the ABM scaffold could promote cell adhesion, growth, proliferation, and chondrogenesis of mesenchymal stem cells. This experiment was performed in a minipig cartilage repair model. The repaired tissue was hyaline-like cartilage according to the morphological and histological results. The mechanical properties of the repaired tissue were similar to those of normal cartilage. The integration of repaired tissue and normal tissue in the ABM+M group was better than those of other two groups. The ABM-based, one-stage, minimally invasive, in situ procedure for cartilage regeneration can potentially improve the treatment of articular cartilage defects.

13.
Nat Commun ; 10(1): 1914, 2019 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-31015473

RESUMO

Degradation of extracellular matrix (ECM) underlies loss of cartilage tissue in osteoarthritis, a common disease for which no effective disease-modifying therapy currently exists. Here we describe BNTA, a small molecule with ECM modulatory properties. BNTA promotes generation of ECM components in cultured chondrocytes isolated from individuals with osteoarthritis. In human osteoarthritic cartilage explants, BNTA treatment stimulates expression of ECM components while suppressing inflammatory mediators. Intra-articular injection of BNTA delays the disease progression in a trauma-induced rat model of osteoarthritis. Furthermore, we identify superoxide dismutase 3 (SOD3) as a mediator of BNTA activity. BNTA induces SOD3 expression and superoxide anion elimination in osteoarthritic chondrocyte culture, and ectopic SOD3 expression recapitulates the effect of BNTA on ECM biosynthesis. These observations identify SOD3 as a relevant drug target, and BNTA as a potential therapeutic agent in osteoarthritis.


Assuntos
Anti-Inflamatórios/farmacologia , Benzamidas/farmacologia , Cartilagem Articular/efeitos dos fármacos , Matriz Extracelular/efeitos dos fármacos , Sequestradores de Radicais Livres/farmacologia , Fatores Imunológicos/farmacologia , Osteoartrite/tratamento farmacológico , Sulfonamidas/farmacologia , Animais , Cartilagem Articular/imunologia , Cartilagem Articular/patologia , Condrócitos/efeitos dos fármacos , Condrócitos/imunologia , Condrócitos/patologia , Citocinas/genética , Citocinas/imunologia , Modelos Animais de Doenças , Progressão da Doença , Matriz Extracelular/imunologia , Matriz Extracelular/patologia , Perfilação da Expressão Gênica , Regulação da Expressão Gênica , Humanos , Injeções Intra-Articulares , Masculino , Osteoartrite/genética , Osteoartrite/imunologia , Osteoartrite/patologia , Cultura Primária de Células , Ratos , Ratos Sprague-Dawley , Superóxido Dismutase/genética , Superóxido Dismutase/imunologia , Superóxidos/antagonistas & inibidores , Superóxidos/metabolismo , Transcriptoma/imunologia
14.
Biochem Biophys Res Commun ; 509(4): 983-987, 2019 02 19.
Artigo em Inglês | MEDLINE | ID: mdl-30654942

RESUMO

Articular cartilage defects are considered a major clinical problem because they cannot heal by themselves. To date, bone marrow-derived mesenchymal stem cells (BMSCs)-based therapy has been widely applied for cartilage repair. However, fibrocartilage was often generated after BMSC therapy; therefore, there is an urgent need to stimulate and maintain BMSCs chondrogenic differentiation. The specific role of superoxide dismutase 3 (SOD3) in chondrogenesis is unknown; therefore, the present study aimed to clarify whether SOD3 could facilitate the chondrogenic differentiation of BMSCs. We first evaluated SOD3 protein levels during chondrogenesis of BMSCs using plate cultures. We then tested whether SOD3 could facilitate chondrogenesis of BMSCs using knockdown or overexpression experiments. Increased SOD3 protein levels were observed during BMSCs chondrogenesis. SOD3 knockdown inhibited collagen type II alpha 1 chain (COL2A1), aggrecan (ACAN), and SRY-box 9 (SOX9) expression. Overexpression of SOD3 increased the levels of chondrogenesis markers (COL2A1, ACAN, and SOX9). Elevated superoxide anions were observed when SOD3 was knocked down. We concluded that SOD3 could facilitate chondrogenesis of BMSCs to improve cartilage regeneration.


Assuntos
Condrogênese , Células-Tronco Mesenquimais/citologia , Superóxido Dismutase/fisiologia , Biomarcadores/metabolismo , Células da Medula Óssea/citologia , Cartilagem Articular/metabolismo , Diferenciação Celular , Células Cultivadas , Humanos , Regeneração/efeitos dos fármacos , Superóxido Dismutase/farmacologia
15.
ACS Biomater Sci Eng ; 5(2): 805-816, 2019 Feb 11.
Artigo em Inglês | MEDLINE | ID: mdl-33405841

RESUMO

Synovium-derived mesenchymal stem cells (SMSCs) are attractive tissue-specific cells for cartilage regeneration because of their easy availability, higher chondrogenic potential, and joint specificity. In the present study, we established a hybrid scaffold to codeliver SMSCs and transforming growth factor beta (TGF-ß), which can integrate the scaffolds, the growth factor, and the autogenous cells within rabbit cartilage defects. A chitosan hydrogel and a decellularized bone matrix were used to fabricate the gel-solid duplex phase biomaterials, which were proven to retain more cells, promote tissue integration, and provide mechanical support. In vitro experiments demonstrated that this hybrid scaffold could release TGF-ß in a controlled biphasic pattern, which can promote cell proliferation and chondrogenic differentiation of loaded rabbit SMSCs. For in vivo experiments, we filled cartilage defects with the hybrid materials, delivering autogenous SMSCs and TGF-ß simultaneously via chitosan's sol-gel transition. Histological analysis, magnetic resonance imaging, and nanoindentation assessment indicated superior cartilage regeneration using this codelivery system compared with that from routine microfracture or control delivery scaffolds. Beyond cartilage regeneration, the easy preparation, favorable biophysical properties, and controlled release ability make this codelivery system applicable to transport other tissue-specific cells or biofactors for tissue engineering.

16.
Sci Rep ; 8(1): 7693, 2018 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-29769537

RESUMO

A meniscus tear often happens during active sports. It needs to be repaired or replaced surgically to avoid further damage to the articular cartilage. To address the shortage of autologous meniscal cells, we designed a co-culture system of synovial stem cells (SMSCs) and meniscal cells (MCs) to produce a large cell number and to maintain characteristics of MCs. Different ratios of SMSCs and MCs at 3:1, 1:1, and 1:3 were tested. Mono-culture of SMSCs or MCs served as control groups. Proliferation and differentiation abilities were compared. The expression of extracellular matrix (ECM) genes in MCs was assessed using an ECM array to reveal the mechanism at the gene level. The co-culture system of SMSCs/MCs at the ratio of 1:3 showed better results than the control groups or those at other ratios. This co-culture system may be a promising strategy for meniscus repair with tissue engineering.


Assuntos
Diferenciação Celular , Proliferação de Células , Condrogênese , Menisco/citologia , Células-Tronco Mesenquimais/citologia , Sinoviócitos/citologia , Engenharia Tecidual/métodos , Animais , Apoptose , Ciclo Celular , Células Cultivadas , Técnicas de Cocultura , Proteínas da Matriz Extracelular/metabolismo , Menisco/metabolismo , Células-Tronco Mesenquimais/metabolismo , Ratos , Ratos Wistar , Sinoviócitos/metabolismo
17.
Bioelectrochemistry ; 120: 138-144, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29253736

RESUMO

The use of polyvinylidene fluoride (PVDF) as a binder was investigated in order to prepare the active carbon catalyst and carbon black diffusion layers of a microbial fuel cell cathode. Compared with other binders, PVDF performed competitively as it did not require a lengthy curing time and high curing temperature. Results of XRD characterization showed that the typical ß-PVDF was enhanced as PVDF content ratio increased. SEM results indicated that the catalyst layer easily peeled off due to the low binder concentration of binder, but the high binder content was deemed undesirable because the large amount of non-conductive PVDF interrupted the percolation path. The optimum binder concentration was double checked using Tafel and EIS tests. Results indicated that the cathode with 10% PVDF is the optimum operated concentration. The cathode can obtain 180mV of cathode potential and the smallest total impedance of 2500Ω, which are consistent with the SEM analysis. Moreover, the cathode with 10% PVDF concentration produced a maximum power density of 1600mWm-2, suggesting that PVDF can compete with other traditional binders.


Assuntos
Fontes de Energia Bioelétrica , Carbono/química , Polivinil/química , Fuligem/química , Fontes de Energia Bioelétrica/economia , Fontes de Energia Bioelétrica/microbiologia , Catálise , Difusão , Impedância Elétrica , Técnicas Eletroquímicas , Eletrodos , Desenho de Equipamento
18.
Sci Rep ; 7(1): 7033, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28765605

RESUMO

Since transplantation of meniscal allograft or artificial menisci is limited by graft sources and a series of adverse events, substitution for meniscus reconstruction still needs to be explored. Natural biomaterials, which can provide a unique 3-D microenvironment, remain a promising alternative for tissue engineering. Among them, autograft is a preferred option for its safety and excellent biocompatibility. In this study, we utilized semitendinosus tendon autograft in meniscus reconstruction to investigate its fibrochondrogenic metaplasticity potential and chondroprotective effect. Tendon-derived stem cells (TDSCs) and synovial-derived mesenchymal stem cells (SMSCs), two most important stem cell sources in our strategy, exhibited excellent viability, distribution, proliferation and fibrochondrogenic differentiation ability in decellularized semitendinosus tendon (DST) scaffolds in vitro. Histologic evaluation of the tendon grafts in vivo suggested endogenous stem cells differentiated into fibrochondrocytes, synthesized proteoglycan, type II collagen and radial type I collagen at 12 weeks and 24 weeks post-surgery. As for elastic modulus and hardness of the grafts, there were no significant differences between native meniscus and regenerated meniscus at 24 weeks. The protection of condylar cartilage from degeneration was significantly better in the reconstruction group comparing to control group. Overall, semitendinosus tendon autograft seems to be a promising substitution in meniscus reconstruction.


Assuntos
Autoenxertos , Tendões dos Músculos Isquiotibiais/cirurgia , Menisco/cirurgia , Transplante de Células-Tronco Mesenquimais/métodos , Células-Tronco Mesenquimais/fisiologia , Transplante Autólogo/métodos , Animais , Coelhos , Resultado do Tratamento
19.
Adv Mater ; 29(29)2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28585319

RESUMO

Articular cartilage repair remains a great challenge for clinicians and researchers. Recently, there emerges a promising way to achieve one-step cartilage repair in situ by combining endogenic bone marrow stem cells (BMSCs) with suitable biomaterials using a tissue engineering technique. To meet the increasing demand for cartilage tissue engineering, a structurally and functionally optimized scaffold is designed, by integrating silk fibroin with gelatin in combination with BMSC-specific-affinity peptide using 3D printing (3DP) technology. The combination ratio of silk fibroin and gelatin greatly balances the mechanical properties and degradation rate to match the newly formed cartilage. This dually optimized scaffold has shown superior performance for cartilage repair in a knee joint because it not only retains adequate BMSCs, due to efficient recruiting ability, and acts as a physical barrier for blood clots, but also provides a mechanical protection before neocartilage formation and a suitable 3D microenvironment for BMSC proliferation, differentiation, and extracellular matrix production. It appears to be a promising biomaterial for knee cartilage repair and is worthy of further investigation in large animal studies and preclinical applications. Beyond knee cartilage, this dually optimized scaffold may also serve as an ideal biomaterial for the regeneration of other joint cartilages.


Assuntos
Materiais Biocompatíveis/química , Cartilagem Articular/fisiologia , Fibroínas/química , Gelatina/química , Impressão Tridimensional , Animais , Materiais Biocompatíveis/farmacologia , Células da Medula Óssea/citologia , Cartilagem Articular/metabolismo , Cartilagem Articular/patologia , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Colágeno/metabolismo , Desenho Assistido por Computador , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Coelhos , Ratos , Ratos Sprague-Dawley , Regeneração/efeitos dos fármacos , Engenharia Tecidual , Tecidos Suporte/química
20.
Acta Biomater ; 53: 279-292, 2017 04 15.
Artigo em Inglês | MEDLINE | ID: mdl-28115294

RESUMO

Due to avascular and hypocellular nature of cartilage, repair of articular cartilage defects within synovial joints still poses a significant clinical challenge. To promote neocartilage properties, we established a functional scaffold named APM-E7 by conjugating a bone marrow-derived mesenchymal stem cell (BM-MSC) affinity peptide (E7) onto the acellular peritoneum matrix (APM). During in vitro culture, the APM-E7 scaffold can support better proliferation as well as better differentiation into chondrocytes of BM-MSCs. After implanting into cartilage defects in rabbits for 24weeks, compared with microfracture and APM groups, the APM-E7 scaffolds exhibited superior quality of neocartilage without transplant rejection, according to general observations, histological assessment, synovial fluid analysis, magnetic resonance imaging (MRI) and nanomechanical properties. This APM-E7 scaffold provided a scaffold for cell attachment, which was crucial for cartilage regeneration. Overall, the APM-E7 is a promising biomaterial with low immunogenicity for one-step cartilage repair by promoting autologous connective tissue progenitor (CTP) attachment. STATEMENT OF SIGNIFICANCE: We report the one-step transplantation of functional acellular peritoneum matrix (APM-E7) with specific mesenchymal stem cell recruitment to repair rabbit cartilage injury. The experimental results illustrated that the APM-E7 scaffold was successfully fabricated, which could specifically recruit MSCs and fill the cartilage defects in the femoral trochlear of rabbits at 24weeks post-surgery. The repaired tissue was hyaline cartilage, which exhibited ideal mechanical stability. The APM-E7 biomaterial could provide scaffold for MSCs and improve cell homing, which are two key factors required for cartilage tissue engineering, thereby providing new insights into cartilage tissue engineering.


Assuntos
Matriz Extracelular/química , Fraturas de Cartilagem/terapia , Fraturas de Estresse/terapia , Transplante de Células-Tronco Mesenquimais/instrumentação , Peritônio/química , Peritônio/citologia , Tecidos Suporte , Animais , Sistema Livre de Células/química , Células Cultivadas , Desenho de Equipamento , Análise de Falha de Equipamento , Fraturas de Cartilagem/patologia , Fraturas de Estresse/patologia , Transplante de Células-Tronco Mesenquimais/métodos , Coelhos , Suínos , Resultado do Tratamento
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...