Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 2.394
Filtrar
1.
J Mater Chem B ; 12(25): 6242-6256, 2024 Jun 27.
Artigo em Inglês | MEDLINE | ID: mdl-38842217

RESUMO

Designing artificial nano-enzymes for scavenging reactive oxygen species (ROS) in chondrocytes (CHOs) is considered the most feasible pathway for the treatment of osteoarthritis (OA). However, the accumulation of ROS due to the amount of nano-enzymatic catalytic site exposure and insufficient oxygen supply seriously threatens the clinical application of this therapy. Although metal-organic framework (MOF) immobilization of artificial nano-enzymes to enhance active site exposure has been extensively studied, artificial nano-enzymes/MOFs for ROS scavenging in OA treatment are still lacking. In this study, a biocompatible lubricating hydrogel-loaded iron-doped zeolitic imidazolate framework-8 (Fe/ZIF-8/Gel) centrase was engineered to scavenge endogenous overexpressed ROS synergistically generating dissolved oxygen and enhancing sustained lubrication for CHOs as a ternary artificial nano-enzyme. This property enabled the nano-enzymatic hydrogels to mitigate OA hypoxia and inhibit oxidative stress damage successfully. Ternary strategy-based therapies show excellent cartilage repair in vivo. The experimental results suggest that nano-enzyme-enhanced lubricating hydrogels are a potentially effective OA treatment and a novel strategy.


Assuntos
Condrócitos , Hidrogéis , Espécies Reativas de Oxigênio , Hidrogéis/química , Hidrogéis/farmacologia , Animais , Condrócitos/metabolismo , Condrócitos/efeitos dos fármacos , Condrócitos/citologia , Espécies Reativas de Oxigênio/metabolismo , Estruturas Metalorgânicas/química , Estruturas Metalorgânicas/farmacologia , Osteoartrite/tratamento farmacológico , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia , Cartilagem/efeitos dos fármacos , Cartilagem/metabolismo , Tamanho da Partícula , Humanos , Zeolitas/química
2.
Int J Biol Macromol ; 272(Pt 1): 132848, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38830491

RESUMO

Collagen-based (COL) hydrogels could be a promising treatment option for injuries to the articular cartilage (AC) becuase of their similarity to AC native extra extracellular matrix. However, the high hydration of COL hydrogels poses challenges for AC's mechanical properties. To address this, we developed a hydrogel platform that incorporating cellulose nanocrystals (CNCs) within COL and followed by plastic compression (PC) procedure to expel the excessive fluid out. This approach significantly improved the mechanical properties of the hydrogels and enhanced the chondrogenic differentiation of mesenchymal stem cells (MSCs). Radially confined PC resulted in higher collagen fibrillar densities together with reducing fibril-fibril distances. Compressed hydrogels containing CNCs exhibited the highest compressive modulus and toughness. MSCs encapsulated in these hydrogels were initially affected by PC, but their viability improved after 7 days. Furthermore, the morphology of the cells and their secretion of glycosaminoglycans (GAGs) were positively influenced by the compressed COL-CNC hydrogel. Our findings shed light on the combined effects of PC and CNCs in improving the physical and mechanical properties of COL and their role in promoting chondrogenesis.


Assuntos
Diferenciação Celular , Celulose , Condrogênese , Colágeno , Hidrogéis , Células-Tronco Mesenquimais , Nanopartículas , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Celulose/química , Celulose/farmacologia , Condrogênese/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Nanopartículas/química , Colágeno/química , Colágeno/farmacologia , Hidrogéis/química , Hidrogéis/farmacologia , Animais , Plásticos/química , Plásticos/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Glicosaminoglicanos/metabolismo , Cartilagem/citologia , Cartilagem/efeitos dos fármacos
3.
Colloids Surf B Biointerfaces ; 239: 113959, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38772085

RESUMO

Cartilage repair remains a major challenge in clinical trials. These current cartilage repair materials can not effectively promote chondrocyte generation, limiting their practical application in cartilage repair. In this work, we develop an implantable scaffold of RADA-16 peptide hydrogel incorporated with TGF-ß1 to provide a microenvironment for stem cell-directed differentiation and chondrocyte adhesion growth. The longest release of growth factor TGF-ß1 release can reach up to 600 h under physiological conditions. TGF-ß1/RADA-16 hydrogel was demonstrated to be a lamellar porous structure. Based on the cell culture with hBMSCs, TGF-ß1/RADA-16 hydrogel showed excellent ability to promote cell proliferation, directed differentiation into chondrocytes, and functional protein secretion. Within 14 days, 80% of hBMSCs were observed to be directed to differentiate into vigorous chondrocytes in the co-culture of TGF-ß1/RADA-16 hydrogels with hBMSCs. Specifically, these newly generated chondrocytes can secrete and accumulate large amounts of collagen II within 28 days, which can effectively promote the formation of cartilage tissue. Finally, the exploration of RADA-16 hydrogel-based scaffolds incorporated with TGF-ß1 bioactive species would further greatly promote the practical clinical trials of cartilage remediation, which might have excellent potential to promote cartilage regeneration in areas of cartilage damage.


Assuntos
Cartilagem , Diferenciação Celular , Condrócitos , Hidrogéis , Regeneração , Alicerces Teciduais , Fator de Crescimento Transformador beta1 , Fator de Crescimento Transformador beta1/metabolismo , Fator de Crescimento Transformador beta1/farmacologia , Regeneração/efeitos dos fármacos , Alicerces Teciduais/química , Hidrogéis/química , Hidrogéis/farmacologia , Humanos , Condrócitos/efeitos dos fármacos , Condrócitos/citologia , Condrócitos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Cartilagem/efeitos dos fármacos , Cartilagem/fisiologia , Cartilagem/metabolismo , Proliferação de Células/efeitos dos fármacos , Engenharia Tecidual/métodos , Células Cultivadas , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Animais , Condrogênese/efeitos dos fármacos , Peptídeos
4.
Mar Drugs ; 22(5)2024 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-38786592

RESUMO

Malnutrition is one of the major factors of bone and cartilage disorders. Pacific cod (Gadus macrocephalus) processing waste is a cheap and highly promising source of bioactive substances, including collagen-derived peptides and amino acids, for bone and cartilage structure stabilization. The addition of these substances to a functional drink is one of the ways to achieve their fast intestinal absorption. Collagen hydrolysate was obtained via enzymatic hydrolysis, ultrafiltration, freeze-drying, and grinding to powder. The lyophilized hydrolysate was a light gray powder with high protein content (>90%), including collagen (about 85% of total protein) and a complete set of essential and non-essential amino acids. The hydrolysate had no observed adverse effect on human mesenchymal stem cell morphology, viability, or proliferation. The hydrolysate was applicable as a protein food supply or a structure-forming food component due to the presence of collagen fiber fragments. An isotonic fitness drink (osmolality 298.1 ± 2.1 mOsm/L) containing hydrolysate and vitamin C as a cofactor in collagen biosynthesis was prepared. The addition of the hydrolysate did not adversely affect its organoleptic parameters. The production of such functional foods and drinks is one of the beneficial ways of fish processing waste utilization.


Assuntos
Osso e Ossos , Cartilagem , Colágeno , Gadiformes , Hidrolisados de Proteína , Animais , Colágeno/metabolismo , Humanos , Cartilagem/efeitos dos fármacos , Cartilagem/metabolismo , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/metabolismo , Hidrolisados de Proteína/farmacologia , Hidrolisados de Proteína/química , Células-Tronco Mesenquimais/efeitos dos fármacos , Bebidas , Alimento Funcional , Hidrólise
5.
Langmuir ; 40(20): 10648-10662, 2024 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-38712915

RESUMO

This study presents new insights into the potential role of polyelectrolyte interfaces in regulating low friction and interstitial fluid pressurization of cartilage. Polymer brushes composed of hydrophilic 3-sulfopropyl methacrylate potassium salt (SPMK) tethered to a PEEK substrate (SPMK-g-PEEK) are a compelling biomimetic solution for interfacing with cartilage, inspired by the natural lubricating biopolyelectrolyte constituents of synovial fluid. These SPMK-g-PEEK surfaces exhibit a hydrated compliant layer approximately 5 µm thick, demonstrating the ability to maintain low friction coefficients (µ ∼ 0.01) across a wide speed range (0.1-200 mm/s) under physiological loads (0.75-1.2 MPa). A novel polyelectrolyte-enhanced tribological rehydration mechanism is elucidated, capable of recovering up to ∼12% cartilage strain and subsequently facilitating cartilage interstitial fluid recovery, under loads ranging from 0.25 to 2.21 MPa. This is attributed to the combined effects of fluid confinement within the contact gap and the enhanced elastohydrodynamic behavior of polymer brushes. Contrary to conventional theories that emphasize interstitial fluid pressurization in regulating cartilage lubrication, this work demonstrates that SPMK-g-PEEK's frictional behavior with cartilage is independent of these factors and provides unabating aqueous lubrication. Polyelectrolyte-enhanced tribological rehydration can occur within a static contact area and operates independently of known mechanisms of cartilage interstitial fluid recovery established for converging or migrating cartilage contacts. These findings challenge existing paradigms, proposing a novel polyelectrolyte-cartilage tribological mechanism not exclusively reliant on interstitial fluid pressurization or cartilage contact geometry. The implications of this research extend to a broader understanding of synovial joint lubrication, offering insights into the development of joint replacement materials that more accurately replicate the natural functionality of cartilage.


Assuntos
Lubrificação , Polímeros , Polímeros/química , Animais , Polieletrólitos/química , Polietilenoglicóis/química , Cartilagem/química , Cartilagem/efeitos dos fármacos , Propriedades de Superfície , Benzofenonas/química , Cartilagem Articular/química , Cartilagem Articular/fisiologia , Cetonas/química
6.
Int J Mol Sci ; 25(9)2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38732122

RESUMO

Osteoarthritis is more prevalent than any other form of arthritis and is characterized by the progressive mechanical deterioration of joints. Glucosamine, an amino monosaccharide, has been used for over fifty years as a dietary supplement to alleviate osteoarthritis-related discomfort. Silibinin, extracted from milk thistle, modifies the degree of glycosylation of target proteins, making it an essential component in the treatment of various diseases. In this study, we aimed to investigate the functional roles of glucosamine and silibinin in cartilage homeostasis using the TC28a2 cell line. Western blots showed that glucosamine suppressed the N-glycosylation of the gp130, EGFR, and N-cadherin proteins. Furthermore, both glucosamine and silibinin differentially decreased and increased target proteins such as gp130, Snail, and KLF4 in TC28a2 cells. We observed that both compounds dose-dependently induced the proliferation of TC28a2 cells. Our MitoSOX and DCFH-DA dye data showed that 1 µM glucosamine suppressed mitochondrial reactive oxygen species (ROS) generation and induced cytosol ROS generation, whereas silibinin induced both mitochondrial and cytosol ROS generation in TC28a2 cells. Our JC-1 data showed that glucosamine increased red aggregates, resulting in an increase in the red/green fluorescence intensity ratio, while all the tested silibinin concentrations increased the green monomers, resulting in decreases in the red/green ratio. We observed increasing subG1 and S populations and decreasing G1 and G2/M populations with increasing amounts of glucosamine, while increasing amounts of silibinin led to increases in subG1, S, and G2/M populations and decreases in G1 populations in TC28a2 cells. MTT data showed that both glucosamine and silibinin induced cytotoxicity in TC28a2 cells in a dose-dependent manner. Regarding endoplasmic reticulum stress, both compounds induced the expression of CHOP and increased the level of p-eIF2α/eIF2α. With respect to O-GlcNAcylation status, glucosamine and silibinin both reduced the levels of O-GlcNAc transferase and hypoxia-inducible factor 1 alpha. Furthermore, we examined proteins and mRNAs related to these processes. In summary, our findings demonstrated that these compounds differentially modulated cellular proliferation, mitochondrial and cytosol ROS generation, the mitochondrial membrane potential, the cell cycle profile, and autophagy. Therefore, we conclude that glucosamine and silibinin not only mediate glycosylation modifications but also regulate cellular processes in human chondrocytes.


Assuntos
Condrócitos , Glucosamina , Homeostase , Fator 4 Semelhante a Kruppel , Espécies Reativas de Oxigênio , Silibina , Glucosamina/farmacologia , Glucosamina/metabolismo , Humanos , Silibina/farmacologia , Glicosilação/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Espécies Reativas de Oxigênio/metabolismo , Fator 4 Semelhante a Kruppel/metabolismo , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Mitocôndrias/metabolismo , Mitocôndrias/efeitos dos fármacos , Cartilagem/metabolismo , Cartilagem/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Osteoartrite/metabolismo , Osteoartrite/tratamento farmacológico
7.
Med Sci Monit ; 30: e943738, 2024 Apr 26.
Artigo em Inglês | MEDLINE | ID: mdl-38664941

RESUMO

BACKGROUND The pathological mechanism of osteoarthritis is still unclear. The regulation of the immune microenvironment has been of growing interest in the progression and treatment of osteoarthritis. Macrophages with different phenotypes, producing different cytokines, have been linked to the mechanism of cartilage injury in osteoarthritis. Copper ions play a role in the immune response and are involved in the pathological mechanisms of osteoarthritis by affecting the metabolism of the cartilage matrix. Bioactive glass (BG) is an osteogenic material with superior biocompatibility. Here, we report on the regulatory behavior of macrophages using a copper-based composite BG material. MATERIAL AND METHODS Cu-BGC powder was prepared by sol-gel method, and scaffolds were fabricated and characterized using 3D printing. Macrophage cultures grown with Cu-BGC were examined for cell culture and proliferation. The effect of Cu-BGC on the degradation metabolism of chondrocytes, cultured in the environment of inflammatory cytokine IL-1ß, was determined. In addition, the morphology of macrophages, secretion of inflammatory cytokines, and expression of surface markers were examined. RESULTS The results show that Cu-BGC promotes macrophage proliferation at a range of concentrations and increases the secretion of anti-inflammatory cytokines while inhibiting proinflammatory cytokines. At the same time, M2-type cell surface markers are definitely expressed and the morphology of macrophages is altered. In addition, Cu-BGC inhibited the degradation metabolism of chondrocytes in the inflammatory environment induced by IL-1ß. CONCLUSIONS These results suggest that Cu-BGC induced macrophage polarization into an M2 type anti-inflammatory phenotype, and inhibition of immune injury response may play a role in delaying cartilage matrix damage in osteoarthritis.


Assuntos
Proliferação de Células , Condrócitos , Cobre , Citocinas , Macrófagos , Osteoartrite , Macrófagos/metabolismo , Macrófagos/efeitos dos fármacos , Osteoartrite/patologia , Osteoartrite/metabolismo , Animais , Condrócitos/metabolismo , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Cobre/metabolismo , Cobre/farmacologia , Citocinas/metabolismo , Camundongos , Proliferação de Células/efeitos dos fármacos , Cartilagem Articular/patologia , Cartilagem Articular/efeitos dos fármacos , Cartilagem Articular/metabolismo , Cartilagem/metabolismo , Cartilagem/efeitos dos fármacos , Cartilagem/patologia , Células RAW 264.7 , Vidro , Alicerces Teciduais
8.
J Cell Physiol ; 239(5): e31224, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38481029

RESUMO

With the prevalence of coronavirus disease 2019, the administration of glucocorticoids (GCs) has become more widespread. Treatment with high-dose GCs leads to a variety of problems, of which steroid-induced osteonecrosis of the femoral head (SONFH) is the most concerning. Since hypoxia-inducible factor 1α (HIF-1α) is a key factor in cartilage development and homeostasis, it may play an important role in the development of SONFH. In this study, SONFH models were established using methylprednisolone (MPS) in mouse and its proliferating chondrocytes to investigate the role of HIF-1α in cartilage differentiation, extracellular matrix (ECM) homeostasis, apoptosis and glycolysis in SONFH mice. The results showed that MPS successfully induced SONFH in vivo and vitro, and MPS-treated cartilage and chondrocytes demonstrated disturbed ECM homeostasis, significantly increased chondrocyte apoptosis rate and glycolysis level. However, compared with normal mice, not only the expression of genes related to collagens and glycolysis, but also chondrocyte apoptosis did not demonstrate significant differences in mice co-treated with MPS and HIF-1α inhibitor. And the effects observed in HIF-1α activator-treated chondrocytes were similar to those induced by MPS. And HIF-1α degraded collagens in cartilage by upregulating its downstream target genes matrix metalloproteinases. The results of activator/inhibitor of endoplasmic reticulum stress (ERS) pathway revealed that the high apoptosis rate induced by MPS was related to the ERS pathway, which was also affected by HIF-1α. Furthermore, HIF-1α affected glucose metabolism in cartilage by increasing the expression of glycolysis-related genes. In conclusion, HIF-1α plays a vital role in the pathogenesis of SONFH by regulating ECM homeostasis, chondrocyte apoptosis, and glycolysis.


Assuntos
Apoptose , Cartilagem , Condrócitos , Glucocorticoides , Glicólise , Homeostase , Subunidade alfa do Fator 1 Induzível por Hipóxia , Metilprednisolona , Animais , Masculino , Camundongos , Apoptose/efeitos dos fármacos , Cartilagem/metabolismo , Cartilagem/patologia , Cartilagem/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Cabeça do Fêmur/patologia , Cabeça do Fêmur/metabolismo , Necrose da Cabeça do Fêmur/induzido quimicamente , Necrose da Cabeça do Fêmur/patologia , Necrose da Cabeça do Fêmur/metabolismo , Necrose da Cabeça do Fêmur/genética , Glucocorticoides/efeitos adversos , Glucocorticoides/farmacologia , Glicólise/efeitos dos fármacos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Metilprednisolona/efeitos adversos , Metilprednisolona/farmacologia , Camundongos Endogâmicos C57BL
9.
Int J Biol Macromol ; 266(Pt 2): 131051, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38556223

RESUMO

In situ-forming hydrogels that possess the ability to be injected in a less invasive manner and mimic the biochemical composition and microarchitecture of the native cartilage extracellular matrix are desired for cartilage tissue engineering. Besides, gelation time and stiffness of the hydrogel are two interdependent factors that affect cells' distribution and fate and hence need to be optimized. This study presented a bioinspired in situ-forming hydrogel composite of hyaluronic acid (HA), chondroitin sulfate (CS), and collagen short nanofiber (CSNF). HA and CS were functionalized with aldehyde and amine groups to form a gel through a Schiff-base reaction. CSNF was fabricated via electrospinning, followed by fragmentation by ultrasonics. Gelation time (11-360 s) and compressive modulus (1.4-16.2 kPa) were obtained by varying the concentrations of CS, HA, CSNFs, and CSNFs length. The biodegradability and biocompatibility of the hydrogels with varying gelation and stiffness were also assessed in vitro and in vivo. At three weeks, the assessment of hydrogels' chondrogenic differentiation also yields varying levels of chondrogenic differentiation. The subcutaneous implantation of the hydrogels in a mouse model indicated no severe inflammation. Results demonstrated that the injectable CS/HA@CSNF hydrogel was a promising hydrogel for tissue engineering and cartilage regeneration.


Assuntos
Sulfatos de Condroitina , Colágeno , Ácido Hialurônico , Hidrogéis , Nanocompostos , Nanofibras , Sulfatos de Condroitina/química , Sulfatos de Condroitina/farmacologia , Ácido Hialurônico/química , Ácido Hialurônico/farmacologia , Nanofibras/química , Animais , Hidrogéis/química , Hidrogéis/farmacologia , Camundongos , Colágeno/química , Nanocompostos/química , Engenharia Tecidual/métodos , Cartilagem/efeitos dos fármacos , Condrogênese/efeitos dos fármacos , Diferenciação Celular/efeitos dos fármacos , Alicerces Teciduais/química , Materiais Biocompatíveis/química , Materiais Biocompatíveis/farmacologia
10.
Small ; 20(25): e2309597, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38279613

RESUMO

Osteoarthritis (OA) is a dynamic condition characterized by cartilage damage and synovial inflammation. Ozone (O3) shows potential therapeutic effects owing to its anti-inflammatory properties; however, its high reactivity and short half-life substantially limit its effectiveness in OA treatment. In this study, an ozone-rich thermosensitive nanocomposite hydrogel loaded with D-mannose is developed for OA treatment. Briefly, O3 is encapsulated in nanoparticles (NPs) composed of perfluorotributylamine and fluorinated hyaluronic acid to improve its stability. Next, D-mannose is conjugated with α-amino of the hydroxypropyl chitin (HPCH) via Schiff base to prepare MHPCH. These nanoparticles are encapsulated in MHPCH to produce O3 NPs@MHPCH. In vitro cell experiments demonstrate that the O3 NPs@MHPCH treatment significantly reduced VEGF and inflammation levels, accompanied by a decrease in inflammatory factors such as IL-1ß, IL-6, TNF-α, and iNOS. Furthermore, O3 NPs@MHPCH promotes the expression of collagen II and aggrecan and stimulates chondrocyte proliferation. Additionally, in vivo studies show that O3 NPs@MHPCH significantly alleviated OA by reducing synovial inflammation, cartilage destruction, and subchondral bone remodeling. O3 NPs@MHPCH offers a promising option for improving the efficacy of O3 therapy and reducing the risk of synovial inflammation and cartilage degeneration in OA.


Assuntos
Anti-Inflamatórios , Hidrogéis , Manose , Nanocompostos , Osteoartrite , Ozônio , Nanocompostos/química , Osteoartrite/tratamento farmacológico , Osteoartrite/patologia , Animais , Ozônio/química , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/química , Anti-Inflamatórios/uso terapêutico , Hidrogéis/química , Manose/química , Cartilagem/efeitos dos fármacos , Cartilagem/patologia , Camundongos , Masculino , Injeções , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo
11.
Jt Dis Relat Surg ; 35(1): 156-168, 2024 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-38108177

RESUMO

OBJECTIVES: In this study, we aimed to determine the bioefficacy of epidermal growth factor (EGF), boric acid (BA), and their combination on cartilage injury in rats. MATERIALS AND METHODS: In in vitro setting, the cytotoxic effects of BA, EGF, and their combinations using mouse fibroblast cell (L929), human bone osteosarcoma cell (Saos-2), and human adipose derived mesenchymal stem cells (hAD-MSCs) were determined by applying MTT [3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide] test. In in vivo setting, 72 rats were randomly divided into four groups. A standard chondral defect was created and microfracture was performed in all groups. Group A was determined as the control group. In addition to the standard procedure, Group B received 100 ng/mL of EGF, Group C received a combination of 100 ng/mL of EGF and 10 µg/mL of BA combination, and Group D 20 µg/mL of BA. RESULTS: The cytotoxic effect of the combinations of EGF dilutions (1, 5, 10, 25, 50, 100, 200 ng/mL) with BA (100, 300, 500 µg/mL) was observed only in the 72-h application period and in Saos-2. The cytotoxic effect of BA was reduced when combined with EGF. There was no significant difference in the histopathological scores among the groups (p=0.13). CONCLUSION: Our study showed that EGF and low-dose BA application had a positive effect on cartilage healing in rats. Significant decreases in recovery scores were observed in the other groups. The combination of EGF and BA promoted osteoblast growth. Detection of lytic lesions in the group treated with 20 µg/mL of BA indicates that BA may have a cytotoxic effect.


Assuntos
Ácidos Bóricos , Cartilagem , Fator de Crescimento Epidérmico , Animais , Humanos , Camundongos , Ratos , Ácidos Bóricos/farmacologia , Ácidos Bóricos/uso terapêutico , Cartilagem/efeitos dos fármacos , Cartilagem/lesões , Fator de Crescimento Epidérmico/farmacologia , Fator de Crescimento Epidérmico/uso terapêutico , Fator de Crescimento Epidérmico/metabolismo , Linhagem Celular
12.
Chem Biol Interact ; 388: 110835, 2024 Jan 25.
Artigo em Inglês | MEDLINE | ID: mdl-38122922

RESUMO

Osteoarthritis (OA) is a common joint degenerative disease, and chondrocyte injury is the main pathological and physiological change. Ruscogenin (Rus), a bioactive compound isolated from Radix Ophiopogon japonicus, exhibits various pharmacological effects. The aim of this research was to test the role and mechanism of Rus on OA both in vivo and in vitro. Destabilized medial meniscus (DMM)-induced OA model was established in vivo and IL-1ß-stimulated mouse chondrocytes was used to explore the role of Rus on OA in vitro. In vivo, Rus exhibited protective effects against DMM-induced OA model. Rus could inhibit MMP1 and MMP3 expression in OA mice. In vitro, IL-1ß-induced inflammation and degradation of extracellular matrix were inhibited by Rus, as confirmed by the inhibition of PGE2, NO, MMP1, and MMP3 by Rus. Also, IL-1ß-induced ferroptosis was suppressed by Rus, as confirmed by the inhibition of MDA, iron, and ROS, as well as the upregulation of GSH, GPX4, Ferritin, Nrf2, and SLC7A11 expression induced by Rus. Furthermore, the suppression of Rus on IL-1ß-induced inflammation, MMPs production, and ferroptosis were reversed when Nrf2 was knockdown. In conclusion, Rus attenuated OA progression through inhibiting chondrocyte ferroptosis via Nrf2/SLC7A11/GPX4 signaling pathway.


Assuntos
Ferroptose , Osteoartrite , Espirostanos , Animais , Camundongos , Cartilagem/efeitos dos fármacos , Cartilagem/metabolismo , Cartilagem/patologia , Condrócitos/efeitos dos fármacos , Ferroptose/efeitos dos fármacos , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Metaloproteinase 1 da Matriz/metabolismo , Metaloproteinase 3 da Matriz/genética , Metaloproteinase 3 da Matriz/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Transdução de Sinais
13.
JCI Insight ; 7(20)2022 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-36194485

RESUMO

No disease-modifying drug exists for osteoarthritis (OA). Despite success in animal models, candidate drugs continue to fail in clinical trials owing to the unmapped interpatient heterogeneity and disease complexity. We used a single-cell platform based on cytometry by time-of-flight (cyTOF) to precisely outline the effects of candidate drugs on human OA chondrocytes. OA chondrocytes harvested from patients undergoing total knee arthroplasty were treated with 2 drugs, an NF-κB pathway inhibitor, BMS-345541, and a chondroinductive small molecule, kartogenin, that showed preclinical success in animal models for OA. cyTOF conducted with 30 metal isotope-labeled antibodies parsed the effects of the drugs on inflammatory, senescent, and chondroprogenitor cell populations. The NF-κB pathway inhibition decreased the expression of p-NF-κB, HIF2A, and inducible NOS in multiple chondrocyte clusters and significantly depleted 4 p16ink4a-expressing senescent populations, including NOTCH1+STRO1+ chondroprogenitor cells. While kartogenin also affected select p16ink4a-expressing senescent clusters, there was a less discernible effect on chondroprogenitor cell populations. Overall, BMS-345541 elicited a uniform drug response in all patients, while only a few responded to kartogenin. These studies demonstrate that a single-cell cyTOF-based drug screening platform can provide insights into patient response assessment and patient stratification.


Assuntos
Cartilagem , Avaliação Pré-Clínica de Medicamentos , Osteoartrite , Humanos , Cartilagem/efeitos dos fármacos , Cartilagem/metabolismo , Avaliação Pré-Clínica de Medicamentos/métodos , Homeostase/efeitos dos fármacos , NF-kappa B/metabolismo , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Transdução de Sinais , Análise de Célula Única/instrumentação , Análise de Célula Única/métodos
14.
J. oral res. (Impresa) ; 11(5): 1-16, nov. 23, 2022. ilus, tab
Artigo em Inglês | LILACS | ID: biblio-1437225

RESUMO

Background: Endocrine Disrupting Chemicals (EDCs) would cause alterations in organs/systems of exposed individuals or their progeny. Objetive: To identify and analyze the main published findings on the effects of exposure to EDCs on teeth, cartilage, and bone. Material and Methods: Two databases were analyzed: Medline and Web of Science. Only observational studies analyzing the effect of EDCs on mineralized tissues published since 2006 were included in the study. Results: 25 articles were selected, most of them involving EDCs pesticides, plasticizers, or personal care products, highlighting organochlorine compounds, bisphenols, phthalates, dioxins, parabens, and perfluoroalkyls. Thirty-six per cent of the studies reported an accumulation of EDCs in teeth or bones, while 64% reported alterations in their development or morphology, mainly at the bone level, primarily affecting their mineral density and size, as well as that of the bones of exposed individuals or their progeny. The type of effect observed was related to the EDCs analyzed, and it seemed to depend on variables such as age, sex, ethnicity/race, and even the metabolic status of the individuals in the different species analyzed. No evidence associated with effects on cartilage was found. Conclusion: EDCs in the environment, at work, or at home, under different exposure routes, are capable of accumulating in teeth and bone, particularly affecting the latter. It is necessary to study the effect of EDCs on mineralized tissues in agro-industrial areas, especially on teeth.


Antecedentes: Los Químicos Disruptores Endocrinos (EDCs) causarían alteraciones en órganos/sistemas de individuos expuestos, o su progenie. Objetivo: Identificar y analizar los principales hallazgos publicados sobre el efecto de la exposición a EDCs en dientes, cartílago y hueso. Material y Métodos: Se analizaron dos bases de datos: Medline y Web of Science, incluyendo solo estudios observacionales publicados desde el 2006, analizando el efecto de los EDCs sobre tejidos mineralizados. Resultados:25 artículos fueron seleccionados, siendo la mayoría de los EDCs pesticidas, plastificantes o productos de cuidado personal, destacando los compuestos Organo-clorados, Bisfenoles, Ftalatos, Dioxinas, Parabenos y los Perfluoroalquilos. Un 36% de los estudios reportaron un acúmulo de EDCs en dientes o huesos, mientras que un 64% informaron de alteraciones en su desarrollo o morfología, particularmente a nivel de huesos, afectando principalmente su densidad mineral y su tamaño, así como el de los individuos expuestos o su progenie. El tipo de efecto observado tuvo relación con el EDCs analizado, pareciendo depender de variables tales como edad, sexo, etnia/raza e incluso el estado metabólico de los individuos, en las diferentes especies analizadas. No se encontraron evidencias asociadas a efectos en el cartílago. Conclusión: Los EDCs en el medio ambiente, ámbito laboral o doméstico, bajo distintas rutas de exposición, son capaces de acumularse en diente y hueso, afectando particularmente a este último. Es necesario estudiar el efecto de los EDCs en los tejidos mineralizados en zonas agroindustriales, particularmente a nivel de dientes.


Assuntos
Humanos , Dente/efeitos dos fármacos , Osso e Ossos/efeitos dos fármacos , Cartilagem/efeitos dos fármacos , Disruptores Endócrinos/toxicidade , Fluorocarbonos , Bioacumulação
15.
BMC Complement Med Ther ; 22(1): 25, 2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35086536

RESUMO

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


Assuntos
Cartilagem/efeitos dos fármacos , Glucosídeos/farmacologia , Inflamação/metabolismo , Interleucina-1beta/metabolismo , Isoflavonas/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Osteoartrite , Animais , Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Cartilagem/citologia , Cartilagem/metabolismo , Cartilagem/patologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Regulação para Baixo , Glucosídeos/uso terapêutico , Inflamação/tratamento farmacológico , Isoflavonas/uso terapêutico , Sistema de Sinalização das MAP Quinases , Masculino , Metaloproteinase 13 da Matriz/metabolismo , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Osteoartrite/patologia , Fitoterapia , Extratos Vegetais/farmacologia , Extratos Vegetais/uso terapêutico , Ratos Sprague-Dawley , Transdução de Sinais , Fator de Necrose Tumoral alfa/metabolismo
16.
Sci China Life Sci ; 65(2): 309-327, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34109475

RESUMO

Significant cellular senescence has been observed in cartilage harvested from patients with osteoarthritis (OA). In this study, we aim to develop a senescence-relevant OA-like cartilage model for developing disease-modifying OA drugs (DMOADs). Specifically, human bone marrow-derived mesenchymal stromal cells (MSCs) were expanded in vitro up to passage 10 (P10-MSCs). Following their senescent phenotype formation, P10-MSCs were subjected to pellet culture in chondrogenic medium. Results from qRT-PCR, histology, and immunostaining indicated that cartilage generated from P10-MSCs displayed both senescent and OA-like phenotypes without using other OA-inducing agents, when compared to that from normal passage 4 (P4)-MSCs. Interestingly, the same gene expression differences observed between P4-MSCs and P10-MSC-derived cartilage tissues were also observed between the preserved and damaged OA cartilage regions taken from human samples, as demonstrated by RNA Sequencing data and other analysis methods. Lastly, the utility of this senescence-initiated OA-like cartilage model in drug development was assessed by testing several potential DMOADs and senolytics. The results suggest that pre-existing cellular senescence can induce the generation of OA-like changes in cartilage. The P4- and P10-MSCs derived cartilage models also represent a novel platform for predicting the efficacy and toxicity of potential DMOADs on both preserved and damaged cartilage in humans.


Assuntos
Antirreumáticos/farmacologia , Cartilagem/efeitos dos fármacos , Senescência Celular/efeitos dos fármacos , Células-Tronco Mesenquimais/citologia , Modelos Biológicos , Osteoartrite/tratamento farmacológico , Antirreumáticos/uso terapêutico , Cartilagem/metabolismo , Cartilagem/patologia , Diferenciação Celular , Células Cultivadas , Senescência Celular/genética , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Células-Tronco Mesenquimais/metabolismo , Osteoartrite/metabolismo , Osteoartrite/patologia , Senoterapia/farmacologia , Engenharia Tecidual , Transcriptoma
17.
Sci Rep ; 11(1): 23120, 2021 11 30.
Artigo em Inglês | MEDLINE | ID: mdl-34848799

RESUMO

The pathophysiology of osteoarthritis (OA) includes the destruction of subchondral bone tissue and inflammation of the synovium. Thus, an effective disease-modifying treatment should act on both of these pathogenetic components. It is known that cSrc kinase is involved in bone and cartilage remodeling, and SYK kinase is associated with the inflammatory component. Thus the aim of this study was to characterize the mechanism of action and efficacy of a small molecule multikinase inhibitor MT-SYK-03 targeting SYK and cSrc kinases among others in different in vitro and in vivo arthritis models. The selectivity of MT-SYK-03 kinase inhibition was assayed on a panel of 341 kinases. The compound was evaluated in a set of in vitro models of OA and in vivo OA and RA models: surgically-induced arthritis (SIA), monosodium iodoacetate-induced arthritis (MIA), collagen-induced arthritis (CIA), adjuvant-induced arthritis (AIA). MT-SYK-03 inhibited cSrc and SYK with IC50 of 14.2 and 23 nM respectively. Only five kinases were inhibited > 90% at 500 nM of MT-SYK-03. In in vitro OA models MT-SYK-03 reduced hypertrophic changes of chondrocytes, bone resorption, and inhibited SYK-mediated inflammatory signaling. MT-SYK-03 showed preferential distribution to joint and bone tissue (in rats) and revealed disease-modifying activity in vivo by halving the depth of cartilage erosion in rat SIA model, and increasing the pain threshold in rat MIA model. Chondroprotective and antiresorptive effects were shown in a monotherapy regime and in combination with methotrexate (MTX) in murine and rat CIA models; an immune-mediated inflammation in rat AIA model was decreased. The obtained preclinical data support inhibition of cSrc and SYK as a viable strategy for disease-modifying treatment of OA. A Phase 2 clinical study of MT-SYK-03 is to be started.


Assuntos
Artrite Reumatoide/tratamento farmacológico , Artrite Reumatoide/enzimologia , Osso e Ossos/efeitos dos fármacos , Proteína Tirosina Quinase CSK/antagonistas & inibidores , Cartilagem/efeitos dos fármacos , Osteoartrite/tratamento farmacológico , Osteoartrite/enzimologia , Quinase Syk/antagonistas & inibidores , Animais , Artrite Experimental/patologia , Reabsorção Óssea/patologia , Condrócitos/patologia , Modelos Animais de Doenças , Avaliação Pré-Clínica de Medicamentos , Inibidores Enzimáticos/farmacologia , Humanos , Inflamação , Concentração Inibidora 50 , Ácido Iodoacético/farmacologia , Receptores de Lipopolissacarídeos/biossíntese , Masculino , Camundongos , Monócitos/citologia , Substâncias Protetoras/farmacologia , Coelhos , Ratos , Ratos Sprague-Dawley , Ratos Wistar , Membrana Sinovial/patologia
18.
Cells ; 10(12)2021 12 17.
Artigo em Inglês | MEDLINE | ID: mdl-34944076

RESUMO

Hydrogels are commonly used for the 3D culture of musculoskeletal cells. Sulfated hydrogels, which have seen a growing interest over the past years, provide a microenvironment that help maintain the phenotype of chondrocytes and chondrocyte-like cells and can be used for sustained delivery of growth factors and other drugs. Sulfated hydrogels are hence valuable tools to improve cartilage and intervertebral disc tissue engineering. To further advance the utilization of these hydrogels, we identify and summarize the current knowledge about different sulfated hydrogels, highlight their beneficial effects in cartilage and disc research, and review the biofabrication processes most suitable to secure best quality assurance through deposition fidelity, repeatability, and attainment of biocompatible morphologies.


Assuntos
Cartilagem/efeitos dos fármacos , Hidrogéis/farmacologia , Disco Intervertebral/efeitos dos fármacos , Pesquisa , Sulfatos/farmacologia , Animais , Humanos , Hidrogéis/química , Sulfatos/química , Engenharia Tecidual
19.
Oxid Med Cell Longev ; 2021: 4139048, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34777685

RESUMO

Osteoarthritis (OA), characterized by chronic systemic low-level inflammation and cartilage degeneration, is a type of arthritis closely associated with aging. Inflammation and aging play a pivotal role in the occurrence and progression of OA. NLRP3 inflammasome is involved in many inflammatory and aging diseases, and NLRP3 inhibitor MCC950 has anti-inflammatory and antisenescence effects on some diseases such as Alzheimer's disease. In the present study, we found that NLRP3 protein was upregulated in human and mouse OA cartilage. Moreover, NLRP3 and Caspase1 expression induced by IL-1ß in chondrocytes was blocked by MCC950. In addition, MCC950 inhibited the expression of inflammatory mediators, matrix-degrading enzymes, senescence marker protein P16 (INK4A), and ß-galactosidase, as well as excessive production of ROS. Meanwhile, MCC950 promoted autophagy-related protein expression and autophagy flux under the inflammatory condition. However, autophagy inhibitor 3-MA reversed anti-inflammatory and anticatabolic effects of MCC950. In in vivo experiments, intra-articular administration of MCC950 further showed its protective effect on cartilage degeneration. Bioinformatic analysis and in vitro experimental results revealed that MCC950 might play a protective role in cartilage by regulating Nrf2/HO-1/NQO1, PI3k/Akt/mTOR, P38/MAPK, and JNK/MAPK pathways. In conclusion, our work demonstrated that NLRP3 inhibitor MCC950 might serve as a promising strategy for OA treatment.


Assuntos
Cartilagem/efeitos dos fármacos , Modelos Animais de Doenças , Furanos/farmacologia , Indenos/farmacologia , Inflamação/prevenção & controle , Proteína 3 que Contém Domínio de Pirina da Família NLR/antagonistas & inibidores , Osteoartrite/complicações , Substâncias Protetoras/farmacologia , Sulfonamidas/farmacologia , Animais , Anti-Inflamatórios/farmacologia , Cartilagem/metabolismo , Cartilagem/patologia , Inflamação/etiologia , Inflamação/metabolismo , Inflamação/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL
20.
J Nanobiotechnology ; 19(1): 343, 2021 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-34702302

RESUMO

OBJECTIVES: This study aimed to investigate the roles of adipose mesenchymal stem cell (AMSC)-derived extracellular vesicles (EVs) binding with chitosan oligosaccharides (COS) in cartilage injury, as well as the related mechanisms. RESULTS: IL-1ß treatment significantly inhibited the viability and migration of chondrocytes and enhanced cell apoptosis (P < 0.05), while chitosan oligosaccharides and extracellular vesicles-chitosan oligosaccharide conjugates (EVs-COS/EVs-COS conjugates) reversed the changes induced by IL-1ß (P < 0.05), and the effects of extracellular vesicles-chitosan oligosaccharide conjugates were better than those of chitosan oligosaccharides (P < 0.05). After cartilage damage, IL-1ß, OPN, and p53 were significantly upregulated, COL1A1, COL2A1, OCN, RUNX2, p-Akt/Akt, PI3K, c-Myc, and Bcl2 were markedly downregulated, and extracellular vesicles-chitosan oligosaccharide conjugates reversed the expression induced by cartilage injury. Through sequencing, 760 differentially expressed genes (DEGs) clustered into four expression patterns were associated with negative regulation of the canonical Wnt, PI3K-Akt, AMPK, and MAPK signaling pathways. CONCLUSION: Extracellular vesicles-chitosan oligosaccharide conjugates may serve as a new cell-free biomaterial to facilitate cartilage injury repair and improve osteoarthritis.


Assuntos
Cartilagem , Quitosana , Transplante de Células-Tronco Mesenquimais , Células-Tronco Mesenquimais , Animais , Cartilagem/efeitos dos fármacos , Cartilagem/lesões , Cartilagem/metabolismo , Células Cultivadas , Quitosana/química , Quitosana/farmacologia , Condrócitos/citologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Vesículas Extracelulares/química , Feminino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/metabolismo , Oligossacarídeos/química , Oligossacarídeos/farmacologia , Osteoartrite/metabolismo , Ratos , Ratos Wistar , Organismos Livres de Patógenos Específicos , Transcriptoma/genética , Cicatrização/efeitos dos fármacos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...