Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros











Intervalo de ano de publicação
1.
ACS Appl Mater Interfaces ; 16(38): 50484-50496, 2024 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-39282962

RESUMO

Characterized by progressive and irreversible degeneration of the articular cartilage (AC), osteoarthritis (OA) is the most common chronic joint disease, and there is no cure for OA at present. Recent studies suggest that enhancing the recruitment of endogenous mesenchymal stem cells (MSCs) to damaged cartilage is a promising therapeutic strategy for cartilage repair. Tetrahedral framework nucleic acid (tFNA) is a novel DNA nanomaterial and has shown great potential in the field of biomedical science. Transforming growth factor-beta 3 (TGF-ß3), a vital member of the highly conserved TGF-ß superfamily, is considered to induce chondrogenesis. A 66-base DNA aptamer named HM69 is reported to identify and recruit MSCs. In this study, aptamer HM69-modified tFNAs were successfully self-assembled and used to load TGF-ß3 when the disulfide bonds combined. We confirmed the successful synthesis of the final composition, HM69-tFNA@TGF-ß3 (HTT), by PAGE, dynamic light scattering, and atomic force microscopy. The results of in vitro experiments showed that HTT effectively induced MSC proliferation, migration, and chondrogenic differentiation. In addition, HTT-treated MSCs were shown to protect the OA chondrocytes. In DMM mice, the injection of HTT improved the therapeutic outcome of mouse pain symptoms and AC degeneration. In conclusion, this study innovatively used the disulfide bonds combined with TGF-ß3 and tFNA, and an additional sequence HM69 was loaded on tFNA for the better-targeted recruitment of MSCs. HTT demonstrated its role in promoting the chondrogenesis of MSCs and cartilage protection, indicating that it might be promising for OA therapy.


Assuntos
Aptâmeros de Nucleotídeos , Diferenciação Celular , Condrogênese , Células-Tronco Mesenquimais , Osteoartrite , Fator de Crescimento Transformador beta3 , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Células-Tronco Mesenquimais/citologia , Fator de Crescimento Transformador beta3/farmacologia , Fator de Crescimento Transformador beta3/química , Fator de Crescimento Transformador beta3/metabolismo , Osteoartrite/patologia , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Animais , Condrogênese/efeitos dos fármacos , Camundongos , Diferenciação Celular/efeitos dos fármacos , Aptâmeros de Nucleotídeos/química , Aptâmeros de Nucleotídeos/farmacologia , Humanos , Cartilagem Articular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/citologia , Proliferação de Células/efeitos dos fármacos
2.
Small Methods ; : e2400660, 2024 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-39036830

RESUMO

Osteoarthritis (OA) is a degenerative whole-joint disease in which the synovium and joint cartilage become inflamed and damaged. The essential role of inflammation in the development of OA has been recognized recently. Accordingly, simultaneous regulation of local inflammation and tissue degeneration is proposed as a promising therapeutic strategy. Herein, multifunctional biomimetic apoptotic nanovesicles (Apo-NVs) are constructed with plasma membrane derived from apoptotic T cells. The anti-inflammatory microRNA-124 is further encapsulated into Apo-NVs in the hope of achieving an enhanced immunomodulatory effect. It is found that apoptotic nanovesicles, including Apo-NVs and Apo-NVs-miR-124, both efficiently promote the M2 repolarization of M1 macrophages and inhibit the degenerative phenotype of chondrocytes. Further in vivo studies show that Apo-NVs and Apo-NVs-miR-124 alleviate synovial inflammation and protect cartilage tissue from degeneration in OA mice. The study highlights the potential of Apo-NVs in treating OA and other inflammation-related diseases.

3.
Phytomedicine ; 129: 155593, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38621329

RESUMO

BACKGROUND: Preventing joint edema is crucial in halting osteoarthritis (OA) progression. Growing clinical evidence indicate that Jianpi-Tongluo Formula (JTF) may have a promising anti-edema effect. However, the therapeutic properties of JTF and the underlying mechanisms remains unclear. MATERIALS AND METHODS: An OA rat model was established and employed to evaluate pharmacological effects of JTF in vivo based on dynamic histopathologic assessments and micro-CT observations. Then, OA-related genes and potential targets of JTF were identified through clinical transcriptomic data analysis and "disease gene-drug target" network analysis, which were verified by a series of in vivo experiments. RESULTS: JTF administration effectively reduced pain and joint edema, inhibited matrix degradation, chondrocyte apoptosis, and aquaporin expression in OA rats. Notably, JTF dose-dependently reversed damage-associated molecular patterns and inflammatory factor upregulation. Mechanically, our "disease gene-drug target" network analysis indicated that the NCOA4-HMGB1-GSK3B-AQPs axis, implicated in ferroptosis and aquaporin dysregulation, may be potentially served as a target of JTF against OA. Accordingly, JTF mitigated NCOA4, HMGB1, and GSK3B expression, oxidative stress, and iron metabolism aberrations in OA rats. Furthermore, JTF treatment significantly attenuated the aberrant upregulation of AQP1, AQP3, and AQP4 proteins observed in cartilage tissues of OA rats. CONCLUSION: Our data reveal for the first time that JTF may exert cartilage protective and anti-edema effects in osteoarthritis therapy by inhibiting NCOA4-HMGB1-driven ferroptosis and aquaporin dysregulation.


Assuntos
Ferroptose , Proteína HMGB1 , Osteoartrite , Ratos Sprague-Dawley , Animais , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Ferroptose/efeitos dos fármacos , Ratos , Masculino , Proteína HMGB1/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Edema/tratamento farmacológico , Aquaporinas/metabolismo , Coativadores de Receptor Nuclear/metabolismo , Modelos Animais de Doenças , Aquaporina 3/metabolismo , Aquaporina 1/metabolismo
4.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-1021673

RESUMO

BACKGROUND:Swimming is an important non-pharmacological treatment for knee osteoarthritis,which can effectively alleviate the disease.However,the effect and mechanism of swimming on senile knee osteoarthritis are still unclear. OBJECTIVE:To investigate the effect of swimming exercise on the articular cartilage of aged mice with knee osteoarthritis. METHODS:Six 3-month-old male C57BL/6 mice were selected as the young group,and twelve 18-month-old male C57BL/6 mice were randomized into the aged group and the swimming group,with six mice in each group.Mice in the swimming group received adaptive swimming for 1 week and formal swimming for 8 weeks.After the intervention,stride length analysis and sampling were performed in each group.The total number of leukocytes and lymphocytes in peripheral blood was detected by blood routine examinations.The morphology of the articular cartilage was observed by hematoxylin-eosin and safranin O-fast green staining.Chondrocyte counts and the modified Mankin's score were used to evaluate the degree of articular cartilage damage.The protein and mRNA expressions of type Ⅱ collagen,aggrecan and matrix metalloproteinase 13 in articular cartilage were detected by immunohistochemical staining and RT-qPCR. RESULTS AND CONCLUSION:Compared with the young group,the mice in the aged group showed significantly decreased stride length(P<0.05),significantly increased numbers of peripheral leukocytes and lymphocytes(P<0.05),significantly decreased count of chondrocytes(P<0.05),significantly increased modified Mankin's score(P<0.05),significantly decreased protein and mRNA expression of type Ⅱ collagen and aggreca(P<0.05),and significantly increased matrix metalloproteinase 13 expression(P<0.05).Moreover,hematoxylin-eosin and safranin O-fast green staining showed the uneven surface of the articular cartilage,abnormal chondrocytes,and proteoglycan loss in the aged group.Compared with the aged group,swimming exercise significantly improved the stride length of mice(P<0.05),decreased the count of peripheral blood lymphocytes(P<0.05),increased the count of chondrocytes(P<0.05),decreased the modified Mankin's score(P<0.05),increased the protein and mRNA expression of type Ⅱ collagen and aggrecan(P<0.05),and decreased the expression of matrix metalloproteinase 13(P<0.05).Hematoxylin-eosin and safranin O-fast green staining showed that the articular surface of mice in the swimming group was smooth,chondrocytes were normal,and proteoglycan loss was less.All these findings indicate that swimming exercise can reduce the number of inflammatory cells in the blood of aged mice,improve articular chondrocytes,matrix composition and cartilage tissue morphology;thus,it has a protective effect on the cartilage of aged mice with knee osteoarthritis.

5.
Acta Pharm Sin B ; 13(12): 5016-5029, 2023 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-38045057

RESUMO

Normalizing inflamed soils including reactive oxygen species (ROS), nitric oxide (NO), cell-free DNA, and regulating inflammation-related seeds such as macrophages, neutrophils, fibroblasts, represent a promising strategy to maintain synovial tissue homeostasis for rheumatoid arthritis (RA) treatment. Herein, ROS scavenging amphiphilic block copolymer PEGylated bilirubin and NO-scavenging PEGylated o-phenylenediamine were fabricated to self-assemble into a dually responsive nanoparticle loaded with JAK inhibitor notopterol (Not@BR/oPDA-PEG, NBOP NPs). The simultaneous ROS and NO depletion combined with JAK-STAT pathway inhibition could not only promote M2 polarization to reduce further ROS and NO generation, but also decrease cytokines and chemokines to prevent immune cell recruitment. Specifically, NBOP NPs responded to high level ROS and NO, and disintegrated to release notopterol in inflamed joints as the hydrophobic heads BR and oPDA were transformed into hydrophilic ones. The released notopterol could inhibit the JAK-STAT pathway of inflammatory cells to reduce the secretion of pro-inflammatory cytokines and chemokines. This strategy represented an effective way to regulate RA soils and seeds through breaking the positive feedback loop of inflammation aggravation, achieving an excellent anti-RA efficacy in a collagen-induced arthritis rat model. Taken together, our work offered a reference to adjust RA soils and seeds for enhanced RA treatment.

6.
ACS Nano ; 17(23): 24308-24319, 2023 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-37975685

RESUMO

Meniscus injuries are associated with the degeneration of cartilage and development of osteoarthritis (OA). It is challenging to protect articular cartilage and improve exercise when a meniscus injury occurs. Herein, inspired by the components and functions of the meniscus, we developed a self-lubricating and friction-responsive hydrogel that contains nanoliposomes loaded with diclofenac sodium (DS) and Kartogenin (KGN) for anti-inflammation and cartilage regeneration. When the hydrogel was injected into the meniscus injury site, the drug-loaded nanoliposomes were released from the hydrogel in a friction-responsive manner and reassembled to form hydration layers that lubricate joints during movement. Meanwhile, DS and KNG were constantly released from the nanoliposomes to mitigate inflammation and promote cartilage regeneration. Additionally, this hydrogel exhibited favorable injectability, mechanical properties, fatigue resistance, and prolonged degradation. In vivo experiments demonstrated that injection of the hydrogel effectively improved exercise performance and protected the articular cartilage of rats, suggesting it as a potential therapeutic approach for meniscal injuries.


Assuntos
Cartilagem Articular , Menisco , Ratos , Animais , Hidrogéis/farmacologia , Fricção , Injeções , Diclofenaco/farmacologia
7.
Molecules ; 28(6)2023 Mar 08.
Artigo em Inglês | MEDLINE | ID: mdl-36985456

RESUMO

Rheumatoid arthritis (RA) is a chronic and autoimmune disease characterized by inflammation, autoimmune dysfunction, and cartilage and bone destruction. In this review, we summarized the available reports on the protective effects of Ganoderma lucidum polysaccharides (GLP) on RA in terms of anti-inflammatory, immunomodulatory, anti-angiogenic and osteoprotective effects. Firstly, GLP inhibits RA synovial fibroblast (RASF) proliferation and migration, modulates pro- and anti-inflammatory cytokines and reduces synovial inflammation. Secondly, GLP regulates the proliferation and differentiation of antigen-presenting cells such as dendritic cells, inhibits phagocytosis by mononuclear macrophages and nature killer (NK) cells and regulates the ratio of M1, M2 and related inflammatory cytokines. In addition, GLP produced activities in balancing humoral and cellular immunity, such as regulating immunoglobulin production, modulating T and B lymphocyte proliferative responses and cytokine release, exhibiting immunomodulatory effects. Thirdly, GLP inhibits angiogenesis through the direct inhibition of vascular endothelial cell proliferation and induction of cell death and the indirect inhibition of vascular endothelial growth factor (VEGF) production in the cells. Finally, GLP can inhibit the production of matrix metalloproteinases and promote osteoblast formation, exerting protective effects on bone and articular cartilage. It is suggested that GLP may be a promising agent for the treatment of RA.


Assuntos
Artrite Reumatoide , Cartilagem Articular , Reishi , Humanos , Reishi/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Artrite Reumatoide/metabolismo , Inflamação/metabolismo , Cartilagem Articular/metabolismo , Citocinas/metabolismo , Anti-Inflamatórios/uso terapêutico , Polissacarídeos/farmacologia , Membrana Sinovial/metabolismo
8.
Acta Pharm Sin B ; 13(2): 787-803, 2023 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-36873164

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease characterized by severe synovial inflammation and cartilage damage. Despite great progress in RA therapy, there still lacks the drugs to completely cure RA patients. Herein, we propose a reprogrammed neutrophil cytopharmaceuticals loading with TNFα-targeting-siRNA (siTNFα) as an alternative anti-inflammatory approach for RA treatment. The loaded siTNFα act as not only the gene therapeutics to inhibit TNFα production by macrophages in inflamed synovium, but also the editors to reprogram neutrophils to anti-inflammatory phenotypes. Leveraging the active tendency of neutrophils to inflammation, the reprogrammed siTNFα/neutrophil cytopharmaceuticals (siTNFα/TP/NEs) can rapidly migrate to the inflamed synovium, transfer the loaded siTNFα to macrophages followed by the significant reduction of TNFα expression, and circumvent the pro-inflammatory activity of neutrophils, thus leading to the alleviated synovial inflammation and improved cartilage protection. Our work provides a promising cytopharmaceutical for RA treatment, and puts forward a living neutrophil-based gene delivery platform.

9.
Adv Sci (Weinh) ; 10(11): e2207490, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36748885

RESUMO

Osteoarthritis (OA) is a progressive joint disease characterized by inflammation and cartilage destruction, and its progression is closely related to imbalances in the M1/M2 synovial macrophages. A two-pronged strategy for the regulation of intracellular/extracellular nitric oxide (NO) and hydrogen protons for reprogramming M1/M2 synovial macrophages is proposed. The combination of carbonic anhydrase IX (CA9) siRNA and NO scavenger in "two-in-one" nanocarriers (NAHA-CaP/siRNA nanoparticles) is developed for progressive OA therapy by scavenging NO and inhibiting CA9 expression in synovial macrophages. In vitro experiments demonstrate that these NPs can significantly scavenge intracellular NO similar to the levels as those in the normal group and downregulate the expression levels of CA9 mRNA (≈90%), thereby repolarizing the M1 macrophages into the M2 phenotype and increasing the expression levels of pro-chondrogenic TGF-ß1 mRNA (≈1.3-fold), and inhibiting chondrocyte apoptosis. Furthermore, in vivo experiments show that the NPs have great anti-inflammation, cartilage protection and repair effects, thereby effectively alleviating OA progression in both monoiodoacetic acid-induced early and late OA mouse models and a surgical destabilization of medial meniscus-induced OA rat model. Therefore, the siCA9 and NO scavenger "two-in-one" delivery system is a potential and efficient strategy for progressive OA treatment.


Assuntos
Anidrase Carbônica IX , Sistemas de Liberação de Fármacos por Nanopartículas , Óxido Nítrico , Osteoartrite , Animais , Camundongos , Ratos , Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Nanomedicina/métodos , Óxido Nítrico/metabolismo , Osteoartrite/terapia , Osteoartrite/metabolismo , RNA Mensageiro/metabolismo , Membrana Sinovial/metabolismo , Reprogramação Celular/efeitos dos fármacos , Sistemas de Liberação de Fármacos por Nanopartículas/farmacologia , Anidrase Carbônica IX/efeitos dos fármacos , Anidrase Carbônica IX/metabolismo
10.
Acta Pharmaceutica Sinica B ; (6): 787-803, 2023.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-971712

RESUMO

Rheumatoid arthritis (RA) is an autoimmune disease characterized by severe synovial inflammation and cartilage damage. Despite great progress in RA therapy, there still lacks the drugs to completely cure RA patients. Herein, we propose a reprogrammed neutrophil cytopharmaceuticals loading with TNFα-targeting-siRNA (siTNFα) as an alternative anti-inflammatory approach for RA treatment. The loaded siTNFα act as not only the gene therapeutics to inhibit TNFα production by macrophages in inflamed synovium, but also the editors to reprogram neutrophils to anti-inflammatory phenotypes. Leveraging the active tendency of neutrophils to inflammation, the reprogrammed siTNFα/neutrophil cytopharmaceuticals (siTNFα/TP/NEs) can rapidly migrate to the inflamed synovium, transfer the loaded siTNFα to macrophages followed by the significant reduction of TNFα expression, and circumvent the pro-inflammatory activity of neutrophils, thus leading to the alleviated synovial inflammation and improved cartilage protection. Our work provides a promising cytopharmaceutical for RA treatment, and puts forward a living neutrophil-based gene delivery platform.

11.
Acta Pharmaceutica Sinica B ; (6): 5016-5029, 2023.
Artigo em Inglês | WPRIM (Pacífico Ocidental) | ID: wpr-1011217

RESUMO

Normalizing inflamed soils including reactive oxygen species (ROS), nitric oxide (NO), cell-free DNA, and regulating inflammation-related seeds such as macrophages, neutrophils, fibroblasts, represent a promising strategy to maintain synovial tissue homeostasis for rheumatoid arthritis (RA) treatment. Herein, ROS scavenging amphiphilic block copolymer PEGylated bilirubin and NO-scavenging PEGylated o-phenylenediamine were fabricated to self-assemble into a dually responsive nanoparticle loaded with JAK inhibitor notopterol (Not@BR/oPDA-PEG, NBOP NPs). The simultaneous ROS and NO depletion combined with JAK-STAT pathway inhibition could not only promote M2 polarization to reduce further ROS and NO generation, but also decrease cytokines and chemokines to prevent immune cell recruitment. Specifically, NBOP NPs responded to high level ROS and NO, and disintegrated to release notopterol in inflamed joints as the hydrophobic heads BR and oPDA were transformed into hydrophilic ones. The released notopterol could inhibit the JAK-STAT pathway of inflammatory cells to reduce the secretion of pro-inflammatory cytokines and chemokines. This strategy represented an effective way to regulate RA soils and seeds through breaking the positive feedback loop of inflammation aggravation, achieving an excellent anti-RA efficacy in a collagen-induced arthritis rat model. Taken together, our work offered a reference to adjust RA soils and seeds for enhanced RA treatment.

12.
Biomater Res ; 26(1): 45, 2022 Sep 17.
Artigo em Inglês | MEDLINE | ID: mdl-36115984

RESUMO

BACKGROUND: The meniscus injury is a common disease in the area of sports medicine. The main treatment for this disease is the pain relief, rather than the meniscal function recovery. It may lead to a poor prognosis and accelerate the progression of osteoarthritis. In this study, we designed a meniscal scaffold to achieve the purposes of meniscal function recovery and cartilage protection. METHODS: The meniscal scaffold was designed using the triply periodic minimal surface (TPMS) method. The scaffold was simulated as a three-dimensional (3D) intact knee model using a finite element analysis software to obtain the results of different mechanical tests. The mechanical properties were gained through the universal machine. Finally, an in vivo model was established to evaluate the effects of the TPMS-based meniscal scaffold on the cartilage protection. The radiography and histological examinations were performed to assess the cartilage and bony structures. Different regions of the regenerated meniscus were tested using the universal machine to assess the biomechanical functions. RESULTS: The TPMS-based meniscal scaffold with a larger volume fraction and a longer functional periodicity demonstrated a better mechanical performance, and the load transmission and stress distribution were closer to the native biomechanical environment. The radiographic images and histological results of the TPMS group exhibited a better performance in terms of cartilage protection than the grid group. The regenerated meniscus in the TPMS group also had similar mechanical properties to the native meniscus. CONCLUSION: The TPMS method can affect the mechanical properties by adjusting the volume fraction and functional periodicity. The TPMS-based meniscal scaffold showed appropriate features for meniscal regeneration and cartilage protection.

13.
Artigo em Inglês | MEDLINE | ID: mdl-35253405

RESUMO

With the change in lifestyle and aging of the population, osteoarthritis (OA) is emerging as a major medical burden globally. OA is a chronic inflammatory and degenerative disease initially manifesting with joint pain and eventually leading to permanent disability. To date, there are no drugs available for the definitive treatment of osteoarthritis and most therapies have been palliative in nature by alleviating symptoms rather than curing the disease. This coupled with the vague understanding of the early symptoms and methods of diagnosis so that the disease continues as a global problem and calls for concerted research efforts. A cascade of events regulates the onset and progression of osteoarthritis starting with the production of proinflammatory cytokines, including interleukin (IL)-1ß, IL-6, tumor necrosis factor (TNF)-α; catabolic enzymes, such as matrix metalloproteinases (MMPs)-1, -3, and -13, culminating into cartilage breakdown, loss of lubrication, pain, and inability to load the joint. Although intra-articular injections of small and macromolecules are often prescribed to alleviate symptoms, low residence times within the synovial cavity severely impair their efficacy. This review will briefly describe the factors dictating the onset and progression of the disease, present the current clinically approved methods for its treatment and diagnosis, and finally elaborate on the main challenges and opportunities for the application of nano/micromedicines in the treatment of osteoarthritis. Thus, future treatment regimens will benefit from simultaneous consideration of the mechanobiological, the inflammatory, and tissue degradation aspects of the disease. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement.


Assuntos
Osteoartrite , Citocinas/metabolismo , Citocinas/uso terapêutico , Humanos , Inflamação/tratamento farmacológico , Osteoartrite/tratamento farmacológico , Osteoartrite/metabolismo , Osteoartrite/patologia
14.
Ann Transl Med ; 9(14): 1120, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34430561

RESUMO

BACKGROUND: We utilized the destabilization of medial meniscus (DMM)-induced mice to illustrate the osteoarthritis (OA) suppressing and pain-relieving effects of a novel prolonged-release intra-articular (IA)-dexamethasone-loaded thermo-sensitive hydrogel (DLTH). METHODS: The effects of temperature and pH on DLTH formation and in vitro DLTH release profile were assessed. C57BL/6J mice were randomly divided into three groups: Ctrl group, Model group and DLTH group. The DLTH group received joint injections of 10 µL DLTH (1 mg/kg) into the right knee once a week from week 2 to week 11. We performed micro-computed tomography (Micro-CT) and histological analyses of safranin O-fast green, hematoxylin and eosin, and tartrate-resistant acid phosphatase in knee joints. We also carried out immunohistochemical (IHC) staining for matrix metalloproteinase-9 (MMP-9), MMP-13, and a disintegrin and metalloproteinase with thrombospondin motifs-5 (ADAMTS-5) in cartilage and Ki-67 in synovia. Pain behavioral testing was carried out in all mice. The serum content of prostaglandin E2 (PGE2) and real-time polymerase chain reaction (PCR) of inflammatory cytokines and pain-related factors in dorsal root ganglia (DRGs) were evaluated. RESULTS: It took 20 minutes to form DLTH at pH 7.0 and 37 °C. The cumulative release profiles of dexamethasone (Dex) from DLTH at 37 °C revealed a rapid release in the first 24 h and a sustained slow release for 7 days. In vivo study illustrated that DLTH attenuated OA bone destruction and ameliorated synovitis and progression of OA in DMM-induced mice. The chondroprotective effects of DLTH were mediated by decreased expressions of MMP-9, MMP-13, and ADAMTS-5. The results showed that IA-DLTH exerted pain-relieving effects in OA mice. Upregulation of nociceptive response time (NRT) and downregulations of serum PGE2, inflammatory factors, and pain-related mediators in DRGs of mice in the DLTH group were recorded. CONCLUSIONS: Data presented in this study elucidated that DLTH exhibited a long and lasting Dex release and it is a potential sustainable drug delivery system (DDS) to treat OA locally. IA-DLTH injection exerted chondroprotective and pain-relieving effects in DMM-induced arthritis. The involvement of MMP-9, MMP-13, ADAMTS-5, and inflammatory and pain-related factors, may account for the suppression of OA progression and pain.

15.
Front Pharmacol ; 12: 678810, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34211396

RESUMO

Osteoarthritis (OA) is a common disease characterized by cartilage degeneration. In recent years much attention has been paid to Traditional Chinese Medicine (TCM) since its treatments have shown efficacy for ameliorating cartilage degradation with mild side effects. Osteoking is a TCM prescription that has long been used in OA treatment. However, the exact mechanism of Osteoking are not fully elucidated. In the current study, destabilization of the medial meniscus (DMM)-induced OA mice was introduced as a wild type animal model. After 8 weeks of administration of Osteoking, histomorphometry, OARSI scoring, gait analysis, micro-CT, and immunohistochemical staining for Col2, MMP-13, TGFßRII and pSmad-2 were conducted to evaluate the chondroprotective effects of Osteoking in vivo. Further in vitro experiments were then performed to detect the effect of Osteoking on chondrocytes. TGFßRIICol2ER transgenic mice were constructed and introduced in the current study to validate whether Osteoking exerts its anti-OA effects via the TGF-ß signaling pathway. Results demonstrated that in wild type DMM mice, Osteoking ameliorated OA-phenotype including cartilage degradation, subchondral bone sclerosis, and gait abnormality. Col2, TGFßRII, and pSmad-2 expressions were also found to be up-regulated after Osteoking treatment, while MMP-13 was down-regulated. In vitro, the mRNA expression of MMP-13 and ADAMTS5 decreased and the mRNA expression of Aggrecan, COL2, and TGFßRII were up-regulated after the treatment of Osteoking in IL-1ß treated chondrocytes. The additional treatment of SB505124 counteracted the positive impact of Osteoking on primary chondrocytes. In TGFßRIICol2ER mice, spontaneous OA-liked phenotype was observed and treatment of Osteoking failed to reverse the OA spontaneous progression. In conclusion, Osteoking ameliorates OA progression by decelerating cartilage degradation and alleviating subchondral bone sclerosis partly via the TGF-ß signaling pathway.

16.
Osteoarthritis Cartilage ; 28(5): 698-707, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-31982563

RESUMO

OBJECTIVE: The alarmin HMGB1 is an endogenous molecule that is released into the extracellular space upon trauma or cell activation. Extracellular HMGB1 initiates innate immune responses and besides mediating inflammation, has osteoclast-activating features and mediates pain, all important features in OA. The aim of this study was to examine the involvement of HMGB1 in experimental OA and to explore the effect of local anti-HMGB1-therapy on disease progression. METHOD: OA was induced in mice by surgical destabilization of knee joints and HMGB1 expression and localization was assessed by immunohistochemistry. For therapy evaluation, HMGB1-neutralizing antibodies were injected intraarticularly, alone or encapsulated in an injectable hyaluronan-based delivery vehicle. Human primary chondrocytes were stimulated with rHMGB1 and analyzed by qPCR and cytometric bead-array. RESULTS: HMGB1 immunostaining of mouse OA joints demonstrated intra- and pericellular expression in chondrocytes, overlapping with proteoglycan depleted areas. Intra-articular injection of anti-HMGB1 antibodies had cartilage-protective effects, comparable to treatment with a TNF inhibitor. Direct and vehicle-based delivery had similar ameliorating effects and the effect of a single, early injection could not be enhanced by repeated injections. In vitro stimulation of chondrocytes with rHMGB1 affected chondrocyte function by inducing protein expression of IL6 and IL8 and downregulating mRNA of COL2A1. CONCLUSIONS: Our results suggest that the alarmin HMGB1 might be a new target for OA therapy development as we could observe an aberrant HMGB1 expression in mouse OA joints, stimulation of chondrocytes with rHMGB1 induced cytokine production and decreased matrix production and finally that HMGB1 blockade suppressed disease progression.


Assuntos
Artrite Experimental/metabolismo , Condrócitos/efeitos dos fármacos , Proteína HMGB1/metabolismo , Imunidade Inata , Inflamação/metabolismo , Osteoartrite/metabolismo , Animais , Ligamento Cruzado Anterior/cirurgia , Anticorpos Neutralizantes/administração & dosagem , Anticorpos Neutralizantes/farmacologia , Cartilagem Articular/citologia , Condrócitos/metabolismo , Colágeno Tipo II/efeitos dos fármacos , Colágeno Tipo II/genética , Géis , Proteína HMGB1/antagonistas & inibidores , Proteína HMGB1/farmacologia , Humanos , Ácido Hialurônico , Imuno-Histoquímica , Injeções Intra-Articulares , Interleucina-6/metabolismo , Interleucina-8/efeitos dos fármacos , Interleucina-8/metabolismo , Camundongos , Osteoartrite/patologia , RNA Mensageiro/metabolismo
17.
Inflammation ; 43(1): 146-154, 2020 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-31659586

RESUMO

Leonurine hydrochloride (LH) has been reported to exhibit a number of biological properties such as suppression of inflammation. This study aimed to examine whether the progression of osteoarthritis (OA) could be delayed by the administration of LH in an OA model. Rat chondrocytes were treated with LH under the condition of TNF-α-induced inflammation. After that, real-time PCR and Western blotting were conducted to evaluate relative gene/protein expression levels. For the in vivo study, rats were randomly allocated to a control group (anterior cruciate ligament transection (ACLT) surgery, treatment with saline) and LH group (ACLT surgery, treatment with LH). Articular cartilage degeneration was assessed by histological evaluation. It was found that LH significantly suppressed the expression of MMP-1, MMP-3, MMP-13, IL-6, and ADAMTS-5 in cells via the NF-κB signaling pathway. In addition, it is revealed that intra-articular injection of LH significantly ameliorated cartilage degeneration in a rat OA model. Taken together, these results indicate that LH attenuates progression of OA by inhibition of inflammation via the NF-κB signaling pathway and represents a potential preventive therapy for OA.


Assuntos
Ligamento Cruzado Anterior/patologia , Anti-Inflamatórios/farmacologia , Cartilagem Articular/patologia , Ácido Gálico/análogos & derivados , Osteoartrite/tratamento farmacológico , Proteína ADAMTS5/biossíntese , Animais , Ligamento Cruzado Anterior/cirurgia , Doenças das Cartilagens/tratamento farmacológico , Linhagem Celular , Condrócitos/efeitos dos fármacos , Condrócitos/patologia , Ácido Gálico/farmacologia , Inflamação/tratamento farmacológico , Interleucina-6/biossíntese , Masculino , Metaloproteinase 1 da Matriz/biossíntese , Metaloproteinase 13 da Matriz/biossíntese , Metaloproteinase 3 da Matriz/biossíntese , NF-kappa B/metabolismo , Osteoartrite/patologia , Ratos
18.
Artigo em Chinês | WPRIM (Pacífico Ocidental) | ID: wpr-847261

RESUMO

BACKGROUND: Cartilage defects due to cartilage lesions such as osteoarthritis are difficult to self-regenerate. How to promote cartilage regeneration and restore smooth wound surface is a hot topic in recent years. OBJECTIVE: To review the general concept, mechanism and effect of Kartogenin as well as the current clinical models, profor the regeneration and repair of damaged cartilage. METHODS: WanFang, CNKI, and PubMed were retrieved for studies on Kartogenin combined with tissue engineering in the regeneration and repair published from January 2010 to January 2020. The keywords were “Kartogenin, cartilage regeneratichondrogenesis, chondroprotection” in English and “Kartogenin, cartilage regeneration and repair, cartilage protection, tissue Chinese. After initial screening by reading titles and abstracts, irrelevant literature was excluded and finally 55 articles were ianalysis according to the inclusion criteria and exclusion criteria. RESULTS AND CONCLUSION: As a newly discovered small molecule, Kartogenin has better stability, lower immunogeniciavailability. In addition, Kartogenin plays a synergistic role with growth factors in cartilage regeneration and has great potentiformation and cartilage protection. Through in vitro experiments or animal experiments, it has been proved that Kartogenin prole in promoting cartilage generation, osteoarthritis treatment, repair of tendon and bone injury, and wound healing, and heldegeneration of cartilage and healing of rotator cuff injury. With more basic research and clinical trials on Kartogenin, a breacartilage regeneration and repair will be made in the near future.

19.
J Ginseng Res ; 39(1): 54-60, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25535477

RESUMO

BACKGROUND: The present study was designed to prepare and find the optimum active preparation or fraction from Korea Red Ginseng inhibiting matrix metalloproteinase-13 (MMP-13) expression, because MMP-13 is a pivotal enzyme to degrade the collagen matrix of the joint cartilage. METHODS: From total red ginseng ethanol extract, n-BuOH fraction (total ginsenoside-enriched fraction), ginsenoside diol-type-enriched fraction (GDF), and ginsenoside triol-type-enriched fraction (GTF) were prepared, and ginsenoside diol type-/F4-enriched fraction (GDF/F4) was obtained from Panax ginseng leaf extract. RESULTS: The n-BuOH fraction, GDF, and GDF/F4 clearly inhibited MMP-13 expression compared to interleukin-1ß-treated SW1353 cells (human chondrosarcoma), whereas the total extract and ginsenoside diol-type-enriched fraction did not. In particular, GDF/F4, the most effective inhibitor, blocked the activation of p38 mitogen-activated protein kinase (p38 MAPK), c-Jun-activated protein kinase (JNK), and signal transducer and activator of transcription-1/2 (STAT-1/2) among the signal transcription pathways involved. Further, GDF/F4 also inhibited the glycosaminoglycan release from interleukin-1α-treated rabbit cartilage culture (30.6% inhibition at 30 µg/mL). CONCLUSION: Some preparations from Korean Red Ginseng and ginseng leaves, particularly GDF/F4, may possess the protective activity against cartilage degradation in joint disorders, and may have potential as new therapeutic agents.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA