Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
1.
Article in English | MEDLINE | ID: mdl-38744373

ABSTRACT

OBJECTIVE: Intervertebral Disc Degeneration (IVDD) is one of the leading causes of low back pain, significantly impacting both individuals and society. This study aimed to investigate the significance of macrophage infiltration and the role of macrophage-secreted platelet-derived growth factor-BB (PDGF-BB) in IVDD progression. METHODS: To confirm the protective function of macrophage-derived PDGF-BB on nucleus pulposus cells (NPCs), we employed Lysm-Cre transgenic mice to genetically ablate PDGF-B within the myeloid cells. Immunohistochemistry was utilized to detect the expression of glycolytic enzymes and pyroptosis-related proteins during the process of IVDD. Western blot, RT-PCR, ELISA and immunofluorescence were used to detect the protective effect of recombinant PDGF-BB on NPCs. RESULTS: Macrophage-derived PDGF-BB deficiency resulted in the loss of NPCs and the increased ossification of cartilage endplates during lumbar disc degeneration. Also, PDGF-BB deficiency triggered the inhibition of glycolytic enzymes' expression and the activation of pathways related to pyroptosis in the nucleus pulposus. Mechanistically, our results suggest that PDGF-BB predominantly conveys its protective influence on NPCs through the PDGF receptor- beta (PDGFR-ß)/ thioredoxin-interacting protein pathway. CONCLUSIONS: The absence of PDGF-BB originating from macrophages expedites the advancement of IVDD, whereas the application of PDGF-BB treatment holds the potential for retarding intervertebral disc degeneration in the human body.

2.
J Am Chem Soc ; 145(44): 24218-24229, 2023 Nov 08.
Article in English | MEDLINE | ID: mdl-37874900

ABSTRACT

Exploring efficient strategies to overcome the performance constraints of oxygen evolution reaction (OER) electrocatalysts is vital for electrocatalytic applications such as H2O splitting, CO2 reduction, N2 reduction, etc. Herein, tunable, wide-range strain engineering of spinel oxides, such as NiFe2O4, is proposed to enhance the OER activity. The lattice strain is regulated by interfacial thermal mismatch during the bonding process between thermally expanding NiFe2O4 nanoparticles and the nonexpanding carbon fiber substrate. The tensile lattice strain causes energy bands to flatten near the Fermi level, lowering eg orbital occupancy, effectively increasing the number of electronic states near the Fermi level, and reducing the pseudoenergy gap. Consequently, the energy barrier of the rate-determining step for strained NiFe2O4 is reduced, achieving a low overpotential of 180 mV at 10 mA/cm2. A total water decomposition voltage range of 1.52-1.56 V at 10 mA/cm2 (without iR correction) was achieved in an asymmetric alkaline electrolytic cell with strained NiFe2O4 nanoparticles, and its robust stability was verified with a voltage retention of approximately 99.4% after 100 h. Furthermore, the current work demonstrates the universality of tuning OER performance with other spinel ferrite systems, including cobalt, manganese, and zinc ferrites.

3.
Cell Biol Int ; 47(5): 954-968, 2023 May.
Article in English | MEDLINE | ID: mdl-36740226

ABSTRACT

Irreversible destruction of joints is the hallmark of rheumatoid arthritis (RA). Osteoclasts are the only bone-resorbing cells and play an important role in joint rebuilding. BML-111 (5(S),6(R),7-trihydroxyheptanoic acid methyl ester, C8 H16 O5 ) is a synthetic lipoxin A4 agonist with antioxidant and anti-inflammatory properties. The present study aimed to investigate the effect of BML-111 on osteoclasts in vivo and in vitro, to investigate its therapeutic effect on joint destruction in RA. Cell Counting Kit-8 assay and flow cytometry were used to exclude cytotoxic effects of BML-111 to bone marrow-derived macrophages (BMMs). Then, osteoclasts were differentiated in vitro from BMMs by used macrophage colony-stimulating factor and receptor activator of nuclear factor-κB ligand, and osteoclasts were observed following tartrate-resistant acid phosphatase staining with or without BML-111 treatment. Meanwhile, absorption pit assay and immunofluorescence staining of the fibrous actin ring were used to observe osteoclast function. Moreover, we examined mitogen-activated protein kinase (MAPK) and nuclear factor-κB (NF-κB) activation. We established collagen-induced arthritis in a rat model and, after treatment with BML-111, joint swelling was measured and the knee joints were processed for histology. We also examined serum and tissue for osteoclastogenesis-related markers. BML-111 inhibited osteoclast formation and differentiation in a time- and concentration-dependent manner, and downregulated the expression levels of MAPK and NF-κB in vitro. Meanwhile, BML-111 effectively alleviated joint structural damage and inhibited osteoclast formation in vivo. BML-111 inhibited osteoclast formation and differentiation in vitro and in vivo, and delayed the progression of joint destruction.


Subject(s)
Bone Resorption , Osteoclasts , Rats , Animals , NF-kappa B/metabolism , Mitogen-Activated Protein Kinases/metabolism , Bone Resorption/metabolism , Bone Resorption/pathology , Cell Differentiation , Knee Joint/metabolism , Knee Joint/pathology , RANK Ligand/metabolism
4.
Bioorg Chem ; 132: 106321, 2023 03.
Article in English | MEDLINE | ID: mdl-36642020

ABSTRACT

Osteoarthritis (OA), a degenerative disease affecting the joint, is characterized by degradation of the joint edge, cartilage injury, and subchondral bone hyperplasia. Treatment of early subchondral bone loss in OA can inhibit subsequent articular degeneration and improve the prognosis of OA. PD0325901, a specific inhibitor of ERK, is widely used in oncology and has potential as a therapeutic agent for osteoarthritis In this study, we investigated the biological function of PD0325901 in bone marrow monocytes/macrophages (BMMs)treated with RANKL and found that it inhibited osteoclast differentiation in vitro in a time- and dose-dependent manner. PD0325901 restrained the expression of osteoclast marker genes, such as c-Fos and NFATc1 induced by RANKL. We tested the biological effects of PD035901 on ATDC5 cells stimulated by IL-1ß and found that it had protective effects on ATDC5 cells. In animal studies, we used a destabilization of the medial meniscus (DMM) model and injected 5 mg/kg or 10 mg/kg of PD0325901 compound into each experimental group of mice. We found that PD0325901 significantly reduced osteochondral pathological changes in post-OA subchondral bone destruction.Finally, we found that PD0325901 down-regulated the pyroptosis level in chondrocytes to rescue cartilage degeneration. Therefore, PD0325901 is expected to be a new generation alternative therapy for OA.


Subject(s)
NF-kappa B , Osteoarthritis , Animals , Mice , NF-kappa B/metabolism , Osteoclasts , Signal Transduction , Inflammation/metabolism , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Cartilage/metabolism , Cartilage/pathology , Chondrocytes
5.
Inflammopharmacology ; 31(1): 369-384, 2023 Feb.
Article in English | MEDLINE | ID: mdl-36401729

ABSTRACT

OBJECTIVES: As one of the major causes of low back pain, intervertebral disc degeneration (IDD) has caused a huge problem for humans. Increasing evidence indicates that NLRP3 inflammasome-mediated pyroptosis of NP cells displays an important role in the progression of IDD. Maltol (MA) is a flavoring agent extracted from red ginseng. Due to its anti-inflammatory and antioxidant effects, MA has been widely considered by researchers. Therefore, we hypothesized that MA may be a potential IVD protective agent by regulating NP cells and their surrounding microenvironment. METHODS: In vitro, qRT-PCR, and Western blot were used to explore the effect of MA on the transcription and protein expression of the anabolic protein (ADAMTS5, MMP3, MMP9) catabolic protein (Aggrecan), and pro-inflammatory factor (iNOS COX-2). Next, the effects of MA on PI3K/AKT/NF-κB pathway and pyroptosis pathway were analyzed by Western blot and immunofluorescence. Molecular docking was used to investigate the relationship between PI3K and MA. Moreover, ELISA was also used to detect the effects of MA on inflammatory factors (TNF-α, PGE2, IL-1ß, and IL-18). In vivo, the effects of MA on the vertebral structure of IDD mice were studied by HE and SO staining and the effects of MA on ECM and PI3K/AKT/NF-κB and pyroptosis pathway of IDD mice were studied by immunohistochemical staining. RESULTS: MA can ameliorate intervertebral disc degeneration in vivo and in vitro. Specifically, the molecular docking results showed that the binding degree of MA and PI3K was significant. Second, in vitro studies showed that MA inhibited the degradation of ECM and inflammatory response by inhibiting the PI3K/AKT/NF-κB pathway and the pyroptosis mediated by NLRP3 inflammasome, which increased the expression of anabolic proteins, decreased the expression of catabolic proteins, and decreased the secretion of inflammatory mediators such as IL-18 and IL-1ß. In addition, according to the study results of the mouse lumbar instability model, MA also improved the tissue disorder and degradation of the intervertebral disc, reduced the loss of proteoglycan and glycosaminoglycan, and inhibited intervertebral disc inflammation, indicating that MA has a protective effect on the intervertebral disc to intervertebral disc in mice. CONCLUSIONS: Our results suggest that MA slowed IDD development through the PI3K/AKT/NF-κB signaling pathway and NLRP3 inflammasome-mediated pyroptosis, indicating that MA appeared to be a viable medication for IDD treatment.


Subject(s)
Intervertebral Disc Degeneration , Nucleus Pulposus , Humans , Mice , Animals , NF-kappa B/metabolism , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/metabolism , Inflammasomes/metabolism , Proto-Oncogene Proteins c-akt/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Interleukin-18/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Pyroptosis , Molecular Docking Simulation , Nucleus Pulposus/metabolism
6.
Bioorg Chem ; 120: 105632, 2022 03.
Article in English | MEDLINE | ID: mdl-35074577

ABSTRACT

Necroptosis of chondrocytes contributes to the progression of osteoarthritis (OA). Recent studies have shown that VX-11e, an ERK inhibitor, exhibited a contrasting expression pattern to RIP3, the key protein of necroptosis. However, its effect on OA remains to be determined. Therefore, we investigated whether VX-11e affected the loss of articular cartilage and subchondral bone during OA. In in vivo experiments, a mouse OA model induced by medial meniscus instability (destabilization of the medial meniscus [DMM]) was used. In in vitro experiments, interleukin-1ß (IL-1ß) was used to simulate the inflammatory microenvironment of chondrocytes, and RANKL was used to induce osteoclast differentiation. Histological analysis, cell viability experiments, high-density cell culture experiments, immunofluorescence assay, western blot assay, quantitative PCR, and molecular docking experiments were conducted to determine the protective effect of VX-11e on articular cartilage during OA. We also performed histological analysis, tartrate-resistant acid phosphatase (TRAP) staining, F-actin ring formation test, quantitative PCR, and western blot assay to study the effect of VX-11e on subchondral bone during OA progression. We found that after the medial meniscus was severed, the articular cartilage of the mice showed pathological changes, accompanied with the loss of subchondral bone. However, an intraperitoneal injection of VX-11e protected the cartilage and subchondral bone of the mouse knee joint. The results of in vitro experiments showed that VX-11e promoted the anabolism of the extracellular matrix of chondrocytes by inhibiting the expression and phosphorylation of RIP3 and MLKL. VX-11e also inhibited RANKL-induced osteoclast differentiation by inhibiting the ERK/RSK signaling pathway, but not the NF-κB pathway. Overall, VX-11e inhibited the loss of articular cartilage and subchondral bone during OA by regulating the RIP1/RIP3/MLKL and MAPK signaling pathways.


Subject(s)
Cartilage, Articular , Osteoarthritis , Animals , Cartilage, Articular/metabolism , Cartilage, Articular/pathology , Chondrocytes/metabolism , Chondrocytes/pathology , Disease Models, Animal , Mice , Molecular Docking Simulation , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Protein Kinases/pharmacology , Pyrimidines , Pyrroles , Signal Transduction
7.
Nanomaterials (Basel) ; 14(11)2024 May 21.
Article in English | MEDLINE | ID: mdl-38869523

ABSTRACT

Electrocatalysts in alkaline electrocatalytic water splitting are required to efficiently produce hydrogen while posing a challenge to show excellent performances. Herein, we have successfully synthesized platinum nanoparticles incorporated in a Co3O4 nanostructure (denoted as Pt-Co3O4) that show superior HER activity and stability in alkaline solutions (the overpotentials of 37 mV to reach 10 mA cm-2). The outstanding electrocatalytic activity originates from synergistic effects between Pt and Co3O4 and increased electron conduction. Theoretical calculations show a significant decrease in the ΔGH* of Co active sites and a remarkable increase in electron transport. Our work puts forward a special and simple synthesized way of adjusting the H* adsorption energy of an inert site for application in HER.

8.
J Colloid Interface Sci ; 650(Pt B): 1174-1181, 2023 Nov 15.
Article in English | MEDLINE | ID: mdl-37473477

ABSTRACT

Achieving efficient hydrogen evolution reaction (HER) catalysts to scale up electrochemical water splitting is desirable but remains a major challenge. Here, nitrogen-doped carbon nanotubes (NCNTs) loaded with PtNi/MoN electrocatalyst (PtNi/MoN@C) is synthesized by a simple strategy to obtain stronger interphase effects and significantly improve HER activity. The surface morphology of the materials is altered by Pt doping and the electronic structure of MoN is changed, which optimizing the electronic environment of the materials, shifting the binding energy and giving the materials a higher electrical conductivity, this ultimately leads to faster proton and electron transfer processes. The synergistic effect of Pt nanoparticles, MoN and the good combination with carbon leads to a high HER activity of 18 mV to reach 10 mA cm-2 in alkaline solution, outperforming that of the commercial Pt/C. Theoretical studies show that the heterostructures can efficiently enhance the electron transport and reduce the △GH*.

9.
Mol Immunol ; 159: 46-57, 2023 07.
Article in English | MEDLINE | ID: mdl-37271009

ABSTRACT

Synovial fibrosis is a driver in the progression of osteoarthritis (OA). Fibroblast growth factor 10 (FGF10) has prominent anti-fibrotic effects in many diseases. Thus, we explored the anti-fibrosis effects of FGF10 in OA synovial tissue. In vitro, fibroblast-like synoviocytes (FLSs) were isolated from OA synovial tissue and stimulated with TGF-ß to establish a cell model of fibrosis. After treatment with FGF10, we assessed the effects on FLS proliferation and migration using CCK-8, EdU, and scratch assays, and collagen production was observed using Sirius Red Stain. The JAK2/STAT3 pathway and expression of fibrotic markers were evaluated through western blotting (WB) and immunofluorescence (IF). In vivo, we treated mice with OA induced by surgical destabilization of the medial meniscus (DMM) with FGF10 and assessed the anti-OA effect using histological and immunohistochemical (IHC) staining of MMP13, and fibrosis was evaluated using HE and Masson's trichrome staining. The expression of IL-6/JAK2/STAT3 pathway components was determined using ELISA, WB, IHC, and IF. In vitro, FGF10 inhibited TGF-ß-induced FLS proliferation and migration, decreased collagen deposition, and improved synovial fibrosis. Moreover, FGF10 mitigated synovial fibrosis and improved the symptoms of OA in DMM-induced OA mice. Overall, FGF10 had promising anti-fibrotic effects on FLSs and improved OA symptoms in mice. The IL-6/STAT3/JAK2 pathway plays key roles in the anti-fibrosis effect of FGF10. This study is the first to demonstrate that FGF10 inhibited synovial fibrosis and attenuated the progression of OA by inhibiting the IL-6/JAK2/STAT3 pathway.


Subject(s)
Fibroblast Growth Factor 10 , Interleukin-6 , Osteoarthritis , Animals , Mice , Fibroblast Growth Factor 10/pharmacology , Fibroblasts , Interleukin-6/metabolism , Osteoarthritis/pathology , Synovial Membrane/pathology , Transforming Growth Factor beta/metabolism
10.
Drug Des Devel Ther ; 17: 2383-2399, 2023.
Article in English | MEDLINE | ID: mdl-37605762

ABSTRACT

Background: Osteoarthritis (OA), a common chronic joint disease, is characterized by cartilage degeneration and subchondral bone reconstruction. NF-κB signaling pathway-activated inflammation and NLRP3-induced pyroptosis play essential roles in the development of OA. In this study, we examine whether paroxetine can inhibit pyroptosis and reduce osteoclast formation, thereby delaying the destruction of knee joints. Methods: We employed high-density cultures, along with quantitative polymerase chain reactions and Western blotting techniques, to investigate the effects of paroxetine on extracellular matrix synthesis and degradation. The expression levels of NF-κB and pyroptosis-related signaling pathway proteins were examined by Western blotting and immunofluorescence. Furthermore, the impact of paroxetine on RANKL-induced osteoclast formation was evaluated through TRAP staining and F-actin ring fluorescence detection. To investigate the role of paroxetine in vivo, we constructed a mouse model with destabilization of the medial meniscus (DMM) surgery. Safranin O-Fast Green staining, Hematoxylin-Eosin staining, and immunohistochemistry were conducted to observe the extent of knee joint cartilage deformation. In addition, TRAP staining was used to observe the formation of osteoclasts in the subchondral bone. Results: In the in vitro experiments with ATDC5, paroxetine treatment attenuated IL-1ß-induced activation of the pyroptosis-related pathway and suppressed extracellular matrix catabolism by inhibiting the NF-kB signaling pathway. In addition, paroxetine treatment decreased the expression of RANKL-induced osteoclast marker genes and reduced osteoclast formation. In animal experiments conducted in vivo, mice treated with paroxetine exhibited thicker knee cartilage with a smoother surface compared to the DMM group. Additionally, the formation of osteoclasts in the subchondral bone was reduced in the paroxetine-treated mice. Further analysis revealed that paroxetine treatment played a role in preserving the balance of the extracellular matrix and delaying knee joint degeneration. Conclusion: Paroxetine can inhibit pyroptosis and reduce osteoclast formation via inhibiting the NF-κB signaling pathway, suggesting that it may have therapeutic effects in patients with OA.


Subject(s)
NF-kappa B , Osteoarthritis, Knee , Animals , Mice , Chondrocytes , Osteoclasts , Paroxetine/pharmacology , Pyroptosis , Signal Transduction
11.
Int Immunopharmacol ; 125(Pt B): 111169, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37948862

ABSTRACT

BACKGROUND: Osteoarthritis (OA) is a prevalent degenerative joint disease [1]. It has come to light that AZD8330 can suppress the generation of proinflammatory factors and deter the inflammatory response [2]. Given that inflammation is a primary causative factor in OA, it is posited that AZD8330 might exhibit superior efficacy in OA management. METHODS: In this study, we investigated the potential of intraperitoneal injection of AZD8330 to retard the progression of osteoarthritis in a murine model with surgically induced medial meniscus destruction (DMM). Concurrently, we employed ATDC5 cartilage cells to dissect the mechanism through which AZD8330 inhibits the TNF-α-induced NF-κB signaling pathway via modulation of RIP1. The findings revealed that AZD8330 mitigated cartilage degradation and the inflammatory response, leading to a substantial reduction in OARSI scores among DMM mice treated with AZD8330. Mechanistically, AZD8330 functioned as a suppressor of the TNF-α-induced NF-κB/p65 signaling pathway by facilitating the phosphorylation activation of cIAP1-mediated RIP1. The combination of data from both in vivo and in vitro experiments supports the conclusion that AZD8330 can attenuate chondrocyte degradation, thereby alleviating OA, by regulating the NF-κB/P65 signaling pathway through modulation of RIP1 activity. Consequently, the utilization of AZD8330 may hold potential in the prophylaxis of osteoarthritis. RESULTS: Our investigation delineates the role of AZD8330 in the regulation of inflammation in the context of OA treatment. Furthermore, we have unveiled that the inhibitory impact of AZD8330 on OA may hinge upon the activation of cIAP1, which in turn downregulates RIP1, thereby restraining the NF-κB/P65 signaling pathway. This study lends credence to the notion that AZD8330 may be a promising contender for osteoarthritis therapy. CONCLUSIONS: Our study provides compelling evidence attesting to the capacity of AZD8330 in managing inflammation within the realm of OA treatment. Likewise, our study has elucidated that the attenuation of OA by AZD8330 relies on the activation of cIAP1 to inhibit RIP1, consequently suppressing the NF-κB signaling pathway. On the strength of our present study, we may have identified a viable drug candidate for OA treatment.


Subject(s)
NF-kappa B , Osteoarthritis , Mice , Animals , NF-kappa B/metabolism , Tumor Necrosis Factor-alpha/metabolism , Osteoarthritis/chemically induced , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Up-Regulation , Signal Transduction , Inflammation/drug therapy , Chondrocytes/metabolism , Menisci, Tibial , Necrosis/metabolism , Interleukin-1beta/metabolism
12.
Biochem Pharmacol ; 211: 115542, 2023 05.
Article in English | MEDLINE | ID: mdl-37028460

ABSTRACT

Osteoarthritis (OA) is a disabling joint disease characterized by cartilage degeneration. Reactive oxygen species (ROS)-induced oxidative stress is an important cause of early chondrocyte death. For this reason, we investigated PD184352, a small molecule inhibitor with potential anti-inflammatory and antioxidant activity. We evaluated the protective effect of PD184352 against destabilized medial meniscus (DMM)-induced OA in mice. The knee joints of the PD184352-treated group had higher Nrf2 expression and milder cartilage damage. Moreover, in in vitro experiments, PD184352 suppressed IL-1ß-induced NO, iNOS, PGE2 production, and attenuated pyroptosis. PD184352 treatment promoted antioxidant protein expression and reduced the accumulation of ROS by activating the Nrf2/HO-1 axis. Finally, the anti-inflammatory and antioxidant effects of PD184352 were shown to be partially dependent on Nrf2 activation. Our study reveals the potential role of PD184352 as an antioxidant and provides a new strategy for OA treatment.


Subject(s)
Antioxidants , Osteoarthritis , Mice , Animals , Antioxidants/metabolism , Reactive Oxygen Species/metabolism , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Anti-Inflammatory Agents/therapeutic use , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Chondrocytes , Interleukin-1beta/metabolism
13.
J Pharm Pharmacol ; 75(5): 612-624, 2023 Apr 17.
Article in English | MEDLINE | ID: mdl-36856818

ABSTRACT

OBJECTIVES: Limonin has received significant attention due to its multiple biological effects, intervertebral disc degeneration (IDD) is also of interest due to the high prevalence of this disease. In this study, we determined the effects of limonin on IDD and the underlying mechanism of action to find novel ways to treat IDD. METHODS: An IL-1ß-induced cell inflammation model and a lumbar instability model inducing IDD were established to assess the progression of IDD with or without limonin treatment. We further evaluated MAPK/NF-κB and necroptosis pathways and alterations in the extracellular matrix specific within the disc. KEY FINDINGS: Limonin suppresses inflammation in the nucleus pulposus in vitro by reducing the production of pro-inflammatory markers such as iNOS and COX-2. Limonin reduced the activation of the MAPK/NF-κB signalling pathway and the RIP1/RIP3/MLKL necroptosis pathway in the NP cells. Moreover, limonin delays the IDD progression in the lumbar instability model. CONCLUSIONS: Limonin could potentially delay IDD by inhibiting NP cell necroptosis and modulating peripheral matrix proteins within the intervertebral disc and is a potential pharmacological research direction for the therapy in patients with IDD.


Subject(s)
Intervertebral Disc Degeneration , Limonins , Inflammation , Intervertebral Disc Degeneration/drug therapy , Limonins/pharmacology , Limonins/therapeutic use , Necroptosis , NF-kappa B/metabolism , Animals , Rats
14.
Front Chem ; 10: 913874, 2022.
Article in English | MEDLINE | ID: mdl-35769446

ABSTRACT

In this study, Cu-based multicomponent metallic compound materials M-Cu (M = Mn, Fe, Co, Ni, and Pt) were studied as electrocatalytic materials for water splitting. Different metal materials attached to the copper foam substrate can change the valence states of copper and oxygen, resulting in the change of electronic structure of the materials, thus changing its catalytic activity.

15.
Nutr Metab (Lond) ; 19(1): 75, 2022 Nov 08.
Article in English | MEDLINE | ID: mdl-36348458

ABSTRACT

BACKGROUND: Senile osteoporosis (SOP) is one of the most prevalent diseases that afflict the elderly population, which characterized by decreased osteogenic ability. Glucosamine (GlcN) is an over-the-counter dietary supplement. Our previous study reported that GlcN promotes osteoblast proliferation by activating autophagy in vitro. The purpose of this study is to determine the effects and mechanisms of GlcN on senile osteoporosis and osteogenic differentiation in vivo. METHODS: Aging was induced by subcutaneous injection of D-Galactose (D-Gal), and treated with GlcN or vehicle. The anti-senile-osteoporosis effect of GlcN was explored by examining changes in micro-CT, serum indicators, body weight, protein and gene expression of aging and apoptosis. Additionally, the effects of GlcN on protein and gene expression of osteogenesis and autophagy were observed by inhibiting autophagy with 3-methyladenine (3-MA). RESULTS: GlcN significantly improved bone mineral density (BMD) and bone micro-architecture, decreased skeletal senescence and apoptosis and increased osteogenesis in D-Gal induced osteoporotic mice. While all effect was reversed with 3-MA. CONCLUSION: GlcN effectively delayed the progression of osteoporosis in senile osteoporotic mice by promoting osteoblast autophagy. This study suggested that GlcN may be a prospective candidate drug for the treatment of SOP.

16.
Nanomaterials (Basel) ; 12(18)2022 Sep 15.
Article in English | MEDLINE | ID: mdl-36144996

ABSTRACT

Graphene exhibits great potential for the corrosion protection of metals, because of its low permeability and high chemical stability. To enhance the anticorrosion ability of Cu, we use plasma-enhanced chemical vapor deposition (PECVD) to prepare a vertically oriented few-layer graphene (VFG) coating on the surface of Cu. The Cu coated with VFG shows superhydrophobic surface with a contact angle of ~150°. The VFG coating is used to significantly increase the anticorrosion ability, enhanced by the chemical stability and the unique geometric structure of vertically oriented graphene. The corrosion rate of VFG-Cu was about two orders of magnitude lower than that of bare Cu. This work highlights the special synthesized way of PECVD and superhydrophobic surface of vertical structures of graphene as coatings for various applications.

17.
Biochem Pharmacol ; 205: 115155, 2022 11.
Article in English | MEDLINE | ID: mdl-35820500

ABSTRACT

Osteoarthritis (OA) is a degenerative disease caused by the progressive destruction of cartilage and subchondral bone [1]. Studies have shown that by inhibiting the degradation of cartilage cells and the loss of subchondral bone, OA can be prevented and treated. Neratinib, as a small molecule compound with anti-inflammatory and anti-tumor properties, is a very effective inhibitor of IL-1ß-induced chondrocyte inflammation and anabolic metabolism. By investigating the effect of neratinib in ATDC5 chondrocytes, the study finds that neratinib reduces inflammation by inhibiting the MAPK and NF-κB signaling pathways, and at the same time reduces pyrolysis (indicated by the results of reverse transcription quantitative PCR and western blotting). For anabolic metabolism, after high-density cell culture, IL-1ß-induced catalytic changes and degradation of the extracellular matrix were evaluated by toluidine blue staining. Since osteoclasts are key participants in the process of subchondral bone remodeling in OA, we also studied the effect of neratinib on the maturation of osteoclasts. The results showed that neratinib also acts as an anti-osteoclast agent in vitro. By inhibiting the NF-κB and MAPK pathways, it reduces the expression of osteoclast-related genes, thereby inhibiting RANKL-induced osteoclastogenesis. The results of in vivo animal experiments supported the conclusions from the experiments in vitro. Neratinib inhibited both the destruction of medial meniscus induced cartilage degradation and osteoclast formation, which proves that neratinib has a dual effect, protecting cartilage and inhibiting osteoclast formation. These results indicate that neratinib can be a brand-new latent strategy for the treatment of OA.


Subject(s)
NF-kappa B , Osteoarthritis , Animals , NF-kappa B/metabolism , Tolonium Chloride/metabolism , Tolonium Chloride/pharmacology , Tolonium Chloride/therapeutic use , Osteoarthritis/pathology , Chondrocytes , Cartilage/metabolism , Interleukin-1beta/metabolism , Signal Transduction , Inflammation/metabolism , Anti-Inflammatory Agents/pharmacology
18.
J Colloid Interface Sci ; 627: 21-27, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35841705

ABSTRACT

The catalyst morphology has a strong impact on the activity of electrocatalytic hydrogen production. Considering the effect, we design and fabricate hollow spherical Ni/MoO2 heterojunction. In addition, an amorphous carbon skeleton is inserted into the hollow sphere, which makes the structure more stable and porous. Compared with other morphological Ni/MoO2, the porous hollow spherical Ni/MoO2 (H-Ni/MoO2) with an internal carbon skeleton shows better hydrogen evolution reaction (HER) activity with a small overpotential of 58 mV to reach 10 mA cm-2 and a tafel value of 44.8 mV dec-1 in alkaline media. The developed HER performance of H-Ni/MoO2 can be attributed to the larger active surface area of porous hollow spherical structure and the faster electron transfer and better stability of carbon skeleton. Undoubtedly, the urea plays a crucial role to construct the hollow spherical morphology and being decomposed to form holes and amorphous carbon in the synthesized steps. The soft-template strategy using urea as the addition for forming the porous hollow structure with carbon skeleton can be extended to explore superior non-noble metal for hydrogen production.

19.
Front Pharmacol ; 13: 938133, 2022.
Article in English | MEDLINE | ID: mdl-36238555

ABSTRACT

Objectives: Osteoarthritis (OA) is a common disease that mainly manifests as inflammation and destruction of cartilage and subchondral bone. Recently, necroptosis has been reported to play an important role in the development of OA. Selumetinib displays a contrasting expression pattern to necroptosis-related proteins. The present study aimed to investigate the potential therapeutic effects of selumetinib in OA process. Methods: In vitro experiments, interleukin-1ß (IL-1ß) was used to induce necroptosis of chondrocytes. We used high-density cell culture, Western Blot and PT-PCR to observe the effect of different concentrations of selumetinib on the extracellular matrix of cartilage. Afterwards, we visualized the effect of selumetinib on osteoclast formation by TRAP staining and F-actin rings. In vivo experiment, we induced experimental osteoarthritis in mice by surgically destabilizing the medial meniscus (DMM) while administering different concentrations of selumetinib intraperitoneally. Results: Selumetinib promoted cartilage matrix synthesis and inhibited matrix decomposition. We found that selumetinib exerted a protective function by inhibiting the activation of RIP1/RIP3/MLKL signaling pathways in chondrocytes. Selumetinib also inhibited the activation of RANKL-induced NF-κB and MAPK signaling pathways in BMMs, thereby interfering with the expression of osteoclast marker genes. In the DMM-induced OA model, a postsurgical injection of selumetinib inhibited cartilage destruction and lessened the formation of TRAP-positive osteoclasts in subchondral bone. Conclusion: Selumetinib can protect chondrocytes by regulating necroptosis to prevent the progression of OA and reduce osteoclast formation. In summary, our findings suggest that selumetinib has potential as a therapeutic agent for OA.

20.
Int Immunopharmacol ; 111: 109085, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35952515

ABSTRACT

As a degenerative disease, the pathogenesis and treatment of osteoarthritis (OA) are still being studied. The prevailing view is that articular cartilage dysfunction plays an essential role in the development of osteoarthritis. Similarly, dynamic bone remodeling dramatically influences the development of osteoarthritis. The inflammatory response is caused by the overexpression of inflammatory factors, among which tumor necrosis factor-α is one of the main causes of OA, and its sources include the secretion of chondrocytes themselves and osteoclast secretion of subchondral bone. Moreover, TNF-α-induced activation of RIP1, RIP3, and MLKL has been shown to play an important role in cell necroptosis and inflammatory responses. In vitro, AZ-628 alleviates chondrocyte inflammation and necroptosis by inhibiting the NF-κB signaling pathway and RIP3 activation instead of RIP1 activation. AZ-628 also reduces osteoclast activity, proliferation and differentiation, and release of inflammatory substances by inhibiting autophagy, MAPK, and NF-κB pathways. Similarly, the in vivo study demonstrated that AZ-628 could inhibit chondrocyte breakdown and lower osteoclast formation and bone resorption, thereby slowing down subchondral bone changes induced by dynamic bone remodeling and reversing the progression of osteoarthritis in mice. The results of this study indicate that AZ-628 could be used to treat OA byinhibiting chondrocyte necroptosis and regulating osteoclast formation.


Subject(s)
Chondrocytes , Osteoarthritis , Animals , Chondrocytes/metabolism , Mice , NF-kappa B/metabolism , Necroptosis , Osteoarthritis/metabolism , Osteoclasts/metabolism , Quinazolines , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL