Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
1.
J Nanobiotechnology ; 20(1): 323, 2022 Jul 14.
Article in English | MEDLINE | ID: mdl-35836178

ABSTRACT

In hypoxic dendritic cells (DCs), a low level of Zn2+ can induce the activation of immunogenic DCs (igDCs), thereby triggering an active T-cell response to propel the immune progression of rheumatoid arthritis (RA). This finding indicates the crucial roles of zinc and oxygen homeostasis in DCs during the pathogenesis of RA. However, very few studies have focused on the modulation of zinc and oxygen homeostasis in DCs during RA treatment. Proposed herein is a DC-targeting immune-regulating strategy to induce igDCs into tolerogenic DCs (tDCs) and inhibit subsequent T-cell activation, referred to as ZnO2/Catalase@liposome-Mannose nanoparticles (ZnCM NPs). ZnCM NPs displayed targeted intracellular delivery of Zn2+ and O2 towards igDCs in a pH-responsive manner. After inactivating OTUB1 deubiquitination, the ZnCM NPs promoted CCL5 degradation via NF-κB signalling, thereby inducing the igDC-tDC transition to further inhibit CD4+ T-cell homeostasis. In collagen-induced arthritis (CIA) mice, this nanoimmunoplatform showed significant accumulation in the spleen, where immature DCs (imDCs) differentiated into igDCs. Splenic tDCs were induced to alleviate ankle swelling, improve walking posture and safely inhibit ankle/spleen inflammation. Our work pioneers the combination of DC-targeting nanoplatforms with RA treatments and highlights the significance of zinc and oxygen homeostasis for the immunoregulation of RA by inducing tDCs with modified ZnO2 NPs, which provides novel insight into ion homeostasis regulation for the treatment of immune diseases with a larger variety of distinct metal or nonmetal ions.


Subject(s)
Arthritis, Rheumatoid , Nanoparticles , Zinc Oxide , Animals , Arthritis, Rheumatoid/metabolism , Dendritic Cells , Mice , Oxygen , Peroxides/metabolism , Zinc
2.
J Cell Mol Med ; 23(6): 4395-4407, 2019 06.
Article in English | MEDLINE | ID: mdl-30983153

ABSTRACT

Increasing evidence indicates that osteoarthritis (OA) is a musculoskeletal disease affecting the whole joint, including both cartilage and subchondral bone. Reactive oxygen species (ROS) have been demonstrated to be one of the important destructive factors during early-stage OA development. The objective of this study was to investigate isorhamnetin (Iso) treatment on osteoclast formation and chondrocyte protection to attenuate OA by modulating ROS. Receptor activator of nuclear factor-kappa B ligand (RANKL) was used to establish the osteoclast differentiation model in bone marrow macrophages (BMMs) in vivo. H2 O2 was used to induce ROS, which could further cause chondrocyte apoptosis. We demonstrated that Iso suppressed RANKL-induced ROS generation, which could mediate osteoclastogenesis. Moreover, we found that Iso inhibited osteoclast formation and function by suppressing the expression of osteoclastogenesis-related genes and proteins. We proved that Iso inhibited RANKL-induced activation of mitogen-activated protein kinase activation of mitogen-activated protein kinase (MAPK), nuclear factor-kappa B (NF-κB) and AKT signalling pathways in BMMs. In addition, Iso inhibited ROS-induced chondrocyte apoptosis by regulating apoptosis-related proteins. Moreover, Iso was administered to an anterior cruciate ligament transection (ACLT)-induced OA mouse model. The results indicated that Iso exerted beneficial effects on inhibiting excessive osteoclast activity and chondrocyte apoptosis, which further remedied cartilage damage. Overall, our data showed that Iso is an effective candidate for treating OA.


Subject(s)
Bone Resorption/drug therapy , Chondrocytes/drug effects , Osteoarthritis, Knee/drug therapy , Osteogenesis/drug effects , Protective Agents/pharmacology , Quercetin/analogs & derivatives , Reactive Oxygen Species/metabolism , Animals , Bone Resorption/etiology , Bone Resorption/metabolism , Bone Resorption/pathology , Chondrocytes/metabolism , Homeostasis , Male , Mice , Mice, Inbred C57BL , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , NFATC Transcription Factors/metabolism , Osteoarthritis, Knee/etiology , Osteoarthritis, Knee/metabolism , Osteoarthritis, Knee/pathology , Quercetin/pharmacology , Signal Transduction
3.
Biochem Biophys Res Commun ; 501(2): 547-555, 2018 06 22.
Article in English | MEDLINE | ID: mdl-29746861

ABSTRACT

Osteolytic diseases are closely associated with osteocyte fate, indicating a more efficient and crucial role of osteocyte-targeting strategy in inhibiting osteoclastogenesis. Here, we investigated the effects of lenalidomide (Lena) on osteocyte fate in order to regulate osteoclastogenesis via effective cascade-controlling response. Our data revealed that lenalidomide treatment notably rescued IL-1ß induced loss of osteocyte viability by inhibiting osteocyte apoptosis with decreased osteoclast-related factors, RANKL and Sclerostin, as demonstrated by the restricted osteoclast formation and reduced bone resorption. Additionally, iTRAQ assay revealed that IL-1ß induced activation of NF-κB inhibitor α/ß were remarkably downregulated by lenalidomide, showing that lenalidomide impaired NF-κB signaling in osteocytes for inhibiting the expression of osteoclast specific genes in osteoclasts, which was further confirmed by KEGG pathway analysis and Western blot. More interestingly, the in vivo analysis of osteocyte apoptosis and osteoclastogenesis in osteoarthritis mice model indicated a role of lenalidomide in the regulation of osteocyte fate and the consequent inhibition of RANKL-induced osteoclastogenesis. Together, these results suggest that lenalidomide regulates osteocyte fate by attenuating IL-1ß/NF-κB signaling, thereby inhibiting RANKL expression for the attenuated osteoclastogenesis both in vitro and vivo, indicating a more efficient remedy among future anti-osteoclastogenesis approaches.


Subject(s)
Immunologic Factors/pharmacology , Interleukin-1beta/immunology , NF-kappa B/immunology , Osteocytes/drug effects , RANK Ligand/immunology , Signal Transduction/drug effects , Thalidomide/analogs & derivatives , Animals , Cell Line , Cells, Cultured , Lenalidomide , Mice, Inbred C57BL , Osteoclasts/cytology , Osteoclasts/drug effects , Osteoclasts/immunology , Osteocytes/cytology , Osteocytes/immunology , Osteogenesis/drug effects , Thalidomide/pharmacology
4.
Pharmazie ; 71(5): 285-91, 2016 May.
Article in English | MEDLINE | ID: mdl-27348974

ABSTRACT

Here we aimed to evaluate the effectiveness of esomeprazole treatment strategies comparing with other proton pump inhibitors (PPI) in clinical practice for six months in the management of patients with symptomatic gastroesophageal reflux disease (GERD). An extensive search of the literature focusing on PPI therapeutic evaluation was performed up to December 2014. Risk ratio (RR) with its corresponding 95% confidence intervals (CIs) in each study was chosen as the effect size. Cochrane's Q statistic and I2 test were both conducted to evaluate heterogeneity across individual studies. Meta-regression was conducted to explore the source of heterogeneity and sensitive analysis was performed to assess the risk bias for the meta-analysis. Totally, eleven trials with high quality enrolled in the meta-analysis. Esomeprazole therapy (20 mg daily) had lower relapse rates than other drugs during six months maintenance treatment (RR = 0.67; 95% CI: 0.55-0.83). Heartburn (RR = 0.72; 95% CI: 0.57-0.92) and epigastric pain (RR = 0.82, 95% Cl: 0.70-0.96) were less likely to happen after esomeprazole treatment, and no significant advantage was found on acid regurgitation and dysphagia. Moreover, lower risk for serious adverse events was observed after esomeprazole therapy (RR = 1.40, 95% CI: 1.04-1.88). Blind method or difference controlled drugs did not influence heterogeneity across studies. Moreover, the conclusion on acid regurgitation, abdominal pain and dysphagia might be unstable. In GERD patients, esomeprazole 20 mg daily is more effective than other PPIs regarding relapse rates, symptoms of epigastric pain and heartburn, and serious adverse events.


Subject(s)
Esomeprazole/therapeutic use , Gastroesophageal Reflux/drug therapy , Proton Pump Inhibitors/therapeutic use , Abdominal Pain/drug therapy , Abdominal Pain/etiology , Deglutition Disorders/drug therapy , Deglutition Disorders/etiology , Esomeprazole/adverse effects , Gastroesophageal Reflux/physiopathology , Humans , Proton Pump Inhibitors/adverse effects , Recurrence , Regression Analysis
5.
J Orthop Translat ; 36: 109-119, 2022 Sep.
Article in English | MEDLINE | ID: mdl-36090821

ABSTRACT

Objective: To evaluate the interbody fusion efficacy and biocompatibility of a graft-free cage made of polyetheretherketone/calcium silicate composite/porous tantalum (PEEK/CS/pTa cage) compared with a PEEK/CS cage with an autogenous bone graft in a goat model. Methods: PEEK/CS/pTa and PEEK/CS cages were prepared through an injection-moulding method. The PEEK/CS composites and porous tantalum were characterized by Fourier transform infrared spectroscopy (FTIR), X-ray photoelectron spectroscopy (XPS), X-ray diffraction (XRD), scanning electron microscopy (SEM), and energy-dispersive spectroscopy (EDS) mapping. Then, adult goats were chosen for C2/C3 and C3/C4 discectomy via the anterior cervical approach and randomly implanted with PEEK/CS/pTa and PEEK/CS/cages with autogenous bone grafts. The fusion performance and osseointegration of the cages were evaluated by X-ray imaging, magnetic resonance imaging (MRI) scanning, and bone histomorphometry analysis. Moreover, the concentrations of Ca and Si in urine, serum, tissue around the fusion segments and major organs of the goats were determined by inductively coupled plasma-optical emission spectrometry (ICP-OES). Histological observation of major organs of the goats was used to evaluate the biosafety of PEEK/CS/pTa and PEEK/CS cages. Results: X-ray and MRI imaging suggested that both PEEK/CS/pTa cages and PEEK/CS cages maintained similar average intervertebral space heights. The tissue volumes in the fusion area were comparable between the two groups of cages at 26 weeks after surgery. Histological morphometric data showed that PEEK/CS/pTa cages and PEEK/CS cages with autogenous bone grafts had similar bone contact and osseointegration at 12 and 26 weeks. Element determination of serum, urine, spinal cord, dura matter, bone and organs showed that the CS/PEEK cages did not cause abnormal systemic metabolism or accumulation of calcium and silicon in local tissues and major organs of goats after implantation. No obvious pathological changes were found in the heart, liver, spleen, liver or kidney tissues. Conclusion: Overall, these results suggested that the graft-free PEEK/CS/pTa cage showed similar bony fusion performance to the PEEK/CS cages with autogenous bone grafts. The cages releasing calcium and silicon had good biological safety in vivo.The translational potential of this article: This study provided a new graft-free interbody fusion solution to patients with degenerative disc diseases, which could avert potential donor-site complications. This study also provided a detailed assessment of element excretion and accumulation of Ca and Si in vivo, which validated the biosafety of this new type of bioactive interbody fusion cage.

6.
ACS Appl Mater Interfaces ; 12(2): 2009-2022, 2020 Jan 15.
Article in English | MEDLINE | ID: mdl-31849213

ABSTRACT

Accumulating evidence suggests that activation of proinflammatory M1-type macrophages in the synovium plays a vital role in the progression of osteoarthritis (OA). Redundant nitric oxide (NO) and hydrogen peroxide (H2O2) are key factors that drive macrophages to polarize to the M1 type. Herein, modified zeolitic imidazolate framework-8 (ZIF-8) nanoparticles (NPs) have been synthesized. By regulating intracellular gases and reprogramming the metabolism phenotype, modified NPs transformed macrophage polarization from proinflammatory M1 to anti-inflammatory M2 phenotype. Specifically, S-methylisothiourea hemisulfate salt was loaded into ZIF-8 NPs to inhibit inducible nitric oxide synthase, hence reducing NO production. Catalase was encapsulated to catalyze the production of oxygen (O2) from H2O2. Results demonstrated that modified NPs were capable of catalyzing H2O2 to produce O2 and eliminate NO, hence inhibiting hypoxia-inducible factor 1α, further rescuing mitochondrial function. Moreover, anti-CD16/32 antibody modification could prolong the retention time of NPs in knee joints of OA mice with anterior cruciate ligament transection. More significantly, modified NPs suppressed M1 macrophages and up-regulated M2 macrophage infiltration in the synovium, further inhibiting cartilage degeneration. This ZIF-8 NP-based gas regulation and metabolic reprogramming strategy may pave a new avenue for OA treatment.


Subject(s)
Imidazoles/chemistry , Macrophages/metabolism , Metabolic Networks and Pathways , Nanoparticles/chemistry , Osteoarthritis/pathology , Synovial Membrane/pathology , Zeolites/chemistry , Adenosine Triphosphate/biosynthesis , Animals , Cell Death , Cell Polarity , Chondrocytes/pathology , Disease Progression , Endocytosis , Gases/metabolism , Hypertrophy , Imidazoles/chemical synthesis , Macrophages/pathology , Mice , Mitochondria/metabolism , Nanoparticles/ultrastructure , RAW 264.7 Cells , Zeolites/chemical synthesis
7.
Int J Nanomedicine ; 15: 6355-6372, 2020.
Article in English | MEDLINE | ID: mdl-32922006

ABSTRACT

BACKGROUND: Cerium oxide nanoparticles (CeO2NPs) are potent scavengers of cellular reactive oxygen species (ROS). Their antioxidant properties make CeO2NPs promising therapeutic agents for bone diseases and bone tissue engineering. However, the effects of CeO2NPs on intracellular ROS production in osteoclasts (OCs) are still unclear. Numerous studies have reported that intracellular ROS are essential for osteoclastogenesis. The aim of this study was to explore the effects of CeO2NPs on osteoclast differentiation and the potential underlying mechanisms. METHODS: The bidirectional modulation of osteoclast differentiation by CeO2NPs was explored by different methods, such as fluorescence microscopy, scanning electron microscopy (SEM), transmission electron microscopy (TEM), quantitative real-time polymerase chain reaction (qRT-PCR), and Western blotting. The cytotoxic and proapoptotic effects of CeO2NPs were detected by cell counting kit (CCK-8) assay, TdT-mediated dUTP nick-end labeling (TUNEL) assay, and flow cytometry. RESULTS: The results of this study demonstrated that although CeO2NPs were capable of scavenging ROS in acellular environments, they facilitated the production of ROS in the acidic cellular environment during receptor activator of nuclear factor kappa-Β ligand (RANKL)-dependent osteoclast differentiation of bone marrow-derived macrophages (BMMs). CeO2NPs at lower concentrations (4.0 µg/mL to 8.0 µg/mL) promoted osteoclast formation, as shown by increased expression of Nfatc1 and C-Fos, F-actin ring formation and bone resorption. However, at higher concentrations (greater than 16.0 µg/mL), CeO2NPs inhibited osteoclast differentiation and promoted apoptosis of BMMs by reducing Bcl2 expression and increasing the expression of cleaved caspase-3, which may be due to the overproduction of ROS. CONCLUSION: This study demonstrates that CeO2NPs facilitate osteoclast formation at lower concentrations while inhibiting osteoclastogenesis in vitro by inducing the apoptosis of BMMs at higher concentrations by modulating cellular ROS levels.


Subject(s)
Cell Differentiation , Cerium/chemistry , Osteoclasts/cytology , Reactive Oxygen Species/metabolism , Actins/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Resorption/metabolism , Cell Differentiation/drug effects , Cell Survival/drug effects , Down-Regulation/drug effects , Gene Expression Regulation/drug effects , Macrophages/cytology , Macrophages/drug effects , Macrophages/ultrastructure , Male , Mice, Inbred C57BL , NF-kappa B/metabolism , Nanoparticles/ultrastructure , Osteoclasts/drug effects , Osteogenesis/drug effects , RANK Ligand/pharmacology , Signal Transduction/drug effects , Up-Regulation/drug effects
8.
Theranostics ; 9(13): 3707-3722, 2019.
Article in English | MEDLINE | ID: mdl-31281508

ABSTRACT

Gouty arthritis (GA) is a form of arthritis caused by uric acid deposition in the joints that result in intense inflammation and pain. Accumulating evidence showed the importance of the sensory neurons signal upon immune cells by releasing neuropeptides and chemokines to regulate associated immune-inflammatory response. In this study, we investigated the significance of sensory neuron neuropeptides and chemokine signals on inflammation-induced macrophages polarization during GA. Methods: We screened the mRNA expression profile during GA in dorsal root ganglion (DRG) neurons to identify the most likely candidate that mediates the neuro-immune communication. Then, we silenced specific gene expression in the DRG by lentiviral vectors in the monosodium urate (MSU)-induced ankle GA mouse model and evaluated alterations in the inflammatory response. In vitro, primary macrophages were used to investigate the neural impact on M1/M2 subtype polarization, proinflammatory cytokine production and downstream endothelial damage. Mechanism by which macrophage inflammation is induced in the DRG was evaluated by Western blot, immunofluorescence, and immunoprecipitation. Results: We found that secreted frizzled-related protein 2 (sFRP2) was the most upregulated gene in dorsal root ganglion (DRG) neurons in response to monosodium urate (MSU) deposition. Injection of LV-sFRP2-shRNA into the L4 and L5 DRG significantly suppressed inflammatory cell infiltration and M1 polarization in the synovial membrane, attenuating hyperalgesia and ankle swelling in the GA mouse model. In vitro, DRG neurons-derived sFRP2 promoted M1 polarization and macrophage migration, thereby upregulating the production of proinflammatory cytokines and preventing endothelial apoptosis. Furthermore, DRG-derived sFRP2 activated the nuclear factor (NF)-κB pathway by destabilizing the ß-catenin and p65 complex. Conclusion: We demonstrated the involvement of a sensory neuron-macrophage axis in GA pathology that was regulated by sFRP2 expression in a paracrine manner. Targeting increased sFRP2 expressions in DRG provide novel insights for future GA research in both pain alleviation and treatment of gout inflammation.


Subject(s)
Arthritis, Gouty/pathology , Ganglia, Spinal/pathology , Human Umbilical Vein Endothelial Cells/pathology , Macrophages/pathology , Membrane Proteins/metabolism , Neurons/pathology , Animals , Apoptosis , Arthritis, Gouty/metabolism , Cell Movement , Cell Polarity , Gene Knockdown Techniques , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Inflammation/pathology , Macrophages/metabolism , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , Neurons/metabolism , RAW 264.7 Cells , RNA, Small Interfering/metabolism , Signal Transduction , Uric Acid
9.
Acta Pharm Sin B ; 9(5): 973-985, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31649847

ABSTRACT

The objective was to investigate the effect of kinsenoside (Kin) treatments on macrophage polarity and evaluate the resulting protection of chondrocytes to attenuate osteoarthritis (OA) progression. RAW264.7 macrophages were polarized to M1/M2 subtypes then administered with different concentrations of Kin. The polarization transitions were evaluated with quantitative real-time polymerase chain reaction (qRT-PCR), confocal observation and flow cytometry analysis. The mechanism of Kin repolarizing M1 macrophages was evaluated by Western blot. Further, macrophage conditioned medium (CM) and IL-1ß were administered to chondrocytes. Micro-CT scanning and histological observations were conducted in vivo on anterior cruciate ligament transection (ACLT) mice with or without Kin treatment. We found that Kin repolarized M1 macrophages to the M2 phenotype. Mechanistically, Kin inhibited the phosphorylation of IκBα, which further reduced the downstream phosphorylation of P65 in nuclear factor-κB (NF-κB) signaling. Moreover, Kin inhibited mitogen-activated protein kinases (MAPK) signaling molecules p-JNK, p-ERK and p-P38. Additionally, Kin attenuated macrophage CM and IL-1ß-induced chondrocyte damage. In vivo, Kin reduced the infiltration of M1 macrophages, promoted M2 macrophages in the synovium, inhibited subchondral bone destruction and reduced articular cartilage damage induced by ACLT. All the results indicated that Kin is an effective therapeutic candidate for OA treatment.

SELECTION OF CITATIONS
SEARCH DETAIL