Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Death Dis ; 14(2): 88, 2023 02 07.
Article in English | MEDLINE | ID: mdl-36750550

ABSTRACT

Osteoblast apoptosis plays an important role in age-related bone loss and osteoporosis. Our previous study revealed that advanced oxidation protein products (AOPPs) could induce nicotinamide adenine dinucleotide phosphate oxidase (NOX)-derived reactive oxygen species (ROS) production, cause mitochondrial membrane potential (ΔΨm) depolarization, trigger the mitochondria-dependent intrinsic apoptosis pathway, and lead to osteoblast apoptosis and ultimately osteopenia and bone microstructural destruction. In this study, we found that AOPPs also induced mitochondrial ROS (mtROS) generation in osteoblastic MC3T3-E1 cells, which was closely related to NOX-derived ROS, and aggravated the oxidative stress condition, thereby further promoting apoptosis. Removing excessive ROS and damaged mitochondria is the key factor in reversing AOPP-induced apoptosis. Here, by in vitro studies, we showed that rapamycin further activated PINK1/Parkin-mediated mitophagy in AOPP-stimulated MC3T3-E1 cells and significantly alleviated AOPP-induced cell apoptosis by eliminating ROS and damaged mitochondria. Our in vivo studies revealed that PINK1/Parkin-mediated mitophagy could decrease the plasma AOPP concentration and inhibit AOPP-induced osteoblast apoptosis, thus ameliorating AOPP accumulation-related bone loss, bone microstructural destruction and bone mineral density (BMD) loss. Together, our study indicated that therapeutic strategies aimed at upregulating osteoblast mitophagy and preserving mitochondrial function might have potential for treating age-related osteoporosis.


Subject(s)
Advanced Oxidation Protein Products , Mitophagy , Advanced Oxidation Protein Products/metabolism , Apoptosis , Osteoblasts/metabolism , Protein Kinases/metabolism , Reactive Oxygen Species/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Mice
2.
J Colloid Interface Sci ; 639: 193-202, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36805744

ABSTRACT

The high entropy alloy is a powerful material due to its high hardness, strength, magnetic performance, corrosion resistance, and temperature stability. Moreover, when combined with reduced graphene oxide (rGO), it formed a novel material for electromagnetic (EM) absorption. In this work, monodisperse high entropy alloy nanocrystals combined with rGO to create a new type of high entropy alloy/rGO EM absorption material. A colloidal synthesis strategy was used to prepare high entropy Pt18Ni26Fe15Co14Cu27 nanocrystals with a small size of around 3.3 nm. These nanocrystals then in situ grew uniformly on the surface of rGO to form Pt18Ni26Fe15Co14Cu27/rGO nanocomposite, which were then characterized and tested for EM absorption performance. Compared to the pure high entropy Pt18Ni26Fe15Co14Cu27 nanocrystals, the composite exhibited an improved EM absorption performance with a minimum reflection loss of -41.8 dB at 4.9 GHz and efficient EM wave absorption up to a bandwidth of 2.5 GHz in the 9.4-11.9 GHz band. This novel high entropy alloy/rGO composite has great potential to be used as an excellent material for EM wave absorption.

3.
Biochem Biophys Res Commun ; 636(Pt 2): 87-96, 2022 12 25.
Article in English | MEDLINE | ID: mdl-36368159

ABSTRACT

To verify the osteoclast differentiation ability of MDSCs from mice of different ages and explore the effect of AOPPs on the osteoclast differentiation of bone marrow MDSCs. Bone marrow cells from C57BL/6 (a.k.a C57) mice of different ages were subjected to flow cytometry, and CD11b+Ly6C+Ly6G+ MDSCs were sorted out. After induction of osteoclast differentiation, these cells were subjected to tartrate-resistant acid phosphatase (TRAP) and F-actin. MDSCs from bone marrows of old mice were injected into the tibial medullary cavity of young mice. One week later, the bone marrows were subjected to histological examination, TRAP, and cell count. MDSCs from bone marrows of old mice were sorted for induction of osteoclast differentiation, intervened with reactive oxygen species (ROS) scavenger, inducible nitric oxide synthase (iNOS) inhibitor, and nitric oxide (NO) scavenger, and then subjected to TRAP. 8-weeks-old C57 mice were injected with the same concentrations of either AOPPs or mouse serum albumin (MSA). Four weeks later, MDSCs from bone marrows were sorted and subjected to induction of osteoclast differentiation, followed by IHC staining and TRAP. MDSCs of 8-weeks-old C57 mice were extracted and subjected to in vitro induction of osteoclast differentiation with different concentrations of AOPPs, followed by TRAP training. The number of MDSCs in the bone marrows of old mice was significantly higher than that in young mice. MDSCs from bone marrows of old mice differentiated into large multinucleated TRAP+ osteoclasts, which were significantly different from those in the middle-aged and young mice in terms of cell quantity and morphology. The actin rings formed in the differentiated osteoclasts from MDSCs of bone marrows were densely distributed in the whole field of view, which were significantly denser than those in the middle-aged and young mice. After injection of MDSCs of old mice, the number of TRAP + osteoclasts in the tibial medullary cavity of young mice was significantly increased. NO inhibitor can significantly inhibit the osteoclast differentiation capacity of MDSCs from bone marrows of old mice. In vivo treatment with AOPPs significantly increased the proportion of MDSCs in the bone marrow, which is up to 55.2%. After injection of AOPPs in 8-week-old mice and induction of osteoclast differentiation from the MDSCs, the ratios of CD11b+ and Gr1+ cells were significantly higher than that in the control and MSA groups but was not significantly different from that in the 15-month-old mice. Upon in vitro treatment with different concentrations of AOPPs, the MDSCs did not show any sign of osteoclast differentiation. MDSCs can directly undergo osteoclast differentiation, the capacity of which is stronger in MDSCs of bone marrows of old mice; the NO pathway is a potential mechanism underlying this phenomenon. In vivo but not in vitro AOPPs treatment can induce osteoclast differentiation of MDSCs, indicating there might be other factors in the body that can interact with AOPPs to induce osteoclast differentiation of MDSCs.


Subject(s)
Myeloid-Derived Suppressor Cells , Osteoclasts , Mice , Animals , Osteoclasts/metabolism , Advanced Oxidation Protein Products/metabolism , Mice, Inbred C57BL , Cell Differentiation , Aging
4.
FASEB J ; 36(9): e22504, 2022 09.
Article in English | MEDLINE | ID: mdl-35980507

ABSTRACT

Vascular calcification is very common in patients with chronic kidney disease (CKD), but so far, there is no effective treatment. Dendrobium officinale polysaccharide (DOP), a natural component of Chinese herbal medicine, has been shown to exert anti-inflammatory and anti-apoptotic activity. Inflammation and apoptosis play an essential role in the progression of vascular calcification. However, the exact role and molecular mechanisms of DOP in vascular calcification remain unclear. In this study, we investigated the effects of DOP on vascular calcification using vascular smooth muscle cells (VSMCs), arterial rings, and CKD rats. Alizarin red staining and gene expression analysis revealed that DOP inhibited calcification and osteogenic differentiation of rat VSMCs in a dose-dependent manner. Similarly, ex vivo studies revealed that DOP inhibited the calcification of rat arterial rings. Furthermore, the administration of DOP alleviated vascular calcification in CKD rats. Moreover, DOP treatment suppressed VSMC inflammation and apoptosis. Finally, DOP treatment upregulated mRNA and protein levels of heme oxygenase-1 (HMOX-1); both pharmacological inhibition of HMOX-1 by the HMOX-1 inhibitor zinc protoporphyrin-9ZnPP9 and knockdown of HMOX-1 by siRNA markedly abrogated the suppression of inflammation and osteogenic differentiation of VSMCs by DOP. Collectively, these results suggest that DOP alleviates vascular calcification in CKD by suppressing apoptosis and inflammation via HMOX-1 activation. These results may provide a promising treatment for vascular calcification in CKD.


Subject(s)
Dendrobium , Renal Insufficiency, Chronic , Vascular Calcification , Animals , Anti-Inflammatory Agents/pharmacology , Inflammation/drug therapy , Inflammation/metabolism , Myocytes, Smooth Muscle/metabolism , Osteogenesis , Polysaccharides/metabolism , Polysaccharides/pharmacology , Rats , Renal Insufficiency, Chronic/drug therapy , Renal Insufficiency, Chronic/metabolism , Vascular Calcification/drug therapy , Vascular Calcification/metabolism , Vascular Calcification/prevention & control
5.
Free Radic Biol Med ; 168: 142-154, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33823244

ABSTRACT

Spinal cord injury (SCI) is a devastating injury that characterized by oxidative stress and inflammatory response. Kaempferol is reported to be an anti-neuroinflammation in neurologic disorders. Nevertheless, the role and mechanism of kaempferol in SCI remains unclear. The present study aims to investigate effects of kaempferol on SCI and its possible underlying mechanisms in in vivo and in vitro models. A C5 hemi-contusion injury was induced in Sprague-Dawley rats to investigate the neuroprotective effects of kaempferol after SCI. For in vitro study, the BV2 microglia cell lines were pretreated with or without kaempferol. A combination of molecular and histological methods was used to clarify the mechanism and explore the signaling pathway both in vivo and in vitro. One-way analysis of variance (ANOVA) was conducted with Bonferroni post hoc tests to examine the differences between groups. The in vivo studies showed that kaempferol could improve the recovery of hindlimb motor function and ameliorate tissue damage in the spinal cord after SCI. Moreover, administration of kaempferol reduced microglia activation and oxidative stress level in the spinal cord. The in vitro studies showed that kaempferol suppressed the microglia activation resulting from the administration of LPS with ATP to BV-2 cells. Pretreated BV2 cells with kaempferol reduced the generation of reactive oxygen species (ROS) by inhibiting NADPH oxidase 4, and then, suppressed the phosphorylation of p38 MAPK and JNK, which subsequently inhibited nuclear translocation of NF-κB p65 to express pro-inflammatory factors. We also observed that kaempferol could inhibite the pyroptosis related proteins (NLRP3 Caspase-1 p10 ASC N-GSDMD) and reduce the release of IL-18 and IL-1ß. In conclusion, kaempferol was able to reduce oxidative stress and inflammatory response through down-regulation of ROS dependent MAPKs- NF-κB and pyroptosis signaling pathway, which suggested that kaempferol might be a novel promising therapeutic agent for SCI.


Subject(s)
NF-kappa B , Spinal Cord Injuries , Animals , Inflammation/drug therapy , Kaempferols/pharmacology , Microglia/metabolism , NF-kappa B/genetics , NF-kappa B/metabolism , Pyroptosis , Rats , Rats, Sprague-Dawley , Signal Transduction , Spinal Cord Injuries/drug therapy
6.
J Neuroinflammation ; 17(1): 90, 2020 Mar 20.
Article in English | MEDLINE | ID: mdl-32192500

ABSTRACT

BACKGROUND: Inflammatory response mediated by oxidative stress is considered as an important pathogenesis of spinal cord injury (SCI). Advanced oxidation protein products (AOPPs) are novel markers of oxidative stress and their role in inflammatory response after SCI remained unclear. This study aimed to investigate the role of AOPPs in SCI pathogenesis and explore the possible underlying mechanisms. METHODS: A C5 hemi-contusion injury was induced in Sprague-Dawley rats to confirm the involvement of AOPPs after SCI. For in vivo study, apocynin, the NADPH oxidase inhibitor was used to study the neuroprotective effects after SCI. For in vitro study, the BV2 microglia cell lines were pretreated with or without the inhibitor or transfected with or without small interference RNA (siRNA) and then stimulated with AOPPs. A combination of molecular and histological methods was used to clarify the mechanism and explore the signaling pathway both in vivo and in vitro. One-way analysis of variance (ANOVA) was conducted with Bonferroni post hoc tests to examine the differences between groups. RESULTS: The levels of AOPPs in plasma and cerebrospinal fluid as well as the contents in the spinal cord showed significant increase after SCI. Meanwhile, apocynin ameliorated tissue damage in the spinal cord after SCI, improving the functional recovery. Immunofluorescence staining and western blot analysis showed activation of microglia after SCI, which was in turn inhibited by apocynin. Pretreated BV2 cells with AOPPs triggered excessive generation of reactive oxygen species (ROS) by activating NADPH oxidase. Increased ROS induced p38 MAPK and JNK phosphorylation, subsequently triggering nuclear translocation of NF-κB p65 to express pro-inflammatory cytokines. Also, treatment of BV2 cells with AOPPs induced NLRP3 inflammasome activation and cleavage of Gasdermin-d (GSDMD), causing pyroptosis. This was confirmed by cleavage of caspase-1, production of downstream mature interleukin (IL)-1ß and IL-18 as well as rupture of rapid cell membrane. CONCLUSIONS: Collectively, these data indicated AOPPs as biomarkers of oxidative stress, modulating inflammatory response in SCI by multiple signaling pathways, which also included the induction of NADPH oxidase dependent ROS, and NLRP3-mediated pyroptosis, and activation of MAPKs and NF-κB.


Subject(s)
Advanced Oxidation Protein Products/metabolism , Microglia/metabolism , Oxidative Stress/physiology , Pyroptosis/physiology , Spinal Cord Injuries/metabolism , Advanced Oxidation Protein Products/pharmacology , Animals , Cell Line , Inflammation/metabolism , Inflammation/pathology , Inflammation/physiopathology , MAP Kinase Signaling System/physiology , Male , Mice , Microglia/drug effects , Microglia/pathology , NF-kappa B/metabolism , Oxidative Stress/drug effects , Pyroptosis/drug effects , Rats , Rats, Sprague-Dawley , Spinal Cord Injuries/pathology , Spinal Cord Injuries/physiopathology
7.
J Orthop Translat ; 20: 73-79, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31908936

ABSTRACT

BACKGROUND AND OBJECTIVE: Existing orthopaedic robotic systems are almost restricted to provide guidance for trajectory direction. In the present study, a novel spinal robotic system with automatic drilling power was introduced. The aim of this study is to evaluate the feasibility and safety in pedicle screw â€‹insertion of posterior lumbar interbody fusion assisted by this novel robotic system. METHODS AND MATERIALS: A randomised controlled trial was conducted for 17 participants who were required posterior lumbar interbody fusion process. Seven (3 M/4 F) were randomly assigned to the robot-assisted group (RA group), and the other ten (4 M/6 F) were assigned to the conventional technique group (FH group). A novel robotic system was used in the RA group. All measurements were based on postoperative computed tomography (CT) data. Accuracy of screw insertion was determined using the Gertzbein and Robbins Scale. Precision was measured by the entry point deviation distance and the trajectory rotation. Other variables included operation time, radiation time, length of stay, and screw-related complications. RESULT: A total of 82 pedicle screws were placed in the 17 participants. In the RA group, 90.6% of screws placed were Grade A, and 9.4% were Grade B. In the FH group, 78.0% of screws were Grade A, 20.0% were Grade B, and 2.0% were Grade C. No statistical difference was found in the operation time, radiation time per case, and length of stay between both groups. The radiation time per screw is significantly lower in the RA group. No screw-related complications or revision occurred in the present study. CONCLUSION: The outcome of screw accuracy of this robotic system was comparable with that of experienced surgeons, and no screw-related complication was found in the RA group during hospitalisation. In addition, radiation time per screw in the robotic group was significantly lower than that in the conventional group, which shows the potential to reduce radiation exposure of pedicle screw fixation assisted by this robotic system. TRANSLATIONAL POTENTIAL: Our study shows that pedicle screw fixation assisted by "Orthbot" system is accurate and safe. It is concluded that this novel robotic system offers a new option for internal implantation in spine surgery.

8.
Redox Biol ; 28: 101306, 2020 01.
Article in English | MEDLINE | ID: mdl-31539804

ABSTRACT

Interleukin (IL)-1ß and tumor necrosis factor (TNF)-α, in particular, control the degeneration of articular cartilage, making them prime targets for osteoarthritis (OA) therapeutic strategies. Advanced oxidation protein products (AOPPs) are prevalent in numerous diseases. Our previous work demonstrates that intra-articular injections of AOPPs accelerate regression of cartilage in OA models. Whether AOPPs exist in the course of OA and their effects on TNF-α and IL-1ß expression in chondrocytes are still unclear. This study confirmed that AOPPs levels in human synovial fluid were positively associated with severity of OA. We also found AOPPs deposition in articular cartilage in anterior cruciate ligament transection (ACLT) induced rodent OA models. AOPPs increased expression of TNF-α and IL-1ß in chondrocytes in vitro, which was inhibited by pre-treatment with SB202190 (p38-MAPK inhibitor) or apocynin (NADPH oxidase inhibitor) or NOX4 knockdown by siRNAs. Subsequently, we further verified in vivo that exogenous injection of AOPPs in OA mice up-regulated expression of TNF-α and IL-1ß in cartilage, which was blocked by treatment with apocynin. In parallel, apocynin attenuated articular cartilage degeneration resulting in substantially lower OARSI scores. Specifically, apocynin reduced NOX4, p-P38, TNF-α and IL-1ß and increased collagen II and glycosaminoglycan (GAG). This study demonstrated that AOPPs increased expression of TNF-α and IL-1ß in chondrocytes via the NADPH oxidase4-dependent and p38-MAPK mediated pathway, and accelerated cartilage degeneration in OA progression. These findings suggest an endogenous pathogenic role of AOPPs in OA progression. Targeting AOPPs-triggered cellular mechanisms might be a promising therapeutic option for patients with OA.


Subject(s)
Advanced Oxidation Protein Products/metabolism , Chondrocytes/cytology , Interleukin-1beta/metabolism , NADPH Oxidase 4/metabolism , Osteoarthritis, Knee/metabolism , Tumor Necrosis Factor-alpha/metabolism , Adult , Advanced Oxidation Protein Products/adverse effects , Aged , Animals , Cells, Cultured , Chondrocytes/metabolism , Disease Models, Animal , Disease Progression , Female , Humans , Male , Mice , Middle Aged , Osteoarthritis, Knee/chemically induced , Severity of Illness Index , Synovial Fluid/metabolism , Up-Regulation
SELECTION OF CITATIONS
SEARCH DETAIL
...