Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
Nat Commun ; 15(1): 5791, 2024 Jul 10.
Article in English | MEDLINE | ID: mdl-38987295

ABSTRACT

Long-term reconstituting haematopoietic stem cells (LT-HSCs) are used to treat blood disorders via stem cell transplantation. The very low abundance of LT-HSCs and their rapid differentiation during in vitro culture hinders their clinical utility. Previous developments using stromal feeder layers, defined media cocktails, and bioengineering have enabled HSC expansion in culture, but of mostly short-term HSCs and progenitor populations at the expense of naive LT-HSCs. Here, we report the creation of a bioengineered LT-HSC maintenance niche that recreates physiological extracellular matrix organisation, using soft collagen type-I hydrogels to drive nestin expression in perivascular stromal cells (PerSCs). We demonstrate that nestin, which is expressed by HSC-supportive bone marrow stromal cells, is cytoprotective and, via regulation of metabolism, is important for HIF-1α expression in PerSCs. When CD34+ve HSCs were added to the bioengineered niches comprising nestin/HIF-1α expressing PerSCs, LT-HSC numbers were maintained with normal clonal and in vivo reconstitution potential, without media supplementation. We provide proof-of-concept that our bioengineered niches can support the survival of CRISPR edited HSCs. Successful editing of LT-HSCs ex vivo can have potential impact on the treatment of blood disorders.


Subject(s)
Extracellular Matrix , Hematopoietic Stem Cells , Hypoxia-Inducible Factor 1, alpha Subunit , Nestin , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Animals , Nestin/metabolism , Nestin/genetics , Extracellular Matrix/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Mice , Stem Cell Niche , Hydrogels/chemistry , Bioengineering/methods , Humans , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/cytology , Hematopoietic Stem Cell Transplantation , Antigens, CD34/metabolism , Collagen Type I/metabolism , Cell Differentiation , Mice, Inbred C57BL
2.
Microbiology (Reading) ; 170(6)2024 Jun.
Article in English | MEDLINE | ID: mdl-38916198

ABSTRACT

Bacterial infection is a dynamic process resulting in a heterogenous population of infected and uninfected cells. These cells respond differently based on their bacterial load and duration of infection. In the case of infection of macrophages with Crohn's disease (CD) associated adherent-invasive Escherichia coli (AIEC), understanding the drivers of pathogen success may allow targeting of cells where AIEC replicate to high levels. Here we show that stratifying immune cells based on their bacterial load identifies novel pathways and therapeutic targets not previously associated with AIEC when using a traditional homogeneous infected population approach. Using flow cytometry-based cell sorting we stratified cells into those with low or high intracellular pathogen loads, or those which were bystanders to infection. Immune cells transcriptomics revealed a diverse response to the varying levels of infection while pathway analysis identified novel intervention targets that were directly related to increasing intracellular AIEC numbers. Chemical inhibition of identified targets reduced AIEC intracellular replication or inhibited secretion of tumour necrosis factor alpha (TNFα), a key cytokine associated with AIEC infection. Our results have identified new avenues of intervention in AIEC infection that may also be applicable to CD through the repurposing of already available inhibitors. Additionally, they highlight the applicability of immune cell stratification post-infection as an effective approach for the study of microbial pathogens.


Subject(s)
Crohn Disease , Escherichia coli Infections , Escherichia coli , Macrophages , Tumor Necrosis Factor-alpha , Crohn Disease/microbiology , Crohn Disease/immunology , Macrophages/microbiology , Macrophages/immunology , Humans , Escherichia coli Infections/microbiology , Escherichia coli Infections/immunology , Escherichia coli/genetics , Tumor Necrosis Factor-alpha/metabolism , Bacterial Load , Bacterial Adhesion , Host-Pathogen Interactions
3.
Biomater Adv ; 148: 213370, 2023 May.
Article in English | MEDLINE | ID: mdl-36931082

ABSTRACT

Tissue engineered cartilage for external ear reconstruction of congenital deformities, such as microtia or resulting from trauma, remains a significant challenge for plastic and reconstructive surgeons. Current strategies involve harvesting autologous costal cartilage or expanding autologous chondrocytes ex vivo. However, these procedures often lead to donor site morbidity and a cell source with limited expansion capacity. Stromal stem cells such as perivascular stem cells (pericytes) offer an attractive alternative cell source, as they can be isolated from many human tissues, readily expanded in vitro and possess chondrogenic differentiation potential. Here, we successfully isolate CD146+ pericytes from the microtia remnant from patients undergoing reconstructive surgery (Microtia pericytes; MPs). Then we investigate their chondrogenic potential using the polymer poly(ethyl acrylate) (PEA) to unfold the extracellular matrix protein fibronectin (FN). FN unfolding exposes key growth factor (GF) and integrin binding sites on the molecule, allowing tethering of the chondrogenic GF transforming growth factor beta 1 (TGFß1). This system leads to solid-phase, matrix-bound, GF presentation in a more physiological-like manner than that of typical chondrogenic induction media (CM) formulations that tend to lead to off-target effects. This simple and controlled material-based approach demonstrates similar chondrogenic potential to CM, while minimising proclivity toward hypertrophy, without the need for complex induction media formulations.


Subject(s)
Congenital Microtia , Humans , Congenital Microtia/surgery , Pericytes , Chondrogenesis , Fibronectins , Cartilage
4.
Nat Commun ; 14(1): 753, 2023 02 10.
Article in English | MEDLINE | ID: mdl-36765065

ABSTRACT

Mesenchymal stromal cells (MSCs) are multipotent progenitor cells that are of considerable clinical potential in transplantation and anti-inflammatory therapies due to their capacity for tissue repair and immunomodulation. However, MSCs rapidly differentiate once in culture, making their large-scale expansion for use in immunomodulatory therapies challenging. Although the differentiation mechanisms of MSCs have been extensively investigated using materials, little is known about how materials can influence paracrine activities of MSCs. Here, we show that nanotopography can control the immunomodulatory capacity of MSCs through decreased intracellular tension and increasing oxidative glycolysis. We use nanotopography to identify bioactive metabolites that modulate intracellular tension, growth and immunomodulatory phenotype of MSCs in standard culture and during larger scale cell manufacture. Our findings demonstrate an effective route to support large-scale expansion of functional MSCs for therapeutic purposes.


Subject(s)
Mesenchymal Stem Cells , Mesenchymal Stem Cells/metabolism , Multipotent Stem Cells/metabolism , Cell Differentiation , Immunomodulation , Phenotype
5.
Mater Today Bio ; 16: 100367, 2022 Dec.
Article in English | MEDLINE | ID: mdl-35937570

ABSTRACT

Mesenchymal stem cell (MSC)-based tissue engineering strategies are of interest in the field of bone tissue regenerative medicine. MSCs are commonly investigated in combination with growth factors (GFs) and biomaterials to provide a regenerative environment for the cells. However, optimizing how biomaterials interact with MSCs and efficiently deliver GFs, remains a challenge. Here, via plasma polymerization, tissue culture plates are coated with a layer of poly (ethyl acrylate) (PEA), which is able to spontaneously permit fibronectin (FN) to form fibrillar nanonetworks. However, vitronectin (VN), another important extracellular matrix (ECM) protein forms multimeric globules on the polymer, thus not displaying functional groups to cells. Interestingly, when FN and VN are co-absorbed onto PEA surfaces, VN can be entrapped within the FN fibrillar nanonetwork in the monomeric form providing a heterogeneous, open ECM network. The combination of FN and VN promote MSC adhesion and leads to enhanced GF binding; here we demonstrate this with bone morphogenetic protein-2 (BMP2). Moreover, MSC differentiation into osteoblasts is enhanced, with elevated expression of osteopontin (OPN) and osteocalcin (OCN) quantified by immunostaining, and increased mineralization observed by von Kossa staining. Osteogenic intracellular signalling is also induced, with increased activity in the SMAD pathway. The study emphasizes the need of recapitulating the complexity of native ECM to achieve optimal cell-material interactions.

6.
Biomaterials ; 286: 121568, 2022 07.
Article in English | MEDLINE | ID: mdl-35580474

ABSTRACT

Hematopoietic stem cells (HSCs) are fundamental to the generation of the body's blood and immune cells. They reside primarily within the bone marrow (BM) niche microenvironment, which provides signals responsible for the regulation of HSC activities. While our understanding of these signalling mechanisms continues to improve, our ability to recapitulate them in vitro to harness the clinical potential of the HSC populations is still lacking. Recent studies have applied novel engineering techniques combined with traditional in vitro work to establish ex vivo BM niche models. These models exhibit promising potential for research and clinical applications. In this review, BM niche factors that regulate the HSCs in vivo are discussed and their applications in the engineering of BM biomaterial-based platforms are considered. Many questions remain regarding the heterogeneity of niche components and the interactions of HSCs with their microenvironment. A greater understanding of the niche would help to elucidate these remaining questions, leading to the development of novel therapeutic tools.


Subject(s)
Bone Marrow , Stem Cell Niche , Bioengineering , Biology , Bone Marrow Cells , Hematopoietic Stem Cells/physiology
7.
Bioact Mater ; 15: 145-159, 2022 Sep.
Article in English | MEDLINE | ID: mdl-35386337

ABSTRACT

Nanotopography presents an effective physical approach for biomaterial cell manipulation mediated through material-extracellular matrix interactions. The extracellular matrix that exists in the cellular microenvironment is crucial for guiding cell behaviours, such as determination of integrin ligation and interaction with growth factors. These interactions with the extracellular matrix regulate downstream mechanotransductive pathways, such as rearrangements in the cytoskeleton and activation of signal cascades. Protein adsorption onto nanotopography strongly influences the conformation and distribution density of extracellular matrix and, therefore, subsequent cell responses. In this review, we first discuss the interactive mechanisms of protein physical adsorption on nanotopography. Secondly, we summarise advances in creating nanotopographical features to instruct desired cell behaviours. Lastly, we focus on the cellular mechanotransductive pathways initiated by nanotopography. This review provides an overview of the current state-of-the-art designs of nanotopography aiming to provide better biomedical materials for the future.

8.
Biomaterials ; 280: 121263, 2022 01.
Article in English | MEDLINE | ID: mdl-34810036

ABSTRACT

Post-operative infection is a major complication in patients recovering from orthopaedic surgery. As such, there is a clinical need to develop biomaterials for use in regenerative surgery that can promote mesenchymal stem cell (MSC) osteospecific differentiation and that can prevent infection caused by biofilm-forming pathogens. Nanotopographical approaches to pathogen control are being identified, including in orthopaedic materials such as titanium and its alloys. These topographies use high aspect ratio nanospikes or nanowires to prevent bacterial adhesion but these features also significantly reduce MSC adhesion and activity. Here, we use a poly (ethyl acrylate) (PEA) polymer coating on titanium nanowires to spontaneously organise fibronectin (FN) and to deliver bone morphogenetic protein 2 (BMP2) to enhance MSC adhesion and osteospecific signalling. Using a novel MSC-Pseudomonas aeruginosa co-culture, we show that the coated nanotopographies protect MSCs from cytotoxic quorum sensing and signalling molecules, enhance MSC adhesion and osteoblast differentiation and reduce biofilm formation. We conclude that the PEA polymer-coated nanotopography can both support MSCs and prevent pathogens from adhering to a biomaterial surface, thus protecting from biofilm formation and bacterial infection, and supporting osteogenic repair.


Subject(s)
Fibronectins , Mesenchymal Stem Cells , Bacterial Adhesion , Biofilms , Cell Adhesion , Cell Differentiation , Fibronectins/metabolism , Humans , Osteogenesis , Virulence Factors/metabolism
9.
Biomed Res Int ; 2020: 4791786, 2020.
Article in English | MEDLINE | ID: mdl-32190665

ABSTRACT

Heightened activity of osteoclast is considered to be the culprit in breaking the balance during bone remodeling in pathological conditions, such as osteoporosis. As a "foe" of skeletal health, many antiosteoporosis therapies aim to inhibit osteoclastogenesis. However, bone remodeling is a dynamic process that requires the subtle coordination of osteoclasts and osteoblasts. Severe suppression of osteoclast differentiation will impair bone formation because of the coupling effect. Thus, understanding the complex roles of osteoclast in maintaining proper bone remodeling is highly warranted to develop better management of osteoporosis. This review aimed to determine the varied roles of osteoclasts in maintaining skeletal health and to highlight the positive roles of osteoclasts in maintaining normal bone remodeling. Generally, osteoclasts interact with osteocytes to initiate targeted bone remodeling and have crosstalk with mesenchymal stem cells and osteoblasts via secreted factors or cell-cell contact to promote bone formation. We believe that a better outcome of bone remodeling disorders will be achieved when proper strategies are made to coordinate osteoclasts and osteoblasts in managing such disorders.


Subject(s)
Bone Remodeling/physiology , Osteoclasts/physiology , Animals , Bone Resorption , Cell Differentiation , Humans , Mesenchymal Stem Cells , Osteoblasts/physiology , Osteocytes , Osteogenesis , Osteoporosis
10.
J Biomed Mater Res A ; 108(9): 1792-1805, 2020 09.
Article in English | MEDLINE | ID: mdl-32198815

ABSTRACT

Chronic inflammation and infection in the tissue surrounding implants after total joint replacement is closely associated with the innate immune response to surgical implants. Wear particles are known to increase apoptosis and impair the innate immunity in macrophages, which can cause immunosuppression around the implants. Excessive autophagy can induce apoptosis. However, the link between autophagy and apoptosis in macrophages during chronic inflammation and infection remains unknown. In this study, we investigated the autophagy and apoptosis induced by titanium particles in RAW264.7 macrophages, and in the interface membrane of patients with late-onset periprosthetic joint infection (PJI). We found that titanium particles stimulated autophagy and apoptosis in macrophages. Inhibition of autophagy significantly reduced titanium particle-induced apoptosis in macrophages, which may be related to the PI3K/Akt signaling pathway. The secretion of inflammatory factors, such as IL-1ß, IL-6, and TNF-α, decreased after inhibition of autophagy in titanium particle-stimulated macrophages, which may be caused by immune dysfunction due to titanium particle-induced autophagy and apoptosis in macrophages. Furthermore, our in vivo mouse calvarial model also showed that autophagy inhibitors lowered the rate of cell apoptosis. Our findings indicate that wear particle-induced apoptosis may be caused by enhanced autophagy in macrophages, which could potentially impair the local innate immunity in periprosthetic tissues and could be a risk factor for PJI. Based on these results, autophagy modulators may act as a new therapeutic option for PJI.


Subject(s)
Apoptosis/drug effects , Biocompatible Materials/adverse effects , Macrophages/drug effects , Signal Transduction/drug effects , Titanium/adverse effects , Animals , Autophagy/drug effects , Macrophages/immunology , Mice , Phosphatidylinositol 3-Kinases/immunology , Prostheses and Implants/adverse effects , Proto-Oncogene Proteins c-akt/immunology , RAW 264.7 Cells
11.
J Orthop Res ; 38(5): 1101-1112, 2020 05.
Article in English | MEDLINE | ID: mdl-31808572

ABSTRACT

The majority of periprosthetic joint infections occur shortly after primary joint replacement (<3 months) and require the removal of all implant components for the treatment period (~4 months). A clinically relevant animal model of periprosthetic infection should, therefore, establish an infection with implant components in place. Here, we describe a joint replacement model in the rat with ultrahigh molecular weight polyethylene (UHMWPE) and titanium components inoculated at the time of surgery by methicillin-sensitive Staphylococcus aureus (S. aureus), which is one of the main causative microorganisms of periprosthetic joint infections. We monitored the animals for 4 weeks by measuring gait, weight-bearing symmetry, von Frey testing, and micro-CT as our primary endpoint analyses. We also assessed the infection ex vivo using colony counts on the implant surfaces and histology of the surrounding tissues. The results confirmed the presence of a local infection for 4 weeks with osteolysis, loosening of the implants, and clinical infection indicators such as redness, swelling, and increased temperature. The utility of specific gait analysis parameters, especially temporal symmetry, hindlimb duty factor imbalance, and phase dispersion was identified in this model for assessing the longitudinal progression of the infection, and these metrics correlated with weight-bearing asymmetry. We propose to use this model to study the efficacy of using different local delivery regimens of antimicrobials on addressing periprosthetic joint infections. Statement of clinical significance: We have established a preclinical joint surgery model, in which postoperative recovery can be monitored over a multi-week course by assessing gait, weight-bearing, and allodynia. This model can be used to study the efficacy of different combinations of implant materials and medication regimens. © 2019 Orthopaedic Research Society. Published by Wiley Periodicals, Inc. J Orthop Res 38:1101-1112, 2020.


Subject(s)
Arthritis, Infectious , Disease Models, Animal , Prosthesis-Related Infections , Animals , Bone and Bones/pathology , Gait Analysis , Hyperalgesia , Male , Pregnancy-Associated alpha 2-Macroglobulins/metabolism , Rats, Sprague-Dawley , X-Ray Microtomography
12.
Inflammation ; 41(4): 1498-1507, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29728804

ABSTRACT

Aseptic implant loosening is a devastating long-term complication of total joint arthroplasty. It is mainly initiated by the interaction of wear debris and macrophages. However, how does the chronic inflammation persist and how to stop it is poorly understood. Sphingosine kinases (SPHKs) are an essential feature of immunosuppressive M2 polarisation in macrophages and a promoter for chronic inflammation. In this study, RAW 264.7 macrophages were exposed to stimulation with titanium particles (0.1 mg/ml), and the subsequent expression of SPHKs and pro-inflammatory cytokines was evaluated. The effect of inhibitors of SPHKs (FTY720, PF543, and ABC294640) on titanium particle-challenged macrophages was analysed. As for results, the amount of sphingosine kinase (SPHK)-1 and SPHK-2 in RAW264.7 macrophages increased in the presence of titanium particles in a time-dependent manner. Two inhibitors of SPHKs (FTY720 and ABC294640) suppressed titanium particle-induced tumour necrosis factor (TNF)-α and interleukin (IL)-6 production in RAW264.7 macrophages. These findings suggest that persistent stimulation with titanium particles may lead to a consistent release of TNF-α and IL-6 via SPHK-2 activity, which may lead to aseptic implant loosening. Appropriate regulation of SPHK-2 may serve as a potential new strategy in the treatment of aseptic implant loosening.


Subject(s)
Inflammation/chemically induced , Interleukin-6/metabolism , Particulate Matter/adverse effects , Phosphotransferases (Alcohol Group Acceptor)/pharmacology , Titanium/adverse effects , Tumor Necrosis Factor-alpha/metabolism , Animals , Mice , Prosthesis Failure/drug effects , RAW 264.7 Cells , Tumor Necrosis Factor-alpha/drug effects
13.
Mol Med Rep ; 18(1): 541-549, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29749508

ABSTRACT

The aim of the present study was to investigate the role of microRNA (miR)­27a­3p in osteoarthritis (OA). Reverse transcription­quantitative polymerase chain reaction and western blotting were performed to determine the expression of miR­27a­3p and aggrecanase­2 (ADAMTS5) in cartilage tissues from patients with OA and healthy controls, and also in interleukin (IL)­1ß­treated primary human chondrocytes. Primary human chondrocytes were transfected with miR­27a­3p. A luciferase reporter assay was used to validate the direct contact between miR­27a­3p and its putative binding site in the 3'­untranslated region ADAMTS5 mRNA. Furthermore, the effects of IL­1ß­induced activation of mitogen­activated protein kinase (MAPK) and nuclear factor (NF)­κB on miR­27a­3p were evaluated using specific inhibitors. The results revealed that the level of miR­27a­3p was reduced in OA cartilage tissues compared with those of normal controls. In addition, decreased miR­27a­3p and increased ADAMTS5 expression was observed in a time­ and dose­dependent manner in chondrocytes treated with IL­1ß. Furthermore, overexpression of miR­27a­3p suppressed the expression of ADAMTS5 in human chondrocytes induced by IL­1ß. miR­27a­3p overexpression also decreased the luciferase activity of the wild­type ADAMTS5 reporter plasmid. Mutation of the miR­27a­3p binding site in the 3'­untranslated region of ADAMTS5 mRNA abolished the miR­27a­3p­mediated repression of reporter activity. Furthermore, the use of specific inhibitors demonstrated that IL­1ß may regulate miR­27a­3p expression via NF­κB and MAPK signaling pathways in chondrocytes. The present study concluded that miR­27a­3p was downregulated in human OA and was suppressed by IL­1ß, and functions as a crucial regulator of ADAMTS5 in OA chondrocytes. In addition, IL­1ß­mediated suppression of miR­27a­3p activity may occur via the MAPK and NF­κB pathways. The present study may provide a novel strategy for clinical treatment of OA caused by upregulation of miR­27a­3p.


Subject(s)
Cartilage, Articular/metabolism , Interleukin-1beta/metabolism , MicroRNAs/genetics , Osteoarthritis, Knee/metabolism , Signal Transduction , ADAMTS5 Protein/genetics , Adolescent , Adult , Aged , Cartilage, Articular/enzymology , Cells, Cultured , Child , Female , Gene Expression Regulation , Humans , Male , MicroRNAs/metabolism , Middle Aged , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Osteoarthritis, Knee/enzymology , Osteoarthritis, Knee/etiology
14.
Inflammation ; 41(2): 614-625, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29294242

ABSTRACT

Macrophages play an essential role in inflammation. Protein disulfide isomerase (PDI) is central to the redox system, which is closely linked with the inflammatory function of macrophages. However, the relationship between PDI and inflammation is still unknown. In this study, we tested the effects of PDI on inflammatory responses in RAW 264.7 macrophages stimulated with lipopolysaccharide (LPS). Using CRISPR/Cas9 system, we found that PDI knockout suppressed migration, M1 polarization, and secretion of tumor necrosis factor-α (TNF-α) and interluekin-6 (IL-6). The repression of these inflammatory processes was accompanied by decreased production of reactive oxygen species (ROS). PDI ablation also inactivated the phosphorylation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) and activated the phosphorylation of NF-κB inhibitor alpha (IκBα). These findings demonstrate that PDI knockout inhibits the inflammatory function of macrophages by decreasing ROS production and inactivating NF-κB pathway.


Subject(s)
Inflammation/genetics , Macrophages/immunology , NF-kappa B/metabolism , Protein Disulfide-Isomerases/genetics , Reactive Oxygen Species/metabolism , Animals , Gene Knockout Techniques , Mice , Oxidation-Reduction , Phosphorylation , RAW 264.7 Cells , Signal Transduction
15.
Inflammation ; 39(2): 775-85, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26878849

ABSTRACT

Aseptic implant loosening is closely associated with chronic inflammation induced by implant wear debris, and reactive oxygen species (ROS) play an important role in this process. Resveratrol, a plant compound, has been reported to act as an antioxidant in many inflammatory conditions; however, its protective effect and mechanism against wear particle-induced oxidative stress remain unknown. In this study, we evaluated resveratrol's protective effects against wear particle-induced oxidative stress in RAW 264.7 macrophages. At non-toxic concentrations, resveratrol showed dose-dependent inhibition of nitric oxide (NO) production, ROS generation, and lipid peroxidation. It also downregulated the gene expression of oxidative enzymes, including inducible nitric oxide synthase (iNOS) and nicotinamide adenine dinucleotide phosphate (NADPH) oxidase (NOX)-1 and NOX-2, and promoted the gene expression and activities of antioxidant enzymes, including catalase (CAT), superoxide dismutase (SOD), glutathione reductase (GR), and glutathione peroxidase (GPx). This protective effect against wear particle-induced oxidative stress was accompanied by a reduction of gene expression and release of tumor necrosis factor-α (TNF-α), and decreased gene expression and phosphorylation of nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB). These findings demonstrate that resveratrol can inhibit wear particle-induced oxidative stress in macrophages, and may exert its antioxidant effect and protect against aseptic implant loosening.


Subject(s)
Antioxidants/therapeutic use , Macrophages/pathology , NF-kappa B/metabolism , Osteolysis/prevention & control , Oxidative Stress/drug effects , Prostheses and Implants/adverse effects , Stilbenes/therapeutic use , Titanium/adverse effects , Animals , Catalase/metabolism , Cell Line , Female , Glutathione Peroxidase/metabolism , Glutathione Reductase/metabolism , Lipid Peroxidation/drug effects , Membrane Glycoproteins/biosynthesis , Mice , Mice, Inbred C57BL , NADH, NADPH Oxidoreductases/biosynthesis , NADPH Oxidase 1 , NADPH Oxidase 2 , NADPH Oxidases/biosynthesis , Nitric Oxide/metabolism , Nitric Oxide Synthase Type II/biosynthesis , Osteolysis/immunology , Osteolysis/pathology , RAW 264.7 Cells , Reactive Oxygen Species/metabolism , Resveratrol , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...