Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
J Orthop Res ; 42(9): 1933-1942, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38520666

ABSTRACT

Osteoarthritis (OA) is a common degenerative joint disease, and subchondral osteosclerosis is an important pathological change that occurs in its late stages. Cardamonin (CD) is a natural flavonoid isolated from Alpinia katsumadai that has anti-inflammatory activity. The objectives of this study were to investigate the therapeutic effects and potential mechanism of CD in regulating OA subchondral osteosclerosis at in vivo and in vitro settings. Eight-week-old male C57BL/6J mice were randomly divided into four groups: sham operation, anterior cruciate ligament transection (ACLT)-induced OA model, low-dose and high-dose CD treated ACLT-OA model groups. Histological assessment and immunohistochemical examinations for chondrocyte metabolism-related markers metalloproteinase-13, ADAMTS-4, Col II, and Sox-9 were performed. Microcomputed tomography was used to assess the sclerosis indicators in subchondral bone. Further, MC3T3-E1 (a mouse calvarial preosteoblast cell line) cells were treated with various concentrations of CD to reveal the influence and potential molecular pathways of CD in osteogenic differentiations. Animal studies suggested that CD alleviated the pathological changes in OA mice such as maintaining integrity and increasing the thickness of hyaline cartilage, decreasing the thickness of calcified cartilage, decreasing the Osteoarthritis Research Society International score, regulating articular cartilage metabolism, and inhibiting subchondral osteosclerosis. In vitro investigation indicated that CD inhibited alkaline phosphatase expression and production of calcium nodules during osteogenic differentiation of MC3T3-E1 cells. In addition, CD inhibited the expression of osteogenic differentiation-related indicators and Wnt/ß-catenin pathway-related proteins. In conclusion, CD inhibits osteogenic differentiation by downregulating Wnt/ß-catenin signaling and alleviating subchondral osteosclerosis in a mouse model of OA.


Subject(s)
Cell Differentiation , Chalcones , Mice, Inbred C57BL , Osteoarthritis , Osteogenesis , Osteosclerosis , Wnt Signaling Pathway , Animals , Male , Chalcones/pharmacology , Chalcones/therapeutic use , Osteogenesis/drug effects , Wnt Signaling Pathway/drug effects , Mice , Osteosclerosis/drug therapy , Osteoarthritis/drug therapy , Osteoarthritis/metabolism , Cell Differentiation/drug effects , Down-Regulation/drug effects , beta Catenin/metabolism
2.
Zhonghua Wei Zhong Bing Ji Jiu Yi Xue ; 33(8): 903-917, 2021 Aug.
Article in Chinese | MEDLINE | ID: mdl-34590555

ABSTRACT

Enteral nutrition plays an irreplaceable role in the nutritional treatment of critically ill patients. In order to help clinical medical staff to manage the common complications during the implementations of enteral nutrition for critically ill patients, the consensus writing team carried out literature retrieval, literature quality evaluation, evidence synthesis. Several topics such as diarrhea, aspiration, high gastric residual volume, abdominal distension, etc. were assessed by evidence-based methodology and Delphi method. After two rounds of expert investigations, Expert consensus on prevention and management of enteral nutrition therapy complications for critically ill patients in China (2021 edition) developed, and provided guidance for clinical medical staff.


Subject(s)
Critical Illness , Enteral Nutrition , China , Consensus , Diarrhea , Enteral Nutrition/adverse effects , Humans
4.
Toxicology ; 442: 152537, 2020 09.
Article in English | MEDLINE | ID: mdl-32663520

ABSTRACT

Long-term exposure to isoflurane may induce long-term developmental neurotoxicity and cognitive impairments in the neonatal brains. Trilobatin, a leaf extract from the Chinese traditional sweet tea Lithocarpus polystachyus Rehd, possesses various biological properties including anti-inflammatory and anti-oxidant properties. Our study aimed to explore the neuroprotective effect of trilobatin on isoflurane-induced neurotoxicity in mouse hippocampal neuronal HT22 cells. The effects of trilobatin on cell viability, LDH release, apoptosis, and caspase-3/7 activity in isoflurane-induced HT22 cells were explored by CCK-8, LDH release assay, flow cytometry analysis, and caspase-3/7 activity assay, respectively. Oxidative stress was evaluated by measuring the levels of reactive oxygen species (ROS) and malonyldialdehyde (MDA) and activities of superoxide dismutase (SOD) and catalase (CAT). The expression of nuclear erythroid-2 related factor 2 (Nrf2), nuclear Nrf2, heme oxygenase-1 (HO-1), and NAD(P)H: quinone oxidoreductase 1 (NQO1) was determined by western blot and qRT-PCR. Results suggested that exposure to isoflurane significantly reduced cell viability and increased LDH release, apoptotic rate and caspase-3/7 activity in HT22 cells, which were abolished by trilobatin. Trilobatin reversed isoflurane-induced increase of ROS and MDA levels and reduction of SOD and CAT activities in HT22 cells. Additionally, trilobatin promoted the nuclear translocation of Nrf2 as well as the mRNA and protein expression of HO-1 and NQO1 in HT22 cells exposed to isoflurane. Nrf2 knockdown attenuated the effects of trilobatin on isoflurane-induced viability reduction, LDH release, apoptosis, and oxidative stress in HT22 cells. Overall, trilobatin protected HT22 cells against isoflurane-induced neurotoxicity via activating the Nrf2/antioxidant response element (ARE) pathway.


Subject(s)
Anesthetics, Inhalation/toxicity , Flavonoids/pharmacology , Isoflurane/antagonists & inhibitors , Isoflurane/toxicity , Major Histocompatibility Complex/drug effects , NF-E2-Related Factor 2/drug effects , Neuroprotective Agents/pharmacology , Neurotoxicity Syndromes/prevention & control , Polyphenols/pharmacology , Signal Transduction/drug effects , Vesicular Transport Proteins/drug effects , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Hippocampus/drug effects , Mice , Plant Extracts/pharmacology , Reactive Oxygen Species/metabolism
5.
J Neurol Sci ; 387: 210-219, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29571866

ABSTRACT

AIMS: Death-associated protein kinase 1 (DAPK1) is a kinase found to promote neuronal apoptosis induced by ischemia. Extracellular signal-regulated kinase (ERK) was identified as a key molecule in DAPK1 signaling. However, the mechanisms of neuronal ischemia reperfusion injury remain unknown. Here, we investigate the influence of DAPK1-ERK signal on neuronal apoptosis following ischemia reperfusion. METHODS: Mouse N2a cells were used in this study and primary cultured neurons along with mice were adopted as supplements. Oxygen glucose deprivation (OGD) or administration of N-methyl-d-aspartate (NMDA) and glycine was performed on cells while middle cerebral artery occlusion (MCAO) model on mice. DAPK1 knocking down was achieved by lentiviral-delivered shRNA. Protein expressions were evaluated by western blots. Protein-protein binding was confirmed by co-immunoprecipitation and immunofluorescent assay. Apoptosis of cells was measured by flow cytometry and lacate dehydrogenase (LDH) leakage assay. RESULTS: Ischemia reperfusion resulted in increased DAPK1 and ERK activation as well as aggravated apoptosis in a time-dependent manner. DAPK1 was proved to bind to ERK during reperfusion following OGD, MCAO and excitotoxicity model. Interception of this binding by knocking down DAPK1 led to nuclear translocation of ERK and reduced apoptosis. CONCLUSION: Our study revealed the DAPK1-ERK signal as a potential mechanism contributing to neuronal apoptosis in response to ischemia reperfusion. Disruption of this signal pathway could be a promising therapeutic target against stroke.


Subject(s)
Apoptosis/physiology , Death-Associated Protein Kinases/metabolism , Glucose/deficiency , Hypoxia/physiopathology , MAP Kinase Signaling System/physiology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cerebral Cortex/cytology , Cytarabine/pharmacology , Death-Associated Protein Kinases/genetics , Disease Models, Animal , Excitatory Amino Acid Agonists/pharmacology , Glycine/pharmacology , Immunosuppressive Agents/pharmacology , Infarction, Middle Cerebral Artery/pathology , MAP Kinase Signaling System/drug effects , Male , Mice , Mice, Inbred C57BL , N-Methylaspartate/pharmacology , Neuroblastoma/pathology , Neurons/drug effects , Protein Biosynthesis/drug effects , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism
6.
Spine J ; 16(10): 1231-1237, 2016 10.
Article in English | MEDLINE | ID: mdl-27208552

ABSTRACT

BACKGROUND: Over half the population of the world will suffer from moderate or severe low back pain (LBP) during their life span. Studies have shown that naringin, a major flavonoid in grapefruit and an active compound extracted from a Chinese herbal medicine (Rhizoma Drynariae) possesses many pharmacological effects. PURPOSE: The aim of this study was to evaluate the influence of naringin on the growth of degenerative human nucleus pulposus (NP) cells, and its repair effects on protein and gene expressions of the cells. STUDY DESIGN/SETTING: This was an in vitro investigation of the human NP cells isolated from degenerated intervertebral discs that were interacted with various concentrated of naringin. METHOD: This study was exempted by the institutional Human Subjects Committee-2, University of Kansas School of Medicine-Wichita. Degenerative human NP cells were isolated from intervertebral discs of patients with discogenic LBP and cultured at 37°C with 5% CO2. The proliferation of NP cells was determined following treatment with various concentrations of naringin. The protein expressions of tumor necrosis factor-α (TNF-α) and Bone morphogenetic protein 2 (BMP-2) were tested using enzyme-linked immunosorbent assay. Aggrecan and type II collagen levels were measured by immunohistological staining. Further examination of the gene expression of aggrecan, Sox6, and MMP3 was performed after intervention with naringin for 3 days. RESULTS: The human NP cells were successfully propagated in culture and stained positive with toluidine blue staining. Naringin effectively enhanced the cell proliferation at an optimal concentration of 20 µg/mL. Naringin treatment resulted in significant inhibition of TNF-α, but elevated protein expressions of BMP-2, collagen II, and aggrecan. Naringin also increased disc matrix gene activity including aggrecan and Sox6, and decreased the gene expression of MMP3. CONCLUSION: Naringin effectively promotes the proliferation of degenerative human NP cells and improves the recuperation of the cells from degeneration by increasing expression of aggrecan, BMP-2, and Sox6 while inhibiting the expression of TNF-α and MMP3. This study suggests that naringin may represent an alternative therapeutic agent for disc degeneration.


Subject(s)
Cell Proliferation/drug effects , Flavanones/pharmacology , Intervertebral Disc Degeneration/pathology , Low Back Pain/pathology , Nucleus Pulposus/drug effects , Aggrecans/metabolism , Bone Morphogenetic Protein 2/metabolism , Cells, Cultured , Collagen Type II/metabolism , Flavanones/therapeutic use , Humans , Intervertebral Disc Degeneration/drug therapy , Intervertebral Disc Degeneration/metabolism , Low Back Pain/drug therapy , Low Back Pain/metabolism , Nucleus Pulposus/metabolism , Nucleus Pulposus/pathology , Tumor Necrosis Factor-alpha/metabolism
7.
Shock ; 44(5): 496-502, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26196843

ABSTRACT

Lung fibrosis is an aggressive disease with uncontrolled fibrotic response and no effective therapeutic treatment. Epithelial-to-mesenchymal transition (EMT) has been proved to be an important pathological feature in lung fibrosis. In this study, we investigated whether MaR1, a kind of proresolving lipid mediators, could inhibit TGF-ß1-induced EMT in vitro and lung fibrosis in vivo. In vitro study, mouse type II alveolar epithelial cells were treated with different does of MaR1 for 30 min and were exposed to TGF-ß1 for 48 h. In vivo study, C57BL/6 mice were administered bleomycin intratracheally. After 14 days, MaR1 was injected intraperitoneally daily for 7 days. In day 28, mice were sacrificed. The results demonstrate that treatment of mouse type II alveolar epithelial cells with MaR1 (10 nM) significantly prevents TGF-ß1-induced fibronectin and α-SMA expression and restores E-Cadherin level. The down-regulation of profibrotic molecules of MaR1 is associated with suppression of Smad2/3 and Akt phosphorylation. In vivo, MaR1 treatment significantly prolongs survival rate and attenuates destruction of lung architecture, as well as collagen deposition after bleomycin inhalation. TGF-ß1 concentration in bronchoalveolar lavage and fibrotic markers (fibronectin and α-SMA) in lung tissues are inhibited by MaR1 administration. These data indicate that MaR1 inhibits TGF-ß1-induced EMT and attenuates bleomycin-induced pulmonary fibrosis. MaR1 may be a promising strategy for alleviation of lung fibrosis.


Subject(s)
Docosahexaenoic Acids/pharmacology , Epithelial-Mesenchymal Transition/drug effects , Pulmonary Fibrosis/prevention & control , Animals , Bleomycin , Cells, Cultured , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/therapeutic use , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Male , Mice, Inbred C57BL , Proto-Oncogene Proteins c-akt/metabolism , Pulmonary Alveoli/drug effects , Pulmonary Alveoli/metabolism , Pulmonary Fibrosis/chemically induced , Pulmonary Fibrosis/metabolism , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Snail Family Transcription Factors , Transcription Factors/metabolism , Transforming Growth Factor beta1/antagonists & inhibitors , Transforming Growth Factor beta1/pharmacology
8.
Shock ; 44(4): 371-80, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26196844

ABSTRACT

Acute lung injury (ALI) is characterized by lung inflammation and diffuse infiltration of neutrophils. Neutrophil apoptosis is recognized as an important control point in the resolution of inflammation. Maresin 1 (MaR1) is a new docosahexaenoic acid-derived proresolving agent that promotes the resolution of inflammation. However, its function in neutrophil apoptosis is unknown. In this study, isolated human neutrophils were incubated with MaR1, the pan-caspase inhibitor z-VAD-fmk, and lipopolysaccharide (LPS) to determine the mechanism of neutrophil apoptosis. Acute lung injury was induced by intratracheal instillation of LPS. In addition, mice were treated with MaR1 intravenously at the peak of inflammation and administered z-VAD-fmk intraperitoneally. We found that culture of isolated human neutrophils with LPS dramatically delayed neutrophil apoptosis through the phosphorylation of AKT, ERK, and p38 to upregulate the expression of the antiapoptotic proteins Mcl-1 and Bcl-2, which was blocked by pretreatment with MaR1 in vitro. In mice, MaR1 accelerated the resolution of inflammation in LPS-induced ALI through attenuation of neutrophil accumulation, pathohistological changes, and pulmonary edema. Maresin 1 promoted resolution of inflammation by accelerating caspase-dependent neutrophil apoptosis. Moreover, MaR1 also reduced the LPS-induced production of proinflammatory cytokines and upregulated the production of the anti-inflammatory cytokine interleukin-10. In contrast, treatment with z-VAD-fmk inhibited the proapoptotic action of MaR1 and attenuated the protective effects of MaR1 in LPS-induced ALI. Taken together, MaR1 promotes the resolution of LPS-induced ALI by overcoming LPS-mediated suppression of neutrophil apoptosis.


Subject(s)
Acute Lung Injury/drug therapy , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Docosahexaenoic Acids/therapeutic use , Neutrophils/drug effects , Acute Lung Injury/pathology , Amino Acid Chloromethyl Ketones/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/antagonists & inhibitors , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Apoptosis/drug effects , Bronchoalveolar Lavage Fluid/cytology , Caspase Inhibitors/pharmacology , Cell Survival/drug effects , Cells, Cultured , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/antagonists & inhibitors , Docosahexaenoic Acids/pharmacology , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/antagonists & inhibitors , Male , Mice, Inbred BALB C , Neutrophils/pathology , Signal Transduction/drug effects
9.
Br J Pharmacol ; 171(14): 3539-50, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24697684

ABSTRACT

BACKGROUND AND PURPOSE: Acute lung injury (ALI) is a severe illness with a high rate of mortality. Maresin 1 (MaR1) was recently reported to regulate inflammatory responses. We used a LPS-induced ALI model to determine whether MaR1 can mitigate lung injury. EXPERIMENTAL APPROACH: Male BALB/c mice were injected, intratracheally, with either LPS (3 mg·kg(-1) ) or normal saline (1.5 mL·kg(-1) ). After this, normal saline, a low dose of MaR1 (0.1 ng per mouse) or a high dose of MaR1 (1 ng per mouse) was given i.v. Lung injury was evaluated by detecting arterial blood gas, pathohistological examination, pulmonary oedema, inflammatory cell infiltration, inflammatory cytokines in the bronchoalveolar lavage fluid and neutrophil-platelet interactions. KEY RESULTS: The high dose of MaR1 significantly inhibited LPS-induced ALI by restoring oxygenation, attenuating pulmonary oedema and mitigating pathohistological changes. A combination of elisa and immunohistochemistry showed that high-dose MaR1 attenuated LPS-induced increases in pro-inflammatory cytokines (TNF-α, IL-1ß and IL-6), chemokines [keratinocyte chemokine, monocyte chemoattractant protein-5, macrophage inflammatory protein (MIP)-1α and MIP-1γ], pulmonary myeloperoxidase activity and neutrophil infiltration in the lung tissues. Consistent with these observations, flow cytometry and Western blotting indicated that MaR1 down-regulated LPS-induced neutrophil adhesions and suppressed the expression of intercellular adhesion molecule (ICAM)-1, P-selection and CD24. CONCLUSIONS AND IMPLICATIONS: High-dose MaR1 mitigated LPS-induced lung injury in mice by inhibiting neutrophil adhesions and decreasing the levels of pro-inflammatory cytokines.


Subject(s)
Acute Lung Injury/drug therapy , Docosahexaenoic Acids/pharmacology , Lipopolysaccharides/antagonists & inhibitors , Acute Lung Injury/chemically induced , Acute Lung Injury/pathology , Administration, Inhalation , Animals , Cell Adhesion/drug effects , Docosahexaenoic Acids/administration & dosage , Inflammation Mediators/metabolism , Lipopolysaccharides/administration & dosage , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred BALB C , Neutrophils/drug effects , Neutrophils/pathology
10.
Shock ; 41(4): 311-6, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24365886

ABSTRACT

Mechanical ventilation can cause structural and functional disturbances in the lung termed ventilator-induced lung injury (VILI). The aim of this study was to evaluate whether BML-111, a lipoxin receptor agonist, could attenuate VILI. Following induction of anesthesia and tracheostomy, Sprague-Dawley rats were ventilated with low tidal volume (6 mL/kg) or high tidal volume (20 mL/kg, HVT) for 4 h. Some rats subjected to HVT ventilation received BML-111 or vehicle (saline) by intraperitoneal injection. Some rats subjected to HVT and BML-111(1 mg/kg) received BOC-2 (a FPR2/ALX antagonist) intraperitoneally 30 min before BML-111. Sham rats were tracheotomized without ventilation. Treatment with BML-111 attenuated VILI, as evidenced by improved oxygenation and reduced histological injury compared with HVT-induced lung injury. BML-111 decreased indices of inflammation such as interleukin 1ß, interleukin 6, tumor necrosis factor α, and bronchoalveolar lavage neutrophil infiltration. Administration with BML-111 suppressed the decrement of the nuclear factor κB (NF-κB) inhibitor IκB-α, diminished NF-κB activation, and reduced activation of mitogen-activated protein kinase in VILI. This study indicates that BML-111 attenuated VILI via a NF-κB and mitogen-activated protein kinase dependent mechanism. BML-111 may be a promising strategy for alleviation of VILI in patients subjected to mechanical ventilation.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Heptanoic Acids/therapeutic use , Receptors, Lipoxin/agonists , Ventilator-Induced Lung Injury/prevention & control , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bronchoalveolar Lavage Fluid/cytology , Cytokines/biosynthesis , Drug Evaluation, Preclinical/methods , Heptanoic Acids/pharmacology , I-kappa B Proteins/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , NF-KappaB Inhibitor alpha , NF-kappa B/metabolism , Peroxidase/metabolism , Phosphorylation , Rats , Rats, Sprague-Dawley , Transcription Factor AP-1/metabolism , Transcription Factor RelA/genetics , Transcription Factor RelA/metabolism , Translocation, Genetic/drug effects , Up-Regulation/drug effects , Ventilator-Induced Lung Injury/metabolism , Ventilator-Induced Lung Injury/pathology
11.
Drug Des Devel Ther ; 8: 1-11, 2014.
Article in English | MEDLINE | ID: mdl-24376342

ABSTRACT

Wear debris associated periprosthetic osteolysis represents a major pathological process associated with the aseptic loosening of joint prostheses. Naringin is a major flavonoid identified in grapefruit. Studies have shown that naringin possesses many pharmacological properties including effects on bone metabolism. The current study evaluated the influence of naringin on wear debris induced osteoclastic bone resorption both in vitro and in vivo. The osteoclast precursor cell line RAW 264.7 was cultured and stimulated with polymethylmethacrylate (PMMA) particles followed by treatment with naringin at several doses. Tartrate resistant acid phosphatase (TRAP), calcium release, and gene expression profiles of TRAP, cathepsin K, and receptor activator of nuclear factor-kappa B were sequentially evaluated. PMMA challenged murine air pouch and the load bearing tibia titanium pin-implantation mouse models were used to evaluate the effects of naringin in controlling PMMA induced bone resorption. Histological analyses and biomechanical pullout tests were performed following the animal experimentation. The in vitro data clearly demonstrated the inhibitory effects of naringin in PMMA induced osteoclastogenesis. The naringin dose of 10 µg/mL exhibited the most significant influence on the suppression of TRAP activities. Naringin treatment also markedly decreased calcium release in the stimulated cell culture medium. The short-term air pouch mouse study revealed that local injection of naringin ameliorated the PMMA induced inflammatory tissue response and subsequent bone resorption. The long-term tibia pin-implantation mouse model study suggested that daily oral gavage of naringin at 300 mg/kg dosage for 30 days significantly alleviated the periprosthetic bone resorption. A significant increase of periprosthetic bone volume and regaining of the pin stability were found in naringin treated mice. Overall, this study suggests that naringin may serve as a potential therapeutic agent to treat wear debris associated osteolysis.


Subject(s)
Flavanones/therapeutic use , Osteoclasts/drug effects , Osteogenesis/drug effects , Osteolysis/prevention & control , Polymethyl Methacrylate/toxicity , Acid Phosphatase/antagonists & inhibitors , Animals , Bone Resorption/prevention & control , Calcium/metabolism , Cells, Cultured , Female , Flavanones/pharmacology , Isoenzymes/antagonists & inhibitors , Mice , Mice, Inbred BALB C , Osteoclasts/physiology , Tartrate-Resistant Acid Phosphatase
12.
ScientificWorldJournal ; 2013: 718061, 2013.
Article in English | MEDLINE | ID: mdl-24222748

ABSTRACT

This study examined the influence of osteoprotegerin (OPG) gene transfer on a murine collagen-induced arthritis model. A single periarticular injection of AAV-OPG or AAV-LacZ on the arthritic paw successfully incorporated the exogenous gene to the local tissue and resulted in marked transgene expression in the joint homogenate for at least three weeks. Clinical disease scores were significantly improved in OPG treated mice starting at 28-day post-treatment (P < 0.05). Histological assessment demonstrated that OPG gene transfer dramatically protected mice from erosive joint changes compared with LacZ controls (P < 0.05), although treatment appeared less effective on the local inflammatory progress. MicroCT data suggested significant protection against subchondral bone mineral density changes in OPG treated CIA mice. Interestingly, mRNA expressions of IFN-g and MMP3 were noticeably diminished following OPG gene transfer. Overall, gene transfer of OPG effectively inhibited the arthritis-associated periarticular bone erosion and preserved the architecture of arthritic joints, and the study provides evidence that the cartilage protection of the OPG gene therapy may be associated with the down-regulation of MMP3 expression.


Subject(s)
Arthritis, Experimental/therapy , Genetic Therapy , Osteoprotegerin/genetics , Animals , Arthritis, Experimental/metabolism , Arthritis, Experimental/pathology , Disease Progression , Interferon-gamma/genetics , Interferon-gamma/metabolism , Joints/pathology , Matrix Metalloproteinase 3/genetics , Matrix Metalloproteinase 3/metabolism , Mice , Osteoprotegerin/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
13.
J Biomed Mater Res A ; 101(10): 2817-25, 2013 Oct.
Article in English | MEDLINE | ID: mdl-24039045

ABSTRACT

This study investigates the roles of orthopedic biomaterial particles [Ti-alloy, poly(methyl methacrylate) (PMMA), ultrahigh-molecular-weight polyethylene (UHMWPE), Co-Cr alloy] on the differentiation and functions of bone marrow stromal cells (BMSCs). Cells were isolated from femurs of BALB/c mice and cultured in complete osteoblast-induction medium in presence of micron-sized biomaterial particles at various doses. 3-(4,5)-Dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide assay and lactate dehydrogenase assay were performed for cell proliferation and cytotoxicity. Differentiation and function of osteoblasts were evaluated by alkaline phosphatase (ALP), osteocalcin, RANKL, OSX, and Runx2 expressions. Murine interleukin-1 (IL-1), IL-6, and tumor necrosis factor-α in culture media were determined by enzyme-linked immunosorbent assay. Challenge with low doses of Ti, UHMWPE, or Co-Cr particles markedly promoted the bone marrow cell proliferation while high dose of Co-Cr significantly inhibited cell growth (p < 0.05). Cells challenged with low dose of PMMA or UHMWPE particles (0.63 mg/mL) exhibited strong ALP activity, whereas Ti and Co-Cr groups showed minimal effects (p < 0.05). UHMWPE and Ti particles also promoted higher expression of proinflammatory cytokines. Real-time polymerase chain reaction data suggested that cells treated with low dose (0.5 mg/mL) particles resulted in distinctly diminished RANKL expression compared to those exposed to high concentrated (3 mg/mL) particles. In conclusion, various types of wear debris particles behaved differently in the differentiation, maturation, and functions of osteogenic cells; and the particulate debris-interacted BMSCs may play an important role in the pathogenesis and process of the debris-associated aseptic prosthetic loosening.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/drug effects , Chromium Alloys/pharmacology , Polyethylenes/pharmacology , Polymethyl Methacrylate/pharmacology , Titanium/pharmacology , Alkaline Phosphatase/metabolism , Animals , Bone Marrow Cells/drug effects , Bone Marrow Cells/enzymology , Cell Death/drug effects , Cell Proliferation/drug effects , Cell Shape/drug effects , Cell Survival/drug effects , Culture Media, Conditioned/chemistry , Cytokines/metabolism , Enzyme-Linked Immunosorbent Assay , Gene Expression Profiling , Immunohistochemistry , Male , Mice , Mice, Inbred BALB C , Osteocalcin/metabolism , RANK Ligand/metabolism , Real-Time Polymerase Chain Reaction , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/enzymology
14.
J Orthop Sci ; 18(3): 478-85, 2013 May.
Article in English | MEDLINE | ID: mdl-23553541

ABSTRACT

BACKGROUND: Osteoporosis is a common pathological condition that influences 20 % of women over 50 years of age. This condition decreases bone strength and increases the risk of bone fracture. Naringin is a major flavonoid found in grapefruit and an active compound extracted from a Chinese herbal medicine (Rhizoma Drynariae). Studies have shown that naringin possesses many pharmacological effects. The current study evaluated the influence of naringin on osteoblastic cell differentiation and proliferation, and assessed its therapeutic effects on a rat osteoporosis model. METHOD: The proliferation, differentiation, and function of rat bone marrow stromal cells (BMSCs) were determined following treatment with various concentrations of naringin. Ovariectomy (OVX)-induced osteoporotic rats were orally administered naringin daily at low, medium, and high dosages, while a control group received PBS for 2 months. Femoral X-ray images and microCT scans were used for bone mineral density (BMD) and BV/TV (bone volume/total volume) analyses, and histological assessments of left tibiae were employed to check for changes in trabecular thickness (Tb.Th) and trabecular space (Tb.Sp) in the groups. RESULTS: Naringin was effective at enhancing the proliferation and osteogenic differentiation of BMSCs, and a concentration of 10 µg/ml prompted the highest levels of osteocalcin expression among the in vitro study groups. There appeared to be a delayed response pattern of BMSCs to the naringin treatment. Naringin also effectively reversed OVX-induced bone loss via increasing BMD, bone volume, and trabecular thickness. The medium dose (300 mg/kg) appeared to be the optimal dosage for delivering satisfactory therapeutic effects. CONCLUSION: Naringin promotes the proliferation and differentiation of BMSCs, and increases osteocalcin expression. Naringin also effectively reverses ovariectomy-induced osteoporosis in rats. The study suggests that naringin administration may represent an effective treatment for osteoporosis.


Subject(s)
Cell Differentiation/drug effects , Flavanones/pharmacology , Flavanones/therapeutic use , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoporosis/prevention & control , Animals , Female , Osteoporosis/etiology , Ovariectomy/adverse effects , Rats , Rats, Inbred Lew
15.
J Orthop Surg Res ; 4: 5, 2009 Feb 26.
Article in English | MEDLINE | ID: mdl-19243637

ABSTRACT

BACKGROUND: Tissue-engineered bone may be developed by seeding the cells capable of both osteogenesis and vascularization on biocompatible composite scaffolds. The current study investigated the performance of mice bone marrow-derived osteogenic cells and endothelial cells as seeded on hydroxyapatite (HA) and poly-epsilon-caprolactone (PCL) composite scaffolds. METHODS: Mononuclear cells were induced to osteoblasts and endothelial cells respectively, which were defined by the expression of osteocalcin, alkaline phosphatase (ALP), and deposits of calcium-containing crystal for osteoblasts, or by the expression of vascular endothelial growth factor receptor-2 (VEGFR-2) and von Willebrand factor (vWF), and the formation of a capillary network in Matrigel for endothelial cells. Both types of cell were seeded respectively on PCL-HA scaffolds at HA to PCL weight ratio of 1:1, 1:4, or 0:1 and were evaluated using scanning electron microscopy, ALP activity (of osteoblasts) and nitric oxide production (of endothelial cells) plus the assessment of cell viability. RESULTS: The results indicated that HA led to a positive stimulation of osteoblasts viability and ALP activity, while HA showed less influence on endothelial cells viability. An elevated nitric oxide production of endothelial cells was observed in HA-containing group. CONCLUSION: Supplement of HA into PCL improved biocompatible for bone marrow-derived osteoblasts and endothelial cells. The PCL-HA composite integrating with two types of cells may provide a useful system for tissue-engineered bone grafts with vascularization.

16.
Article in English | MEDLINE | ID: mdl-17357487

ABSTRACT

To investigate the protective effect of curcumin on endotoxin-induced acute lung injury in rats, and explore the underlying mechanisms, 24 male Wistar rats were randomly divided into 4 experimental groups: sham-vehicle (S), sham-curcumin (C), lipopolysaccharide (LPS)-vehicle (L), and curcumin-lipopolysaccharide (C-L) groups. The wet/dry (W/D) weight ratio of the lung and bronchoalveolar lavage (BAL) fluid protein content were used as measures of lung injury. Neutrophil recruitment and activation were evaluated by BAL fluid cellularity and myeloperoxidase (MPO) activity in cell-free BAL and lung tissue. The levels of cytokine-induced neutrophil chemoattractant-I (CINC-1) in lung tissues were measured by ELISA. The histopathological changes of lung tissues were observed by using the HE staining. Our results showed that lung injury parameters, including the wet/dry weight ratio and protein content in BALF, were significantly higher in the L group than in the S group (P<0.01). In the L group, higher numbers of neutrophils and greater MPO activity in cell-free BAL and lung homogenates were observed when compared with the S group (P<0.01). There was a marked increase in CINC-1 levels in lung tissues in response to LPS challenge (P<0.01, L group vs S group). Curcumin pretreatment significantly attenuated LPS-induced changes in these indices. LPS caused extensive morphological lung damage, which was also lessened after curcumin pretreatment. All the above-mentioned parameters in the C group were not significantly different from those of the S group. It is concluded that curcumin pretreatment attenuates LPS-induced lung injury in rats. This beneficial effect of curcumin may involves, in part, inhibition of neutrophilic recruitment and activity, possibly through inhibition of lung CINC-1 expression.


Subject(s)
Acute Lung Injury/drug therapy , Curcumin/therapeutic use , Acute Lung Injury/chemically induced , Acute Lung Injury/metabolism , Animals , Chemokine CXCL1/metabolism , Disease Models, Animal , Endotoxins/toxicity , Lung/metabolism , Lung/pathology , Male , Neutrophils/cytology , Rats , Rats, Wistar
SELECTION OF CITATIONS
SEARCH DETAIL