Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 678
Filter
1.
Int J Mol Sci ; 23(21)2022 Oct 29.
Article in English | MEDLINE | ID: mdl-36361965

ABSTRACT

Mineralization-competent cells like osteoblasts and chondrocytes release matrix vesicles (MVs) which accumulate Ca2+ and Pi, creating an optimal environment for apatite formation. The mineralization process requires the involvement of proteins, such as annexins (Anx) and tissue-nonspecific alkaline phosphatase (TNAP), as well as low molecular-weight compounds. Apigenin, a flavonoid compound, has been reported to affect bone metabolism, but there are doubts about its mechanism of action under physiological and pathological conditions. In this report, apigenin potency to modulate annexin A6 (AnxA6)- and TNAP-mediated osteoblast mineralization was explored using three cell lines: human fetal osteoblastic hFOB 1.19, human osteosarcoma Saos-2, and human coronary artery smooth muscle cells HCASMC. We compared the mineralization competence, the morphology and composition of minerals, and the protein distribution in control and apigenin-treated cells and vesicles. The mineralization ability was monitored by AR-S/CPC analysis, and TNAP activity was determined by ELISA assay. Apigenin affected the mineral structure and modulated TNAP activity depending on the concentration. We also observed increased mineralization in Saos-2 cells. Based on TEM-EDX, we found that apigenin influenced the mineral composition. This flavonoid also disturbed the intracellular distribution of AnxA6 and TNAP, especially blocking AnxA6 aggregation and TNAP attachment to the membrane, as examined by FM analysis of cells and TEM-gold analysis of vesicles. In summary, apigenin modulates the mineralization process by regulating AnxA6 and TNAP, as well as through various effects on normal and cancer bone tissues or atherosclerotic soft tissue.


Subject(s)
Apigenin , Calcification, Physiologic , Humans , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Annexin A6/drug effects , Annexin A6/metabolism , Apigenin/pharmacology , Apigenin/metabolism , Calcification, Physiologic/drug effects , Calcification, Physiologic/physiology , Osteoblasts/drug effects , Osteoblasts/metabolism
2.
Acta Pharmacol Sin ; 43(3): 624-633, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34163023

ABSTRACT

Vascular calcification (VC) is characterized by pathological depositions of calcium and phosphate in the arteries and veins via an active cell-regulated process, in which vascular smooth muscle cells (VSMCs) transform into osteoblast/chondrocyte-like cells as in bone formation. VC is associated with significant morbidity and mortality in chronic kidney disease (CKD) and cardiovascular disease, but the underlying mechanisms remain unclear. In this study we investigated the role of large-conductance calcium-activated potassium (BK) channels in 3 experimental VC models. VC was induced in vascular smooth muscle cells (VSMCs) by ß-glycerophosphate (ß-GP), or in rats by subtotal nephrectomy, or in mice by high-dosage vitamin D3. We showed that the expression of BK channels in the artery of CKD rats with VC and in ß-GP-treated VSMCs was significantly decreased, which was functionally confirmed by patch-clamp recording. In ß-GP-treated VSMCs, BK channel opener NS1619 (20 µM) significantly alleviated VC by decreasing calcium content and alkaline phosphatase activity. Furthermore, NS1619 decreased mRNA expression of ostoegenic genes OCN and OPN, as well as Runx2 (a key transcription factor involved in preosteoblast to osteoblast differentiation), and increased the expression of α-SMA protein, whereas BK channel inhibitor paxilline (10 µM) caused the opposite effects. In primary cultured VSMCs from BK-/- mice, BK deficiency aggravated calcification as did BK channel inhibitor in normal VSMCs. Moreover, calcification was more severe in thoracic aorta rings of BK-/- mice than in those of wild-type littermates. Administration of BK channel activator BMS191011 (10 mg· kg-1 ·d-1) in high-dosage vitamin D3-treated mice significantly ameliorated calcification. Finally, co-treatment with Akt inhibitor MK2206 (1 µM) or FoxO1 inhibitor AS1842856 (3 µM) in calcified VSMCs abrogated the effects of BK channel opener NS1619. Taken together, activation of BK channels ameliorates VC via Akt/FoxO1 signaling pathways. Strategies to activate BK channels and/or enhance BK channel expression may offer therapeutic avenues to control VC.


Subject(s)
Large-Conductance Calcium-Activated Potassium Channels/drug effects , Muscle, Smooth, Vascular/drug effects , Proto-Oncogene Proteins c-akt/drug effects , Vascular Calcification/pathology , Alkaline Phosphatase/drug effects , Animals , Aorta, Thoracic/drug effects , Benzimidazoles/pharmacology , Cholecalciferol/pharmacology , Disease Models, Animal , Glycerophosphates/pharmacology , Male , Mice , Mice, Inbred C57BL , Nephrectomy , Osteocalcin/drug effects , Osteopontin/drug effects , Peptide Fragments/drug effects , RNA, Messenger/drug effects , Random Allocation , Rats , Rats, Sprague-Dawley
3.
Pak J Pharm Sci ; 34(3): 933-942, 2021 May.
Article in English | MEDLINE | ID: mdl-34602416

ABSTRACT

The intention to conduct this study was to evaluate the hepatoprotective effects of Fenugreek seeds' extract supplementation in thioacetamide induced liver damage in male Sprague Dawley rats. For this study, 24 male Sprague Dawley rats (200-264gm) were distributed randomly into four groups. Group I remained untreated as control rats, group II received thioacetamide (200mg/Kg b.w i.p, administered on alternative days for 8 weeks), group III received thioacetamide (200mg/Kg b.w i.p administered on alternative days for 8 weeks) as well as 2ml of 2% extract of fenugreek seeds (orally administered daily from 4th week till 8th week of the experiment. Group IV only received 2ml of 2% extract of Fenugreek seeds daily for 4 weeks respectively. At the end of the experiment, blood was sampled to obtain plasma that was used for the analysis of liver markers and liver was used for analysis of antioxidant enzymes (catalase and SOD). Increase in total bilirubin, direct bilirubin, ALT and ALP levels, catalase activity and decrease in SOD activity was found in TAA-treated groups which assured liver damage. Whereas, treatment with Fenugreek seeds extract restored the altered levels of total bilirubin, direct bilirubin, ALT, ALP, catalase and SOD activities in the Test + Supp group. The results of this study confirmed the hepatoprotective role of Fenugreek seeds extract in thioacetamide induced liver damage.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Liver/drug effects , Plant Extracts/pharmacology , Thioacetamide/toxicity , Alanine Transaminase/drug effects , Alanine Transaminase/metabolism , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Antioxidants/metabolism , Bilirubin/metabolism , Body Weight/drug effects , Catalase/drug effects , Catalase/metabolism , Chemical and Drug Induced Liver Injury/pathology , Liver/metabolism , Liver/pathology , Organ Size/drug effects , Random Allocation , Rats , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Trigonella
4.
Drug Deliv ; 28(1): 1649-1663, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34338560

ABSTRACT

Existing biologically inert or unmodified implants to treat infectious bone defects or osteomyelitis still cannot effectively solve bacterial infection and osseointegration. In this work, a simple co-deposition strategy was developed to modify porous polyetheretherketone (PEEK) with improved antibacterial activity and controllable immunoregulatory ability. After PEEK was treated by H2SO4 to obtain porous PEEK (SPEEK), the self-polymerization of dopamine was operated on SPEEK in the solution of dopamine and gentamicin sulfate (GS) to prepare polydopamine (pDA) and GS layer-modified SPEEK (labeled as SPEEK-pDA-GS). The morphology, surface property, and molecular structure of SPEEK-pDA-GS were investigated. Besides the antibacterial property of SPEEK-pDA-GS ascribed to the successful immobilization of GS, SPEEK-pDA-GS exhibited promoted osseointegration through the results of mineralization, alkaline phosphatase (ALP) levels and osteogenic gene expression. Furthermore, the evaluation of the cell proliferation suggested that SPEEK-pDA-GS possessed the biocompatibility and the immunoregulatory ability that induced macrophages to anti-inflammatory M2 phenotype. Using rat as model, in vivo results containing X-ray, µ-CT, immunohistochemistry, and pathological analysis showed the excellent healing effect of SPEEK-pDA-GS on bone defect with infection with biological safety. This work illustrates a new insight into the simple and effective modification of PEEK and other implants with antibacterial, immunoregulatory, and osseointegration abilities for clinical requirement.


Subject(s)
Anti-Bacterial Agents/pharmacology , Benzophenones/pharmacology , Drug Implants/chemistry , Gentamicins/pharmacology , Indoles/chemistry , Polymers/chemistry , Polymers/pharmacology , Alkaline Phosphatase/drug effects , Animals , Anti-Bacterial Agents/administration & dosage , Benzophenones/administration & dosage , Biocompatible Materials , Cell Differentiation/drug effects , Cell Line , Cell Proliferation/drug effects , Drug Liberation , Escherichia coli/drug effects , Gentamicins/administration & dosage , Osteogenesis/drug effects , Polymers/administration & dosage , Porosity , Rats , Rats, Sprague-Dawley , Staphylococcus aureus/drug effects , Surface Properties
5.
Pak J Pharm Sci ; 34(2): 649-656, 2021 Mar.
Article in English | MEDLINE | ID: mdl-34275842

ABSTRACT

Studied the optimum extraction process of polysaccharide from Phaeoporus obliquus and the effect of Phaeoporus obliquus polysaccharide on carbon tetrachloride (CCl4)- or alcohol-induced acute liver injury in mice. The main factor in influencing the extraction rate of Phaeoporus obliquus polysaccharide were extraction power and time, which was a kind of pyran glucose by infrared spectroscopy. CCl4 and alcohol were employed respectively to establish CCl4 and alcohol-induced acute liver injury mouse models. Compared with model groups mice, Phaeoporus obliquus polysaccharide treatment at the doses of 100mg/kg and 200mg/kg exhibited an obvious reduction liver index, ALP, ALT, AST levels, MDA content and TNF-α level (p<0.01) and SOD activity was increased, which was in a dose-dependent manner. Compared with the model group, the necrosis degree of hepatocytes was obviously reduced and the small fat droplets were formed in some cytoplasm, especially in high dose group, which the liver cells recovered to the level of normal group. Rt-PCR results showed that the expression of CYP2E1 mRNA in liver tissues of Phaeoporus obliquus polysaccharide groups were significantly reduced, and the difference were statistically significant compared with the model group (p<0.05). These results demonstrated that Phaeoporus obliquus polysaccharide has significantly hepatoprotective effect on CCl4 and alcohol-induced acute liver injury in mice.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Fungal Polysaccharides/pharmacology , Hepatocytes/drug effects , Inonotus , Liver Diseases, Alcoholic/metabolism , Liver/drug effects , Alanine Transaminase/drug effects , Alanine Transaminase/metabolism , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Aspartate Aminotransferases/drug effects , Aspartate Aminotransferases/metabolism , Carbon Tetrachloride/toxicity , Central Nervous System Depressants/toxicity , Cytochrome P-450 CYP2E1/drug effects , Cytochrome P-450 CYP2E1/genetics , Ethanol/toxicity , Hepatocytes/metabolism , Hepatocytes/pathology , Liver/metabolism , Liver/pathology , Malondialdehyde/metabolism , Mice , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Superoxide Dismutase/drug effects , Superoxide Dismutase/metabolism , Tumor Necrosis Factor-alpha/drug effects , Tumor Necrosis Factor-alpha/metabolism
6.
Eur J Clin Invest ; 51(7): e13534, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33656763

ABSTRACT

BACKGROUND: Statins are 3-Hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase inhibitors blocking cholesterol biosynthesis in hepatic cells, thereby causing an increase in low-density lipoprotein (LDL) receptors resulting in enhanced uptake and clearance of atherogenic LDL-cholesterol (LDL-C) from the blood. Accordingly, statins decrease the risk of developing atherosclerosis and its acute complications, such as acute myocardial infarction and ischaemic stroke. Besides the LDL-C-lowering impact, statins also have other so-called pleiotropic effects. Among them, the ability to modulate differentiation and function of bone cells and exert direct effects on osteosynthesis factors. Specifically, earlier studies have shown that statins cause in vitro and in vivo osteogenic differentiation. DESIGN: The most relevant papers on the bone-related 'pleiotropic' effects of statins were selected following literature search in databases and were reveiwed. RESULTS: Statins increase the expression of many mediators involved in bone metabolism including bone morphogenetic protein-2 (BMP-2), glucocorticoids, transforming growth factor-beta (TGF-ß), alkaline phosphatase (ALP), type I collagen and collagenase-1. As a result, they enhance bone formation and improve bone mineral density by modulating osteoblast and osteoclast differentiation. CONCLUSION: This review summarizes the literature exploring bone-related 'pleiotropic' effects of statins and suggests an anabolic role in the bone tissue for this drug class. Accordingly, current knowledge encourages further clinical trials to assess the therapeutic potential of statins in the treatment of bone disorders, such as arthritis and osteoporosis.


Subject(s)
Bone Density/drug effects , Cell Differentiation/drug effects , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Osteoblasts/drug effects , Osteoclasts/drug effects , Osteogenesis/drug effects , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Bone Morphogenetic Protein 2/drug effects , Bone Morphogenetic Protein 2/metabolism , Collagen Type I/drug effects , Collagen Type I/metabolism , Collagenases/drug effects , Collagenases/metabolism , Glucocorticoids/metabolism , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/therapeutic use , Transforming Growth Factor beta/drug effects , Transforming Growth Factor beta/metabolism
7.
Curr Pharm Biotechnol ; 22(5): 609-621, 2021.
Article in English | MEDLINE | ID: mdl-33198615

ABSTRACT

BACKGROUND: Osteoporosis, characterized by bone loss, usually occurs with the increased bone resorption and decreased bone formation. H2O2-induced MC3T3-E1 cells are commonly used for the study of osteoblastic activities, which play a crucial role in bone formation. OBJECTIVE: This study aimed to investigate the effects of Phosphocreatine (PCr) on the osteoblastic activities in H2O2-induced MC3T3-E1 cells and elaborate on the possible molecular mechanism. METHODS: The Osteoprotegerin (OPG)/Receptor Activator of NF-κB Ligand (RANKL) ratio and osteogenic markers were detected to investigate the effects of PCr on osteoblastic activities, and the osteoblastic apoptosis was detected using Hochest staining. Moreover, oxidative stress, Adenosine Triphosphate (ATP) generation and the expression of Sirtuin 1 (SIRT1), Forkhead Box O 1 (FOXO1) and Peroxisome Proliferator-Activated Receptor Γ Coactivator-1α (PGC-1α) were also examined to uncover the possible molecular mechanism in H2O2-induced MC3T3-E1 cells. RESULT: The results showed that PCr promoted the osteoblastic differentiation by increasing the expression levels of osteogenic markers of Alkaline Phosphatase (ALP) and Runt-related transcription factor 2 (Runx2), as well as increased the OPG/RANKL ratio and suppressed the osteoblastic apoptosis in H2O2-induced MC3T3-E1 cells. Moreover, treatment with PCr suppressed reactive oxygen species (ROS) over-generation and promoted the ATP production as well as increased the PGC-1α, FOXO1 and SIRT1 protein expression levels in H2O2-induced MC3T3-E1 cells. CONCLUSION: PCr treatment could promote osteoblastic activities via suppressing oxidative stress and increasing the ATP generation in H2O2-induced MC3T3-E1 cells. In addition, the positive effects of PCr on osteoblasts might be regulated by SIRT1/FOXO1/ PGC-1α signaling pathway.


Subject(s)
Forkhead Box Protein O1/drug effects , Hydrogen Peroxide/pharmacology , Osteoblasts/drug effects , Osteogenesis/drug effects , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/drug effects , Phosphocreatine/pharmacology , Signal Transduction/drug effects , Sirtuin 1/drug effects , 3T3 Cells , Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/drug effects , Animals , Apoptosis/drug effects , Core Binding Factor Alpha 1 Subunit/biosynthesis , Core Binding Factor Alpha 1 Subunit/drug effects , Mice , Osteoprotegerin/drug effects , Osteoprotegerin/metabolism , Oxidative Stress , RANK Ligand/drug effects , RANK Ligand/metabolism , Reactive Oxygen Species
8.
Aging (Albany NY) ; 13(4): 4976-4985, 2020 11 10.
Article in English | MEDLINE | ID: mdl-33196456

ABSTRACT

Osteogenic differentiation is critical to bone homeostasis, and its imbalance plays a key role in the progression of osteoporosis. Osteoblast cells are responsible for synthesizing new bone tissue, and understanding how to control osteoblastic differentiation is vital to the treatment of osteoporosis. Herein, we show that GPR173 signaling is involved in the regulation of osteoblastic differentiation in MC3T3-E1 cells. Our data reveals that GPR173 is abundantly expressed in MC3T3-E1 cells, and its expression is inducible upon the introduction of osteogenic media. The activation of GPR173 by its selective agonist phoenixin 20 induces the expression of several osteoblast signature genes including collagen type 1 alpha 1 (Col-I), osteocalcin (OCN), alkaline phosphatase (ALP) as well as increased matrix mineralization and ALP activity, suggesting that the activation of GPR173 promotes osteoblastic differentiation. Moreover, we show that the effect of phoenixin 20 is mediated by its induction on the key regulator runt-Related Transcription Factor 2 (Runx2). Mechanistically, we display that the action of phoenixin 20 requires the activation of MAPK kinase p38, and deactivation of p38 by its inhibitor SB203580 weakens the phoenixin 20-mediated induction of RUNX-2, ALP, and matrix mineralization. Silencing of GPR173 attenuates phoenixin 20-mediated osteoblastic differentiation, indicating its dependence on the receptor. Collectively, our study reveals a new role of GPR173 and its agonist phoenixin 20 in osteoblastic differentiation.


Subject(s)
Calcification, Physiologic/genetics , Cell Differentiation/physiology , Osteoblasts/physiology , Receptors, G-Protein-Coupled/metabolism , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Calcification, Physiologic/drug effects , Calcification, Physiologic/physiology , Cell Differentiation/drug effects , Collagen Type I/drug effects , Collagen Type I/genetics , Collagen Type I, alpha 1 Chain , Core Binding Factor Alpha 1 Subunit/drug effects , Core Binding Factor Alpha 1 Subunit/genetics , Enzyme Inhibitors/pharmacology , Gene Expression , Gene Knockdown Techniques , Imidazoles/pharmacology , Mice , Osteoblasts/cytology , Osteoblasts/drug effects , Osteocalcin/drug effects , Osteocalcin/genetics , Peptide Hormones/pharmacology , Pyridines/pharmacology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/physiology , Transcriptome , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
9.
Medicine (Baltimore) ; 99(40): e22542, 2020 Oct 02.
Article in English | MEDLINE | ID: mdl-33019463

ABSTRACT

BACKGROUND: The goal of this study was to review relevant randomized controlled trials or case-control studies to determine the clinical efficacy of minodronate in the treatment of osteoporosis. METHOD: The relevant studies were identified on PubMed, Cochrane, and Embase databases using appropriate keywords. Pertinent sources in the literature were also reviewed, and all articles published through October 2019 were considered for inclusion. For each study, we assessed odds ratios, mean difference, and 95% confidence interval (95% CI) to evaluate and synthesize outcomes. RESULT: Thirteen studies comprising 3740 patients were included in this study. Compared with other drugs, minodronate significantly decreased N-telopeptide of type I collagen/creatinine (weighted mean difference [WMD]: -13.669, 95% confidence interval [CI]: -23.108 to -4.229), bone alkaline phosphatase (BAP) (WMD: -1.26, 95% CI: -2.04 to -0.47) and tartrate-resistant acid phosphatase 5b (WMD: -154.11, 95% CI: -277.85 to -30.37). Minodronate combined with other drugs would significantly decrease BAP (WMD: -3.10, 95% CI: -5.20 to -1.00) than minodronate. Minodronate-naïve would significantly decrease BAP (WMD: -3.00, 95% CI: -5.47 to 0.53) and tartrate-resistant acid phosphatase 5b (WMD: -128.20, 95% CI: -198.11 to -58.29) than minodronate-switch. The incidence of vertebral fracture was significantly decreased in the minodronate group than the other drugs (relative risk: 0.520, 95% CI: 0.363-0.744). CONCLUSION: Minodronate has better clinical efficacy in the treatment of osteoporosis than other drugs (alendronate, risedronate, raloxifene, or eldecalcitol).


Subject(s)
Diphosphonates/therapeutic use , Imidazoles/therapeutic use , Osteoporosis/drug therapy , Aged , Aged, 80 and over , Alendronate/therapeutic use , Alkaline Phosphatase/drug effects , Bone Density Conservation Agents/therapeutic use , Case-Control Studies , Collagen Type I/drug effects , Creatinine , Drug Therapy, Combination/statistics & numerical data , Female , Humans , Male , Middle Aged , Raloxifene Hydrochloride/therapeutic use , Randomized Controlled Trials as Topic , Risedronic Acid/therapeutic use , Spinal Fractures/epidemiology , Tartrate-Resistant Acid Phosphatase/drug effects , Treatment Outcome , Vitamin D/analogs & derivatives , Vitamin D/therapeutic use
10.
Complement Ther Med ; 52: 102398, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32951697

ABSTRACT

Several randomized clinical trials (RCTs) evaluated the effect of melatonin supplementation on liver enzymes in patients with non-alcoholic fatty liver disease (NAFLD) and reported conflicting results. To meet these discrepancies, a meta-analysis was conducted to evaluate the eff ;ect of melatonin on liver indices in patients with NAFLD. To collect the required data, a thorough search was conducted through Web of science, Pubmed, Cochrane database, Embase, Google Scholar, ProQuest, and Scopus databases. The aim was to find clinical trials over the effect of melatonin supplementation on liver indices up to 16 May 2019. As a result, five eligible articles were selected and analysed in this meta-analysis using a fixed-effects model. Heterogeneity test was performed by I2 statistics and Cochrane Q test. The results showed that melatonin had a significant effect on aspartate aminoteransferase (AST) (WMD = 2.29, [95 %CI: 1.14, 3.43] IU/L, p = <0.001), alkaline phosphatase (ALP) (WMD = -8.40, [95 %CI -11.33, -5.48] IU/L, p < 0.001), and gamma-glutamyltransferase (GGT) (WMD = -33.37, [95 %CI: -37.24, -29.49] IU/L, p= < 0.001). Melatonin had no significant effect on alanine aminotransferase (ALT) regarding the overall effect size. Based on this meta-analysis, melatonin supplementation can improve liver indices. However, more RCTs are required with larger sample sizes and better control of confounding variables such as weight, body mass index, and gender to determine the effect of melatonin on patients with non-alcoholic fatty acid disease.


Subject(s)
Alanine Transaminase/drug effects , Alkaline Phosphatase/drug effects , Aspartate Aminotransferases/drug effects , Melatonin/therapeutic use , Non-alcoholic Fatty Liver Disease/drug therapy , gamma-Glutamyltransferase/drug effects , Biomarkers/blood , Humans , Non-alcoholic Fatty Liver Disease/enzymology , Randomized Controlled Trials as Topic
11.
Mol Med Rep ; 22(2): 1119-1128, 2020 08.
Article in English | MEDLINE | ID: mdl-32626952

ABSTRACT

The Indian hedgehog (IHH) signaling pathway is an important pathway for bone growth and development. The aim of the present study was to examine the role of the IHH signaling pathway in the development of the ossification of ligamentum flavum (OLF) at the cellular and tissue levels. The expression levels and localization of the osteogenic genes Runt-related transcription factor 2 (RUNX2), Osterix, alkaline phosphatase (ALP), osteocalcin (OCN) and IHH were evaluated in OLF tissues by reverse transcription-quantitative PCR (RT-qPCR) and immunohistochemistry. Non-ossified ligamentum flavum (LF) sections were used as control samples. The tissue explant method was used to obtain cultured LF cells. In addition, OLF cells were subjected to cyclic stretch application for 0, 6, 12 or 24 h. The expression levels of osteogenic genes, and the IHH signaling pathway genes IHH, Smoothened (SMO), GLI family zinc finger 1 (GLI1), GLI2 and GLI3 were evaluated with RT-qPCR and western blotting. Osteogenic differentiation was further evaluated by assessing ALP activity and staining. Moreover, the effect of cyclopamine (Cpn), an IHH signaling inhibitor, on osteogenic differentiation was examined. The RT-qPCR and immunohistochemical results indicated that the mRNA and protein expression levels of RUNX2, Osterix, ALP, OCN and IHH were significantly higher in the OLF group compared with the LF group. Furthermore, application of cyclic stretch to OLF cells resulted in greater ALP activity, and significant increases in mRNA and protein expression levels of RUNX2, Osterix, ALP and OCN in a time-d00ependent manner. Cyclic stretch application also led to significant increases in IHH signaling pathway genes, including IHH, SMO, GLI1 and GLI2, while no significant effect was found on GLI3 expression level. In addition, it was found that Cpn significantly reversed the effect of cyclic stretch on the ALP activity, and the expression levels of RUNX2, Osterix, ALP, OCN, GLI1 and GLI2. Collectively, the present results suggested that the IHH signaling pathway may mediate the effect of cyclic stretch on the OLF cells.


Subject(s)
Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Ligamentum Flavum/metabolism , Ossification, Heterotopic/genetics , Ossification, Heterotopic/metabolism , Signal Transduction , Stress, Mechanical , Adult , Aged , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Cell Differentiation , Cells, Cultured , Core Binding Factor Alpha 1 Subunit/drug effects , Core Binding Factor Alpha 1 Subunit/metabolism , Female , Hedgehog Proteins/antagonists & inhibitors , Humans , Ligamentum Flavum/pathology , Male , Middle Aged , Nerve Tissue Proteins/metabolism , Nuclear Proteins/metabolism , Osteocalcin/drug effects , Osteocalcin/metabolism , Signal Transduction/drug effects , Smoothened Receptor/genetics , Smoothened Receptor/metabolism , Sp7 Transcription Factor/drug effects , Sp7 Transcription Factor/metabolism , Veratrum Alkaloids/pharmacology , Zinc Finger Protein GLI1/metabolism , Zinc Finger Protein Gli2/metabolism , Zinc Finger Protein Gli3/metabolism
12.
J Med Case Rep ; 14(1): 87, 2020 Jul 03.
Article in English | MEDLINE | ID: mdl-32616027

ABSTRACT

BACKGROUND: Teriparatide is a homolog of human parathyroid hormone (1-34), which is approved for the treatment of postmenopausal and glucocorticoid-induced osteoporosis. Several minor and transient side effects have been reported for teriparatide. However, controversial findings showed an increased risk of more significant adverse effects, including osteosarcoma in humans, although this finding has been demonstrated primarily in murine models. CASE PRESENTATION: We present a case of a 22-year-old Persian man with a previous history of systemic lupus erythematosus and glucocorticoid-induced osteoporosis. He had a previous history of joint hypermobility, idiopathic kyphoscoliosis, mitral valve prolapse, and bilateral congenital inguinal hernia, which were probably compatible with an inherited connective tissue disease. He was treated with teriparatide for 7 months because of glucocorticoid-induced osteoporosis. He was referred with a complaint of generalized bone pain and an extremely elevated serum alkaline phosphatase concentration of 6480 U/L (normal range, 80-306). A whole-body bone scan revealed a diffuse increased osseous uptake. Furthermore, the patient's systemic lupus erythematosus was clinically inactive on the basis of laboratory findings during this period. The medication was discontinued, and the patient's serum alkaline phosphatase level began to decline. CONCLUSIONS: To the best of our knowledge, this is the first case of an osteoblast hyperactivation state observed during treatment with teriparatide. It appears that the osteoblastogenic effect of teriparatide might induce this condition and, most likely, osteosarcoma in certain populations. However, the potential influence of the patient's young age, systemic lupus erythematosus, underlying inherited connective tissue disease, and medication use cannot be ignored. The potential risk factors of this side effect shall be studied in specific subpopulations of patients with osteoporosis in future studies.


Subject(s)
Alkaline Phosphatase , Bone Density Conservation Agents/adverse effects , Osteoblasts/drug effects , Osteoporosis/drug therapy , Teriparatide/adverse effects , Alkaline Phosphatase/blood , Alkaline Phosphatase/drug effects , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/pharmacology , Humans , Male , Teriparatide/administration & dosage , Whole Body Imaging , Young Adult
13.
Pediatr Rheumatol Online J ; 18(1): 46, 2020 Jun 09.
Article in English | MEDLINE | ID: mdl-32517762

ABSTRACT

BACKGROUND: The effects of C-type natriuretic peptide (CNP) and fibroblast growth factor (FGF)-23 appear to oppose each other during the process of bone formation, whereas few studies exist on the interaction between CNP and FGF-23. The main objective of the present study is to probe whether CNP is directly responsible for the regulation of osteoblast or via antagonizing FGF-23. METHODS: Osteoblasts were cultured in the absence or presence of CNP (0, 10, and 100 pmol/L) for 24 h, 48 h and 72 h, respectively. RESULTS: The findings of the present study indicated that: (1) CNP significantly stimulated osteoblastic proliferation and collagen (Col)-X expression; (2) both osteoblastic (osteocalcin, procollagen type I carboxy-terminal propeptide, total alkaline phosphatase and bone-specific alkaline phosphatase) and osteolytic (tartrate-resistant acid phosphatase and cross-linked carboxyterminal telopeptide of type I collagen) bone turnover biomarkers were up-regulated by CNP in osteoblasts; (3) FGF-23 mRNA and protein were significantly down-regulated at 24 h by CNP in osteoblasts, but the expression of FGF receptor-1/Klotho had no significant change. CONCLUSIONS: CNP stimulates osteoblastic proliferation and Col-X expression via the down-regulation of FGF-23 possibly in vitro. However, the specific mechanisms of the interaction between CNP and FGF-23 in osteoblasts are still unclear according to our findings. A further study on osteoblasts cultured with CNP and FGF-23 inhibitor will be undertaken in our laboratory.


Subject(s)
Cell Proliferation/genetics , Fibroblast Growth Factors/genetics , Natriuretic Peptide, C-Type/metabolism , Osteoblasts/metabolism , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Blotting, Western , Bone Remodeling/drug effects , Bone Remodeling/genetics , Cell Proliferation/drug effects , Collagen Type I/drug effects , Collagen Type I/metabolism , Collagen Type X/drug effects , Collagen Type X/genetics , Collagen Type X/metabolism , Enzyme-Linked Immunosorbent Assay , Fibroblast Growth Factors/drug effects , Fibroblast Growth Factors/metabolism , Fluorescent Antibody Technique , Gene Expression , Gene Expression Regulation , Glucuronidase/drug effects , Glucuronidase/genetics , Glucuronidase/metabolism , In Vitro Techniques , Klotho Proteins , Natriuretic Peptide, C-Type/pharmacology , Osteoblasts/drug effects , Osteocalcin/drug effects , Osteocalcin/metabolism , Osteogenesis/genetics , Peptide Fragments/drug effects , Peptide Fragments/metabolism , Primary Cell Culture , Procollagen/drug effects , Procollagen/metabolism , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Rats , Real-Time Polymerase Chain Reaction , Receptor, Fibroblast Growth Factor, Type 1/drug effects , Receptor, Fibroblast Growth Factor, Type 1/genetics , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Tartrate-Resistant Acid Phosphatase/drug effects , Tartrate-Resistant Acid Phosphatase/metabolism
14.
Biologicals ; 66: 9-16, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32561214

ABSTRACT

Bone tissue-derive biomaterials have become of great interest to treat diseases of the skeletal system. Biological scaffolds of demineralized and decellularized extracellular matrices (ECM) have been developed and one of these options are ECM hydrogels derived from bovine bone. Nanomaterials may be able to regulate stem cell differentiation due to their unique physical-chemical properties. The present work aimed to evaluate the osteoinductive effects of ECM hydrogels associated with barium titanate nanoparticles (BTNP) on dental pulp cells derived from exfoliated teeth. The addition of BTNP in the ECM derived hydrogel did not affect cell proliferation and the formation of bone nodules. Furthermore, it increased the expression of bone alkaline phosphatase. The results demonstrated that the nanobiocomposites were able to promote the osteogenic differentiation, even in the absence of chemical inducing factors for osteogenic differentiation. In conclusion, bovine bone ECM hydrogel combined with BTNP presented and increased expression of markers of osteogenic differentiation in the absence of chemical inducing factors.


Subject(s)
Barium Compounds/pharmacology , Cell Proliferation/drug effects , Extracellular Matrix , Hydrogels/pharmacology , Osteogenesis/drug effects , Stem Cells/drug effects , Titanium/pharmacology , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/genetics , Animals , Bone Demineralization Technique , Bone Morphogenetic Protein 2/drug effects , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 4/drug effects , Bone Morphogenetic Protein 4/genetics , Cattle , Dental Pulp/cytology , Glycosaminoglycans/metabolism , Humans , Metal Nanoparticles , Microscopy, Electron, Scanning , Osteogenesis/genetics , Rheology , Spectrum Analysis, Raman , Stem Cells/metabolism , Stem Cells/ultrastructure , Tissue Engineering/methods , Tissue Scaffolds
15.
Climacteric ; 23(4): 404-409, 2020 08.
Article in English | MEDLINE | ID: mdl-32410477

ABSTRACT

Objective: This study aims to investigate the beneficial effects of 17ß-estradiol supplementation on the function of osteoblastic cells through the Sirtuin-1/nuclear transcription factor-κB/matrix metalloproteinase-8 (Sirt1/NF-κB/MMP-8) pathway.Methods: Mouse primary osteoblasts were obtained from neonatal mouse calvaria, and the cells were treated with or without 17ß-estradiol. We first detected the effect of 17ß-estradiol on the function of osteoblastic cells. Then, the changes in estrogen receptor-α (ERα), Sirt1, NF-κB, and MMP-8 were determined after the osteoblasts were treated with 17ß-estradiol. During supplementation with 17ß-estradiol, knockdown of Sirt1 in osteoblasts was used to further measure the changes of NF-κB and MMP-8 and observe the cell function.Results: In primary osteoblastic cells, exposure to 17ß-estradiol improved cell viability and increased the levels of bone formation biomarkers, including osteocalcin, osteoprotegerin (OPG), procollagen type 1 N-terminal propeptide (P1NP), and alkaline phosphatase (ALP). In addition, 17ß-estradiol supplement activated ERα and Sirt1 expression and inhibited NF-κB and MMP-8 expression. Moreover, these effects induced by 17ß-estradiol were reversed by knockdown of Sirt1 in mouse primary osteoblasts.Conclusion: 17ß-Estradiol replacement therapy may treat postmenopausal osteoporosis by improving osteoblastic cell function via the Sirt1/NF-κB/MMP-8 pathway.


Subject(s)
Estradiol/pharmacology , Matrix Metalloproteinase 8/metabolism , NF-kappa B/metabolism , Osteoblasts/drug effects , Signal Transduction/drug effects , Sirtuin 1/metabolism , Alkaline Phosphatase/drug effects , Animals , Biomarkers/metabolism , Cell Survival/drug effects , Estrogen Replacement Therapy/methods , Female , Humans , Mice , Models, Animal , Osteoblasts/metabolism , Osteocalcin/drug effects , Osteoporosis, Postmenopausal/drug therapy , Osteoprotegerin/drug effects , Peptide Fragments/drug effects , Procollagen/drug effects
16.
Complement Ther Clin Pract ; 39: 101173, 2020 May.
Article in English | MEDLINE | ID: mdl-32379697

ABSTRACT

OBJECTIVE: Existing evidence on the possible effects of ginseng on liver function has not been fully established. Therefore, the present review was undertaken to evaluate the overall effects of ginseng supplementation on liver enzymes in adults. METHODS: A systematic computerized literature search of PubMed, Scopus, Web of Science, Cochrane Library and Google scholar databases was conducted up to May 2019. All RCTs using ginseng supplements in adults were included in this systematic review and meta-analysis. RESULTS: Overall, 14 randomized trials (with 20 arms) including 992 subjects were identified. Pooled analysis did not illustrate any significant changes in alanine aminotransferase (ALT), aspartate aminotransferase (AST), gamma-glutamyl transferase (GGT), alkaline phosphatase (ALP), and albumin (ALB) levels, however, it showed a minor significant increase in bilirubin (BIL) levels. Subgroup analysis by dosage and study population revealed significant increase of bilirubin after ginseng supplementation ≥3 g/day or in unhealthy individuals. CONCLUSION: Ginseng appears to have neither hepatoprotective nor hepatotoxic effects in conventional doses and duration. It is noteworthy that this seems applicable only for individuals with healthy liver function. Further largescale studies are warranted to confirm present findings.


Subject(s)
Complementary Therapies/adverse effects , Dietary Supplements/adverse effects , Liver/drug effects , Panax/chemistry , Plant Exudates/adverse effects , Plant Exudates/therapeutic use , Adult , Aged , Aged, 80 and over , Alanine Transaminase/drug effects , Alkaline Phosphatase/drug effects , Aspartate Aminotransferases/drug effects , Bilirubin/analysis , Female , Humans , Liver Function Tests , Male , Middle Aged , gamma-Glutamyltransferase/drug effects
17.
Ann Agric Environ Med ; 27(1): 66-75, 2020 Mar 17.
Article in English | MEDLINE | ID: mdl-32208582

ABSTRACT

OBJECTIVE: The aim of the study was to determine the effect of nesfatin-1 on bone properties in female rats in the conditions of developing osteopenia induced by ovariectomy (OVX). MATERIAL AND METHODS: The experiment was performed on 21 female Wistar rats assigned to 3 groups receiving intraperitoneally physiological saline (SHO, OVX-PhS) and nesfatin-1 in dose 2 µg/kg BW of (OVX-NES) once a day for 8 wks. At the end of the experiment, the rats were scanned using the DXA method to determine the body composition, tBMC, and tBMD. The isolated femora and tibia were tested with the DXA method for BMD and BMC, and with the pQCT method for separate analysis of the cortical and trabecular bone tissue. The bone strength parameters were also determined. The immunohistochemical method was used for determination of nesfatin-1 localization in growth cartilage. Bone metabolism markers (osteocalcin, bALP, and NTx) were identified using an ELISA kit. RESULTS: OVX exerts a negative effect on bone tissue. The nesfatin-1 administration influenced positively the DXA parameters of tibia. TvBMD and TbvBMD measured by pQCT in metaphysis of bones were significantly higher in the OVX-NES group than in OVX-PhS. No differences were found in the values of bone strength parameters between SHO and OVX-NES females. Extra- and intracellular immunohistochemical reaction for nesfatin-1 was observed in all zones of growth cartilage, with the strongest reaction detected in the calcifying zone. Nesfatin-1 administration caused a significant increase in the osteocalcin and bALP concentration in relation to the OVX-PhS animals. CONCLUSIONS: The results of the experiment indicate that nesfatin-1 exerts a protective effect on bone tissue properties and can be used in the prevention of osteoporosis.


Subject(s)
Bone Density/drug effects , Bone Diseases, Metabolic/drug therapy , Nucleobindins/pharmacology , Absorptiometry, Photon , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Bone Diseases, Metabolic/diagnostic imaging , Bone Diseases, Metabolic/metabolism , Cartilage/drug effects , Cartilage/metabolism , Female , Femur/diagnostic imaging , Femur/drug effects , Osteocalcin/metabolism , Ovariectomy , Rats, Wistar , Tibia/diagnostic imaging , Tibia/drug effects
20.
Eur J Trauma Emerg Surg ; 46(2): 287-299, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31028428

ABSTRACT

PURPOSE: Treatment of complex fractures in the elderly is a challenge for operative reconstruction due to degraded bone structure. Early peri-operative bone anabolic treatment could improve new bone formation, avoid implant loosening and accelerate fracture healing. METHODS: To compare the osteoanabolic potential of different drugs after distraction osteogenesis, 168 female Sprague-Dawley rats underwent lengthening of the right femur using a monolateral external fixator. Animals were randomly divided into six groups: vehicle-injected group, PTH(1-34), raloxifen, strontium ranelate, alendronate and simvastatin. Histomorphometry, CT-scanning, DEXA- and biomechanical analysis were performed to evaluate new bone formation, callus volume, mineralisation and biomechanical strength. Expression of bone metabolic mediators and differentiation indicators of distracted and intact bone were examined by RT-PCR and western blot. RESULTS: Histological analysis showed significant increase of the bone mass after treatment with PTH(1-34), raloxifen and strontium ranelate (p = 0.02). Raloxifen increased bone mineral content (BMC) of the whole distracted femur significantly (p = 0.007). Callus volume was significantly larger in the PTH(1-34), raloxifen and simvastatin groups (p = 0.001) compared to control. Ultimate load of distracted new formed bone was increased in PTH(1-34) and raloxifen groups. It seems that PTH(1-34) and raloxifen have a stronger effect on bone where a repair response is activated. Strontium ranelate demonstrates similar effects to PTH regarding new bone formation but shows low values for mineralisation and biomechanical strength. CONCLUSION: This study suggests that peri-operative treatment of complex and/or osteoporotic fractures with PTH(1-34) and raloxifen might be useful as a stimulator of bone formation and mineralisation to shorten the consolidation time in humans.


Subject(s)
Bone Density Conservation Agents/pharmacology , Bone Density/drug effects , Bone Regeneration/drug effects , Femur/drug effects , Osteogenesis/drug effects , Absorptiometry, Photon , Alendronate/pharmacology , Alkaline Phosphatase/drug effects , Alkaline Phosphatase/metabolism , Animals , Biomechanical Phenomena/drug effects , Blotting, Western , Bone Morphogenetic Protein 2/drug effects , Bone Morphogenetic Protein 2/genetics , Bony Callus/diagnostic imaging , Bony Callus/metabolism , Bony Callus/pathology , Calcium-Regulating Hormones and Agents/pharmacology , Collagen Type I/drug effects , Collagen Type I/metabolism , Female , Femur/diagnostic imaging , Femur/pathology , Femur/surgery , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Interleukin-6/genetics , Macrophage Colony-Stimulating Factor/drug effects , Macrophage Colony-Stimulating Factor/genetics , Osteocalcin/drug effects , Osteocalcin/genetics , Osteogenesis/genetics , Osteogenesis, Distraction , Parathyroid Hormone/pharmacology , RANK Ligand/drug effects , RANK Ligand/genetics , Raloxifene Hydrochloride/pharmacology , Rats , Reverse Transcriptase Polymerase Chain Reaction , Simvastatin/pharmacology , Thiophenes/pharmacology , Tomography, X-Ray Computed
SELECTION OF CITATIONS
SEARCH DETAIL
...