Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.883
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Mol Cell ; 81(2): 340-354.e5, 2021 01 21.
Article in English | MEDLINE | ID: mdl-33450210

ABSTRACT

In addition to its role as an electron transporter, mitochondrial nicotinamide adenine dinucleotide (NAD+) is an important co-factor for enzymatic reactions, including ADP-ribosylation. Although mitochondria harbor the most intra-cellular NAD+, mitochondrial ADP-ribosylation remains poorly understood. Here we provide evidence for mitochondrial ADP-ribosylation, which was identified using various methodologies including immunofluorescence, western blot, and mass spectrometry. We show that mitochondrial ADP-ribosylation reversibly increases in response to respiratory chain inhibition. Conversely, H2O2-induced oxidative stress reciprocally induces nuclear and reduces mitochondrial ADP-ribosylation. Elevated mitochondrial ADP-ribosylation, in turn, dampens H2O2-triggered nuclear ADP-ribosylation and increases MMS-induced ARTD1 chromatin retention. Interestingly, co-treatment of cells with the mitochondrial uncoupler FCCP decreases PARP inhibitor efficacy. Together, our results suggest that mitochondrial ADP-ribosylation is a dynamic cellular process that impacts nuclear ADP-ribosylation and provide evidence for a NAD+-mediated mitochondrial-nuclear crosstalk.


Subject(s)
ADP-Ribosylation , Cell Nucleus/enzymology , Mitochondria/enzymology , NAD/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , ADP-Ribosylation/drug effects , Animals , Antimycin A/analogs & derivatives , Antimycin A/pharmacology , Cell Line , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Chromatin/chemistry , Chromatin/metabolism , Electron Transport/drug effects , HeLa Cells , Humans , Hydrogen Peroxide/pharmacology , Methacrylates/pharmacology , Mice , Mice, Inbred C57BL , Mitochondria/drug effects , Mitochondria/genetics , Myoblasts/cytology , Myoblasts/drug effects , Myoblasts/enzymology , Oligomycins/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/enzymology , Poly (ADP-Ribose) Polymerase-1/genetics , Rotenone/pharmacology , Thiazoles/pharmacology
2.
J Biol Chem ; 298(3): 101639, 2022 03.
Article in English | MEDLINE | ID: mdl-35090892

ABSTRACT

Phosphatidylinositol-4-phosphate 5-kinase type-1 gamma (Pip5k1c) is a lipid kinase that plays a pivotal role in the regulation of receptor-mediated calcium signaling in multiple tissues; however, its role in the skeleton is not clear. Here, we show that while deleting Pip5k1c expression in the mesenchymal stem cells using Prx1-Cre transgenic mice does not impair the intramembranous and endochondral ossification during skeletal development, it does cause osteopenia in adult mice, but not rapidly growing young mice. We found Pip5k1c loss dramatically decreases osteoblast formation and osteoid and mineral deposition, leading to reduced bone formation. Furthermore, Pip5k1c loss inhibits osteoblastic, but promotes adipogenic, differentiation of bone marrow stromal cells. Pip5k1c deficiency also impairs cytoplasmic calcium influx and inactivates the calcium/calmodulin-dependent protein kinase, which regulates levels of transcription factor Runx2 by modulating its stability and subsequent osteoblast and bone formation. In addition, Pip5k1c loss reduces levels of the receptor activator of nuclear factor-κB ligand, but not that of osteoprotegerin, its decoy receptor, in osteoblasts in bone and in sera. Finally, we found Pip5k1c loss impairs the ability of bone marrow stromal cells to support osteoclast formation of bone marrow monocytes and reduces the osteoclast precursor population in bone marrow, resulting in reduced osteoclast formation and bone resorption. We conclude Pip5k1c deficiency causes a low-turnover osteopenia in mice, with impairment of bone formation being greater than that of bone resorption. Collectively, we uncover a novel function and mechanism of Pip5k1c in the control of bone mass and identify a potential therapeutic target for osteoporosis.


Subject(s)
Bone Diseases, Metabolic , Bone Resorption , Mesenchymal Stem Cells , Phosphotransferases (Alcohol Group Acceptor) , Animals , Bone Diseases, Metabolic/genetics , Bone Diseases, Metabolic/metabolism , Bone Remodeling/physiology , Bone Resorption/enzymology , Bone Resorption/metabolism , Calcium/metabolism , Cell Differentiation/physiology , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/enzymology , Mesenchymal Stem Cells/metabolism , Mice , Osteoblasts/cytology , Osteoblasts/enzymology , Osteoblasts/metabolism , Osteoclasts/cytology , Osteoclasts/enzymology , Osteoclasts/metabolism , Osteogenesis , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Phosphotransferases (Alcohol Group Acceptor)/metabolism , RANK Ligand/metabolism
3.
Hum Mol Genet ; 29(2): 274-285, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31816052

ABSTRACT

Bone differentiation defects have been recently tied to Wnt signaling alterations occurring in vitro and in vivo Gaucher disease (GD) models. In this work, we provide evidence that the Wnt signaling multi-domain intracellular transducers Dishevelled 1 and 2 (DVL1 and DVL2) may be potential upstream targets of impaired beta glucosidase (GBA1) activity by showing their misexpression in different type 1 GD in vitro models. We also show that in Gba mutant fish a miR-221 upregulation is associated with reduced dvl2 expression levels and that in type I Gaucher patients single-nucleotide variants in the DVL2 3' untranslated region are related to variable canonical Wnt pathway activity. Thus, we strengthen the recently outlined relation between bone differentiation defects and Wnt/ß-catenin dysregulation in type I GD and further propose novel mechanistic insights of the Wnt pathway impairment caused by glucocerebrosidase loss of function.


Subject(s)
Dishevelled Proteins/metabolism , Gaucher Disease/metabolism , Glucosylceramidase/metabolism , Wnt Signaling Pathway/genetics , Zebrafish/metabolism , 3' Untranslated Regions , Adaptor Proteins, Vesicular Transport/genetics , Adaptor Proteins, Vesicular Transport/metabolism , Animals , Animals, Genetically Modified , Cell Line , Disease Models, Animal , Dishevelled Proteins/genetics , Gaucher Disease/enzymology , Gaucher Disease/genetics , Glucosylceramidase/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/genetics , Membrane Proteins/metabolism , MicroRNAs/genetics , MicroRNAs/metabolism , Osteoblasts/enzymology , Osteoblasts/metabolism , Osteoblasts/pathology , Transcription, Genetic , Zebrafish/genetics , Zebrafish Proteins/genetics , Zebrafish Proteins/metabolism
4.
Horm Metab Res ; 54(1): 42-49, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34986499

ABSTRACT

Incretins including glucose-dependent insulinotropic polypeptide (GIP) and glucagon-like peptide-1 (GLP-1), which are secreted from the small intestine after oral food ingestion, are currently well-known to stimulate insulin secretion from pancreatic ß-cells and used for the treatment of type 2 diabetes mellitus. We have previously reported that prostaglandin F2α (PGF2α) stimulates the synthesis of interleukin-6 (IL-6) and osteoprotegerin in osteoblast-like MC3T3-E1 cells, and that IL-6 and osteoprotegerin release are mediated through the p44/p42 mitogen-activated protein (MAP) kinase, p38 MAP kinase or stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) pathways. In the present study, we investigated the effects of incretins including GLP-1 and GIP, on the PGF2α-induced synthesis of IL-6 and osteoprotegerin and examined the detailed mechanism in osteoblast-like MC3T3-E1 cells. We found that GIP and GLP-1 significantly stimulated the PGF2α-induced synthesis of IL-6 in osteoblast-like MC3T3-E1 cells. In addition, GIP and GLP-1 significantly enhanced the PGF2α-induced mRNA expression levels of IL-6. On the other hand, GIP and GLP-1 markedly stimulated the PGF2α-induced synthesis of osteoprotegerin. However, the phosphorylation of p44/p42 MAP kinase, p38 MAP kinase, or JNK induced by PGF2α was not affected by GIP or GLP-1. Therefore, these results strongly suggest that incretins enhance the PGF2α-induced synthesis of IL-6 and osteoprotegerin in osteoblast-like MC3T3-E1 cells. However, these syntheses are not mediated through p44/p42 MAP kinase, p38 MAP kinase, or JNK pathways.


Subject(s)
Dinoprost/pharmacology , Gastric Inhibitory Polypeptide/pharmacology , Glucagon-Like Peptide 1/pharmacology , Incretins/metabolism , Interleukin-6/biosynthesis , Osteoblasts/metabolism , Osteoprotegerin/biosynthesis , Animals , Cell Line , Gene Expression Regulation/drug effects , Interleukin-6/genetics , MAP Kinase Signaling System/drug effects , Mice , Osteoblasts/drug effects , Osteoblasts/enzymology , Phosphorylation/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism
5.
J Cell Physiol ; 236(6): 4614-4624, 2021 06.
Article in English | MEDLINE | ID: mdl-33305372

ABSTRACT

Supraphysiological levels of the osteoblast-enriched mineralization regulator ectonucleotide pyrophosphatase or phosphodiesterase-1 (NPP1) is associated with type 2 diabetes mellitus. We determined the impact of osteoblast-specific Enpp1 ablation on skeletal structure and metabolic phenotype in mice. Female, but not male, 6-week-old mice lacking osteoblast NPP1 expression (osteoblast-specific knockout [KO]) exhibited increased femoral bone volume or total volume (17.50% vs. 11.67%; p < .01), and reduced trabecular spacing (0.187 vs. 0.157 mm; p < .01) compared with floxed (control) mice. Furthermore, an enhanced ability of isolated osteoblasts from the osteoblast-specific KO to calcify their matrix in vitro compared to fl/fl osteoblasts was observed (p < .05). Male osteoblast-specific KO and fl/fl mice showed comparable glucose and insulin tolerance despite increased levels of insulin-sensitizing under-carboxylated osteocalcin (195% increase; p < .05). However, following high-fat-diet challenge, osteoblast-specific KO mice showed impaired glucose and insulin tolerance compared with fl/fl mice. These data highlight a crucial local role for osteoblast NPP1 in skeletal development and a secondary metabolic impact that predominantly maintains insulin sensitivity.


Subject(s)
Bone and Bones/enzymology , Diet, High-Fat/adverse effects , Insulin Resistance , Osteoblasts/enzymology , Osteogenesis , Phosphoric Diester Hydrolases/deficiency , Pyrophosphatases/deficiency , Animals , Biomarkers/blood , Blood Glucose/metabolism , Bone and Bones/pathology , Cancellous Bone/enzymology , Cancellous Bone/pathology , Cells, Cultured , Disease Models, Animal , Female , Femur/enzymology , Femur/pathology , Insulin/blood , Male , Mice, Knockout , Osteoblasts/pathology , Osteocalcin/blood , Phosphoric Diester Hydrolases/genetics , Pyrophosphatases/genetics , Sex Factors , Skull/enzymology , Skull/pathology , Tibia/enzymology , Tibia/pathology
6.
J Cell Physiol ; 236(6): 4764-4777, 2021 06.
Article in English | MEDLINE | ID: mdl-33275302

ABSTRACT

Primary cilia have been found to function as mechanosensors in low-magnitude high-frequency vibration (LMHFV)-induced osteogenesis. The PGE2 also regulates bone homeostasis and mechanical osteogenesis through its receptor EP4 signaling, but its involvement in LMHFV-induced or in primary cilia-induced osteogenesis has not been investigated. We hypothesized that LMHFV stimulates osteoblast (OB) differentiation by activating the COX2-PGE2-EP pathway in a manner dependent on primary cilia and that primary cilia are also affected by the PGE2 pathway. In this study, through western blot analysis, RNA interference, enzyme-linked immunosorbent assay, real-time quantitative polymerase chain reaction, and cytochemical staining, we observed that COX2, mPGES-1, and PGE2 levels were markedly elevated in cells treated with LMHFV and were greatly decreased in LMHFV-treated cells following IFT88 silencing. EP4 expression was significantly increased in OBs following LMHFV treatment, but IFT88 silencing significantly blocked this increase. EP4 localized to the bases of primary cilia. LMHFV reduced the length and abundance of primary cilia, but the cells could self-repair their primary cilia after mechanical damage. EP4 antagonism significantly blocked the LMHFV-induced increase in IFT88 expression and blocked the recovery of primary cilia length and the proportion of cells with primary cilia. In addition, COX2 or EP4 antagonism disrupted LMHFV-induced osteogenesis. These results demonstrate the integration of and crosstalk between primary cilia and the COX2-PGE2-EP4 signaling pathway under mechanical stimulation.


Subject(s)
Cell Differentiation , Cilia/enzymology , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Mechanotransduction, Cellular , Osteoblasts/enzymology , Osteogenesis , Receptors, Prostaglandin E, EP4 Subtype/metabolism , 3T3 Cells , Animals , Cell Differentiation/drug effects , Cilia/drug effects , Cyclooxygenase 2/genetics , Cyclooxygenase 2 Inhibitors/pharmacology , Mice , Osteoblasts/drug effects , Osteogenesis/drug effects , Physical Stimulation , Prostaglandin Antagonists/pharmacology , Prostaglandin-E Synthases/genetics , Prostaglandin-E Synthases/metabolism , Receptors, Prostaglandin E, EP4 Subtype/antagonists & inhibitors , Receptors, Prostaglandin E, EP4 Subtype/genetics , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Vibration
7.
J Pathol ; 250(1): 30-41, 2020 01.
Article in English | MEDLINE | ID: mdl-31509234

ABSTRACT

Medial arterial calcification (MAC) is a major complication of chronic kidney disease (CKD) and an indicator of poor prognosis. Aortic overexpression of tissue-nonspecific alkaline phosphatase (TNAP) accelerates MAC formation. The present study aimed to assess whether a TNAP inhibitor, SBI-425, protects against MAC and improves survival probability in a CKD-mineral and bone disorder (MBD) mouse model. CKD-MBD mice were divided in three groups: vehicle, SBI-10, and SBI-30. They were fed a 0.2% adenine and 0.8% phosphorus diet from 14 to 20 weeks of age to induce CKD, followed by a high-phosphorus (0.2% adenine and 1.8% phosphorus) diet for another 6 weeks. At 14-20 weeks of age, mice in the SBI-10 and SBI-30 groups were given 10 and 30 mg/kg SBI-425 by gavage once a day, respectively, while vehicle-group mice were given distilled water as vehicle. Control mice were fed a standard chow (0.8% phosphorus) between the ages of 8 and 20 weeks. Computed tomography imaging, histology, and aortic tissue calcium content revealed that, compared to vehicle animals, SBI-425 nearly halted the formation of MAC. Mice in the control, SBI-10 and SBI-30 groups exhibited 100% survival, which was significantly better than vehicle-treated mice (57.1%). Aortic mRNA expression of Alpl, encoding TNAP, as well as plasma and aortic tissue TNAP activity, were suppressed by SBI-425 administration, whereas plasma pyrophosphate increased. We conclude that a TNAP inhibitor successfully protected the vasculature from MAC and improved survival rate in a mouse CKD-MBD model, without causing any adverse effects on normal skeletal formation and residual renal function. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Alkaline Phosphatase/antagonists & inhibitors , Aorta/drug effects , Aortic Diseases/prevention & control , Chronic Kidney Disease-Mineral and Bone Disorder/drug therapy , Enzyme Inhibitors/pharmacology , Niacinamide/analogs & derivatives , Sulfonamides/pharmacology , Vascular Calcification/prevention & control , Alkaline Phosphatase/genetics , Alkaline Phosphatase/metabolism , Animals , Aorta/enzymology , Aorta/pathology , Aortic Diseases/enzymology , Aortic Diseases/etiology , Aortic Diseases/pathology , Chronic Kidney Disease-Mineral and Bone Disorder/complications , Chronic Kidney Disease-Mineral and Bone Disorder/enzymology , Chronic Kidney Disease-Mineral and Bone Disorder/pathology , Disease Models, Animal , Gene Expression Regulation , Male , Mice, Inbred C57BL , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/drug effects , Myocytes, Smooth Muscle/enzymology , Myocytes, Smooth Muscle/pathology , Niacinamide/pharmacology , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteoblasts/pathology , Time Factors , Vascular Calcification/enzymology , Vascular Calcification/etiology , Vascular Calcification/pathology
8.
Stem Cells ; 37(6): 766-778, 2019 06.
Article in English | MEDLINE | ID: mdl-30786091

ABSTRACT

Aberrant wound healing presents as inappropriate or insufficient tissue formation. Using a model of musculoskeletal injury, we demonstrate that loss of transforming growth factor-ß activated kinase 1 (TAK1) signaling reduces inappropriate tissue formation (heterotopic ossification) through reduced cellular differentiation. Upon identifying increased proliferation with loss of TAK1 signaling, we considered a regenerative approach to address insufficient tissue production through coordinated inactivation of TAK1 to promote cellular proliferation, followed by reactivation to elicit differentiation and extracellular matrix production. Although the current regenerative medicine paradigm is centered on the effects of drug treatment ("drug on"), the impact of drug withdrawal ("drug off") implicit in these regimens is unknown. Because current TAK1 inhibitors are unable to phenocopy genetic Tak1 loss, we introduce the dual-inducible COmbinational Sequential Inversion ENgineering (COSIEN) mouse model. The COSIEN mouse model, which allows us to study the response to targeted drug treatment ("drug on") and subsequent withdrawal ("drug off") through genetic modification, was used here to inactivate and reactivate Tak1 with the purpose of augmenting tissue regeneration in a calvarial defect model. Our study reveals the importance of both the "drug on" (Cre-mediated inactivation) and "drug off" (Flp-mediated reactivation) states during regenerative therapy using a mouse model with broad utility to study targeted therapies for disease. Stem Cells 2019;37:766-778.


Subject(s)
Bone Regeneration/drug effects , Fractures, Bone/genetics , MAP Kinase Kinase Kinases/genetics , Mesenchymal Stem Cells/enzymology , Osteoblasts/enzymology , Wound Healing/genetics , Animals , Bone Regeneration/genetics , Cell Differentiation/drug effects , Cell Proliferation/drug effects , DNA Nucleotidyltransferases/genetics , DNA Nucleotidyltransferases/metabolism , Female , Founder Effect , Fractures, Bone/drug therapy , Fractures, Bone/enzymology , Fractures, Bone/pathology , Gene Expression Regulation , Integrases/genetics , Integrases/metabolism , MAP Kinase Kinase Kinases/antagonists & inhibitors , MAP Kinase Kinase Kinases/deficiency , Male , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Transgenic , Osteoblasts/cytology , Osteoblasts/drug effects , Primary Cell Culture , Protein Kinase Inhibitors/pharmacology , Signal Transduction , Skull/drug effects , Skull/injuries , Skull/metabolism , Wound Healing/drug effects
9.
Biosci Biotechnol Biochem ; 84(3): 526-535, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31743080

ABSTRACT

Lysyl oxidase (LOX) is required for the formation of bone collagen cross-links. Inactivation of the LOX gene in osteoblasts by DNA methylation and JAK signaling has been reported to cause loss of cross-links and an increased risk of fractures. Tocotrienols (T3s) have proven benefits on bone strength, but their potential effects on LOX remain largely unknown. Thus, the present study investigates the in vitro effects of T3s on LOX expression in human osteoblastic MG-63 cells. Results indicated that Tocotrienol-Rich Fraction (TRF), the δ-T3 rich oil extracted from Annatto was the most effective and significantly increased LOX expression. TRF treatment decreased de-novo methyltransferases (DNMTs), DNMT3A and DNMT3B levels. In addition, TRF significantly inhibited JAK2 activation and decreased expression of Fli1, a transcription factor of DNMTs. We conclude that TRF induced an increase in LOX expression via inhibition of de-novo methylation and reduction of Fli1 expression by the inactivation of JAK2.Abbreviations: CpG: cytosine-guanine dinucleotide; DNMT: DNA methyltransferase; Fli1: friend leukemia virus integration 1; JAK: janus kinase; LOX: lysyl oxidase; PCR: polymerase chain reaction; STAT: signal transducers and activators of transcription; T3s: tocotrienols; TPs: tocopherols; TRF: Tocotrienol-Rich Fraction.


Subject(s)
Bixaceae/metabolism , Carotenoids/metabolism , Osteoblasts/metabolism , Plant Extracts/metabolism , Protein-Lysine 6-Oxidase/metabolism , Tocotrienols/metabolism , Cell Line , Humans , Osteoblasts/enzymology
10.
J Orthop Sci ; 25(2): 331-337, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31072650

ABSTRACT

BACKGROUND: Mechanical unloading induces bone loss in human weight-loaded bones. The findings of recent studies have revealed that cluster of differentiation 38 knockout mice display bone loss similar to that observed in osteoporosis. This study aimed to determine whether the expression of cluster of differentiation 38 is implicated in skeletal unloading and reloading. METHODS: Eight-week-old male C57BL/6J mice were assigned to control, tail-suspension, or reloading after tail-suspension groups. In the tail-suspension group, tail suspension elevated the hind limbs for 1 week. The bilateral femurs and tibias from the groups were evaluated for cluster of differentiation 38 immunocytochemistry, and the cluster of differentiation 38 messenger ribonucleic acid levels and the expression of cluster of differentiation 38 and other cell-surface antigens were evaluated using quantitative real-time polymerase chain reaction and flow cytometric analyses. RESULTS: In the tail-suspension group, the alkaline phosphatase reactivity, cluster of differentiation 38 immunoreactivity in the bone marrow and osteoblasts, and the expression of cluster of differentiation 38 messenger ribonucleic acid and that of other cell-surface antigens were significantly lower than those in the control group. In the reloading after tail-suspension group, the level of cluster of differentiation 38 expression was restored to the same level as that in the control group. CONCLUSIONS: Cluster of differentiation 38 expression declined after skeletal unloading and recovered to normal levels after reloading. In the bone marrow, cluster of differentiation 38 expression plays a crucial role in bone formation in response to mechanical stress.


Subject(s)
ADP-ribosyl Cyclase 1/physiology , Bone Diseases, Metabolic/enzymology , Bone Marrow Cells/enzymology , Cyclic ADP-Ribose/metabolism , Osteoblasts/enzymology , Weight-Bearing , Animals , Femur/metabolism , Hindlimb Suspension , Male , Mice , Mice, Inbred C57BL
11.
Int J Mol Sci ; 21(1)2020 Jan 02.
Article in English | MEDLINE | ID: mdl-31906530

ABSTRACT

Fused deposit modeling (FDM) 3D printing technology cannot generate scaffolds with high porosity while maintaining good integrity, anatomical-surface detail, or high surface area-to-volume ratio (S/V). Solvent casting and particulate leaching (SCPL) technique generates scaffolds with high porosity and high S/V. However, it is challenging to generate complex-shaped scaffolds; and solvent, particle and residual water removal are time consuming. Here we report techniques surmounting these problems, successfully generating a highly porous scaffold with the anatomical-shape characteristics of a human femur by polylactic acid polymer (PLA) and PLA-hydroxyapatite (HA) casting and salt leaching. The mold is water soluble and is easily removable. By perfusing with ethanol, water, and dry air sequentially, the solvent, salt, and residual water were removed 20 fold faster than utilizing conventional methods. The porosities are uniform throughout the femoral shaped scaffold generated with PLA or PLA-HA. Both scaffolds demonstrated good biocompatibility with the pre-osteoblasts (MC3T3-E1) fully attaching to the scaffold within 8 h. The cells demonstrated high viability and proliferation throughout the entire time course. The HA-incorporated scaffolds demonstrated significantly higher compressive strength, modulus and osteoinductivity as evidenced by higher levels of alkaline-phosphatase activity and calcium deposition. When 3D printing a 3D model at 95% porosity or above, our technology preserves integrity and surface detail when compared with FDM-generated scaffolds. Our technology can also generate scaffolds with a 31 fold larger S/V than FDM. We have developed a technology that is a versatile tool in creating personalized, patient-specific bone graft scaffolds efficiently with high porosity, good scaffold integrity, high anatomical-shaped surface detail and large S/V.


Subject(s)
Biocompatible Materials/chemistry , Osteoblasts/chemistry , Printing, Three-Dimensional , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Biocompatible Materials/chemical synthesis , Calcium/analysis , Cell Differentiation , Cell Proliferation , Cell Survival , Compressive Strength , Durapatite/chemistry , Femur , Humans , Materials Testing , Osteoblasts/enzymology , Osteoblasts/metabolism , Perfusion , Polyesters/chemistry , Porosity , Tissue Scaffolds/adverse effects
12.
Int J Mol Sci ; 21(3)2020 Jan 23.
Article in English | MEDLINE | ID: mdl-31979313

ABSTRACT

Titanium (Ti) is an osteoconductive material that is routinely used as a bulk implant to fix and restore bones and teeth. This study explored the effective use of Ti as a bone engineering scaffold. Challenges to overcome were: (1) difficult liquid/cell infiltration into Ti microfiber scaffolds due to the hydrophobic nature of Ti; and (2) difficult cell attachment on thin and curved Ti microfibers. A recent discovery of UV-photofunctionalization of Ti prompted us to examine its effect on Ti microfiber scaffolds. Scaffolds in disk form were made by weaving grade 4 pure Ti microfibers (125 µm diameter) and half of them were acid-etched to roughen the surface. Some of the scaffolds with original or acid-etched surfaces were further treated by UV light before cell culture. Ti microfiber scaffolds, regardless of the surface type, were hydrophobic and did not allow glycerol/water liquid to infiltrate, whereas, after UV treatment, the scaffolds became hydrophilic and immediately absorbed the liquid. Osteogenic cells from two different origins, derived from the femoral and mandibular bone marrow of rats, were cultured on the scaffolds. The number of cells attached to scaffolds during the early stage of culture within 24 h was 3-10 times greater when the scaffolds were treated with UV. The development of cytoplasmic projections and cytoskeletal, as well as the expression of focal adhesion protein, were exclusively observed on UV-treated scaffolds. Osteoblastic functional phenotypes, such as alkaline phosphatase activity and calcium mineralization, were 2-15 times greater on UV-treated scaffolds, with more pronounced enhancement on acid-etched scaffolds compared to that on the original scaffolds. These effects of UV treatment were associated with a significant reduction in atomic carbon on the Ti microfiber surfaces. In conclusion, UV treatment of Ti microfiber scaffolds tunes their physicochemical properties and effectively enhances the attachment and function of osteoblasts, proposing a new strategy for bone engineering.


Subject(s)
Osseointegration , Osteoblasts/metabolism , Tissue Scaffolds/chemistry , Titanium/radiation effects , Animals , Bone Marrow Cells/cytology , Calcification, Physiologic/physiology , Cell Culture Techniques , Cells, Cultured , Femur/cytology , Hydrophobic and Hydrophilic Interactions , Male , Mandible/cytology , Microscopy, Electron, Scanning , Osteoblasts/chemistry , Osteoblasts/enzymology , Osteogenesis/physiology , Rats , Rats, Sprague-Dawley , Surface Properties/radiation effects , Tissue Engineering , Titanium/chemistry , Ultraviolet Rays
13.
J Cell Physiol ; 234(4): 4472-4490, 2019 04.
Article in English | MEDLINE | ID: mdl-30192012

ABSTRACT

Glucocorticoid-induced osteoporosis (GIO) is a secondary osteoporosis with extensive use of glucocorticoids (GCs). GCs can increase bone fragility and fracture via inhibiting osteoblastic proliferation and differentiation. Luteolin (LUT), a kind of plant flavonoid, has been reported to exhibit the antioxidant activity, but the effects of LUT on GIO still remain unclear. This study aimed to investigate the effects of LUT on GIO both in vivo and in vitro and elaborate the potential molecular mechanisms. LUT increased the superoxide dismutase activity, glutathione level and decreased reactive oxygen species (ROS) level and lactate dehydrogenase release in GIO. Meanwhile, LUT decreased caspase-3, caspase-9, and Bax protein expressions and increased Bcl-2 protein expression in GIO. LUT increased the ratio of osteoprotegerin (OPG)/receptor activator of nuclear factor-κB Ligand (RANKL) messenger RNA (mRNA) expression and mRNA expression levels of osteogenic markers, including runt-related transcription factor 2, osterix, collagen type I, and osteocalcin. LUT also enhanced the extracellular signal-regulated kinases (ERK) phosphorylation, glycogen synthase kinase 3ß (GSK-3ß) phosphorylation, mRNA expression levels of lipoprotein-receptor-related protein 5 (Lrp-5) and ß-catenin. Further study revealed that Lrp-5 small interfering RNA (siRNA )and ERK-siRNA reduced the effects of LUT on GSK-3ß phosphorylation, alkaline phosphatase (ALP) activity and the ratio of OPG/RANKL mRNA expression. Moreover, ERK-siRNA decreased Lrp-5 mRNA expression in vitro. These results indicated that LUT promoted proliferation by attenuating oxidative stress and promoted osteoblastic differentiation by regulating the ERK/Lrp-5/GSK-3ß pathway in GIO. This study may bring to light the possible mechanisms involved in the action of LUT in GIO treatment, and benefit for further research on GIO.


Subject(s)
Dexamethasone , Extracellular Signal-Regulated MAP Kinases/metabolism , Femur/drug effects , Glucocorticoids , Glycogen Synthase Kinase 3 beta/metabolism , Low Density Lipoprotein Receptor-Related Protein-5/metabolism , Luteolin/pharmacology , Osteoblasts/drug effects , Osteoporosis/prevention & control , Signal Transduction/drug effects , 3T3 Cells , Animals , Apoptosis/drug effects , Bone Density/drug effects , Cancellous Bone/drug effects , Cancellous Bone/enzymology , Cancellous Bone/pathology , Cell Proliferation/drug effects , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Femur/enzymology , Femur/pathology , Low Density Lipoprotein Receptor-Related Protein-5/genetics , Mice , Osteoblasts/enzymology , Osteoblasts/pathology , Osteogenesis/drug effects , Osteoporosis/chemically induced , Osteoporosis/enzymology , Osteoporosis/pathology , Oxidative Stress/drug effects , Phosphorylation
14.
J Cell Biochem ; 120(9): 15069-15082, 2019 09.
Article in English | MEDLINE | ID: mdl-31020682

ABSTRACT

Modular bone tissue engineering is touted as an alternative approach to replace the damaged bone tissue. Hydrogel microcapsules could promote therapeutic properties by providing 3D condition and an increased cell-to-cell interaction. We investigated the osteogenic properties of alginate-nano-silica hydrogels enriched with collagen and gelatin on human osteoblast-like MG-63 cells. For encapsulation, cells were divided into three groups; control (alginate+ nano-silica), collagen (alginate + collagen + nano-silica), and gelatin (alginate + gelatin + nano-silica) and expanded for 28 days. Cell survival was determined by trypan blue staining and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay. To confirm the osteogenic potential, we measured the alkaline phosphatase activity. Alizarin red S staining was used to reveal the existence of hydroxyapatite and transcription BMP-2, osteocalcin and osteonectin evaluated by the real-time polymerase chain reaction. Collagen substrate caused a reduced swelling ratio compared with the control and gelatin groups (P < 0.05). Compared with other groups, collagen had potential to improve mechanical strength and generate porous membrane structure. The addition of collagen (4-fold) and gelatin (1.5-fold) increased cell proliferation rate compared with the control (P < 0.05). Biochemical analysis and Alizarin red S staining showed that collagen-induced osteogenesis by induction of alkaline phosphatase and matrix mineralization. The expression of osteocalcin and BMP-2 was increased in cells from the collagen group. As a result, the combination of natural polymers collagen and gelatin with alginate + nano-silica can increase the osteogenic potential of human osteoblasts.


Subject(s)
Alginates/pharmacology , Collagen/pharmacology , Microspheres , Osteoblasts/metabolism , Osteogenesis/drug effects , Silicon Dioxide/pharmacology , Alkaline Phosphatase/metabolism , Animals , Biocompatible Materials/pharmacology , Bone Morphogenetic Protein 2/genetics , Bone Morphogenetic Protein 2/metabolism , Calcium/metabolism , Cattle , Cell Line , Cell Survival/drug effects , Gelatin/pharmacology , Humans , Hydrogels/chemistry , Mechanical Phenomena , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteocalcin/genetics , Osteocalcin/metabolism , Osteogenesis/genetics , Osteonectin/genetics , Osteonectin/metabolism , Tissue Scaffolds/chemistry
15.
J Cell Mol Med ; 22(2): 1127-1137, 2018 02.
Article in English | MEDLINE | ID: mdl-29063674

ABSTRACT

Bisphosphonates impair function of osteoclasts and prevent bone resorption, the mechanism of which has been studied extensively. However, the possible effects of bisphosphonates on chondroblast differentiation and calcium deposition by osteoblasts have only been demonstrated recently. Moreover, cells from monocytic lineage are capable of stimulating osteoblast proliferation. Hence, susceptibility of osteoblasts to various factors requires further investigation. A primary culture of bone marrow-derived stromal cells was treated with liposomal clodronate (0.1, 0.5, or 1.0 mg/ml) or conditioned medium from liposomal clodronate. Liposomal clodronate (0.25 mg) was injected into mouse femur for in vivo experiments. The effects of liposomal clodronate were examined by alkaline phosphatase staining and/or activity assay, and real-time RT-PCR was used for studying the effect on osteogenic gene expression. Administration of liposomal clodronate to bone marrow-derived mesenchymal stromal cell culture enhanced alkaline phosphatase activity and mRNA levels of Runx2 and Dlx5. In addition, conditioned medium from liposomal clodronate also stimulated osteogenic characteristics similar to those of observed in vitro, and the number of exosomes in the conditioned medium was highest when pre-treated with liposomal clodronate. Western blot analysis revealed the presence of RANK proteins in exosomes collected from conditioned medium of liposomal clodronate. Identical observations were obtained in vivo, as liposomal clodronate-injected mouse femur showed increased alkaline phosphatase activity and Runx2 and Dlx5 mRNA expressions, even though the numbers of monocytes and macrophages were reduced. In conclusion, osteoblast differentiation was promoted via soluble RANK-containing exosomes in response to clodronates.


Subject(s)
Cell Differentiation/drug effects , Clodronic Acid/pharmacology , Osteoblasts/cytology , Alkaline Phosphatase/metabolism , Animals , Cell Count , Cell Survival/drug effects , Cells, Cultured , Culture Media, Conditioned/pharmacology , Exosomes/metabolism , Female , Femur/cytology , Gene Expression Regulation/drug effects , Liposomes , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/enzymology , Mice, Inbred ICR , Monocytes/cytology , Osteoblasts/drug effects , Osteoblasts/enzymology , Osteoblasts/metabolism , Osteogenesis/drug effects , Osteogenesis/genetics
16.
Dev Biol ; 431(2): 252-262, 2017 11 15.
Article in English | MEDLINE | ID: mdl-28899668

ABSTRACT

Sp7 is a zinc finger transcription factor that is essential for osteoblast differentiation in mammals. To verify the characteristic features of osteoblast-lineage cells in teleosts, we established medaka sp7 mutants using a transcription activator-like effector nuclease (TALEN) genome editing system. These mutants showed severe defects in the formation of skeletal structures. In particular, the neural and the hemal arches were not formed, although the chordal centra were formed. Analysis of the transgenic medaka revealed that sp7 mutant had normal distribution of type X collagen a1 a (col10a1a)-positive osteoblast-like cells around the centrum and at the proximal region of the vertebral arch. The sp7 mutant phenotype could be rescued by exogenous sp7 expression in col10a1a-positive cells, as well as in sp7-positive osteoblast cells. Furthermore, runx2-positive osteoblast progenitors were observed on the vertebral arches, but not on the centrum, during vertebral column development. In addition, these osteoblast progenitors differentiated into the col10a1a-positive cells. In sp7 mutant, the runx2-positive cells were normally distributed at the region of unformed vertebral arch but failed to differentiate into col10a1a-positive cells. These results indicate that osteoblast-lineage cells undergo two distinct differentiation processes during development of the vertebral arch and the centrum. Nevertheless, our results verified that sp7 gene expression in osteoblast-lineage cells is required for differentiation into mature osteoblasts to form the vertebral column and other skeletal structures.


Subject(s)
Cell Differentiation/genetics , Cell Lineage/genetics , Oryzias/embryology , Oryzias/genetics , Osteoblasts/cytology , Spine/cytology , Spine/embryology , Transcription Factors/genetics , Alkaline Phosphatase/metabolism , Animals , Core Binding Factor Alpha 1 Subunit/metabolism , Green Fluorescent Proteins/metabolism , Larva/cytology , Larva/metabolism , Mutation/genetics , Osteoblasts/enzymology , Osteoblasts/metabolism , Phenotype , Spine/metabolism , Transcription Factors/metabolism
17.
J Cell Physiol ; 233(3): 2549-2559, 2018 Mar.
Article in English | MEDLINE | ID: mdl-28786489

ABSTRACT

Bone marrow stromal cells (BMSCs) are multipotent cells that can differentiate into adipocytes and osteoblasts. Inadequate BMSC differentiation is occasionally implicated in chronic bone metabolic disorders. However, specific signaling pathways directing BMSC differentiation have not been elucidated. Here, we explored the roles of spleen tyrosine kinase (Syk) in BMSC differentiation into adipocytes and osteoblasts. We found that Syk phosphorylation was increased in the early stage, whereas its protein expression was gradually decreased during the adipogenic and osteogenic differentiation of two mouse mesenchymal stromal cell lines, ST2 and 10T(1/2), and a human BMSC line, UE6E-7-16. Syk inactivation with either a pharmacological inhibitor or Syk-specific siRNA suppressed adipogenic differentiation, characterized by decreased lipid droplet appearance and the gene expression of fatty acid protein 4 (Fabp4), peroxisome proliferator-activated receptor γ2 (Pparg2), CCAAT/enhancer binding proteins α (C/EBPα), and C/EBPß. In contrast, Syk inhibition promoted osteogenic differentiation, represented by increase in matrix mineralization and alkaline phosphatase (ALP) activity, as well as the expression levels of osteocalcin, runt-related transcription factor 2 (Runx2), and distal-less homeobox 5 (Dlx5) mRNAs. We also found that Syk-induced signals are mediated by phospholipase C γ1 (PLCγ1) in osteogenesis and PLCγ2 in adipogenesis. Notably, Syk-activated PLCγ2 signaling was partly modulated through B-cell linker protein (BLNK) in adipogenic differentiation. On the other hand, growth factor receptor-binding protein 2 (Grb2) was involved in Syk-PLCγ1 axis in osteogenic differentiation. Taken together, these results indicate that Syk-PLCγ signaling has a dual role in regulating the initial stage of adipogenic and osteogenic differentiation of BMSCs.


Subject(s)
Adipocytes/enzymology , Adipogenesis , Cell Lineage , Mesenchymal Stem Cells/enzymology , Osteoblasts/enzymology , Osteogenesis , Phospholipase C gamma/metabolism , Syk Kinase/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Biomarkers/metabolism , Cell Line , GRB2 Adaptor Protein/genetics , GRB2 Adaptor Protein/metabolism , Gene Expression Regulation , Humans , Mice, Inbred C3H , Phenotype , Phospholipase C gamma/genetics , Phosphorylation , RNA Interference , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Syk Kinase/genetics , Time Factors , Transfection
18.
J Cell Physiol ; 233(2): 1752-1762, 2018 Feb.
Article in English | MEDLINE | ID: mdl-28681925

ABSTRACT

Wnt5a, a non-canonical Wnt protein, is known to play important roles in several cell functions. However, little is known about the effects of Wnt5a on osteoblastic differentiation of periodontal ligament (PDL) cells. Here, we examined the effects of Wnt5a on osteoblastic differentiation and associated intracellular signaling in human PDL stem/progenitor cells (HPDLSCs). We found that Wnt5a suppressed expression of bone-related genes (ALP, BSP, and Osterix) and alizarin red-positive mineralized nodule formation in HPDLSCs under osteogenic conditions. Immunohistochemical analysis revealed that a Wnt5a-related receptor, receptor tyrosine kinase-like orphan receptor 2 (Ror2), was expressed in rat PDL tissue. Interestingly, knockdown of Ror2 by siRNA inhibited the Wnt5a-induced downregulation of bone-related gene expression in HPDLSCs. Moreover, Western blotting analysis showed that phosphorylation of the intracellular signaling molecule, c-Jun N-terminal kinase (JNK) was upregulated in HPDLSCs cultured in osteoblast induction medium with Wnt5a, but knockdown of Ror2 by siRNA downregulated the phosphorylation of JNK. We also examined the effects of JNK inhibition on Wnt5a-induced suppression of osteoblastic differentiation of HPDLSCs. The JNK inhibitor, SP600125 inhibited the Wnt5a-induced downregulation of bone-related gene expression in HPDLSCs. Additionally, SP600125 inhibited the Wnt5a-induced suppression of the alizarin red-positive reaction in HPDLSCs. These results suggest that Wnt5a suppressed osteoblastic differentiation of HPDLSCs through Ror2/JNK signaling. Non-canonical Wnt signaling, including Wnt5a/Ror2/JNK signaling, may function as a negative regulator of mineralization, preventing the development of non-physiological mineralization in PDL tissue.


Subject(s)
Cell Differentiation , JNK Mitogen-Activated Protein Kinases/metabolism , Multipotent Stem Cells/enzymology , Osteoblasts/enzymology , Osteogenesis , Periodontal Ligament/enzymology , Receptor Tyrosine Kinase-like Orphan Receptors/metabolism , Wnt Signaling Pathway , Wnt-5a Protein/metabolism , Animals , Cell Differentiation/drug effects , Cell Line , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Male , Multipotent Stem Cells/drug effects , Osteoblasts/drug effects , Osteogenesis/drug effects , Periodontal Ligament/cytology , Periodontal Ligament/drug effects , Phosphorylation , Protein Kinase Inhibitors/pharmacology , RNA Interference , Rats, Sprague-Dawley , Receptor Tyrosine Kinase-like Orphan Receptors/genetics , Transfection , Wnt Signaling Pathway/drug effects
19.
J Cell Physiol ; 233(1): 596-606, 2018 Jan.
Article in English | MEDLINE | ID: mdl-28322449

ABSTRACT

High incidence of osteoporotic fractures emphasizes the necessity of developing effective measures to promote osteogenesis. In our study, we investigated a possible role of MAPK-ERK signaling in the TGF-ß-mediated osteoblastic differentiation. Our results indicated that TGF-ß activated the MAPK-ERK pathway and inhibited osteogenesis in mesenchymal pluripotent cell line, C3H10T1/2, and preosteoblastic cell line, MC3T3 cells. And the downregulation of MAPK-ERK signaling using pharmacological inhibitor U0126 and RNA interference rescued osteoblast differentiation suppressed by TGF-ß, which was confirmed by Alkaline phosphatase (ALP) staining and alizarrn red staining, and the enhanced expression of osteogenesic markers. Western blotting analysis indicated that TGF-ß induced protein expression of E3 ubiquitin-protein ligase SMURF1, which contributed to the degradation of RUNX2 and SMAD1 as evidenced by SMURF1 inhibition using RNA interference and proteasome inhibitor MG132. Moreover, we observed that the expression of SMURF1 was decreased, while that of SMAD1 and RUNX2 increased by MAPK-ERK inhibitor U0126 in TGF-ß-treated differentiating preosteoblasts, suggesting that MAPK-ERK regulated the transcription of osteogenesis-related genes. Furthermore, a synergistic effect between U0126 and bone morphogenic protein (BMP)-2 on osteoblast differentiation and bone formation was observed both in cell cultures and experimental animals. In conclusion, our results revealed that TGF-ß inhibited osteoblastic differentiation by inducing the MAPK-ERK pathway which upregulated the expression of ubiquitin ligase SMURF1 and resulted in reduced presence of osteogenic proteins. In addition, the potentiation of BMP-2 on osteogenic activity by ERK1/2 inhibitor U0126 suggests that it may have potential clinical utility for promoting osteogenesis in bone fracture repair.


Subject(s)
Cell Differentiation/drug effects , Extracellular Signal-Regulated MAP Kinases/metabolism , Osteoblasts/drug effects , Osteogenesis/drug effects , Transforming Growth Factor beta/pharmacology , Ubiquitin-Protein Ligases/metabolism , 3T3 Cells , Animals , Bone Morphogenetic Protein 2/pharmacology , Core Binding Factor Alpha 1 Subunit/genetics , Core Binding Factor Alpha 1 Subunit/metabolism , Dose-Response Relationship, Drug , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , MAP Kinase Kinase 1/antagonists & inhibitors , MAP Kinase Kinase 1/genetics , MAP Kinase Kinase 1/metabolism , Male , Mice , Mice, Inbred C57BL , Osteoblasts/enzymology , Phosphorylation , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors/pharmacology , Protein Kinase Inhibitors/pharmacology , RNA Interference , Signal Transduction/drug effects , Smad1 Protein/metabolism , Transfection , Ubiquitin-Protein Ligases/genetics , Up-Regulation
20.
J Cell Physiol ; 233(4): 3230-3243, 2018 04.
Article in English | MEDLINE | ID: mdl-28976001

ABSTRACT

Arterial medial calcification (AMC) is thought to share some outward similarities to skeletal mineralization and has been associated with the transdifferentiation of vascular smooth muscle cells (VSMCs) to an osteoblast-like phenotype. ATP and UTP have previously been shown to inhibit bone mineralization. This investigation compared the effects of extracellular nucleotides on calcification in VSMCs with those seen in osteoblasts. ATP, UTP and the ubiquitous mineralization inhibitor, pyrophosphate (PPi ), dose dependently inhibited VSMC calcification by ≤85%. Culture of VSMCs in calcifying conditions was associated with an increase in apoptosis; treatment with ATP, UTP, and PPi reduced apoptosis to levels seen in non-calcifying cells. Extracellular nucleotides had no effect on osteoblast viability. Basal alkaline phosphatase (TNAP) activity was over 100-fold higher in osteoblasts than VSMCs. ATP and UTP reduced osteoblast TNAP activity (≤50%) but stimulated VSMC TNAP activity (≤88%). The effects of extracellular nucleotides on VSMC calcification, cell viability and TNAP activity were unchanged by deletion or inhibition of the P2Y2 receptor. Conversely, the actions of ATP/UTP on bone mineralization and TNAP activity were attenuated in osteoblasts lacking the P2Y2 receptor. Ecto-nucleotide pyrophosphatase/phosphodiesterase 1 (NPP1) hydrolyses ATP and UTP to produce PPi . In both VSMCs and osteoblasts, deletion of NPP1 blunted the inhibitory effects of extracellular nucleotides suggesting involvement of P2 receptor independent pathways. Our results show that although the overall functional effect of extracellular nucleotides on AMC and bone mineralization is similar there are clear differences in the cellular mechanisms mediating these actions.


Subject(s)
Calcification, Physiologic , Extracellular Space/metabolism , Nucleotides/pharmacology , Tunica Media/pathology , Vascular Calcification/pathology , Adenosine Triphosphate/pharmacology , Alkaline Phosphatase/metabolism , Animals , Apoptosis/drug effects , Calcification, Physiologic/drug effects , Cell Survival/drug effects , Diphosphates/pharmacology , Mice , Models, Biological , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/enzymology , Muscle, Smooth, Vascular/pathology , Myocytes, Smooth Muscle/metabolism , Osteoblasts/drug effects , Osteoblasts/enzymology , Phosphoric Diester Hydrolases/deficiency , Phosphoric Diester Hydrolases/metabolism , Pyrophosphatases/deficiency , Pyrophosphatases/metabolism , Receptors, Purinergic P2/metabolism , Uridine Triphosphate/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL