Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Sci Rep ; 14(1): 4136, 2024 02 19.
Artículo en Inglés | MEDLINE | ID: mdl-38374135

RESUMEN

While the relationship between circulating osteoprotegerin (OPG) and cardiovascular events is well-established in the general population, its association with cardiovascular risks in chronic kidney disease (CKD) patients remains less robust. This study hypothesized that elevated circulating OPG levels might be associated with an increased risk of major adverse cardiac events (MACE) in CKD patients, a total of 2,109 patients with CKD stages 1 through pre-dialysis 5 from the KNOW-CKD cohort were categorized into quartiles based on serum OPG levels. The primary outcome of the study was 3-point MACE, defined as a composite of nonfatal myocardial infarction, nonfatal stroke, or cardiac death. The median follow-up duration was 7.9 years. The cumulative incidence of 3-point MACE significantly varied across serum OPG levels in Kaplan-Meier curve analysis (P < 0.001, log-rank test), with the highest incidence observed in the 4th quartile. Cox regression analysis indicated that, relative to the 1st quartile, the risk of 3-point MACE was significantly higher in the 3rd (adjusted hazard ratio 2.901, 95% confidence interval 1.009 to 8.341) and the 4th quartiles (adjusted hazard ratio 4.347, 95% confidence interval 1.410 to 13.395). In conclusion, elevated circulating OPG levels are associated with adverse cardiovascular outcomes in pre-dialysis CKD patients.


Asunto(s)
Enfermedades Cardiovasculares , Infarto del Miocardio , Insuficiencia Renal Crónica , Humanos , Enfermedades Cardiovasculares/etiología , Enfermedades Cardiovasculares/metabolismo , Sistema Cardiovascular , Diálisis , Infarto del Miocardio/epidemiología , Infarto del Miocardio/complicaciones , Osteoprotegerina/sangre , Osteoprotegerina/química , Insuficiencia Renal Crónica/complicaciones , Factores de Riesgo
2.
Sci Rep ; 12(1): 15279, 2022 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-36088403

RESUMEN

Recently, a human mutation of OPG was identified to be associated with familial forms of osteoarthritis. This missense mutation (c.1205A = > T; p.Stop402Leu) occurs on the stop codon of OPG, which results in a 19-residue appendage to the C-terminus (OPG+19). The biochemical consequence of this unusual sequence alteration remains unknown. Here we expressed OPG+19 in 293 cells and the mutant OPG was purified to homogeneity by heparin affinity chromatography and size exclusion chromatography. We found that in sharp contrast to wildtype OPG, which mainly exists in dimeric form, OPG+19 had a strong tendency to form higher-order oligomers. To our surprise, the hyper-oligomerization of OPG+19 had no impact on how it binds cell surface heparan sulfate, how it inhibits RANKL-induced osteoclastogenesis and TRAIL-induced chondrocytes apoptosis. Our data suggest that in biological contexts where OPG is known to play a role, OPG+19 functions equivalently as wildtype OPG. The disease-causing mechanism of OPG+19 likely involves an unknown function of OPG in cartilage homeostasis and mineralization. By demonstrating the biochemical nature of this disease-causing OPG mutant, our study will likely help elucidating the biological roles of OPG in cartilage biology.


Asunto(s)
Osteoprotegerina/química , Ligando Inductor de Apoptosis Relacionado con TNF , Apoptosis , Condrocitos/metabolismo , Heparitina Sulfato/metabolismo , Humanos , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
3.
Molecules ; 26(21)2021 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-34770897

RESUMEN

Receptor activator of nuclear factor κB (RANK) and its ligand (RANKL) play key roles in bone metabolism and the immune system. The RANK/RANKL complex has also been shown to be critical in the formation of mammary epithelia cells. The female hormones estradiol and progesterone closely control the action of RANKL with RANK. Blood concentration of these sex hormones in the postmenopausal period leads to an increase in RANK/RANKL signaling and are a major cause of women's osteoporosis, characterized by altered bone mineralization. Knowledge of the biochemical relationships between hormones and RANK/RANKL signaling provides the opportunity to design novel therapeutic agents to inhibit bone loss, based on the anti-RANKL treatment and inhibition of its interaction with the RANK receptor. The new generation of both anti- and mesoprogestins that inhibit the NF-κB-cyclin D1 axis and blocks the binding of RANKL to RANK can be considered as a potential source of new RANK receptor ligands with anti-RANKL function, which may provide a new perspective into osteoporosis treatment itself as well as limit the osteoporosis rise during breast cancer metastasis to the bone.


Asunto(s)
Osteoporosis/etiología , Osteoporosis/metabolismo , Osteoprotegerina/farmacología , Animales , Biomarcadores , Huesos/metabolismo , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Desarrollo de Medicamentos , Homeostasis , Humanos , Osteogénesis/efectos de los fármacos , Osteoporosis/tratamiento farmacológico , Osteoporosis/prevención & control , Osteoprotegerina/química , Osteoprotegerina/uso terapéutico , Unión Proteica , Ligando RANK , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/efectos de los fármacos
5.
FEBS J ; 286(18): 3582-3593, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31081236

RESUMEN

Fibrosis is characterized by the progressive alteration of the tissue structure due to the excessive production of extracellular matrix (ECM). The signaling system encompassing Receptor Activator of Nuclear factor NF-κB Ligand (RANKL)/RANK/Osteoprotegerin (OPG) was discovered to play an important role in the regulation of ECM formation and degradation in bone tissue. However, whether and how this signaling pathway plays a role in liver or pulmonary ECM degradation is unclear up to now. Interestingly, increased decoy receptor OPG levels are found in fibrotic tissues. We hypothesize that RANKL can stimulate RANK on macrophages and initiate the process of ECM degradation. This process may be inhibited by highly expressed OPG in fibrotic conditions. In this case, RANKL mutants that can bind to RANK without binding to OPG might become promising therapeutic candidates. In this study, we built a structure-based library containing 44 RANKL mutants and found that the Q236 residue of RANKL is important for OPG binding. We show that RANKL_Q236D can activate RAW cells to initiate the process of ECM degradation and is able to escape from the obstruction by exogenous OPG. We propose that the generation of RANKL mutants with reduced affinity for OPG is a promising strategy for the exploration of new therapeutics against fibrosis.


Asunto(s)
Fibrosis/genética , Osteoprotegerina/química , Ligando RANK/química , Receptor Activador del Factor Nuclear kappa-B/química , Animales , Matriz Extracelular/química , Matriz Extracelular/genética , Matriz Extracelular/ultraestructura , Fibrosis/patología , Humanos , Hígado/metabolismo , Hígado/patología , Pulmón/metabolismo , Pulmón/patología , Macrófagos/química , Macrófagos/metabolismo , Macrófagos/patología , Ratones , FN-kappa B/genética , Osteoprotegerina/genética , Osteoprotegerina/ultraestructura , Unión Proteica/genética , Conformación Proteica , Ligando RANK/ultraestructura , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/ultraestructura , Transducción de Señal/genética
6.
Int J Mol Sci ; 20(3)2019 Feb 06.
Artículo en Inglés | MEDLINE | ID: mdl-30736365

RESUMEN

The superfamily of tumor necrosis factor (TNF) receptors includes osteoprotegerin (OPG) and its ligands, which are receptor activators of nuclear factor kappa-B ligand (RANKL) and TNF-related apoptosis-inducing ligand (TRAIL). The OPG/RANKL/RANK system plays an active role in pathological angiogenesis and inflammation as well as cell survival. It has been demonstrated that there is crosstalk between endothelial cells and osteoblasts during osteogenesis, thus establishing a connection between angiogenesis and osteogenesis. This OPG/RANKL/RANK/TRAIL system acts on specific cell surface receptors, which are then able to transmit their signals to other intracellular components and modify gene expression. Cytokine production and activation of their receptors induce mechanisms to recruit monocytes and neutrophils as well as endothelial cells. Data support the role of an increased OPG/RANKL ratio as a possible marker of progression of endothelial dysfunction in metabolic disorders in relationship with inflammatory marker levels. We review the role of the OPG/RANKL/RANK triad in vascular function as well as molecular mechanisms related to the etiology of vascular diseases. The potential therapeutic strategies may be very promising in the future.


Asunto(s)
Vasos Sanguíneos/fisiología , Endotelio/metabolismo , Ligandos , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Animales , Biomarcadores , Senescencia Celular , Susceptibilidad a Enfermedades , Células Endoteliales/metabolismo , Humanos , Miocardio/metabolismo , Neovascularización Fisiológica , Osteoprotegerina/química , Complejo de la Endopetidasa Proteasomal/metabolismo , Unión Proteica , Transporte de Proteínas , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal , Relación Estructura-Actividad , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo
7.
J Biol Chem ; 293(45): 17523-17535, 2018 11 09.
Artículo en Inglés | MEDLINE | ID: mdl-30254073

RESUMEN

Previous structural studies of osteoprotegerin (OPG), a crucial negative regulator of bone remodeling and osteoclastogenesis, were mostly limited to the N-terminal ligand-binding domains. It is now known that the three C-terminal domains of OPG also play essential roles in its function by mediating OPG dimerization, OPG-heparan sulfate (HS) interactions, and formation of the OPG-HS-receptor activator of nuclear factor κB ligand (RANKL) ternary complex. Employing hydrogen-deuterium exchange MS methods, here we investigated the structure of full-length OPG in complex with HS or RANKL in solution. Our data revealed two noteworthy aspects of the OPG structure. First, we found that the interconnection between the N- and C-terminal domains is much more rigid than previously thought, possibly because of hydrophobic interactions between the fourth cysteine-rich domain and the first death domain. Second, we observed that two hydrophobic clusters located in two separate C-terminal domains directly contribute to OPG dimerization, likely by forming a hydrophobic dimerization interface. Aided by site-directed mutagenesis, we further demonstrated that an intact dimerization interface is essential for the biological activity of OPG. Our study represents an important step toward deciphering the structure-function relationship of the full-length OPG protein.


Asunto(s)
Medición de Intercambio de Deuterio , Espectrometría de Masas , Osteoprotegerina/química , Multimerización de Proteína , Animales , Heparitina Sulfato/química , Heparitina Sulfato/genética , Heparitina Sulfato/metabolismo , Ratones , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Dominios Proteicos , Ligando RANK/química , Ligando RANK/genética , Ligando RANK/metabolismo
8.
J Mater Sci Mater Med ; 29(8): 119, 2018 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-30030632

RESUMEN

Strontium (Sr) has shown effectiveness for stimulating bone remodeling. Nevertheless, the exact therapeutic values are not established yet. Authors hypothesized that local application of Sr-enriched ceramics would enhance bone remodeling in constant osteoporosis of rabbits' femoral neck bone. Seven different bone conditions were analyzed: ten healthy rabbits composed a control group, while other twenty underwent ovariectomy and were divided into three groups. Bone defect was filled with hydroxyapatite 30% (HAP) and tricalcium phosphate 70% (TCP) granules in 7 rabbits, 5% of Sr-enriched HAP/TCP granules in 7, but sham defect was left unfilled in 6 rabbits. Bone samples were obtained from operated and non-operated legs 12 weeks after surgery and analyzed by histomorphometry and immunohistochemistry (IMH). Mean trabecular bone area in control group was 0.393 mm2, in HAP/TCP - 0.226 mm2, in HAP/TCP/Sr - 0.234 mm2 and after sham surgery - 0.242 mm2. IMH revealed that HAP/TCP/Sr induced most noticeable increase of nuclear factor kappa beta 105 (NFkB 105), osteoprotegerin (OPG), osteocalcin (OC), bone morphogenetic protein 2/4 (BMP 2/4), collagen type 1α (COL-1α), interleukin 1 (IL-1) with comparison to intact leg; NFkB 105 and OPG rather than pure HAP/TCP or sham bone. We concluded that Sr-enriched biomaterials induce higher potential to improve bone regeneration than pure bioceramics in constant osteoporosis of femoral neck bone. Further studies on bigger osteoporotic animals using Sr-substituted orthopedic implants for femoral neck fixation should be performed to confirm valuable role in local treatment of osteoporotic femoral neck fractures in humans.


Asunto(s)
Regeneración Ósea/efectos de los fármacos , Sustitutos de Huesos/química , Cerámica/química , Fémur/química , Osteoporosis/terapia , Estroncio/química , Animales , Materiales Biocompatibles/química , Remodelación Ósea , Fosfatos de Calcio/química , Durapatita/química , Femenino , Cabeza Femoral/patología , Inmunohistoquímica , Inflamación , Osteoprotegerina/química , Conejos
9.
Acta Neuropathol Commun ; 6(1): 31, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29699580

RESUMEN

Although there is a strong association between osteoporosis and skeletal muscle atrophy/dysfunction, the functional relevance of a particular biological pathway that regulates synchronously bone and skeletal muscle physiopathology is still elusive. Receptor-activator of nuclear factor κB (RANK), its ligand RANKL and the soluble decoy receptor osteoprotegerin (OPG) are the key regulators of osteoclast differentiation and bone remodelling. We thus hypothesized that RANK/RANKL/OPG, which is a key pathway for bone regulation, is involved in Duchenne muscular dystrophy (DMD) physiopathology. Our results show that muscle-specific RANK deletion (mdx-RANK mko ) in dystrophin deficient mdx mice improves significantly specific force [54% gain in force] of EDL muscles with no protective effect against eccentric contraction-induced muscle dysfunction. In contrast, full-length OPG-Fc injections restore the force of dystrophic EDL muscles [162% gain in force], protect against eccentric contraction-induced muscle dysfunction ex vivo and significantly improve functional performance on downhill treadmill and post-exercise physical activity. Since OPG serves a soluble receptor for RANKL and as a decoy receptor for TRAIL, mdx mice were injected with anti-RANKL and anti-TRAIL antibodies to decipher the dual function of OPG. Injections of anti-RANKL and/or anti-TRAIL increase significantly the force of dystrophic EDL muscle [45% and 17% gains in force, respectively]. In agreement, truncated OPG-Fc that contains only RANKL domains produces similar gains, in terms of force production, than anti-RANKL treatments. To corroborate that full-length OPG-Fc also acts independently of RANK/RANKL pathway, dystrophin/RANK double-deficient mice were treated with full-length OPG-Fc for 10 days. Dystrophic EDL muscles exhibited a significant gain in force relative to untreated dystrophin/RANK double-deficient mice, indicating that the effect of full-length OPG-Fc is in part independent of the RANKL/RANK interaction. The sarco/endoplasmic reticulum Ca2+ ATPase (SERCA) activity is significantly depressed in dysfunctional and dystrophic muscles and full-length OPG-Fc treatment increased SERCA activity and SERCA-2a expression. These findings demonstrate the superiority of full-length OPG-Fc treatment relative to truncated OPG-Fc, anti-RANKL, anti-TRAIL or muscle RANK deletion in improving dystrophic muscle function, integrity and protection against eccentric contractions. In conclusion, full-length OPG-Fc represents an efficient alternative in the development of new treatments for muscular dystrophy in which a single therapeutic approach may be foreseeable to maintain both bone and skeletal muscle functions.


Asunto(s)
Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/patología , Distrofias Musculares/terapia , Osteoprotegerina/uso terapéutico , Receptor Activador del Factor Nuclear kappa-B/deficiencia , Animales , Creatina Quinasa/metabolismo , Modelos Animales de Enfermedad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos mdx , Ratones Transgénicos , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Distrofias Musculares/genética , Osteoprotegerina/química , Osteoprotegerina/metabolismo , ARN Mensajero/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología
10.
Eur J Med Chem ; 145: 661-672, 2018 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-29348072

RESUMEN

Osteoporosis is a metabolic bone disease characterized by low bone mass and micro-architectural deterioration of bone, for which the underlying mechanism is an imbalance between bone resorption and bone remodeling. The protein-protein interactions between receptor activator of nuclear factor-κB ligand (RANKL), RANK (its receptor), and osteoprotegerin (OPG), are known to mediate the development and activation of osteoclasts in bone remodeling, and are regarded as a pivotal therapeutic target for the treatment of osteoporosis. Herein, we disclose the successful development of a novel glycopeptide (OM-2), the structure of which is based on the key interacting sites of the reported RANKL and OPG crystal structure. OM-2 exhibited potent binding affinity with RANKL and resistance to degradation by protease enzymes. It also blocked RANKL/RANK interactions, and inhibited osteoclastogenesis in vitro. In vivo studies confirmed that OM-2 could effectively reduce bone loss and inhibit osteoclast activation in ovariectomized (OVX) mice at a dosage of 20.0 mg/kg/day. Accordingly, OM-2 is suggested as a therapeutic candidate for postmenopausal osteoporosis (PMOP) and osteoclastogenesis-related diseases like rheumatoid arthritis (RA). More importantly, its identification validates our structure-based strategy for the development of drugs that target the RANKL/RANK/OPG system.


Asunto(s)
Glicopéptidos/farmacología , Osteoporosis/tratamiento farmacológico , Osteoprotegerina/farmacología , Ovariectomía , Ligando RANK/antagonistas & inhibidores , Receptores Citoplasmáticos y Nucleares/antagonistas & inhibidores , Animales , Huesos/efectos de los fármacos , Huesos/metabolismo , Huesos/patología , Relación Dosis-Respuesta a Droga , Femenino , Glicopéptidos/síntesis química , Glicopéptidos/química , Masculino , Ratones , Ratones Endogámicos C57BL , Simulación del Acoplamiento Molecular , Estructura Molecular , Osteoporosis/metabolismo , Osteoprotegerina/química , Unión Proteica/efectos de los fármacos , Ligando RANK/metabolismo , Receptores Citoplasmáticos y Nucleares/metabolismo , Relación Estructura-Actividad
11.
J Phys Chem B ; 121(39): 9141-9148, 2017 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-28945380

RESUMEN

Recent studies have revealed the importance and the active contribution of the RANKL/OPG/RANK pathway in many bone diseases including different forms of common osteoporosis. In this study, we present an extensive atomistic molecular dynamic study of the OPG/RANKL system. Within the molecular models, we varied the number of OPG molecules bound to the RANKL trimer and carried out a study to determine how the binding affinity of the OPG/RANKL system changes as a function of OPG concentration. The molecular mechanics Poisson-Boltzmann surface area method was used to analyze binding free energies. It is shown that the binding affinity decreases with increasing numbers of OPG molecules. Additionally, conformational changes of RANKL, interactions between the N-terminus outlier module of OPG with RANKL, and residues that play an important role in the binding of OPG to RANKL trimer were investigated. A probable cause for unfavorable binding for a third OPG molecule was found. Along with the currently available experimental studies, this computational study will be valuable for the comprehensive understanding of OPG/RANKL at the atomistic level.


Asunto(s)
Simulación de Dinámica Molecular , Osteoprotegerina/química , Ligando RANK/química , Humanos , Modelos Biológicos , Unión Proteica , Termodinámica
12.
Appl Microbiol Biotechnol ; 101(12): 4923-4933, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28303296

RESUMEN

As a natural inhibitor of the receptor activator of nuclear factor-кB ligand (RANKL), osteprotegerin (OPG) is considered a promising treatment for metabolic bone diseases. Typical approaches for preparing recombinant OPG or its derivatives employ eukaryotic expression systems. Due to the advantages of a prokaryotic expression system, which include its convenience, low cost, and abundant production, in this study, we establish a strategy for preparing functional OPG using the Escherichia coli expression system. After initial failures in preparation of OPG and its truncation, OPG cysteine-rich domain (OPG-CRD/OPGT) by using pET and pGEX vectors, we constructed a sortase A (SrtA)-aided E. coli expression system, in which the expressed protein was a self-cleaving SrtA fusion protein. Using this system, we successfully prepared the recombinant OPGT protein. The BIAcore analyses indicated that the prepared OPGT had high affinities in binding with RANKL and TRAIL. Cell experiments confirmed the inhibitory effects of the prepared OPGT on RANKL-induced osteoclast differentiation and TRAIL-induced tumor cell apoptosis. The sortase A-aided E. coli expression system for OPGT established in this study may contribute to further studies and commercial applications of OPG.


Asunto(s)
Aminoaciltransferasas/metabolismo , Proteínas Bacterianas/metabolismo , Cisteína Endopeptidasas/metabolismo , Cisteína/química , Escherichia coli/genética , Osteoprotegerina/química , Osteoprotegerina/genética , Aminoaciltransferasas/genética , Animales , Apoptosis/efectos de los fármacos , Proteínas Bacterianas/genética , Diferenciación Celular/efectos de los fármacos , Cisteína/genética , Cisteína Endopeptidasas/genética , Escherichia coli/enzimología , Vectores Genéticos , Humanos , Ratones , Osteoclastos/efectos de los fármacos , Osteoprotegerina/biosíntesis , Osteoprotegerina/farmacología , Unión Proteica , Dominios Proteicos , Ligando RANK/farmacología , Células RAW 264.7 , Proteínas Recombinantes/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/metabolismo , Ligando Inductor de Apoptosis Relacionado con TNF/farmacología
13.
J Biomed Mater Res A ; 105(2): 398-407, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27684563

RESUMEN

The osteoprotegerin (OPG) system plays a critical role in bone remodelling by regulating osteoclast formation and activity. The study aimed to determine the physicochemical properties and biocompatibility of a newly formulated OPG-chitosan gel. The OPG-chitosan gel was formulated using human OPG protein and water-soluble chitosan. The physicochemical properties were determined using Fourier transform infrared (FTIR) spectroscopy, thermogravimetric analysis (TGA) and differential scanning calorimetry (DSC). Gel morphology was determined using scanning electron microscopy (SEM) and then it was subjected to a protein release assay and biodegradability test. An in vitro cytotoxicity test on normal human periodontal ligament (NHPL) fibroblasts and normal human (NH) osteoblasts was carried out using the AlamarBlue assay. In vivo evaluation in a rabbit model involved creating critical-sized defects in calvarial bone, filling with the OPG-chitosan gel and sacrificing at 12 weeks. In vitro results demonstrated that the 25 kDa OPG-chitosan gel had the highest rate of protein release and achieved 90% degradation in 28 days. At 12 weeks, the defects filled with 25 kDa OPG-chitosan gel showed significant (p < 0.05) new bone formation and the highest expression of osteocalcin and osteopontin compared to controls. Thus, the 25 kDa OPG-chitosan gel could be a promising new biomaterial for tissue engineering. © 2016 Wiley Periodicals, Inc. J Biomed Mater Res Part A: 105A: 398-407, 2017.


Asunto(s)
Regeneración Ósea , Quitosano , Osteoblastos/metabolismo , Osteoprotegerina , Cráneo , Línea Celular , Quitosano/química , Quitosano/farmacología , Clero , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Hidrogeles/química , Hidrogeles/farmacología , Osteoblastos/patología , Osteoprotegerina/química , Osteoprotegerina/farmacología , Ligamento Periodontal/metabolismo , Ligamento Periodontal/patología , Cráneo/lesiones , Cráneo/metabolismo , Cráneo/patología
14.
J Biol Chem ; 291(46): 24160-24171, 2016 Nov 11.
Artículo en Inglés | MEDLINE | ID: mdl-27697839

RESUMEN

Osteoprotegerin (OPG), a decoy receptor secreted by osteoblasts, is a major negative regulator of bone resorption. It functions by neutralizing the receptor activator of nuclear factor κB ligand (RANKL), which plays a central role in promoting osteoclastogenesis. OPG is known to be a high-affinity heparan sulfate (HS)-binding protein. Presumably, HS could regulate the function of OPG and affect how it inhibits RANKL. However, the molecular detail of HS-OPG interaction remains poorly understood, which hinders our understanding of how HS functions in osteoclastogenesis. Here we report mapping of the HS-binding site of OPG. The HS-binding site, identified by mutagenesis study, consists of eight basic residues that are located mostly at the junction of the second death domain and the C-terminal domain. We further show that heparin-derived dodecasaccharide is able to induce dimerization of OPG monomers with a stoichiometry of 1:1. Small-angle X-ray scattering analysis revealed that upon binding of HS, OPG undergoes a dramatic conformational change, resulting in a more compact and less flexible structure. Importantly, we present here three lines of evidence that HS, OPG, and RANKL form a stable ternary complex. Using a HS binding-deficient OPG mutant, we further show that in an osteoblast/bone marrow macrophage co-culture system, immobilization of OPG by HS at the osteoblast cell surface substantially lowers the inhibitory threshold of OPG toward RANKL. These discoveries strongly suggest that HS plays an active role in regulating OPG-RANKL interaction and osteoclastogenesis.


Asunto(s)
Heparitina Sulfato/metabolismo , Macrófagos/metabolismo , Osteoblastos/metabolismo , Osteoclastos/metabolismo , Osteoprotegerina/metabolismo , Animales , Sitios de Unión , Línea Celular , Técnicas de Cocultivo , Heparitina Sulfato/química , Heparitina Sulfato/genética , Macrófagos/citología , Ratones , Osteoblastos/citología , Osteoclastos/citología , Osteoprotegerina/química , Osteoprotegerina/genética , Ligando RANK/química , Ligando RANK/genética , Ligando RANK/metabolismo
15.
Orthod Craniofac Res ; 19(4): 198-208, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27647720

RESUMEN

OBJECTIVES: To test the following two hypotheses: 1) different types of retainers result in distinct levels of biomarkers in gingival crevicular fluid (GCF) and 2) the retainer bonded to all mandibular anterior teeth induces more detrimental outcomes to the periodontium. SETTING AND SAMPLE POPULATION: The Department of Orthodontics at the University of Florida. The population consisted of individuals in the retention phase of orthodontic treatment. MATERIAL AND METHODS: This was a cross-sectional study that enrolled 36 individuals. Subjects in group 1 had retainers bonded to the mandibular canines only. Group 2 consisted of individuals having retainers bonded to all mandibular anterior teeth. Group 3 included patients using mandibular removable retainers. After clinical examination, GCF was collected from the mandibular incisor and biomarker levels were compared between the groups. RESULTS: Plaque accumulation and gingivitis differed significantly among groups, with the highest median values in group 2 subjects. Pairwise comparison of the groups with respect to gingivitis showed significant differences between groups 1 and 2. Significant differences among groups were detected for RANKL, OPG, OPN, M-CSF, MMP-3, and MMP-9. The ratio RANKL/OPG was significantly higher in group 2 subjects, with pairwise comparisons indicating that groups 1 and 2 differed from group 3. CONCLUSION: An association was found between orthodontic retention groups and GCF biomarker levels, which should be further explored in longitudinal studies. The presence of retainers bonded to all anterior teeth seems to increase plaque accumulation and gingivitis.


Asunto(s)
Biomarcadores/química , Recubrimiento Dental Adhesivo/efectos adversos , Recubrimiento Dental Adhesivo/métodos , Placa Dental/etiología , Líquido del Surco Gingival/química , Recesión Gingival/etiología , Gingivitis/etiología , Incisivo/patología , Incisivo/fisiopatología , Retenedores Ortodóncicos/efectos adversos , Adolescente , Adulto , Estudios Transversales , Diente Canino , Índice de Placa Dental , Femenino , Humanos , Proteína Antagonista del Receptor de Interleucina 1/química , Interleucina-1beta/química , Interleucina-6/química , Interleucina-8/química , Factor Estimulante de Colonias de Macrófagos/química , Masculino , Mandíbula , Metaloproteinasa 3 de la Matriz/química , Metaloproteinasa 9 de la Matriz/química , Persona de Mediana Edad , Diseño de Aparato Ortodóncico , Osteopontina/química , Osteoprotegerina/química , Índice Periodontal , Ligando RANK/química
16.
Med Hypotheses ; 94: 40-2, 2016 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-27515196

RESUMEN

Chronic inflammatory processes in periapical tissues caused by etiological agents of endodontic origin lead to apical periodontitis. Apart from bacteria, two herpesviruses, Epstein-Barr virus (EBV) and Human cytomegalovirus (HCMV) are recognized as putative pathogens in apical periodontitis. Although previous reports suggest the involvement of EBV in the pathogenesis of apical periodontitis, its exact role in periapical bone resorption has not yet been fully elucidated. We hypothesize that EBV infection in apical periodontitis is capable of inducing periapical bone resorption via stimulation of reactive oxygen species (ROS) overproduction. Increased levels of ROS induce expression of receptor activator of nuclear factor kappa B (NF-κB) ligand (RANKL). RANKL binding to receptor activator of nuclear factor κB (RANK) present on the surface of preosteoclasts induces their maturation and activation which consequently leads to bone resorption. The potential benefit of antiviral and antioxidant-based therapies in periapical bone resorption treatment remains to be assessed.


Asunto(s)
Resorción Ósea , Infecciones por Virus de Epstein-Barr/fisiopatología , Periodontitis Periapical/patología , Especies Reactivas de Oxígeno/metabolismo , Animales , Linfocitos B/citología , Células Epiteliales/citología , Herpesvirus Humano 4 , Humanos , Inflamación , Modelos Teóricos , Osteoclastos/citología , Osteoprotegerina/química , Periodontitis Periapical/fisiopatología , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo
17.
Angle Orthod ; 86(2): 187-92, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26154939

RESUMEN

OBJECTIVE: To carry out an immunoassay analysis of biomarkers expressed in gingival crevicular fluid (GCF) with the main goal of finding a useful diagnostic pattern to distinguish between resorbing deciduous teeth and nonresorbing controls. MATERIALS AND METHODS: A split-mouth design was used in this study with a total of 22 GCF samples collected from 11 patients in the mixed dentition. For each child, one deciduous molar with radiographic evidence of root resorption was used as the test tooth whereas the contralateral first permanent molar with formed roots was used as the control tooth. Samples were processed with immunoassays using a panel of selected biomarkers including interleukin-1 beta (IL-1b), interleukin-1 receptor antagonist (IL-1RA), nuclear factor kappa B ligand (RANKL), osteoprotegerin (OPG), matrix metalloproteinase-9 (MMP-9), and dentin sialoprotein (DSP). RESULTS: There were no statistically significant differences in levels of IL-1b, OPG, and MMP-9 between test and control sites (P > .05). IL-1RA was the only biomarker to show a significant down-regulation (P  =  .04) in GCF samples collected from resorbing teeth. RANKL data showed a heavily skewed distribution and was deemed unreliable. Only one deciduous GCF sample had detectable levels of DSP; therefore, no further statistical calculation was applicable because of the limited amount of data for this biomarker. CONCLUSIONS: This study indicated that IL1-RA is down-regulated in GCF from resorbing primary molars, thus suggesting this cytokine as a potential analyte to be included in a panel that can discriminate between resorbing and nonresorbing teeth.


Asunto(s)
Biomarcadores/química , Líquido del Surco Gingival/química , Inmunoensayo , Proteína Antagonista del Receptor de Interleucina 1/química , Resorción Radicular/diagnóstico , Niño , Proteínas de la Matriz Extracelular/química , Humanos , Interleucina-1beta/química , Metaloproteinasa 9 de la Matriz/química , Diente Molar , Osteoprotegerina/química , Fosfoproteínas/química , Ligando RANK/química , Sialoglicoproteínas/química
18.
ACS Appl Mater Interfaces ; 7(42): 23787-97, 2015 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-26452150

RESUMEN

Replicating the biocomplexity of native extracellular matrices (ECM) is critical for a deeper understanding of biochemical signals influencing bone homeostasis. This will foster the development of bioinspired biomaterials with adjustable bone-inducing properties. Collagen-based coatings containing single HA derivatives have previously been reported to promote osteogenic differentiation and modulate osteoclastogenesis and resorption depending on their sulfation degree. However, the potential impact of different GAG concentrations as well as the interplay of multiple GAGs in these coatings is not characterized in detail to date. These aspects were addressed in the current study by integrating HA and different sulfate-modified HA derivatives (sHA) during collagen in vitro fibrillogenesis. Besides cellular microenvironments with systematically altered single-GAG concentrations, matrices containing both low and high sHA (sHA1, sHA4) were characterized by biochemical analysis such as agarose gel electrophoresis, performed for the first time with sHA derivatives. The morphology and composition of the collagen coatings were altered in a GAG sulfation- and concentration-dependent manner. In multi-GAG microenvironments, atomic force microscopy revealed intermediate collagen fibril structures with thin fibrils and microfibrils. GAG sulfation altered the surface charge of the coatings as demonstrated by ζ-potential measurements revealed for the first time as well. This highlights the prospect of GAG-containing matrices to adjust defined surface charge properties. The sHA4- and the multi-GAG coatings alike significantly enhanced the viability of murine osteoclast-precursor-like RAW264.7 cells. Although in single-GAG matrices there was no dose-dependent effect on cell viability, osteoclastogenesis was significantly suppressed only on sHA4-coatings in a dose-dependent fashion. The multi-GAG coatings led to an antiosteoclastogenic effect in-between those with single-GAGs which cannot simply be attributed to the overall content of sulfate groups. These data suggest that the interplay of sGAGs influences bone cell behavior. Whether these findings translate into favorable biomaterial properties needs to be validated in vivo.


Asunto(s)
Materiales Biomiméticos/química , Matriz Extracelular/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteoprotegerina/química , Animales , Colágeno/química , Matriz Extracelular/ultraestructura , Ácido Hialurónico/química , Ratones , Microscopía de Fuerza Atómica , Osteoclastos/ultraestructura , Osteogénesis/efectos de los fármacos , Osteoprotegerina/farmacología
19.
Methods Enzymol ; 562: 501-27, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26412665

RESUMEN

This chapter illustrates how analytical ultracentrifugation methods, coupled with the fluorescence detection system, are an excellent approach to characterizing and comparing protein-binding interactions in dilute solution and concentrated, crowded solutions like serum. We show that in serum, the binding and assembly states for a pair of endogenous protein ligands and an antibody inhibitor are dramatically different than those observed in dilute, simple buffers. This type of analysis approach may be helpful in research efforts intent at discerning the underpinnings to a therapeutic's activity and pharmacokinetic properties in vivo.


Asunto(s)
Ligando RANK/aislamiento & purificación , Anticuerpos Monoclonales/química , Anticuerpos Monoclonales/aislamiento & purificación , Tampones (Química) , Humanos , Osteoprotegerina/química , Osteoprotegerina/aislamiento & purificación , Unión Proteica , Multimerización de Proteína , Ligando RANK/química , Suero/química , Albúmina Sérica/química , Albúmina Sérica/aislamiento & purificación , Soluciones , Ultracentrifugación/métodos
20.
Biomacromolecules ; 16(8): 2374-81, 2015 Aug 10.
Artículo en Inglés | MEDLINE | ID: mdl-26151628

RESUMEN

Polymers of similar molecular weights and chemical constitution but varying in their macromolecular architectures were conjugated to osteoprotegerin (OPG) to determine the effect of polymer topology on protein activity in vitro and in vivo. OPG is a protein that inhibits bone resorption by preventing the formation of mature osteoclasts from the osteoclast precursor cell. Accelerated bone loss disorders, such as osteoporosis, rheumatoid arthritis, and metastatic bone disease, occur as a result of increased osteoclastogenesis, leading to the severe weakening of the bone. OPG has shown promise as a treatment in bone disorders; however, it is rapidly cleared from circulation through rapid liver uptake, and frequent, high doses of the protein are necessary to achieve a therapeutic benefit. We aimed to improve the effectiveness of OPG by creating OPG-polymer bioconjugates, employing reversible addition-fragmentation chain transfer polymerization to create well-defined polymers with branching densities varying from linear, loosely branched to densely branched. Polymers with each of these architectures were conjugated to OPG using a "grafting-to" approach, and the bioconjugates were characterized by sodium dodecyl sulfate polyacrylamide gel electrophoresis. The OPG-polymer bioconjugates showed retention of activity in vitro against osteoclasts, and each bioconjugate was shown to be nontoxic. Preliminary in vivo studies further supported the nontoxic characteristics of the bioconjugates, and measurement of the bone mineral density in rats 7 days post-treatment via peripheral quantitative computed tomography suggested a slight increase in bone mineral density after administration of the loosely branched OPG-polymer bioconjugate.


Asunto(s)
Artritis Reumatoide/tratamiento farmacológico , Resorción Ósea/tratamiento farmacológico , Osteoporosis/tratamiento farmacológico , Osteoprotegerina/química , Animales , Artritis Reumatoide/patología , Densidad Ósea/efectos de los fármacos , Resorción Ósea/patología , Humanos , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteoporosis/patología , Osteoprotegerina/administración & dosificación , Polímeros/administración & dosificación , Polímeros/química , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...