Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Int J Mol Sci ; 23(18)2022 Sep 06.
Artigo em Inglês | MEDLINE | ID: mdl-36142172

RESUMO

Attention deficit hyperactivity disorder (ADHD) is one of the most common worldwide mental disorders in children, young and adults. If left untreated, the disorder can continue into adulthood. The abuse of ADHD-related drugs to improve mental performance for studying, working and everyday life is also rising. The potentially high number of subjects with controlled or uncontrolled use of such substances increases the impact of possible side effects. It has been shown before that the early ADHD drug methylphenidate influences bone metabolism negatively. This study focused on the influence of three more recent cognitive enhancers, modafinil, atomoxetine and guanfacine, on the differentiation of mesenchymal stem cells to osteoblasts and on their cell functions, including migration. Human mesenchymal stem cells (hMSCs) were incubated with a therapeutic plasma dosage of modafinil, atomoxetine and guanfacine. Gene expression analyses revealed a high beta-2 adrenoreceptor expression in hMSC, suggesting it as a possible pathway to stimulate action. In bone formation assays, all three cognitive enhancers caused a significant decrease in the mineralized matrix and an early slight reduction of cell viability without triggering apoptosis or necrosis. While there was no effect of the three substances on early differentiation, they showed differing effects on the expression of osterix (OSX), receptor activator of NF-κB ligand (RANKL) and osteoprotegerin (OPG) in the later stages of osteoblast development, suggesting alternative modes of action. All three substances significantly inhibited hMSC migration. This effect could be rescued by a selective beta-blocker (Imperial Chemical Industries ICI-118,551) in modafinil and atomoxetine, suggesting mediation via beta-2 receptor stimulation. In conclusion, modafinil, atomoxetine and guanfacine negatively influence hMSC differentiation to bone-forming osteoblasts and cell migration through different intracellular pathways.


Assuntos
Transtorno do Deficit de Atenção com Hiperatividade , Estimulantes do Sistema Nervoso Central , Metilfenidato , Nootrópicos , Adulto , Cloridrato de Atomoxetina/farmacologia , Transtorno do Deficit de Atenção com Hiperatividade/tratamento farmacológico , Diferenciação Celular , Estimulantes do Sistema Nervoso Central/farmacologia , Criança , Guanfacina/farmacologia , Humanos , Ligantes , Metilfenidato/uso terapêutico , Modafinila/farmacologia , Modafinila/uso terapêutico , Nootrópicos/uso terapêutico , Osteoprotegerina/uso terapêutico , Receptor Ativador de Fator Nuclear kappa-B
2.
J Diabetes Res ; 2022: 8260111, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35845316

RESUMO

Background: The relationship between diabetes and periodontitis is bidirectional, and there is now consensus that periodontitis and diabetes are comorbid. There is a quest for a drug that can be used to treat both conditions simultaneously. This study evaluated the anti-inflammatory and osteoprotective effects of liraglutide (LIRA) on periodontitis in diabetic rats. Methods: Male Wistar rats (n = 46) were randomly divided into four groups: control group (n = 8), LIRA group (n = 8), diabetes-associated periodontitis+0.9% saline group (diabetic periodontitis (DP)+NaCl group, n = 15), and diabetes-associated periodontitis+LIRA group (DP+LIRA group, n = 15). LIRA treatment lasted for 4 weeks (300 µg/kg/d) after establishment of a rat model of DP. The expression of IL-6, TNF-α, and IL-1ß was detected by enzyme-linked immunosorbent assay. The morphological changes of periodontal tissues were observed by hematoxylin-eosin staining. The absorption of alveolar bone and its ultrastructural changes were observed by histomorphometry and microcomputed tomography. The expression of receptor activator of NF-κB ligand (RANKL) and osteoprotegerin (OPG) in alveolar bone was detected by immunohistochemistry. The levels of Runx2 mRNA and ALP mRNA in the gingival epithelium were examined by quantitative real-time polymerase chain reaction. Results: LIRA decreased alveolar bone resorption, improved the microstructure of alveolar bone, and reduced periodontal inflammation and damage (P < 0.05). LIRA also reduced blood glucose level and inhibited the secretion of serum IL-6, TNF-α, and IL-1ß (P < 0.05). In addition, after treatment with LIRA, the ratio of RANKL/OPG was reduced, and the expression levels of ALP mRNA and Runx2 mRNA were upregulated (P < 0.05). Conclusions: LIRA not only controls blood glucose level but also reduces inflammation and bone loss and enhances osteogenic differentiation in diabetes-associated periodontitis. Those indicate that LIRA may be used as a potential medicine for the adjunctive therapy of diabetes-periodontitis comorbidity.


Assuntos
Perda do Osso Alveolar , Diabetes Mellitus Experimental , Periodontite , Perda do Osso Alveolar/tratamento farmacológico , Animais , Glicemia , Comorbidade , Subunidade alfa 1 de Fator de Ligação ao Core , Diabetes Mellitus Experimental/complicações , Diabetes Mellitus Experimental/tratamento farmacológico , Inflamação , Interleucina-6/metabolismo , Liraglutida/farmacologia , Liraglutida/uso terapêutico , Masculino , Osteogênese , Osteoprotegerina/genética , Osteoprotegerina/uso terapêutico , Periodontite/complicações , Periodontite/tratamento farmacológico , Periodontite/genética , Ligante RANK , RNA Mensageiro , Ratos , Ratos Wistar , Fator de Necrose Tumoral alfa/metabolismo , Microtomografia por Raio-X
3.
Comput Math Methods Med ; 2022: 2011625, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35669373

RESUMO

Epimedium is a traditional Chinese medicine that is most commonly prescribed by practitioners of Chinese medicine for the clinical treatment of malignant tumor bone metastasis. The main component of Epimedium is icariin (ICA). Studies have shown that ICA inhibits bone resorption of osteoclasts through the OPG/RANKL/RANK signaling pathway. Osteoclasts are the only cells in the body that have a bone-destroying capability. The OPG/RANKL/RANK system consists of cytokines that play major roles in osteoclast formation. Therefore, our study selected the OPG/RANKL/RANK system as the research target to investigate the effect of ICA on nude mice with lung cancer bone metastasis. We established the model of bone metastasis in nude mice, intervened the model with icariin and zoledronic acid, and detected the levels of OPG and RANKL by ELISA and western blot. The results showed that ICA had a significant inhibitory effect on bone metastases in nude mice. ICA achieved its antibone metastasis effect in nude mice with lung cancer via inhibiting RANKL expression and simultaneously increasing OPG expression. ICA not only alleviated osteolytic bone destruction caused by bone metastases, but it also reduced weight loss in tumor-bearing nude mice at the late stage of the experiment. The role of ICA in preventing bone metastasis of lung cancer merits further investigation.


Assuntos
Neoplasias Ósseas , Neoplasias Pulmonares , Animais , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/metabolismo , Flavonoides/farmacologia , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/patologia , Camundongos , Camundongos Nus , Osteoprotegerina/metabolismo , Osteoprotegerina/uso terapêutico , Ligante RANK/metabolismo , Ligante RANK/uso terapêutico
4.
Heart Fail Rev ; 27(4): 1395-1411, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-34313900

RESUMO

Osteoprotegerin (OPG) is a glycoprotein involved in the regulation of bone remodelling. OPG regulates osteoclast activity by blocking the interaction between the receptor activator of nuclear factor kappa B (RANK) and its ligand (RANKL). More and more studies confirm the relationship between OPG and cardiovascular diseases. Numerous studies have confirmed that a high plasma concentration of OPG and a low concentration of tumour necrosis factor-related apoptosis inducing ligand (TRAIL) together with a high OPG/TRAIL ratio are predictors of poor prognosis in patients with myocardial infarction. A high plasma OPG concentration and a high ratio of OPG/TRAIL in the acute myocardial infarction are a prognostic indicator of adverse left ventricular remodelling and of the development of heart failure. Ever more data indicates the participation of OPG in the regulation of the function of vascular endothelial cells and the initiation of the atherosclerotic process in the arteries. Additionally, it has been shown that TRAIL has a protective effect on blood vessels and exerts an anti-atherosclerotic effect. The mechanisms of action of both OPG and TRAIL within the cells of the vascular wall are complex and remain largely unclear. However, these mechanisms of action as well as their interaction in the local vascular environment are of great interest to researchers. This article presents the current state of knowledge on the mechanisms of action of OPG and TRAIL in the circulatory system and their role in cardiovascular diseases. Understanding these mechanisms may allow their use as a therapeutic target in cardiovascular diseases in the future.


Assuntos
Aterosclerose , Doenças Cardiovasculares , Insuficiência Cardíaca , Infarto do Miocárdio , Aterosclerose/tratamento farmacológico , Doenças Cardiovasculares/tratamento farmacológico , Células Endoteliais , Insuficiência Cardíaca/tratamento farmacológico , Humanos , Ligantes , Infarto do Miocárdio/tratamento farmacológico , Osteoprotegerina/uso terapêutico , Receptor Ativador de Fator Nuclear kappa-B , Ligante Indutor de Apoptose Relacionado a TNF
5.
Molecules ; 26(21)2021 Oct 27.
Artigo em Inglês | MEDLINE | ID: mdl-34770897

RESUMO

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.


Assuntos
Osteoporose/etiologia , Osteoporose/metabolismo , Osteoprotegerina/farmacologia , Animais , Biomarcadores , Osso e Ossos/metabolismo , Gerenciamento Clínico , Suscetibilidade a Doenças , Desenvolvimento de Medicamentos , Homeostase , Humanos , Osteogênese/efeitos dos fármacos , Osteoporose/tratamento farmacológico , Osteoporose/prevenção & controle , Osteoprotegerina/química , Osteoprotegerina/uso terapêutico , Ligação Proteica , Ligante RANK , Receptor Ativador de Fator Nuclear kappa-B/metabolismo , Transdução de Sinais/efeitos dos fármacos
6.
Joint Bone Spine ; 88(6): 105243, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34166796

RESUMO

INTRODUCTION: Juvenile Paget's Disease (JPD) is an ultra-rare inherited osteopathy featuring markedly accelerated bone turnover. Several clinical characteristics have been reported, including bone deformities developing in childhood and hearing loss. CASE REPORT: We report the case of a 2 ¾-year-old girl that presented with progressive bowing of both legs since the age of 2, lower limb pain and frequent falls with one consequent femur fracture. Plain radiographs revealed osteoectasia of the long bone's diaphysis, and laboratory tests showed extremely high serum total alkaline phosphatase levels. A missense mutation on the gene TNFRSF11B was identified in homozygosity, and the diagnosis of JPD was made. Treatment with bisphosphonates was initiated early and markedly improved lower limb bowing and pain. The patient reached adulthood with normal height, minor bone deformities, and no functional impairment. Despite the good skeletal symptom's response, bisphosphonates failed to prevent or improve sensorineural hearing loss. CONCLUSIONS: In this clinical case, early treatment with bisphosphonates was effective for the treatment of JPD skeletal deformities. New therapeutic strategies need to be developed to better control the extraskeletal manifestations of JPD.


Assuntos
Mutação de Sentido Incorreto , Osteíte Deformante , Adulto , Difosfonatos/uso terapêutico , Feminino , Homozigoto , Humanos , Osteíte Deformante/diagnóstico , Osteíte Deformante/tratamento farmacológico , Osteíte Deformante/genética , Osteoprotegerina/genética , Osteoprotegerina/uso terapêutico , Adulto Jovem
7.
Int Immunopharmacol ; 98: 107821, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34118644

RESUMO

This study aimed to discuss the expression of angiogenesis-related proteins in bone marrow mesenchymal stem cells (BMSCs) induced by osteoprotegerin (OGP) during osteogenic differentiation in rats, and to analyze the effect of fracture healing inflammatory factor TNF-ɑ on the osteogenic differentiation of BMSCs of rats. BMSCs isolated and cultured from the third generation rats were taken as the research object. According to the addition amount of OGP, BMSCs were divided into control group, OGP (10-7 mol/L) group, OGP (10-8 mol/L) group, and OGP (10-9 mol/L) group. The cell growth and morphological characteristics of each group were observed by inverted phase contrast microscope, the cell proliferation rate was measured by MTT method, angiogenesis-related markers (platelet growth factor (VEGF), cingulate protein 5 (Fbln5), and angiogenin-like protein 4 (Angptl4)) were quantitatively detected by Western blot, and the effect of TNF-ɑ on osteogenic differentiation was detected by CCK. Compared with the control group, MTT results showed that the value-added rate of cells in the OGP (10-8 mol/L) group reached the maximum at 9 days (P < 0.05). The ALP activity in osteoblasts in the OGP (10-8 mol/L) group reached the maximum at 9 days (P < 0.01). The OGP (10-8 mol/L) group had the highest expression of vascular regeneration proteins (VEGF, Fbln5, and Angptl4) (P < 0.05). CCK analysis showed that the TNF-ɑ (1.0 ng/mL) group showed a significant increase in absorbance compared with the control group on 6 days (P < 0.05), and the OD value of the TNF-ɑ (10 ng/mL) group decreased at all time points (P < 0.05). Overall, 10-8 mol/L OGP can induce the proliferation and osteogenic differentiation of MSCs, and promote the expression of angiogenesis-related proteins (VEGF, Fbln5, and Angptl4) during osteogenic differentiation. Besides, 1.0 ng/mL of TNF-ɑ can also promote osteogenesis differentiation of BMSCs in the short term.


Assuntos
Regeneração Óssea/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Osteogênese/efeitos dos fármacos , Osteoprotegerina/farmacologia , Animais , Regeneração Óssea/genética , Regeneração Óssea/imunologia , Diferenciação Celular/imunologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Consolidação da Fratura/efeitos dos fármacos , Consolidação da Fratura/imunologia , Regulação da Expressão Gênica/imunologia , Humanos , Células-Tronco Mesenquimais/imunologia , Modelos Animais , Neovascularização Fisiológica/genética , Osteogênese/imunologia , Osteoporose/tratamento farmacológico , Osteoprotegerina/uso terapêutico , Cultura Primária de Células , Ratos
8.
Photodiagnosis Photodyn Ther ; 34: 102220, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33610736

RESUMO

OBJECTIVE: The aim was to assess the effect of scaling and root planing (SRP) with and without adjunct photodynamic therapy (PDT) on the levels of osteoprotegerin (OPG) receptor activator of NF-kappa B ligand (RANKL) in the unstimulated whole saliva (UWS) of type-2 diabetic and normoglycemic individuals with chronic periodontitis (CP). METHODS: Type-2 diabetic and normoglycemic subjects with CP (Groups 1 and 2, respectively) were divided into test- (SRP + PDT) and control (SRP only) groups. Patient demographics were recorded; and periodontal parameters (marginal bone loss [MBL], probing depth [P.D], plaque index [PI], gingival index [GI], and clinical attachment loss [CAL]) were assessed at baseline and at 3-months-follow-up. Rate of flow of unstimulated whole saliva and levels of RANKL and osteoprotegerin were measured at both time intervals. P < 0.05 was considered statistically significant. RESULTS: Eighty-four persons with CP (42 with and 42 without type-2 DM) were included. At baseline, clinicoradiographic parameters were comparable in all groups. At 3-months of follow-up, there was no significant difference in the clinicoradiographic parameters in all groups. At 3-months of follow-up, there was no significant reduction in whole salivary RANKL and osteoprotegerin levels among individuals in the test and control groups among CP patients with and without CP. CONCLUSION: The whole salivary RANKL/OPG ratio remains high in patients with poorly-controlled type-2 DM after SRP with or without adjunct PDT.


Assuntos
Periodontite Crônica , Diabetes Mellitus Tipo 2 , Fotoquimioterapia , Periodontite Crônica/tratamento farmacológico , Raspagem Dentária , Diabetes Mellitus Tipo 2/tratamento farmacológico , Humanos , Osteoprotegerina/uso terapêutico , Fotoquimioterapia/métodos , Fármacos Fotossensibilizantes/uso terapêutico , Aplainamento Radicular
9.
Clin Exp Metastasis ; 37(1): 13-30, 2020 02.
Artigo em Inglês | MEDLINE | ID: mdl-31578655

RESUMO

More than 2 decades ago, the discovery of osteoprotegerin (OPG) as inhibitor of the receptor of activator of nuclear factor Kb (RANK) ligand (RANKL) revolutionized our understanding of bone biology and oncology. Besides acting as decoy receptor for RANKL, OPG acts as decoy receptor for tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). OPG, RANKL, and TRAIL are ubiquitously expressed, stimulating per se pivotal signaling cascades implicated in cancer. In the context of cancer cell-bone cell interactions, cancer cells skew the OPG/RANKL/RANK (RANKL cognate receptor) balance towards bone destruction and tumor growth through favoring the RANKL/RANK interface, circumventing OPG. Numerous preclinical and clinical studies demonstrate the dual role of OPG in cancer: antitumor and tumor-promoting. OPG potentially conveys an antitumor signal through inhibiting the tumor-promoting RANKL signaling-both the osteoclast-dependent and the osteoclast-independent-and the tumor-promoting TRAIL signaling. On the other hand, the presumed tumor-promoting functions of OPG are: (i) abrogation of TRAIL-induced apoptosis of cancer cells; (ii) abrogation of RANKL-induced antitumor immunity; and (iii) stimulation of oncogenic and prometastatic signaling cascades downstream of the interaction of OPG with diverse proteins. The present review dissects the role of OPG in bone oncology. It presents the available preclinical and clinical data sustaining the dual role of OPG in cancer and focuses on the imbalanced RANKL/RANK/OPG interplay in the landmark "vicious cycle" of skeletal metastatic disease, osteosarcoma, and multiple myeloma. Finally, current challenges and future perspectives in exploiting OPG signaling in bone oncology therapeutics are discussed.


Assuntos
Antineoplásicos/uso terapêutico , Neoplasias Ósseas/tratamento farmacológico , Imunoconjugados/uso terapêutico , Osteoprotegerina/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Ósseas/patologia , Osso e Ossos/citologia , Osso e Ossos/efeitos dos fármacos , Osso e Ossos/patologia , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Imunoconjugados/farmacologia , Metástase Neoplásica/patologia , Metástase Neoplásica/prevenção & controle , Osteoblastos/efeitos dos fármacos , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoclastos/efeitos dos fármacos , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteólise/tratamento farmacológico , Osteoprotegerina/uso terapêutico , Ligante RANK/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo
10.
Eur J Orthod ; 41(1): 1-8, 2019 01 23.
Artigo em Inglês | MEDLINE | ID: mdl-29608684

RESUMO

Background: Because orthodontic tooth movement is dependent upon osteoclast-mediated resorption of alveolar bone adjacent to the pressure side of tooth roots, biologic mediators that regulate osteoclasts can be utilized to control tooth movement. Objectives: To develop a novel method to locally enhance orthodontic anchorage. Methods: We encapsulated osteoprotegerin (OPG) in polymer microspheres and tested the effectiveness of microsphere encapsulated versus non-encapsulated OPG for enhancing orthodontic anchorage in a rodent model of tooth movement. A single injection of 1 mg/kg non-encapsulated or microsphere encapsulated OPG was delivered into the palatal mucosa mesial to the first maxillary molar 1 day prior to tooth movement. A positive control group received injections of 5 mg/kg non-encapsulated OPG every 3 days during tooth movement. After 28 days of tooth movement, hemi-maxillae and femurs were dissected. Molar mesial and incisor distal tooth movement was measured using stone casts that were scanned and magnified. Local alveolar, distant femur bone, and tooth root volumes were analyzed by micro computed tomography. Serum OPG levels were measured by ELISA. Osteoclast numbers were quantified by histomorphometry. Results: The single injection of microsphere encapsulated OPG significantly enhanced orthodontic anchorage, while the single injection of non-encapsulated OPG did not. Injection of encapsulated OPG inhibited molar mesial movement but did not inhibit incisor tooth movement, and did not alter alveolar or femur bone volume fraction, density, or mineral content. Multiple injections of 5 mg/kg non-encapsulated OPG enhanced orthodontic anchorage, but also inhibited incisor retraction and altered alveolar and femur bone quality parameters. Increased OPG levels were found only in animals receiving multiple injections of non-encapsulated 5 mg/kg OPG. Osteoclast numbers were higher upon tooth movement in animals that did not receive OPG. Osteoclast numbers in OPG injected animals were variable within groups. Conclusions: Microsphere encapsulation of OPG allows for controlled drug release, and enhances site-specific orthodontic anchorage without systemic side effects. With additional refinements, this drug delivery system could be applicable to a broad array of potential biologic orthodontic therapeutics.


Assuntos
Reabsorção Óssea/prevenção & controle , Procedimentos de Ancoragem Ortodôntica/métodos , Osteoprotegerina/administração & dosagem , Técnicas de Movimentação Dentária/métodos , Animais , Reabsorção Óssea/diagnóstico por imagem , Sistemas de Liberação de Medicamentos , Avaliação Pré-Clínica de Medicamentos , Fêmur/diagnóstico por imagem , Fêmur/efeitos dos fármacos , Incisivo/diagnóstico por imagem , Incisivo/efeitos dos fármacos , Masculino , Microesferas , Dente Molar/diagnóstico por imagem , Dente Molar/efeitos dos fármacos , Osteoclastos/efeitos dos fármacos , Osteoprotegerina/uso terapêutico , Ratos Sprague-Dawley , Microtomografia por Raio-X
11.
Acta Neuropathol Commun ; 6(1): 31, 2018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29699580

RESUMO

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.


Assuntos
Músculo Esquelético/metabolismo , Distrofias Musculares/metabolismo , Distrofias Musculares/patologia , Distrofias Musculares/terapia , Osteoprotegerina/uso terapêutico , Receptor Ativador de Fator Nuclear kappa-B/deficiência , Animais , Creatina Quinase/metabolismo , Modelos Animais de Doenças , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos mdx , Camundongos Transgênicos , Contração Muscular/efeitos dos fármacos , Contração Muscular/fisiologia , Distrofias Musculares/genética , Osteoprotegerina/química , Osteoprotegerina/metabolismo , RNA Mensageiro/metabolismo , Receptor Ativador de Fator Nuclear kappa-B/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
12.
Int J Mol Sci ; 19(3)2018 Mar 16.
Artigo em Inglês | MEDLINE | ID: mdl-29547583

RESUMO

BACKGROUND: We examined the effects of recombinant human osteoclastogenesis inhibitory factor (hOCIF) on osteolysis, proliferation of mammary tumor cells, and induction of cancer stem cells (CSCs) in the tumor-bone and tumor-subcutaneous microenvironments (TB- and TS-microE). METHODS: Mouse mammary tumor cells were transplanted onto the calvaria or into a subcutaneous lesion of female mice, creating a TB-microE and a TS-microE, and the mice were then treated with hOCIF. To investigate the preventive effects of hOCIF, mice were treated with hOCIF before tumor cell implantation onto the calvaria (Pre), after (Post), and both before and after (Whole). The number of CSCs and cytokine levels were evaluated by IHC and ELISA assay, respectively. RESULTS: hOCIF suppressed osteolysis, and growth of mammary tumors in the TB-microE, but not in the TS-microE. In the Pre, Post, and Whole groups, hOCIF suppressed osteolysis, and cell proliferation. hOCIF increased mouse osteoprotegrin (mOPG) levels in vivo, which suppressed mammary tumor cell proliferation in vitro. These preventive effects were observed in the dose-dependent. hOCIF did not affect the induction of CSCs in either microenvironment. CONCLUSION: While receptor activator of NF-κB ligand (RANKL) targeting therapy may not affect the induction of CSCs, RANKL is a potential target for prevention as well as treatment of breast cancer bone metastasis.


Assuntos
Neoplasias Ósseas/prevenção & controle , Neoplasias Mamárias Experimentais/terapia , Células-Tronco Neoplásicas/efeitos dos fármacos , Osteólise/prevenção & controle , Osteoprotegerina/uso terapêutico , Proteínas Recombinantes/uso terapêutico , Microambiente Tumoral/efeitos dos fármacos , Animais , Neoplasias Ósseas/secundário , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citocinas/análise , Relação Dose-Resposta a Droga , Feminino , Humanos , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Endogâmicos BALB C , Metástase Neoplásica , Osteoprotegerina/análise , Osteoprotegerina/metabolismo , Osteoprotegerina/farmacologia , Proteínas Recombinantes/farmacologia
13.
Semin Dial ; 31(1): 72-81, 2018 01.
Artigo em Inglês | MEDLINE | ID: mdl-28608927

RESUMO

Cardiovascular events make up the primary cause of death in hemodialysis patients, and the risk for cardiovascular mortality is significantly increased by vascular calcification, a condition observed frequently in this patient population. The mechanisms underlying the pathogenesis of vascular calcification are complex, and many factors facilitate or hinder the development of calcification. In this review, we first summarize the main factors contributing to the pathogenesis of vascular calcification in patients with end-stage renal disease. We then explore the role of calcification inhibitors in the calcification process, as well as their effect on vascular dysfunction and mortality in hemodialysis patients.


Assuntos
Doenças Cardiovasculares/etiologia , Doenças Cardiovasculares/prevenção & controle , Causas de Morte , Falência Renal Crônica/terapia , Diálise Renal/efeitos adversos , Calcificação Vascular/tratamento farmacológico , Calcificação Vascular/etiologia , Cardiotônicos/uso terapêutico , Feminino , Seguimentos , Humanos , Falência Renal Crônica/diagnóstico , Falência Renal Crônica/mortalidade , Masculino , Osteopontina/uso terapêutico , Osteoprotegerina/uso terapêutico , Diálise Renal/métodos , Diálise Renal/mortalidade , Medição de Risco , Análise de Sobrevida , Resultado do Tratamento , Calcificação Vascular/fisiopatologia , alfa-2-Glicoproteína-HS/uso terapêutico
14.
Am J Pathol ; 187(3): 498-504, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28041995

RESUMO

Our recent work showed that daily injections of osteoprotegerin (OPG)-immunoglobulin fragment complex (OPG-Fc) completely restore the function of fast-twitch extensor digitorum longus muscles in dystrophic mdx mice, a murine model of Duchenne muscular dystrophy. However, despite marked improvements, OPG-Fc was not as effective in preventing the loss of function of slow-twitch soleus and diaphragm muscles. Because ß2-agonists enhance the function of slow- and fast-twitch dystrophic muscles and because their use is limited by their adverse effects on bone and cardiac tissues, we hypothesized that OPG-Fc, a bone and skeletal muscle protector, acts synergistically with ß2-agonists and potentiates their positive effects on skeletal muscles. We observed that the content of ß2-adrenergic receptors, which are mainly expressed in skeletal muscle, is significantly reduced in dystrophic muscles but is rescued by the injection of OPG-Fc. Most important, OPG-Fc combined with a low dose of formoterol, a member of a new generation of ß2-agonists, histologically and functionally rescued slow-twitch dystrophic muscles. This combination of therapeutic agents, which have already been tested and approved for human use, may open up new therapeutic avenues for Duchenne muscular dystrophy and possibly other neuromuscular diseases.


Assuntos
Agonistas de Receptores Adrenérgicos beta 2/uso terapêutico , Fibras Musculares de Contração Rápida/patologia , Fibras Musculares de Contração Lenta/patologia , Osteoprotegerina/uso terapêutico , Agonistas de Receptores Adrenérgicos beta 2/farmacologia , Animais , Fumarato de Formoterol/farmacologia , Fumarato de Formoterol/uso terapêutico , Inflamação/patologia , Masculino , Camundongos Endogâmicos C57BL , Modelos Biológicos , Fibras Musculares de Contração Rápida/efeitos dos fármacos , Fibras Musculares de Contração Lenta/efeitos dos fármacos , Distrofia Muscular Animal/tratamento farmacológico , Distrofia Muscular Animal/patologia , Osteoprotegerina/farmacologia , Receptores Fc/metabolismo
15.
Vascul Pharmacol ; 82: 30-40, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26924459

RESUMO

Vascular calcification (VC), a disorder that causes blood vessel hardening and dysfunction, is a significant risk factor for type-2 diabetes mellitus (T2DM), which invariably manifests associated cardiovascular complications. Although the clinical effects of VC have been well-documented, the precise cellular events underlying the manifestation and progression of VC are only now coming to light. Current research models indicate that VC likely involves signalling pathways traditionally associated with bone remodelling, such as the OPG/RANKL/TRAIL signalling system. In this respect, receptor activator of NF-κB ligand (RANKL) promotes VC whilst osteoprotegerin (OPG) acts as a RANKL decoy receptor to block this effect, events that contrast with the known functional influence of these proteins during bone metabolism. Moreover, evidence suggests that tumour necrosis factor-related apoptosis-inducing ligand (TRAIL), an alternative decoy ligand for OPG, may exert an anti-calcific influence within the vasculature. In the current review, we conduct a timely examination of this complex VC pathology from both mechanistic and therapeutic perspectives. Our objectives are twofold: (i) to critically assess our current understanding of both osteogenic and vascular calcification pathways, with particular focus on the co-interactive roles of OPG, RANKL, and TRAIL. Extensive in vitro, in vivo, and clinical studies will therefore be reviewed and critical findings highlighted; and (ii) to examine a range of therapeutic approaches of potential relevance to VC pathology. In this regard, a clear focus on VC as it applies to T2DM and cardiovascular disease (and particularly atherosclerosis) will be maintained.


Assuntos
Artérias/metabolismo , Aterosclerose/metabolismo , Diabetes Mellitus Tipo 2/complicações , Angiopatias Diabéticas/metabolismo , Osteogênese , Osteoprotegerina/metabolismo , Ligante RANK/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/metabolismo , Calcificação Vascular/metabolismo , Animais , Artérias/efeitos dos fármacos , Artérias/patologia , Aterosclerose/tratamento farmacológico , Aterosclerose/etiologia , Aterosclerose/patologia , Fármacos Cardiovasculares/uso terapêutico , Angiopatias Diabéticas/tratamento farmacológico , Angiopatias Diabéticas/etiologia , Angiopatias Diabéticas/patologia , Humanos , Osteoblastos/metabolismo , Osteoblastos/patologia , Osteoclastos/metabolismo , Osteoclastos/patologia , Osteogênese/efeitos dos fármacos , Osteoprotegerina/uso terapêutico , Placa Aterosclerótica , Ligante RANK/antagonistas & inibidores , Transdução de Sinais , Ligante Indutor de Apoptose Relacionado a TNF/uso terapêutico , Calcificação Vascular/tratamento farmacológico , Calcificação Vascular/etiologia , Calcificação Vascular/patologia
16.
Dental Press J Orthod ; 20(5): 58-65, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26560822

RESUMO

INTRODUCTION: Orthodontic anchorage is one of the most challenging aspects of Orthodontics. Preventing undesired movement of teeth could result in safer and less complicated orthodontic treatment. Recently, several reviews have been published about the effects of different molecules on bone physiology and the clinical side effects in Orthodontics. However, the effects of local application of these substances on the rate of orthodontic tooth movement have not been assessed. OBJECTIVES: The aim of this research was to analyze the scientific evidence published in the literature about the effects of different molecules on orthodontic anchorage. METHODS: The literature was systematically reviewed using PubMed/Medline, Scopus and Cochrane databases from 2000 up to July 31st, 2014. Articles were independently selected by two different researchers based on previously established inclusion and exclusion criteria, with a concordance Kappa index of 0.86. The methodological quality of the reviewed papers was performed. RESULTS: Search strategy identified 270 articles. Twenty-five of them were selected after application of inclusion/exclusion criteria, and only 11 qualified for final analysis. Molecules involved in orthodontic anchorage were divided into three main groups: osteoprotegerin (OPG), bisphosphonates (BPs) and other molecules (OMs). CONCLUSIONS: Different drugs are able to alter the bone remodeling cycle, influencing osteoclast function and, therefore, tooth movement. Thus, they could be used in order to provide maximal anchorage while preventing undesired movements. OPG was found the most effective molecule in blocking the action of osteoclasts, thereby reducing undesired movements.


Assuntos
Anti-Inflamatórios/farmacologia , Anti-Inflamatórios/uso terapêutico , Antioxidantes/farmacologia , Antioxidantes/uso terapêutico , Difosfonatos/farmacologia , Difosfonatos/uso terapêutico , Procedimentos de Ancoragem Ortodôntica/métodos , Osteoprotegerina/farmacologia , Osteoprotegerina/uso terapêutico , Mobilidade Dentária/tratamento farmacológico , Mobilidade Dentária/prevenção & controle , Acetilcisteína/farmacologia , Acetilcisteína/uso terapêutico , Animais , Remodelação Óssea/efeitos dos fármacos , Celecoxib/farmacologia , Celecoxib/uso terapêutico , Ácido Clodrônico/farmacologia , Ácido Clodrônico/uso terapêutico , Diclofenaco/farmacologia , Diclofenaco/uso terapêutico , Humanos , Imidazóis/farmacologia , Imidazóis/uso terapêutico , Interferon gama/farmacologia , Interferon gama/uso terapêutico , Isoxazóis/farmacologia , Isoxazóis/uso terapêutico , Lactonas/farmacologia , Lactonas/uso terapêutico , Camundongos , Osteoclastos/efeitos dos fármacos , Pamidronato , Ratos , Resveratrol , Estilbenos/farmacologia , Estilbenos/uso terapêutico , Sulfonas/farmacologia , Sulfonas/uso terapêutico , Técnicas de Movimentação Dentária , Ácido Zoledrônico
17.
PLoS One ; 10(8): e0133917, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26247358

RESUMO

Langerhans cell histiocytosis (LCH) is a rare disease caused by the clonal accumulation of dendritic Langerhans cells, which is often accompanied by osteolytic lesions. It has been reported that osteoclast-like cells play a major role in the pathogenic bone destruction seen in patients with LCH and these cells are postulated to originate from the fusion of DCs. However, due to the lack of reliable animal models the pathogenesis of LCH is still poorly understood. In this study, we have established a mouse model of histiocytosis- recapitulating human disease for osteolytic lesions seen in LCH patients. At 12 weeks after birth, severe bone lesions were observed in our multisystem histiocytosis (Mushi) model, when CD8α conventional dendritic cells (DCs) are transformed (MuTuDC) and accumulate. Most importantly, our study demonstrates that bone loss in LCH can be accounted for the transdifferentiation of MuTuDCs into functional osteoclasts both in vivo and in vitro. Moreover, we have shown that injected MuTuDCs reverse the osteopetrotic phenotype of oc/oc mice in vivo. In conclusion, our results support a crucial role of DCs in bone lesions in histiocytosis patients. Furthermore, our new model of LCH based on adoptive transfer of MuTuDC lines, leading to bone lesions within 1-2 weeks, will be an important tool for investigating the pathophysiology of this disease and ultimately for evaluating the potential of anti-resorptive drugs for the treatment of bone lesions.


Assuntos
Osso e Ossos/patologia , Células Dendríticas/patologia , Histiocitose de Células de Langerhans/patologia , Células de Langerhans/patologia , Osteólise/patologia , Animais , Conservadores da Densidade Óssea/uso terapêutico , Osso e Ossos/efeitos dos fármacos , Linhagem Celular , Transdiferenciação Celular , Difosfonatos/uso terapêutico , Modelos Animais de Doenças , Histiocitose de Células de Langerhans/complicações , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Osteoclastos/patologia , Osteólise/complicações , Osteólise/prevenção & controle , Osteoprotegerina/uso terapêutico
18.
Endocrinology ; 156(8): 2762-73, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25961842

RESUMO

Lactation is associated with increased bone turnover and rapid bone loss, which liberates skeletal calcium used for milk production. Previous studies suggested that an increase in the skeletal expression of receptor activator of nuclear factor kappa-light-chain-enhancer of activated B cells ligand (RANKL) coupled with a decrease in osteoprotegerin (OPG) levels likely triggered bone loss during lactation. In this study, we treated lactating mice with recombinant OPG to determine whether bone loss during lactation was dependent on RANKL signaling and whether resorption of the maternal skeleton was required to support milk production. OPG treatment lowered bone resorption rates and completely prevented bone loss during lactation but, surprisingly, did not decrease osteoclast numbers. In contrast, OPG was quite effective at lowering osteoblast numbers and inhibiting bone formation in lactating mice. Furthermore, treatment with OPG during lactation prevented the usual anabolic response associated with reversal of lactational bone loss after weaning. Preventing bone loss had no appreciable effect on milk production, milk calcium levels, or maternal calcium homeostasis when mice were on a standard diet. However, when dietary calcium was restricted, treatment with OPG caused maternal hypocalcemia, maternal death, and decreased milk production. These studies demonstrate that RANKL signaling is a requirement for bone loss during lactation, and suggest that osteoclast activity may be required to increase osteoblast numbers during lactation in preparation for the recovery of bone mass after weaning. These data also demonstrate that maternal bone loss is not absolutely required to supply calcium for milk production unless dietary calcium intake is inadequate.


Assuntos
Reabsorção Óssea/prevenção & controle , Cálcio/metabolismo , Lactação/efeitos dos fármacos , Leite/efeitos dos fármacos , Leite/metabolismo , Osteoprotegerina/uso terapêutico , Animais , Animais Lactentes , Densidade Óssea/efeitos dos fármacos , Cálcio da Dieta/farmacologia , Feminino , Lactação/fisiologia , Camundongos , Mães , Osteoprotegerina/farmacologia , Desmame
19.
Orthod Craniofac Res ; 18 Suppl 1: 187-95, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25865548

RESUMO

OBJECTIVES: To determine minimal dose levels required for local inhibition of orthodontic relapse by recombinant OPG protein (OPG-Fc), while also determining effects of injected OPG-Fc on alveolar bone and long bone. SETTING AND SAMPLE POPULATION: The Department of Orthodontics and Pediatric Dentistry at the University of Michigan. Eighteen male Sprague Dawley rats. MATERIALS & METHODS: Maxillary molars were moved with nickel-titanium springs and then allowed to relapse in Sprague Dawley rats. Upon appliance removal, animals were injected with a single dose of 1.0 mg/kg OPG-Fc, 0.1 mg/kg OPG-Fc, or phosphate-buffered saline (vehicle) just distal to the molar teeth. Tooth movement measurements were made from stone casts, which were scanned and digitally measured. Alveolar tissues were examined by histology. Micro-computed tomography was used to quantify changes in alveolar and femur bone. RESULTS: Local injection of OPG-Fc inhibited molar but not incisor relapse, when compared to vehicle-injected animals. No significant differences in alveolar or femur bone were seen between the three treatment groups after 24 days of relapse. CONCLUSIONS: Our results demonstrate that a single local injection of OPG-Fc effectively inhibits orthodontic relapse, with minimal systemic bone metabolic effects. Our results also show that a single injection of OPG-Fc will influence tooth movement only in teeth close to the injection site. These findings indicate that OPG-Fc has potential as a safe and effective pharmacological means to locally control osteoclasts, for uses such as maintaining anchorage during orthodontic tooth movement and preventing orthodontic relapse in humans.


Assuntos
Processo Alveolar/efeitos dos fármacos , Reabsorção Óssea/prevenção & controle , Osteoprotegerina/uso terapêutico , Técnicas de Movimentação Dentária/métodos , Processo Alveolar/patologia , Animais , Densidade Óssea/efeitos dos fármacos , Remodelação Óssea/efeitos dos fármacos , Reabsorção Óssea/patologia , Fêmur/efeitos dos fármacos , Fêmur/patologia , Incisivo/efeitos dos fármacos , Injeções , Masculino , Maxila/efeitos dos fármacos , Maxila/patologia , Modelos Dentários , Dente Molar/efeitos dos fármacos , Fios Ortodônticos , Osteoclastos/efeitos dos fármacos , Osteoclastos/patologia , Osteoprotegerina/administração & dosagem , Veículos Farmacêuticos , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes , Recidiva , Técnicas de Movimentação Dentária/instrumentação , Microtomografia por Raio-X/métodos
20.
J Periodontal Res ; 50(4): 434-43, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25203865

RESUMO

OBJECTIVE: The objective of this study was to investigate the effects of osteoprotegerin (OPG) gene therapy on alveolar bone resorption caused by experimental periodontitis in rats, thus forming a foundation for potential clinical applications of OPG gene therapy in the treatment of periodontitis and peri-implantitis. MATERIAL AND METHODS: To study the effects of OPG on alveolar bone protection, an experimental periodontitis model was used by placing a bacterial plaque retentive silk ligature in the gingival sulcus around the maxillary second molar tooth, injection of Porphyromonas gingivalis and high carbohydrate diet. A total of 30 Sprague-Dawley rats were randomly divided into three groups, with 10 rats in each group: group I (control) was treated with 10 µL normal saline injection; group II with 10 µL mock vector; and group III with 10 µL local OPG gene transfer by transfection with in vitro constructed pcDNA3.1-human OPG (pcDNA3.1-hOPG). A subperiosteal injection was done adjacent to the second molars on days 0, 7, 14 and 21. Four weeks later, all animals were killed and radiographic, histological and immunohistochemical examinations were performed. Statistical analysis included ANOVA and LSD-Bonferroni test. RESULTS: Group III (OPG gene therapy) had significantly enhanced (p < 0.05) integrated optical density of OPG, had significantly decreased alveolar bone resorption volume and active osteoclast number (p < 0.05) through descriptive histological examination when compared with the other two groups at week 4. CONCLUSION: Local recombinant OPG plasmid-mediated gene therapy suppresses osteoclastogenesis in vivo and inhibits alveolar bone height reduction caused by experimental periodontitis in rats. OPG gene therapy may be beneficial in preventing progressive periodontal bone loss.


Assuntos
Perda do Osso Alveolar/prevenção & controle , Terapia Genética/métodos , Osteoprotegerina/genética , Periodontite/complicações , Perda do Osso Alveolar/microbiologia , Animais , Linhagem Celular , Feminino , Técnicas de Transferência de Genes , Vetores Genéticos/uso terapêutico , Humanos , Microscopia Eletrônica de Varredura , Mioblastos/fisiologia , Osteoclastos/patologia , Osteoprotegerina/uso terapêutico , Periodontite/microbiologia , Plasmídeos/genética , Porphyromonas gingivalis/fisiologia , Distribuição Aleatória , Ratos , Ratos Sprague-Dawley , Transfecção/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...