Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Biochem Pharmacol ; 188: 114579, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33895161

RESUMEN

Osteolytic diseases, including breast cancer-induced osteolysis and postmenopausal osteoporosis, are attributed to excessive bone resorption by osteoclasts. Spleen tyrosine kinase (SYK) is involved in osteoclastogenesis and bone resorption, whose role in breast cancer though remains controversial. Effects of PRT062607 (PRT), a highly specific inhibitor of SYK, on the osteoclast and breast cancer functionalities are yet to be clarified. This study demonstrated the in vitro inhibitory actions of PRT on the osteoclast-specific gene expression, bone resorption, and osteoclastogenesis caused by receptor activator of nuclear factor kappa B ligand (RANKL), as well as its in vitro suppressive effects on the growth, migration and invasion of breast carcinoma cell line MDA-MB-231, which were achieved through PLCγ2 and PI3K-AKT-mTOR pathways. Further, we proved that PRT could prevent post-ovariectomy (OVX) loss of bone and breast cancer-induced bone destruction in vivo, which agreed with the in vitro outcomes. In conclusion, our findings suggest the potential value of PRT in managing osteolytic diseases mediated by osteoclasts.


Asunto(s)
Neoplasias de la Mama/enzimología , Ciclohexilaminas/uso terapéutico , Osteólisis/enzimología , Ovariectomía/efectos adversos , Pirimidinas/uso terapéutico , Quinasa Syk/antagonistas & inhibidores , Quinasa Syk/metabolismo , Animales , Resorción Ósea/enzimología , Resorción Ósea/patología , Resorción Ósea/prevención & control , Neoplasias de la Mama/patología , Neoplasias de la Mama/prevención & control , Línea Celular Tumoral , Ciclohexilaminas/farmacología , Relación Dosis-Respuesta a Droga , Femenino , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Desnudos , Osteólisis/patología , Osteólisis/prevención & control , Pirimidinas/farmacología
2.
Mol Med Rep ; 23(1)2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33236155

RESUMEN

Matrix metalloproteinase 2 (MMP2) is a well­characterized protein that is indispensable for extracellular matrix remodeling and other pathological processes, such as tumor progression and skeletal dysplasia. Excessive activation of MMP2 promotes osteolytic metastasis and bone destruction in late­stage cancers, while its loss­of­function mutations result in the decreased bone mineralization and generalized osteolysis occurring progressively in skeletal developmental disorders, particularly in multicentric osteolysis, nodulosis and arthropathy (MONA). Either upregulation or downregulation of MMP2 activity can result in the same osteolytic effects. Thus, different functions of MMP2 have been recently identified that could explain this observation. While MMP2 can degrade bone matrix, facilitate osteoclastogenesis and amplify various signaling pathways that enhance osteolysis in bone metastasis, its role in maintaining the number of bone cells, supporting osteocytic canalicular network formation and suppressing leptin­mediated inhibition of bone formation has been implicated in osteolytic disorders caused by MMP2 deficiency. Furthermore, the proangiogenic activity of MMP2 is one of the potential mechanisms that are associated with both pathological situations. In the present article, the latest research on MMP2 in bone homeostasis is reviewed and the mechanisms underlying the role of this protein in skeletal metastasis and developmental osteolysis are discussed.


Asunto(s)
Neoplasias Óseas , Huesos , Metaloproteinasa 2 de la Matriz , Proteínas de Neoplasias , Osteocondrodisplasias , Osteólisis , Animales , Neoplasias Óseas/enzimología , Neoplasias Óseas/genética , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Huesos/enzimología , Huesos/patología , Humanos , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metástasis de la Neoplasia , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Osteocondrodisplasias/enzimología , Osteocondrodisplasias/genética , Osteocondrodisplasias/patología , Osteólisis/enzimología , Osteólisis/genética , Osteólisis/patología
3.
Biosci Rep ; 40(6)2020 06 26.
Artículo en Inglés | MEDLINE | ID: mdl-32478376

RESUMEN

Bone metastasis of colorectal cancer (CRC) cells leads to osteolysis. Aberrant activation of osteoclasts is responsible for bone resorption in tumor. In general, bone marrow-derived monocytes (BMMs) differentiate into osteoclasts, however, how CRC cells interact with BMMs and how to regulate the differentiation is elusive. We here report that CRC cells promote bone resorption in bone metastasis. Transcriptomic profiling revealed CCL3 up-regulated in MC-38 conditional medium treated BMMs. Further investigation demonstrated that CCL3 produced by BMMs facilitated cell infusion and thus promoted the osteoclastogenesis. In addition, CRC cells derived EGF stimulated the production of CCL3 in BMMs through activation of ERK/CREB pathway. Blockage of EGF or CCL3 can efficiently attenuate the osteolysis in bone metastasis of CRC.


Asunto(s)
Neoplasias Óseas/enzimología , Quimiocina CCL3/metabolismo , Neoplasias Colorrectales/enzimología , Factor de Crecimiento Epidérmico/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Osteoclastos/enzimología , Osteogénesis , Osteólisis/enzimología , Tibia/enzimología , Animales , Neoplasias Óseas/genética , Neoplasias Óseas/secundario , Comunicación Celular , Línea Celular Tumoral , Quimiocina CCL3/genética , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/genética , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Masculino , Ratones Endogámicos C57BL , Osteoclastos/patología , Osteólisis/genética , Osteólisis/patología , Transducción de Señal , Tibia/patología
4.
Cell Death Dis ; 9(5): 498, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29703893

RESUMEN

Osteolysis is an osteolytic lesion featured by enhanced osteoclast formation and potent bone erosion. Lacking of effective regimen for treatment of the pathological process highlights the importance of identifying agents that can suppress the differentiation and function of osteoclast. Artemether is a natural compound derived from Artemisia annua L. and it is popularized for the treatment of malaria. In present study, we demonstrated that artemether could suppress RANKL-induced osteoclastogenesis and expression of osteoclast marker genes such as tartrate-resistant acid phosphatase, cathepsin K, matrix metalloproteinase 9, nuclear factor of activated T-cell cytoplasmic 1, and dendritic cell-specific transmembrane protein. It inhibited the osteoclastic bone resorption in a dose-dependent manner in vitro. Furthermore, artemether attenuated RANKL-induced MAPKs (ERK, JNK, p-38) activity. In addition, we have showed that artemether was able to mitigate bone erosion in a murine model of LPS-induced inflammatory bone loss. Taken together, these findings suggest that artemether reduces inflammatory bone loss via inhibition of MAPKs activation during osteoclast differentiation, and it might be a potential candidate for the treatment of osteoclast-related disorders.


Asunto(s)
Arteméter/farmacología , Conservadores de la Densidad Ósea/farmacología , Remodelación Ósea/efectos de los fármacos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteólisis/prevención & control , Animales , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Lipopolisacáridos , Masculino , Ratones , Ratones Endogámicos C57BL , Osteoclastos/enzimología , Osteoclastos/patología , Osteogénesis/genética , Osteólisis/inducido químicamente , Osteólisis/enzimología , Osteólisis/patología , Ligando RANK/farmacología , Células RAW 264.7
5.
J Cell Physiol ; 233(3): 2398-2408, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-28731198

RESUMEN

Currently, there are no medications available to treat aseptic loosening of orthopedic implants. Using osteoprotegerin fusion protein (OPG-Fc), we previously blocked instability-induced osteoclast differentiation and peri-prosthetic osteolysis. Wnt/ß-catenin signaling, which regulates OPG secretion from osteoblasts, also modulates the bone tissue response to mechanical loading. We hypothesized that activating Wnt/ß-catenin signaling by inhibiting glycogen synthase kinase-3ß (GSK-3ß) would reduce instability-induced bone loss through regulation of both osteoblast and osteoclast differentiation. We examined effects of GSK-3ß inhibition on regulation of RANKL and OPG in a rat model of mechanical instability-induced peri-implant osteolysis. The rats were treated daily with a GSK-3ß inhibitor, AR28 (20 mg/kg bw), for up to 5 days. Bone tissue and blood serum were assessed by qRT-PCR, immunohistochemistry, and ELISA on days 3 and 5, and by micro-CT on day 5. After 3 days of treatment with AR28, mRNA levels of ß-catenin, Runx2, Osterix, Col1α1, and ALP were increased leading to higher osteoblast numbers compared to vehicle-treated animals. BMP-2 and Wnt16 mRNA levels were downregulated by mechanical instability and this was rescued by GSK-3ß inhibition. Osteoclast numbers were decreased significantly after 3 days of GSK-3ß inhibition, which correlated with enhanced OPG mRNA expression. This was accompanied by decreased serum levels of TRAP5b on days 3 and 5. Treatment with AR28 upregulated osteoblast differentiation, while osteoclastogenesis was blunted, leading to increased bone mass by day 5. These data suggest that GSK-3ß inactivation suppresses osteolysis through regulating both osteoblast and osteoclast differentiation in a rat model of instability-induced osteolysis.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Osteoblastos/efectos de los fármacos , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Osteólisis/prevención & control , Falla de Prótesis , Inhibidores de Proteínas Quinasas/farmacología , Tibia/efectos de los fármacos , Fosfatasa Alcalina/genética , Fosfatasa Alcalina/metabolismo , Animales , Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 2/metabolismo , Placas Óseas , Proliferación Celular/efectos de los fármacos , Colágeno Tipo I/genética , Colágeno Tipo I/metabolismo , Cadena alfa 1 del Colágeno Tipo I , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 1 del Factor de Unión al Sitio Principal/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Glucógeno Sintasa Quinasa 3 beta/metabolismo , Masculino , Osteoblastos/enzimología , Osteoblastos/patología , Osteoclastos/enzimología , Osteoclastos/patología , Osteólisis/enzimología , Osteólisis/genética , Osteólisis/patología , Osteoprotegerina/genética , Osteoprotegerina/metabolismo , Implantación de Prótesis/instrumentación , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas Sprague-Dawley , Fosfatasa Ácida Tartratorresistente/sangre , Tibia/enzimología , Tibia/patología , Tibia/cirugía , Factores de Tiempo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , beta Catenina/genética , beta Catenina/metabolismo
6.
J Cell Physiol ; 232(3): 617-624, 2017 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-27312515

RESUMEN

Bone homeostasis is maintained by a balance between resorption of the bone matrix and its replacement by new bone. Osteoclasts play a crucially important role in bone metabolism. They are responsible for bone resorption under pathophysiological conditions. Differentiation of these cells, which are derived from bone marrow cells, depends on receptor activator of NF-κB ligand (RANKL). RANKL-induced osteoclastogenesis is regulated by the phosphoinositide (PI) signaling pathway, in which diacylglycerol (DG) serves as a second messenger in signal transduction. In this study, we examined the functional implications of DG kinase (DGK), an enzyme family responsible for DG metabolism, for osteoclast differentiation and activity. Of DGKs, DGKζ is most abundantly expressed in osteoclast precursors such as bone marrow-derived monocytes/macrophages. During osteoclast differentiation from precursor cells, DGKζ is downregulated at the protein level. In this regard, we found that DGKζ deletion enhances osteoclast differentiation and bone resorption activity under inflammatory conditions in an animal model of osteolysis. Furthermore, DGKζ deficiency upregulates RANKL expression in response to TNFα stimulation. Collectively, results suggest that DGKζ is silent under normal conditions, but it serves as a negative regulator in osteoclast function under inflammatory conditions. Downregulation of DGKζ might be one factor predisposing a person to osteolytic bone destruction in pathological conditions. J. Cell. Physiol. 232: 617-624, 2017. © 2016 Wiley Periodicals, Inc.


Asunto(s)
Resorción Ósea/enzimología , Resorción Ósea/patología , Diferenciación Celular , Diacilglicerol Quinasa/metabolismo , Regulación hacia Abajo , Inflamación/patología , Osteoclastos/patología , Animales , Biomarcadores/metabolismo , Densidad Ósea/efectos de los fármacos , Resorción Ósea/complicaciones , Diferenciación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Fibroblastos/metabolismo , Inflamación/complicaciones , Inflamación/enzimología , Isoenzimas/metabolismo , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones Noqueados , Osteoclastos/efectos de los fármacos , Osteólisis/complicaciones , Osteólisis/enzimología , Osteólisis/patología , Ligando RANK/genética , Ligando RANK/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Factores de Tiempo , Tomografía Computarizada por Rayos X , Factor de Necrosis Tumoral alfa/farmacología , Regulación hacia Arriba/efectos de los fármacos
7.
Sci Transl Med ; 8(353): 353ra113, 2016 08 24.
Artículo en Inglés | MEDLINE | ID: mdl-27559096

RESUMEN

Myelomatous bone disease is characterized by the development of lytic bone lesions and a concomitant reduction in bone formation, leading to chronic bone pain and fractures. To understand the underlying mechanism, we investigated the contribution of myeloma-expressed thymidine phosphorylase (TP) to bone lesions. In osteoblast progenitors, TP up-regulated the methylation of RUNX2 and osterix, leading to decreased bone formation. In osteoclast progenitors, TP up-regulated the methylation of IRF8 and thereby enhanced expression of NFATc1 (nuclear factor of activated T cells, cytoplasmic 1 protein), leading to increased bone resorption. TP reversibly catalyzes thymidine into thymine and 2-deoxy-d-ribose (2DDR). Myeloma-secreted 2DDR bound to integrin αVß3/α5ß1 in the progenitors, activated PI3K (phosphoinositide 3-kinase)/Akt signaling, and increased DNMT3A (DNA methyltransferase 3A) expression, resulting in hypermethylation of RUNX2, osterix, and IRF8 This study elucidates an important mechanism for myeloma-induced bone lesions, suggesting that targeting TP may be a viable approach to healing resorbed bone in patients. Because TP overexpression is common in bone-metastatic tumors, our findings could have additional mechanistic implications.


Asunto(s)
Neoplasias Óseas/enzimología , Neoplasias Óseas/patología , Resorción Ósea/enzimología , Resorción Ósea/patología , Mieloma Múltiple/enzimología , Mieloma Múltiple/patología , Osteogénesis/fisiología , Timidina Fosforilasa/metabolismo , Neoplasias Óseas/fisiopatología , Resorción Ósea/fisiopatología , Línea Celular Tumoral , Subunidad alfa 1 del Factor de Unión al Sitio Principal/genética , Islas de CpG , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN , ADN Metiltransferasa 3A , Regulación hacia Abajo , Humanos , Factores Reguladores del Interferón/genética , Mieloma Múltiple/fisiopatología , Osteoblastos/patología , Osteoblastos/fisiología , Osteoclastos/patología , Osteoclastos/fisiología , Osteólisis/enzimología , Osteólisis/patología , Osteólisis/prevención & control , Ligando RANK/metabolismo , Factor de Transcripción Sp7/genética , Timidina Fosforilasa/antagonistas & inhibidores , Regulación hacia Arriba
8.
J Clin Invest ; 126(5): 1759-72, 2016 05 02.
Artículo en Inglés | MEDLINE | ID: mdl-27043283

RESUMEN

Multiple myeloma (MM) cells secrete osteoclastogenic factors that promote osteolytic lesions; however, the identity of these factors is largely unknown. Here, we performed a screen of human myeloma cells to identify pro-osteoclastogenic agents that could potentially serve as therapeutic targets for ameliorating MM-associated bone disease. We found that myeloma cells express high levels of the matrix metalloproteinase MMP-13 and determined that MMP-13 directly enhances osteoclast multinucleation and bone-resorptive activity by triggering upregulation of the cell fusogen DC-STAMP. Moreover, this effect was independent of the proteolytic activity of the enzyme. Further, in mouse xenograft models, silencing MMP-13 expression in myeloma cells inhibited the development of osteolytic lesions. In patient cohorts, MMP-13 expression was localized to BM-associated myeloma cells, while elevated MMP-13 serum levels were able to correctly predict the presence of active bone disease. Together, these data demonstrate that MMP-13 is critical for the development of osteolytic lesions in MM and that targeting the MMP-13 protein - rather than its catalytic activity - constitutes a potential approach to mitigating bone disease in affected patients.


Asunto(s)
Metaloproteinasa 13 de la Matriz/metabolismo , Mieloma Múltiple/enzimología , Proteínas de Neoplasias/metabolismo , Osteoclastos/enzimología , Osteólisis/enzimología , Animales , Fusión Celular , Femenino , Xenoinjertos , Humanos , Masculino , Metaloproteinasa 13 de la Matriz/genética , Ratones , Ratones Noqueados , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Mieloma Múltiple/terapia , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Osteoclastos/patología , Osteólisis/genética , Osteólisis/patología , Osteólisis/terapia
9.
Am J Med Genet A ; 170A(2): 410-417, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26601801

RESUMEN

​Multicentric osteolysis nodulosis and arthropathy (MONA) is an infrequently described autosomal recessive skeletal dysplasia characterized by progressive osteolysis and arthropathy. Inactivating mutations in MMP2, encoding matrix metalloproteinase-2, are known to cause this disorder. Fifteen families with mutations in MMP2 have been reported in literature. In this study we screened thirteen individuals from eleven families for MMP2 mutations and identified eight mutations (five novel and three known variants). We characterize the clinical, radiographic and molecular findings in all individuals with molecularly proven MONA from the present cohort and previous reports, and provide a comprehensive review of the MMP2 related disorders.


Asunto(s)
Metaloproteinasa 2 de la Matriz/genética , Mutación/genética , Osteólisis/genética , Adolescente , Secuencia de Aminoácidos , Niño , Preescolar , Estudios de Cohortes , Femenino , Homocigoto , Humanos , Lactante , Recién Nacido , Masculino , Datos de Secuencia Molecular , Osteólisis/enzimología , Osteólisis/patología , Pronóstico , Homología de Secuencia de Aminoácido
10.
Biomaterials ; 69: 12-21, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26275858

RESUMEN

Aseptic loosening is associated with the development of wear debris-induced peri-implant osteolytic bone disease caused by an increased osteoclastic bone resorption and decreased osteoblastic bone formation. However, no effective measures for the prevention and treatment of peri-implant osteolysis currently exist. The aim of this study was to determine whether lithium chloride (LiCl), a selective inhibitor of glycogen synthetase kinase 3 beta (GSK-3ß), mitigates wear debris-induced osteolysis in a murine calvarial model of osteolysis. GSK-3ß is activated by titanium (Ti) particles, and implantation of Ti particles on the calvarial surface in C57BL/6 mice resulted in osteolysis caused by an increase in the number of osteoclasts and a decrease in the number of osteoblasts. Mice implanted with Ti particles were gavage-fed LiCl (50 or 200 mg kg(-1)d(-1)), 6 days per week for 2 weeks. The LiCl treatment significantly inhibited GSK-3ß activity and increased ß-catenin and axin-2 expression in a dose-dependent manner, dramatically mitigating the Ti particle-induced suppression of osteoblast numbers and the expression of bone formation markers. Finally, we demonstrated that inhibition of GSK-3ß suppresses osteoclast differentiation and reduces the severity of Ti particle-induced osteolysis. The results of this study indicate that Ti particle-induced osteolysis is partly dependent on GSK-3ß and, therefore, the canonical Wnt signaling pathway. This suggests that selective inhibitors of GSK-3ß such as LiCl may help prevent and treat wear debris-induced osteolysis.


Asunto(s)
Glucógeno Sintasa Quinasa 3/antagonistas & inhibidores , Cloruro de Litio/uso terapéutico , Osteólisis/tratamiento farmacológico , Osteólisis/enzimología , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Femenino , Glucógeno Sintasa Quinasa 3/metabolismo , Glucógeno Sintasa Quinasa 3 beta , Ratones , Ratones Endogámicos C57BL , Osteólisis/etiología , Osteólisis/patología , Prótesis e Implantes/efectos adversos , Cráneo/efectos de los fármacos , Cráneo/enzimología , Cráneo/patología , Titanio/efectos adversos
11.
Int J Mol Sci ; 15(12): 21913-34, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25464380

RESUMEN

The aim of this study was to assess the effect of naringenin on osteoclastogenesis and titanium particle-induced osteolysis. Osteolysis from wear-induced particles and aseptic loosening are the most frequent late complications of total joint arthroplasty leading to revision of the prosthesis. Osteolysis during aseptic loosening is most likely due to increased bone resorption by osteoclasts. Through in vitro studies, we demonstrated that naringenin, a naturally occurring flavanone in grapefruit and tomatoes, exerts potent inhibitory effects on the ligand of the receptor activator of nuclear factor-κB (RANKL)-induced osteoclastogenesis and revealed that the mechanism of action of naringenin, which inhibited osteoclastogenesis by suppression of the p38 signaling pathway. Through in vivo studies, we proved that naringenin attenuated titanium particle-induced osteolysis in a mouse calvarial model. In general, we demonstrated that naringenin inhibited osteoclastogenesis via suppression of p38 signaling in vitro and attenuated titanium particle-induced osteolysis in vivo. This study also suggested that naringenin has significant potential for the treatment of osteolysis-related diseases caused by excessive osteoclast formation and activity.


Asunto(s)
Flavanonas/farmacología , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Osteoclastos/enzimología , Osteogénesis/efectos de los fármacos , Osteólisis/enzimología , Ligando RANK/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Actinas/metabolismo , Animales , Anisomicina/farmacología , Resorción Ósea/complicaciones , Resorción Ósea/patología , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Flavanonas/química , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones Endogámicos C57BL , Modelos Biológicos , FN-kappa B/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoclastos/efectos de los fármacos , Osteoclastos/patología , Osteólisis/complicaciones , Osteólisis/patología , Titanio
12.
J Biol Chem ; 288(47): 33634-33641, 2013 Nov 22.
Artículo en Inglés | MEDLINE | ID: mdl-24081142

RESUMEN

Phospholipase C γ2 (PLCγ2) is a critical regulator of innate immune cells and osteoclasts (OCs) during inflammatory arthritis. Both the catalytic domain and the adaptor motifs of PLCγ2 are required for OC formation and function. Due to the high homology between the catalytic domains of PLCγ2 and the ubiquitously expressed PLCγ1, molecules encompassing the adaptor motifs of PLCγ2 were designed to test the hypothesis that uncoupling the adaptor and catalytic functions of PLCγ2 could specifically inhibit osteoclastogenesis and bone erosion. Wild-type (WT) bone marrow macrophages (BMM) that overexpress the tandem Src homology 2 (SH2) domains of PLCγ2 (SH2(N+C)) failed to form mature OCs and resorb bone in vitro. Activation of the receptor activator of NF-κB (RANK) signaling pathway, which is critical for OC development, was impaired in cells expressing SH2(N+C). Arrest in OC differentiation was evidenced by a reduction of p38 and Iκ-Bα phosphorylation as well as decreased NFATc1 and c-Fos/c-Jun levels. Consistent with our hypothesis, SH2(N+C) abrogated formation of the RANK-Gab2 complex, which mediates NF-κB and AP-1 activation following RANK ligand (RANKL) stimulation. Furthermore, the ability of SH2(N+C) to prevent inflammatory osteolysis was examined in vivo following RANKL or LPS injections over the calvaria. Both models induced osteolysis in the control group, whereas the SH2(N+C)-treated cohort was largely protected from bone erosion. Collectively, these data indicate that inflammatory osteolysis can be abrogated by treatment with a molecule composed of the tandem SH2 domains of PLCγ2.


Asunto(s)
Células de la Médula Ósea/enzimología , Osteoclastos/enzimología , Osteólisis/enzimología , Fosfolipasa C gamma/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Animales , Células de la Médula Ósea/patología , Proteínas I-kappa B/genética , Proteínas I-kappa B/metabolismo , Inflamación/inducido químicamente , Inflamación/tratamiento farmacológico , Inflamación/enzimología , Inflamación/genética , Inflamación/patología , Lipopolisacáridos/toxicidad , Ratones , Inhibidor NF-kappaB alfa , Factores de Transcripción NFATC/genética , Factores de Transcripción NFATC/metabolismo , Osteoclastos/patología , Osteólisis/tratamiento farmacológico , Osteólisis/genética , Osteólisis/patología , Fosfolipasa C gamma/antagonistas & inhibidores , Fosfolipasa C gamma/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fosforilación/efectos de los fármacos , Fosforilación/genética , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Ligando RANK/genética , Ligando RANK/metabolismo , Receptor Activador del Factor Nuclear kappa-B/genética , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Dominios Homologos src
13.
J Biol Chem ; 288(42): 30399-30410, 2013 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-24005670

RESUMEN

Abnormal osteoclast formation and osteolysis are the hallmarks of multiple myeloma (MM) bone disease, yet the underlying molecular mechanisms are incompletely understood. Here, we show that the AKT pathway was up-regulated in primary bone marrow monocytes (BMM) from patients with MM, which resulted in sustained high expression of the receptor activator of NF-κB (RANK) in osteoclast precursors. The up-regulation of RANK expression and osteoclast formation in the MM BMM cultures was blocked by AKT inhibition. Conditioned media from MM cell cultures activated AKT and increased RANK expression and osteoclast formation in BMM cultures. Inhibiting AKT in cultured MM cells decreased their growth and ability to promote osteoclast formation. Of clinical significance, systemic administration of the AKT inhibitor LY294002 blocked the formation of tumor tissues in the bone marrow cavity and essentially abolished the MM-induced osteoclast formation and osteolysis in SCID mice. The level of activating transcription factor 4 (ATF4) protein was up-regulated in the BMM cultures from multiple myeloma patients. Adenoviral overexpression of ATF4 activated RANK expression in osteoclast precursors. These results demonstrate a new role of AKT in the MM promotion of osteoclast formation and bone osteolysis through, at least in part, the ATF4-dependent up-regulation of RANK expression in osteoclast precursors.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Mieloma Múltiple/enzimología , Osteoclastos/enzimología , Osteólisis/enzimología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Regulación hacia Arriba , Factor de Transcripción Activador 4/metabolismo , Animales , Cromonas/farmacología , Inhibidores Enzimáticos/farmacología , Femenino , Xenoinjertos , Humanos , Masculino , Ratones , Ratones SCID , Morfolinas/farmacología , Mieloma Múltiple/patología , Trasplante de Neoplasias , Osteoclastos/patología , Osteólisis/patología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Células Tumorales Cultivadas
14.
Biomed Res Int ; 2013: 230805, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23862137

RESUMEN

The leading complication of total joint replacement is periprosthetic osteolysis, which often results in aseptic loosening of the implant, leading to revision surgery. Extracellular matrix degradation and connective tissue remodeling around implants have been considered as major biological events in the periprosthetic loosening. Critical mediators of wear particle-induced inflammatory osteolysis released by periprosthetic synovial cells (mainly macrophages) are inflammatory cytokines, chemokines, and proteolytic enzymes, mainly matrix metalloproteinases (MMPs). Numerous studies reveal a strong interdependence of MMP expression and activity with the molecular mechanisms that control the composition and turnover of periprosthetic matrices. MMPs can either actively modulate or be modulated by the molecular mechanisms that determine the debris-induced remodeling of the periprosthetic microenvironment. In the present study, the molecular mechanisms that control the composition, turnover, and activity of matrix macromolecules within the periprosthetic microenvironment exposed to wear debris are summarized and presented. Special emphasis is given to MMPs and their endogenous tissue inhibitors (TIMPs), as well as to the proteasome pathway, which appears to be an elegant molecular regulator of specific matrix macromolecules (including specific MMPs and TIMPs). Furthermore, strong rationale for potential clinical applications of the described molecular mechanisms to the treatment of periprosthetic loosening and osteolysis is provided.


Asunto(s)
Matriz Extracelular/metabolismo , Metaloproteinasas de la Matriz/metabolismo , Osteólisis/enzimología , Osteólisis/etiología , Falla de Prótesis/efectos adversos , Complejo de la Endopetidasa Proteasomal/metabolismo , Inhibidores Tisulares de Metaloproteinasas/metabolismo , Humanos , Osteólisis/terapia
15.
Exp Hematol ; 41(6): 547-557.e2, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23435312

RESUMEN

Multiple myeloma (MM) cells typically grow in focal lesions, stimulating osteoclasts that destroy bone and support MM. Osteoclasts and MM cells are hypermetabolic. The coenzyme nicotinamide adenine dinucleotide (NAD(+)) is not only essential for cellular metabolism; it also affects activity of NAD-dependent enzymes, such as PARP-1 and SIRT-1. Nicotinamide phosphoribosyltransferase (NAMPT/PBEF/visfatin, encoded by PBEF1) is a rate-limiting enzyme in NAD(+) biosynthesis from nicotinamide. Coculture of primary MM cells with osteoclasts induced PBEF1 upregulation in both cell types. PBEF1 expression was higher in experimental myelomatous bones than in nonmyelomatous bone and higher in MM patients' plasma cells than in healthy donors' counterparts. APO866 is a specific PBEF1 inhibitor known to deplete cellular NAD(+). APO866 at low nanomolar concentrations inhibited growth of primary MM cells or MM cell lines cultured alone or cocultured with osteoclasts and induced apoptosis in these cells. PBEF1 activity and NAD(+) content were reduced in MM cells by APO866, resulting in lower activity of PARP-1 and SIRT-1. The inhibitory effect of APO866 on MM cell growth was abrogated by supplementation of extracellular NAD(+) or NAM. APO866 inhibited NF-κB activity in osteoclast precursors and suppressed osteoclast formation and activity. PBEF1 knockdown similarly inhibited MM cell growth and osteoclast formation. In the SCID-rab model, APO866 inhibited growth of primary MM and H929 cells and prevented bone disease. These findings indicate that MM cells and osteoclasts are highly sensitive to NAD(+) depletion and that PBEF1 inhibition represents a novel approach to target cellular metabolism and inhibit PARP-1 and bone disease in MM.


Asunto(s)
Citocinas/fisiología , Mieloma Múltiple/enzimología , Proteínas de Neoplasias/fisiología , Nicotinamida Fosforribosiltransferasa/fisiología , Osteoclastos/enzimología , Osteólisis/enzimología , Acrilamidas/farmacología , Animales , Huesos/patología , Diferenciación Celular/efectos de los fármacos , Técnicas de Cocultivo , Citocinas/antagonistas & inhibidores , Inducción Enzimática , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones SCID , Mieloma Múltiple/complicaciones , Mieloma Múltiple/patología , NAD/metabolismo , FN-kappa B/antagonistas & inhibidores , Proteínas de Neoplasias/antagonistas & inhibidores , Niacinamida/metabolismo , Mononucleótido de Nicotinamida/análogos & derivados , Mononucleótido de Nicotinamida/farmacología , Nicotinamida Fosforribosiltransferasa/antagonistas & inhibidores , Osteoclastos/fisiología , Osteólisis/etiología , Osteólisis/patología , Piperidinas/farmacología , Poli(ADP-Ribosa) Polimerasa-1 , Poli(ADP-Ribosa) Polimerasas/metabolismo , Conejos , Sirtuina 1/metabolismo , Células Tumorales Cultivadas/metabolismo , Regulación hacia Arriba
16.
Leukemia ; 26(9): 2114-23, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22425892

RESUMEN

Bone destruction is a hallmark of multiple myeloma and affects more than 80% of patients. However, current therapy is unable to completely cure and/or prevent bone lesions. Although it is accepted that myeloma cells mediate bone destruction by inhibition of osteoblasts and activation of osteoclasts, the underlying mechanism is still poorly understood. This study demonstrates that constitutive activation of p38 mitogen-activated protein kinase in myeloma cells is responsible for myeloma-induced osteolysis. Our results show that p38 is constitutively activated in most myeloma cell lines and primary myeloma cells from patients. Myeloma cells with high/detectable p38 activity, but not those with low/undetectable p38 activity, injected into severe combined immunodeficient (SCID) or SCID-hu mice caused bone destruction. Inhibition or knockdown of p38 in human myeloma reduced or prevented myeloma-induced osteolytic bone lesions without affecting tumor growth, survival, or homing to bone. Mechanistic studies showed that myeloma cell p38 activity inhibited osteoblastogenesis and bone formation and activated osteoclastogenesis and bone resorption in myeloma-bearing SCID mice. This study elucidates a novel molecular mechanism-activation of p38 signaling in myeloma cells-by which myeloma cells induce osteolytic bone lesions, and indicates that targeting myeloma cell p38 may be a viable approach to treating or preventing myeloma bone disease.


Asunto(s)
Enfermedades Óseas/etiología , Mieloma Múltiple/complicaciones , Mieloma Múltiple/enzimología , Osteólisis/etiología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Apoptosis , Western Blotting , Enfermedades Óseas/enzimología , Enfermedades Óseas/patología , Estudios de Casos y Controles , Comunicación Celular , Proliferación Celular , Humanos , Técnicas para Inmunoenzimas , Ratones , Ratones SCID , Mieloma Múltiple/patología , Osteólisis/enzimología , Osteólisis/patología , ARN Interferente Pequeño/genética , Transducción de Señal , Células Tumorales Cultivadas , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas p38 Activadas por Mitógenos/genética
17.
Acta Neurochir (Wien) ; 154(4): 611-20; discussion 620, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22327326

RESUMEN

OBJECT: Although bone invasion and hyperostosis are common phenomena in patients with intracranial meningiomas, the basic pathomechanism is not fully understood. Based on an immunohistochemical study of surgically resected samples with hyperostosis, we postulate a possible mechanism of hyperostosis in patients with intracranial meningiomas. MATERIALS AND METHODS: Forty-six meningiomas were evaluated in this study. Twenty-six meningiomas associated with hyperostosis specimens served as the study group, and 20 meningiomas without any bony changes served as controls. An immunohistochemical staining technique was used to detect the expression of matrix metalloproteinase (MMP)-2, -9, and -13, membrane type (MT)1-MMP, estrogen receptor (ER), and progesterone receptor (PR) in the main tumor and hyperostotic portions of the studied samples. RESULTS: In the non-hyperostosis group, expression of MMP-13, MT1-MMP, and ER was significantly less than in the main tumor portion of hyperostotic meningiomas, while there was no difference in the expression of MMP-2 and -9 and PR in the main tumor between the two groups. In the hyperostosis group, the immunoreactivity of MMP-2 in the hyperostotic portion revealed a higher pattern of expression than the main tumor (p < 0.002). The expression of MMP-9, MT1-MMP, ER, and PR had relatively positive immunoreactivity in the main tumor portion (P < 0.05). CONCLUSIONS: Increased expression of MMP-13 and MT1-MMP in the tumor portion of hyperostosis of meningiomas might contribute to the initiation of osteolysis. Activated MMP-2 in hyperostotic lesions may change the physiological metabolism of the skull bone, thus playing an important role in hyperostosis formation.


Asunto(s)
Hiperostosis/enzimología , Metaloproteinasas de la Matriz/fisiología , Neoplasias Meníngeas/enzimología , Meningioma/enzimología , Cráneo/enzimología , Biomarcadores de Tumor/fisiología , Femenino , Humanos , Hiperostosis/patología , Hiperostosis/fisiopatología , Masculino , Metaloproteinasa 13 de la Matriz/fisiología , Metaloproteinasa 14 de la Matriz/fisiología , Metaloproteinasa 2 de la Matriz/fisiología , Metaloproteinasa 9 de la Matriz/fisiología , Neoplasias Meníngeas/patología , Neoplasias Meníngeas/fisiopatología , Meningioma/patología , Meningioma/fisiopatología , Invasividad Neoplásica/patología , Invasividad Neoplásica/fisiopatología , Osteólisis/enzimología , Osteólisis/patología , Osteólisis/fisiopatología , Cráneo/patología , Cráneo/fisiopatología
18.
PLoS One ; 7(1): e29615, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22253746

RESUMEN

We investigated the effects of the matrix metalloproteinase 13 (MMP13)-selective inhibitor, 5-(4-{4-[4-(4-fluorophenyl)-1,3-oxazol-2-yl]phenoxy}phenoxy)-5-(2-methoxyethyl) pyrimidine-2,4,6(1H,3H,5H)-trione (Cmpd-1), on the primary tumor growth and breast cancer-associated bone remodeling using xenograft and syngeneic mouse models. We used human breast cancer MDA-MB-231 cells inoculated into the mammary fat pad and left ventricle of BALB/c Nu/Nu mice, respectively, and spontaneously metastasizing 4T1.2-Luc mouse mammary cells inoculated into mammary fat pad of BALB/c mice. In a prevention setting, treatment with Cmpd-1 markedly delayed the growth of primary tumors in both models, and reduced the onset and severity of osteolytic lesions in the MDA-MB-231 intracardiac model. Intervention treatment with Cmpd-1 on established MDA-MB-231 primary tumors also significantly inhibited subsequent growth. In contrast, no effects of Cmpd-1 were observed on soft organ metastatic burden following intracardiac or mammary fat pad inoculations of MDA-MB-231 and 4T1.2-Luc cells respectively. MMP13 immunostaining of clinical primary breast tumors and experimental mice tumors revealed intra-tumoral and stromal expression in most tumors, and vasculature expression in all. MMP13 was also detected in osteoblasts in clinical samples of breast-to-bone metastases. The data suggest that MMP13-selective inhibitors, which lack musculoskeletal side effects, may have therapeutic potential both in primary breast cancer and cancer-induced bone osteolysis.


Asunto(s)
Neoplasias de la Mama/complicaciones , Neoplasias de la Mama/patología , Inhibidores de la Metaloproteinasa de la Matriz , Osteólisis/etiología , Osteólisis/patología , Inhibidores de Proteasas/farmacología , Animales , Neoplasias Óseas/patología , Neoplasias Óseas/secundario , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/prevención & control , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Creatina Quinasa/metabolismo , Modelos Animales de Enfermedad , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Metaloproteinasa 13 de la Matriz/metabolismo , Ratones , Osteoblastos/efectos de los fármacos , Osteoblastos/enzimología , Osteoblastos/patología , Osteólisis/enzimología , Inhibidores de Proteasas/sangre , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Curr Protein Pept Sci ; 13(2): 164-79, 2012 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-22044155

RESUMEN

Over the last three decades, V-ATPases have emerged from the obscurity of poorly understood membrane proton transport phenomena to being recognized as ubiquitous proton pumps that underlie vital cellular processes in all eukaryotic and many prokaryotic cells. These exquisitely complex molecular motors also engage in diverse specialized roles contributing to development, tissue function and pH homeostasis within complex organisms. Increasingly, mutations and misappropriation of V-ATPase function have been linked to diseases, ranging from sclerosing bone pathologies and renal tubular acidosis to bone-loss disorders and cancer metastasis. Much remains to be learned about the details of V-ATPase cell and molecular biology; nevertheless, interest in V-ATPases as potential therapeutic targets has burgeoned in recent years. In this review, we present a history of our involvement and contributions to the understanding of V-ATPase structure and function and our nascent and ongoing contributions to translating the knowledge gained from basic research on the nature of V-ATPases into tools for drug discovery. We focus here primarily on the treatment of bone-loss pathologies, like osteoporosis, and present proof-of-concept for a drug screening strategy based on targeting a3-B2 subunit interactions within the V-ATPase complex.


Asunto(s)
Descubrimiento de Drogas/métodos , Osteoporosis/tratamiento farmacológico , Osteoporosis/enzimología , Mapas de Interacción de Proteínas/efectos de los fármacos , ATPasas de Translocación de Protón Vacuolares/metabolismo , Animales , Humanos , Modelos Moleculares , Osteólisis/tratamiento farmacológico , Osteólisis/enzimología , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , ATPasas de Translocación de Protón Vacuolares/química
20.
Artículo en Inglés | MEDLINE | ID: mdl-20971661

RESUMEN

OBJECTIVE: The aim of this study was to investigate the relationship between the immunohistochemical expression of MMP-1 and MMP-9 with the clinical behavior of central giant cell lesions (CGCLs) of the jaws. STUDY DESIGN: Paraffin-embedded tissue from 30 aggressive and 12 nonaggressive CGCLs was assessed for the expression of MMP-1 and MMP-9 using immunohistochemistry. RESULTS: Although cellular immunolocalization patterns of MMP-1 and MMP-9 were similar, mean values of expression estimation/SID scores of each protease were significantly higher in aggressive CGCLs in comparison with nonaggressive lesions. Moreover, linear regression analysis showed that there was a reasonably good correlation not only between the expression estimation but also among SID scores of the 2 proteolytic enzymes. CONCLUSION: The findings of this study suggest a role for MMP-1 and MMP-9 in the resorptive activity of different cellular groups in CGCLs and indicate that differences in immunoreactivity of these 2 proteolytic enzymes may underlie the distinct clinical behavior.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/genética , Granuloma de Células Gigantes/enzimología , Enfermedades Maxilomandibulares/enzimología , Metaloproteinasa 1 de la Matriz/análisis , Metaloproteinasa 9 de la Matriz/análisis , Adolescente , Adulto , Recuento de Células , Forma de la Célula , Niño , Preescolar , Citoplasma/enzimología , Citoplasma/ultraestructura , Progresión de la Enfermedad , Femenino , Células Gigantes/enzimología , Células Gigantes/patología , Granuloma de Células Gigantes/patología , Humanos , Inmunohistoquímica , Enfermedades Maxilomandibulares/patología , Masculino , Enfermedades Mandibulares/enzimología , Enfermedades Mandibulares/patología , Enfermedades Maxilares/enzimología , Enfermedades Maxilares/patología , Persona de Mediana Edad , Osteólisis/enzimología , Osteólisis/patología , Recurrencia , Células del Estroma/enzimología , Células del Estroma/patología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA