Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
1.
Cell ; 184(5): 1330-1347.e13, 2021 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-33636130

RESUMEN

Osteoclasts are large multinucleated bone-resorbing cells formed by the fusion of monocyte/macrophage-derived precursors that are thought to undergo apoptosis once resorption is complete. Here, by intravital imaging, we reveal that RANKL-stimulated osteoclasts have an alternative cell fate in which they fission into daughter cells called osteomorphs. Inhibiting RANKL blocked this cellular recycling and resulted in osteomorph accumulation. Single-cell RNA sequencing showed that osteomorphs are transcriptionally distinct from osteoclasts and macrophages and express a number of non-canonical osteoclast genes that are associated with structural and functional bone phenotypes when deleted in mice. Furthermore, genetic variation in human orthologs of osteomorph genes causes monogenic skeletal disorders and associates with bone mineral density, a polygenetic skeletal trait. Thus, osteoclasts recycle via osteomorphs, a cell type involved in the regulation of bone resorption that may be targeted for the treatment of skeletal diseases.


Asunto(s)
Resorción Ósea/patología , Osteoclastos/patología , Ligando RANK/metabolismo , Animales , Apoptosis , Resorción Ósea/metabolismo , Fusión Celular , Células Cultivadas , Humanos , Macrófagos/citología , Ratones , Osteocondrodisplasias/tratamiento farmacológico , Osteocondrodisplasias/genética , Osteocondrodisplasias/metabolismo , Osteocondrodisplasias/patología , Osteoclastos/metabolismo , Transducción de Señal
2.
Bone ; 139: 115493, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32569873

RESUMEN

Bisphosphonates (BP) are a class of calcium-binding drug used to prevent bone resorption in skeletal disorders such as osteoporosis and metastatic bone disease. They act by selectively targeting bone-resorbing osteoclasts and can be grouped into two classes depending on their intracellular mechanisms of action. Simple BPs cause osteoclast apoptosis after cytoplasmic conversion into toxic ATP analogues. In contrast, nitrogen-containing BPs potently inhibit FPP synthase, an enzyme of the mevalonate (cholesterol biosynthesis) pathway. This results in production of a toxic metabolite (ApppI) and the loss of long-chain isoprenoid lipids required for protein prenylation, a process necessary for the function of small GTPase proteins essential for the survival and activity of osteoclasts. In this review we provide a state-of-the-art overview of these mechanisms of action and a historical perspective of how they were discovered. Finally, we challenge the long-held dogma that BPs act only in the skeleton and highlight recent studies that reveal insights into hitherto unknown effects on tumour-associated and tissue-resident macrophages.


Asunto(s)
Resorción Ósea , Difosfonatos , Resorción Ósea/tratamiento farmacológico , Huesos , Difosfonatos/farmacología , Humanos , Osteoclastos , Prenilación de Proteína
3.
Oncotarget ; 8(40): 68047-68058, 2017 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-28978095

RESUMEN

Melphalan is a cytotoxic chemotherapy used to treat patients with multiple myeloma (MM). Bone resorption by osteoclasts, by remodeling the bone surface, can reactivate dormant MM cells held in the endosteal niche to promote tumor development. Dormant MM cells can be reactivated after melphalan treatment; however, it is unclear whether melphalan treatment increases osteoclast formation to modify the endosteal niche. Melphalan treatment of mice for 14 days decreased bone volume and the endosteal bone surface, and this was associated with increases in osteoclast numbers. Bone marrow cells (BMC) from melphalan-treated mice formed more osteoclasts than BMCs from vehicle-treated mice, suggesting that osteoclast progenitors were increased. Melphalan also increased osteoclast formation in BMCs and RAW264.7 cells in vitro, which was prevented with the cell stress response (CSR) inhibitor KNK437. Melphalan also increased expression of the osteoclast regulator the microphthalmia-associated transcription factor (MITF), but not nuclear factor of activated T cells 1 (NFATc1). Melphalan increased expression of MITF-dependent cell fusion factors, dendritic cell-specific transmembrane protein (Dc-stamp) and osteoclast-stimulatory transmembrane protein (Oc-stamp) and increased cell fusion. Expression of osteoclast stimulator receptor activator of NFκB ligand (RANKL) was unaffected by melphalan treatment. These data suggest that melphalan stimulates osteoclast formation by increasing osteoclast progenitor recruitment and differentiation in a CSR-dependent manner. Melphalan-induced osteoclast formation is associated with bone loss and reduced endosteal bone surface. As well as affecting bone structure this may contribute to dormant tumor cell activation, which has implications for how melphalan is used to treat patients with MM.

4.
Nat Commun ; 6: 8983, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26632274

RESUMEN

Multiple myeloma is largely incurable, despite development of therapies that target myeloma cell-intrinsic pathways. Disease relapse is thought to originate from dormant myeloma cells, localized in specialized niches, which resist therapy and repopulate the tumour. However, little is known about the niche, and how it exerts cell-extrinsic control over myeloma cell dormancy and reactivation. In this study, we track individual myeloma cells by intravital imaging as they colonize the endosteal niche, enter a dormant state and subsequently become activated to form colonies. We demonstrate that dormancy is a reversible state that is switched 'on' by engagement with bone-lining cells or osteoblasts, and switched 'off' by osteoclasts remodelling the endosteal niche. Dormant myeloma cells are resistant to chemotherapy that targets dividing cells. The demonstration that the endosteal niche is pivotal in controlling myeloma cell dormancy highlights the potential for targeting cell-extrinsic mechanisms to overcome cell-intrinsic drug resistance and prevent disease relapse.


Asunto(s)
Remodelación Ósea/fisiología , Mieloma Múltiple/metabolismo , Osteoclastos/fisiología , Anciano , Anciano de 80 o más Años , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Femenino , Humanos , Masculino , Ratones , Ratones Endogámicos , Persona de Mediana Edad , Osteoblastos/fisiología
5.
Cancer Discov ; 5(1): 35-42, 2015 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-25312016

RESUMEN

UNLABELLED: Recent clinical trials have shown that bisphosphonate drugs improve breast cancer patient survival independent of their antiresorptive effects on the skeleton. However, because bisphosphonates bind rapidly to bone mineral, the exact mechanisms of their antitumor action, particularly on cells outside of bone, remain unknown. Here, we used real-time intravital two-photon microscopy to show extensive leakage of fluorescent bisphosphonate from the vasculature in 4T1 mouse mammary tumors, where it initially binds to areas of small, granular microcalcifications that are engulfed by tumor-associated macrophages (TAM), but not tumor cells. Importantly, we also observed uptake of radiolabeled bisphosphonate in the primary breast tumor of a patient and showed the resected tumor to be infiltrated with TAMs and to contain similar granular microcalcifications. These data represent the first compelling in vivo evidence that bisphosphonates can target cells in tumors outside the skeleton and that their antitumor activity is likely to be mediated via TAMs. SIGNIFICANCE: Bisphosphonates are assumed to act solely in bone. However, mouse models and clinical trials show that they have surprising antitumor effects outside bone. We provide unequivocal evidence that bisphosphonates target TAMs, but not tumor cells, to exert their extraskeletal effects, offering a rationale for use in patients with early disease.


Asunto(s)
Conservadores de la Densidad Ósea/metabolismo , Difosfonatos/metabolismo , Macrófagos/metabolismo , Neoplasias/diagnóstico , Tomografía Computarizada de Emisión de Fotón Único , Tomografía Computarizada por Rayos X , Animales , Conservadores de la Densidad Ósea/uso terapéutico , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/inmunología , Calcinosis , Carbocianinas , Difosfonatos/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Ratones , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Neoplasias/tratamiento farmacológico , Fagocitosis/inmunología , Ensayos Antitumor por Modelo de Xenoinjerto
7.
Biochim Biophys Acta ; 1841(4): 569-73, 2014 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-24369118

RESUMEN

Nitrogen-containing bisphosphonates (N-BPs) such as zoledronic acid (ZOL) are the gold standard treatment for diseases of excessive bone resorption. N-BPs inactivate osteoclasts via inhibition of farnesyl diphosphate synthase (FPPS), thereby preventing the prenylation of essential small GTPases. Not all patients respond to N-BP therapy to the same extent, and some patients, for example with tumour-associated bone disease or Paget's disease, appear to develop resistance to N-BPs. The extent to which upregulation of FPPS might contribute to these phenomena is not clear. Using quantitative PCR and western blot analysis we show that levels of FPPS mRNA and protein can be upregulated in HeLa cells by culturing in lipoprotein deficient serum (LDS) or by over-expression of SREBP-1a. Upregulated, endogenous FPPS was predominantly localised to the cytosol and did not co-localise with peroxisomal or mitochondrial markers. Upregulation of endogenous FPPS conferred resistance to the inhibitory effect of low concentrations of ZOL on the prenylation of the small GTPase Rap1a. These observations suggest that an increase in the expression of endogenous FPPS could confer at least partial resistance to the pharmacological effect of N-BP drugs such as ZOL in vivo.


Asunto(s)
Resorción Ósea/genética , Difosfonatos/farmacología , Geraniltranstransferasa/genética , Prenilación de Proteína/efectos de los fármacos , Resorción Ósea/patología , Regulación de la Expresión Génica/efectos de los fármacos , Geraniltranstransferasa/antagonistas & inhibidores , Geraniltranstransferasa/biosíntesis , Células HeLa , Humanos , Imidazoles/farmacología , Osteoclastos/efectos de los fármacos , Osteoclastos/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/biosíntesis , Ácido Zoledrónico
8.
Arthritis Rheum ; 64(10): 3334-43, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22674221

RESUMEN

OBJECTIVE: CD248 (tumor endothelial marker 1/endosialin) is found on stromal cells and is highly expressed during malignancy and inflammation. Studies have shown a reduction in inflammatory arthritis in CD248-knockout (CD248(-/-) ) mice. The aim of the present study was to investigate the functional effect of genetic deletion of CD248 on bone mass. METHODS: Western blotting, polymerase chain reaction, and immunofluorescence were used to investigate the expression of CD248 in humans and mice. Micro-computed tomography and the 3-point bending test were used to measure bone parameters and mechanical properties of the tibiae of 10-week-old wild-type (WT) or CD248(-/-) mice. Human and mouse primary osteoblasts were cultured in medium containing 10 mM ß-glycerophosphate and 50 µg/ml ascorbic acid to induce mineralization, and then treated with platelet-derived growth factor BB (PDGF-BB). The mineral apposition rate in vivo was calculated by identifying newly formed bone via calcein labeling. RESULTS: Expression of CD248 was seen in human and mouse osteoblasts, but not osteoclasts. CD248(-/-) mouse tibiae had higher bone mass and superior mechanical properties (increased load required to cause fracture) compared to WT mice. Primary osteoblasts from CD248(-/-) mice induced increased mineralization in vitro and produced increased bone over 7 days in vivo. There was no decrease in bone mineralization and no increase in proliferation of osteoblasts in response to stimulation with PDGF-BB, which could be attributed to a defect in PDGF signal transduction in the CD248(-/-) mice. CONCLUSION: There is an unmet clinical need to address rheumatoid arthritis-associated bone loss. Genetic deletion of CD248 in mice results in high bone mass due to increased osteoblast-mediated bone formation, suggesting that targeting CD248 in rheumatoid arthritis may have the effect of increasing bone mass in addition to the previously reported effect of reducing inflammation.


Asunto(s)
Antígenos CD/metabolismo , Huesos/metabolismo , Proteínas de Neoplasias/metabolismo , Osteoblastos/metabolismo , Osteogénesis/fisiología , Animales , Antígenos CD/genética , Becaplermina , Huesos/efectos de los fármacos , Calcificación Fisiológica/efectos de los fármacos , Calcificación Fisiológica/fisiología , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Proteínas Proto-Oncogénicas c-sis/farmacología
9.
Cancer ; 118(3): 750-60, 2012 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-21751201

RESUMEN

BACKGROUND: Osteosarcoma is the most frequent malignant primary bone tumor that occurs mainly in the young, with an incidence peak observed at age 18 years. Both apomine and lovastatin have antitumor activity in a variety of cancer cell lines. Apomine, a 1,1-bisphosphonate-ester, increases the rate of degradation of 3-hydroxy-3 methylglutaryl-coenzyme A (HMG-CoA) reductase, the rate-limiting enzyme in the mevalonate pathway, whereas lovastatin competitively inhibits HMG-CoA reductase enzyme activity, thereby preventing protein prenylation and cholesterol synthesis. METHODS: The authors of this report investigated the effect of combined treatment with apomine and lovastatin in vitro on human and murine osteosarcoma cell lines and in vivo using a murine syngeneic model of osteosarcoma. Apomine and lovastatin synergistically decreased viability and induced apoptosis in both murine and human osteosarcoma cell lines. RESULTS: Combined apomine and lovastatin strongly decreased HMG-CoA reductase enzyme levels compared with lovastatin treatment alone. Consequently, the accumulation of unprenylated ras-related protein 1A induced by lovastatin was enhanced in the presence of apomine. All synergistic effects on cell viability, apoptosis, and protein prenylation were overcome by the addition of mevalonate or geranylgeraniol, 2 mevalonate pathway intermediates downstream from the target enzyme, HMG-CoA reductase. This confirmed that the mechanism of synergy in osteosarcoma cells is through augmented inhibition of HMG-CoA reductase. Finally, treatment of POS-1 osteosarcoma-bearing mice with a combination of apomine and lovastatin significantly reduced tumor progression in these mice compared with single treatments, which had no effect at the doses used. CONCLUSIONS: The results from this study revealed that combination therapy with apomine and lovastatin may be a novel treatment strategy for osteosarcoma.


Asunto(s)
Apoptosis/efectos de los fármacos , Neoplasias Óseas/tratamiento farmacológico , Proliferación Celular/efectos de los fármacos , Difosfonatos/farmacología , Inhibidores de Hidroximetilglutaril-CoA Reductasas/farmacología , Lovastatina/farmacología , Osteosarcoma/tratamiento farmacológico , Animales , Protocolos de Quimioterapia Combinada Antineoplásica , Neoplasias Óseas/patología , Ciclo Celular/efectos de los fármacos , Sinergismo Farmacológico , Humanos , Masculino , Ratones , Ratones Endogámicos C3H , Osteosarcoma/patología , Prenilación de Proteína/efectos de los fármacos , Ratas , Células Tumorales Cultivadas
10.
J Bone Miner Res ; 26(8): 1926-38, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21472776

RESUMEN

Familial expansile osteolysis and related disorders are caused by heterozygous tandem duplication mutations in the signal peptide region of the gene encoding receptor activator of NF-κB (RANK), a receptor critical for osteoclast formation and function. Previous studies have shown that overexpression of these mutant proteins causes constitutive activation of NF-κB signaling in vitro, and it has been assumed that this accounts for the focal osteolytic lesions that are seen in vivo. We show here that constitutive activation of NF-κB occurred in HEK293 cells overexpressing wild-type or mutant RANK but not in stably transfected cell lines expressing low levels of each RANK gene. Importantly, only cells expressing wild-type RANK demonstrated ligand-dependent activation of NF-κB. When overexpressed, mutant RANK did not localize to the plasma membrane but localized to extensive areas of organized smooth endoplasmic reticulum, whereas, as expected, wild-type RANK was detected at the plasma membrane and in the Golgi apparatus. This intracellular accumulation of the mutant proteins is probably the result of lack of signal peptide cleavage because, using two in vitro translation systems, we demonstrate that the mutations in RANK prevent cleavage of the signal peptide. In conclusion, signal peptide mutations lead to accumulation of RANK in the endoplasmic reticulum and prevent direct activation by RANK ligand. These results strongly suggest that the increased osteoclast formation/activity caused by these mutations cannot be explained by studying the homozygous phenotype alone but requires further detailed investigation of the heterozygous expression of the mutant RANK proteins.


Asunto(s)
Mutación/genética , FN-kappa B/metabolismo , Señales de Clasificación de Proteína/genética , Receptor Activador del Factor Nuclear kappa-B/genética , Secuencia de Bases , Línea Celular , ADN Nucleotidiltransferasas/metabolismo , Regulación de la Expresión Génica , Células HEK293 , Humanos , Datos de Secuencia Molecular , Peso Molecular , Proteínas Mutantes/metabolismo , Proteínas Mutantes/ultraestructura , Osteoclastos/metabolismo , Osteoclastos/ultraestructura , Transporte de Proteínas , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Receptor Activador del Factor Nuclear kappa-B/ultraestructura , Reproducibilidad de los Resultados , Fracciones Subcelulares/metabolismo , Transfección
11.
Bone ; 49(1): 111-21, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21419243

RESUMEN

The described ability of phosphonocarboxylate analogues of bisphosphonates (BPs) to inhibit Rab geranylgeranyl transferase (RGGT) is thought to be the mechanism underlying their cellular effects, including their ability to reduce macrophage cell viability and to inhibit osteoclast-mediated resorption. However, until now the possibility that at least some of the effects of these drugs may be mediated through other targets has not been excluded. Since RGGT is the most distal enzyme in the process of Rab prenylation, it has not proved possible to confirm the mechanism underlying the effects of these drugs by adding back downstream intermediates of the mevalonate pathway, the approach used to demonstrate that bisphosphonates act through this pathway. We now confirm that RGGT is the major pharmacological target of phosphonocarboxylates by using several alternative approaches. Firstly, analysis of several different phosphonocarboxylate drugs demonstrates a very good correlation between the ability of these drugs to inhibit RGGT with their ability to: (a) reduce macrophage cell viability; (b) induce apoptosis; and (c) induce vacuolation in rabbit osteoclasts. Secondly, we have found that cells from the gunmetal (gm/gm) mouse, which bear a homozygous mutation in RGGT that results in ~80% reduced activity of this enzyme compared to wild-type or heterozygous mice, are more sensitive to the effects of active phosphonocarboxylates (including reducing macrophage cell viability, inhibiting osteoclast formation and inhibiting fluid-phase endocytosis), confirming that these effects are mediated through inhibition of RGGT. In conclusion, these data demonstrate that all of the pharmacological effects of phosphonocarboxylates found thus far appear to be mediated through the specific inhibition of RGGT, highlighting the potential therapeutic value of this class of drugs.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Difosfonatos/metabolismo , Difosfonatos/farmacología , Transferasas Alquil y Aril/metabolismo , Animales , Apoptosis/efectos de los fármacos , Recuento de Células , Línea Celular , Supervivencia Celular/efectos de los fármacos , Diterpenos/farmacología , Endocitosis/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Heterocigoto , Macrófagos/efectos de los fármacos , Macrófagos/enzimología , Ratones , Ratones Endogámicos , Osteoblastos/efectos de los fármacos , Osteoblastos/enzimología , Osteoclastos/efectos de los fármacos , Osteoclastos/enzimología , Prenilación de Proteína/efectos de los fármacos , Transporte de Proteínas/efectos de los fármacos , Piridinas/farmacología , Conejos , Vacuolas/efectos de los fármacos , Vacuolas/metabolismo
12.
Bone ; 49(1): 34-41, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21111853

RESUMEN

This review describes the key discoveries over the last 15 years that have led to a clearer understanding of the molecular mechanisms by which bisphosphonate drugs inhibit bone resorption. Once released from bone mineral surfaces during bone resorption, these agents accumulate intracellularly in osteoclasts. Simple bisphosphonates such as clodronate are incorporated into non-hydrolysable analogues of adenosine triphosphate, which induce osteoclast apoptosis. The considerably more potent nitrogen-containing bisphosphonates are not metabolised but potently inhibit farnesyl pyrophosphate (FPP) synthase, a key enzyme of the mevalonate pathway. This prevents the synthesis of isoprenoid lipids necessary for the post-translational prenylation of small GTPases, thereby disrupting the subcellular localisation and normal function of these essential signalling proteins. Inhibition of FPP synthase also results in the accumulation of the upstream metabolite isopentenyl diphosphate, which is incorporated into the toxic nucleotide metabolite ApppI. Together, these properties explain the ability of bisphosphonate drugs to inhibit bone resorption by disrupting osteoclast function and survival. These discoveries are also giving insights into some of the adverse effects of bisphosphonates, such as the acute phase reaction that is triggered by inhibition of FPP synthase in peripheral blood monocytes.


Asunto(s)
Difosfonatos/química , Difosfonatos/farmacología , Animales , Huesos/efectos de los fármacos , Dimetilaliltranstransferasa/antagonistas & inhibidores , Dimetilaliltranstransferasa/metabolismo , Humanos , Espacio Intracelular/efectos de los fármacos , Espacio Intracelular/metabolismo , Redes y Vías Metabólicas/efectos de los fármacos , Prenilación de Proteína/efectos de los fármacos
13.
Br J Pharmacol ; 160(3): 762-71, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20590578

RESUMEN

BACKGROUND AND PURPOSE: Increased circulating levels of L-alpha-lysophosphatidylinositol (LPI) are associated with cancer and LPI is a potent, ligand for the G-protein-coupled receptor GPR55. Here we have assessed the modulation of breast cancer cell migration, orientation and polarization by LPI and GPR55. EXPERIMENTAL APPROACH: Quantitative RT-PCR was used to measure GPR55 expression in breast cancer cell lines. Cell migration and invasion were measured using a Boyden chamber chemotaxis assay and Cultrex invasion assay, respectively. Cell polarization and orientation in response to the microenvironment were measured using slides containing nanometric grooves. KEY RESULTS: GPR55 expression was detected in the highly metastatic MDA-MB-231 breast cancer cell line. In these cells, LPI stimulated binding of [(35)S]GTPgammaS to cell membranes (pEC(50) 6.47 +/- 0.45) and significantly enhanced cell chemotaxis towards serum. MCF-7 cells expressed low levels of GPR55 and did not migrate or invade towards serum factors. When GPR55 was over-expressed in MCF-7 cells, serum induced a robust migratory and invasive response, which was further enhanced by LPI and prevented by siRNA to GPR55. The physical microenvironment has been identified as a key factor in determining breast tumour cell metastatic fate. LPI endowed MDA-MB-231 cells with the capacity to detect shallow (40 nm deep) grooved slides and induced marked cancer cell polarization on both flat and grooved surfaces. CONCLUSIONS AND IMPLICATIONS: LPI and GPR55 play a role in the modulation of migration, orientation and polarization of breast cancer cells in response to the tumour microenvironment.


Asunto(s)
Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Movimiento Celular/fisiología , Polaridad Celular/fisiología , Quimiotaxis/fisiología , Lisofosfolípidos/fisiología , Invasividad Neoplásica , Metástasis de la Neoplasia/fisiopatología , Receptores Acoplados a Proteínas G/fisiología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Polaridad Celular/efectos de los fármacos , Quimiotaxis/efectos de los fármacos , Femenino , Guanosina 5'-O-(3-Tiotrifosfato)/metabolismo , Humanos , Lisofosfolípidos/antagonistas & inhibidores , Lisofosfolípidos/farmacología , ARN Interferente Pequeño/farmacología , Receptores de Cannabinoides , Receptores Acoplados a Proteínas G/metabolismo
14.
J Bone Miner Res ; 25(3): 606-16, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20422624

RESUMEN

Bisphosphonates are effective antiresorptive agents owing to their bone-targeting property and ability to inhibit osteoclasts. It remains unclear, however, whether any non-osteoclast cells are directly affected by these drugs in vivo. Two fluorescent risedronate analogues, carboxyfluorescein-labeled risedronate (FAM-RIS) and Alexa Fluor 647-labeled risedronate (AF647-RIS), were used to address this question. Twenty-four hours after injection into 3-month-old mice, fluorescent risedronate analogues were bound to bone surfaces. More detailed analysis revealed labeling of vascular channel walls within cortical bone. Furthermore, fluorescent risedronate analogues were present in osteocytic lacunae in close proximity to vascular channels and localized to the lacunae of newly embedded osteocytes close to the bone surface. Following injection into newborn rabbits, intracellular uptake of fluorescently labeled risedronate was detected in osteoclasts, and the active analogue FAM-RIS caused accumulation of unprenylated Rap1A in these cells. In addition, CD14(high) bone marrow monocytes showed relatively high levels of uptake of fluorescently labeled risedronate, which correlated with selective accumulation of unprenylated Rap1A in CD14(+) cells, as well as osteoclasts, following treatment with risedronate in vivo. Similar results were obtained when either rabbit or human bone marrow cells were treated with fluorescent risedronate analogues in vitro. These findings suggest that the capacity of different cell types to endocytose bisphosphonate is a major determinant for the degree of cellular drug uptake in vitro as well as in vivo. In conclusion, this study shows that in addition to bone-resorbing osteoclasts, bisphosphonates may exert direct effects on bone marrow monocytes in vivo.


Asunto(s)
Células de la Médula Ósea/metabolismo , Difosfonatos/farmacocinética , Ácido Etidrónico/análogos & derivados , Monocitos/metabolismo , Osteocitos/metabolismo , Animales , Western Blotting , Conservadores de la Densidad Ósea/química , Ácido Etidrónico/síntesis química , Ácido Etidrónico/química , Femenino , Colorantes Fluorescentes/síntesis química , Colorantes Fluorescentes/química , Ratones , Ratones Endogámicos C57BL , Prenilación , Conejos , Ácido Risedrónico , Proteínas de Unión al GTP rap1/metabolismo
15.
Int J Cancer ; 126(1): 239-46, 2010 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-19621390

RESUMEN

Nitrogen-containing bisphosphonates (N-BPs) are effective antiosteolytic agents in patients with multiple myeloma. Preclinical studies have also demonstrated that these agents have direct antitumor effects in vitro and can reduce tumor burden in a variety of animal models, although it is not clear whether such effects are caused by direct actions on tumor cells or by inhibition of bone resorption. N-BPs prevent bone destruction in myeloma by inhibiting the enzyme farnesyl pyrophosphate synthase in osteoclasts, thereby preventing the prenylation of small GTPase signaling proteins. In this study, utilizing a plasmacytoma xenograft model without complicating skeletal lesions, treatment with zoledronic acid (ZOL) led to significant prolongation of survival in severe combined immunodeficiency mice inoculated with human INA-6 plasma cells. Following treatment with a clinically relevant dose of ZOL, histological analysis of INA-6 tumors from the peritoneal cavity revealed extensive areas of apoptosis associated with poly (ADP-ribose) polymerase cleavage. Furthermore, Western blot analysis of tumor homogenates demonstrated the accumulation of unprenylated Rap1A, indicative of the uptake of ZOL by nonskeletal tumors and inhibition of farnesyl pyrophosphate synthase. These studies provide, for the first time, clear evidence that N-BPs have direct antitumor effects in plasma cell tumors in vivo and this is executed by a molecular mechanism similar to that observed in osteoclasts.


Asunto(s)
Conservadores de la Densidad Ósea/uso terapéutico , Difosfonatos/uso terapéutico , Imidazoles/uso terapéutico , Mieloma Múltiple/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Conservadores de la Densidad Ósea/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Difosfonatos/farmacología , Modelos Animales de Enfermedad , Femenino , Humanos , Imidazoles/farmacología , Ratones , Ratones SCID , Mieloma Múltiple/metabolismo , Prenilación de Proteína , Trasplante Heterólogo , Ácido Zoledrónico
16.
Proc Natl Acad Sci U S A ; 106(38): 16511-6, 2009 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-19805329

RESUMEN

GPR55 is a G protein-coupled receptor recently shown to be activated by certain cannabinoids and by lysophosphatidylinositol (LPI). However, the physiological role of GPR55 remains unknown. Given the recent finding that the cannabinoid receptors CB(1) and CB(2) affect bone metabolism, we examined the role of GPR55 in bone biology. GPR55 was expressed in human and mouse osteoclasts and osteoblasts; expression was higher in human osteoclasts than in macrophage progenitors. Although the GPR55 agonists O-1602 and LPI inhibited mouse osteoclast formation in vitro, these ligands stimulated mouse and human osteoclast polarization and resorption in vitro and caused activation of Rho and ERK1/2. These stimulatory effects on osteoclast function were attenuated in osteoclasts generated from GPR55(-/-) macrophages and by the GPR55 antagonist cannabidiol (CBD). Furthermore, treatment of mice with this non-psychoactive constituent of cannabis significantly reduced bone resorption in vivo. Consistent with the ability of GPR55 to suppress osteoclast formation but stimulate osteoclast function, histomorphometric and microcomputed tomographic analysis of the long bones from male GPR55(-/-) mice revealed increased numbers of morphologically inactive osteoclasts but a significant increase in the volume and thickness of trabecular bone and the presence of unresorbed cartilage. These data reveal a role of GPR55 in bone physiology by regulating osteoclast number and function. In addition, this study also brings to light an effect of both the endogenous ligand, LPI, on osteoclasts and of the cannabis constituent, CBD, on osteoclasts and bone turnover in vivo.


Asunto(s)
Densidad Ósea , Huesos/metabolismo , Osteoclastos/metabolismo , Receptores de Cannabinoides/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Animales , Animales Recién Nacidos , Resorción Ósea/prevención & control , Huesos/citología , Cannabidiol/farmacología , Línea Celular Tumoral , Células Cultivadas , Relación Dosis-Respuesta a Droga , Femenino , Técnica del Anticuerpo Fluorescente , Humanos , Lisofosfolípidos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Osteoblastos/citología , Osteoblastos/efectos de los fármacos , Osteoblastos/metabolismo , Osteoclastos/citología , Osteoclastos/efectos de los fármacos , Osteogénesis/efectos de los fármacos , Receptores de Cannabinoides/genética , Receptores Acoplados a Proteínas G/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
Arthritis Res Ther ; 11(2): R58, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19405951

RESUMEN

INTRODUCTION: Bisphosphonates are the most widely used class of drug for inhibiting osteoclast-mediated bone loss, but their effectiveness at preventing joint destruction in rheumatoid arthritis has generally been disappointing. We examined whether the ability of bisphosphonates to induce osteoclast apoptosis and inhibit bone resorption in vitro is influenced by the cytokine receptor activator of nuclear factor-kappa B ligand (RANKL), an important mediator of inflammation-induced bone loss. METHODS: Rabbit osteoclasts were treated with the bisphosphonates clodronate or alendronate for up to 48 hours in the absence or presence of RANKL. Changes in cell morphology and induction of apoptosis were examined by scanning electron microscopy, whilst resorptive activity was determined by measuring the area of resorption cavities. Changes in the level of anti-apoptotic proteins, including Mcl-1, Bcl-2, and Bcl-x>L, were determined in rabbit osteoclasts and in cytokine-starved mouse osteoclasts by Western blotting. RESULTS: RANKL significantly attenuated the ability of both clodronate and alendronate to induce osteoclast apoptosis and inhibit bone resorption. Treatment of rabbit osteoclasts with RANKL was associated with an increase in the anti-apoptotic protein Mcl-1 but not Bcl-2. A role for Mcl-1 in osteoclast survival was suggested using osteoclasts generated from mouse bone marrow macrophages in the presence of RANKL + macrophage colony-stimulating factor (M-CSF) since cytokine deprivation of mouse osteoclasts caused a rapid loss of Mcl-1 (but not Bcl-2 or Bcl-xL), which preceded the biochemical and morphological changes associated with apoptosis. Loss of Mcl-1 from mouse osteoclasts could be prevented by factors known to promote osteoclast survival (RANKL, M-CSF, tumour necrosis factor-alpha [TNF-alpha], or lipopolysaccharide [LPS]). CONCLUSIONS: RANKL protects osteoclasts from the apoptosis-inducing and anti-resorptive effects of bisphosphonates in vitro. The ability of RANKL (and other pro-inflammatory factors such as TNF-alpha and LPS) to increase the level of Mcl-1 in osteoclasts may explain the lack of effectiveness of some bisphosphonates in preventing inflammation-induced bone loss.


Asunto(s)
Apoptosis/efectos de los fármacos , Difosfonatos/farmacología , Osteoclastos/efectos de los fármacos , Ligando RANK/metabolismo , Alendronato/farmacología , Animales , Apoptosis/fisiología , Western Blotting , Ácido Clodrónico/farmacología , Ciclina D1/metabolismo , Técnicas In Vitro , Ratones , Microscopía Electrónica de Rastreo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides , Osteoclastos/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Conejos , Proteína bcl-X/metabolismo
18.
Bone ; 44(5): 908-16, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-19442620

RESUMEN

Bisphosphonates (BPs), bone targeted drugs that disrupt osteoclast function, are routinely used to treat complications of bone metastasis. Studies in preclinical models of cancer have shown that BPs reduce skeletal tumor burden and increase survival. Similarly, we observed in the present study that administration of the Nitrogen-containing BP (N-BP), zoledronic acid (ZA) to osteolytic tumor-bearing Tax+ mice beginning at 6 months of age led to resolution of radiographic skeletal lesions. N-BPs inhibit farnesyl diphosphate (FPP) synthase, thereby inhibiting protein prenylation and causing cellular toxicity. We found that ZA decreased Tax+ tumor and B16 melanoma viability and caused the accumulation of unprenylated Rap1a proteins in vitro. However, it is presently unclear whether N-BPs exert anti-tumor effects in bone independent of inhibition of osteoclast (OC) function in vivo. Therefore, we evaluated the impact of treatment with ZA on B16 melanoma bone tumor burden in irradiated mice transplanted with splenic cells from src(-/-) mice, which have non-functioning OCs. OC-defective mice treated with ZA demonstrated a significant 88% decrease in tumor growth in bone compared to vehicle-treated OC-defective mice. These data support an osteoclast-independent role for N-BP therapy in bone metastasis.


Asunto(s)
Neoplasias Óseas/tratamiento farmacológico , Difosfonatos/farmacología , Imidazoles/farmacología , Osteoclastos/patología , Animales , Neoplasias Óseas/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Difosfonatos/administración & dosificación , Activación Enzimática/efectos de los fármacos , Geraniltranstransferasa/metabolismo , Imidazoles/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Metástasis de la Neoplasia/prevención & control , Osteoclastos/efectos de los fármacos , Prenilación de Proteína/efectos de los fármacos , Ácido Zoledrónico
19.
Br J Pharmacol ; 157(3): 427-35, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19371349

RESUMEN

BACKGROUND AND PURPOSE: Bisphosphonates (BPs) are highly effective inhibitors of bone resorption. Nitrogen-containing bisphosphonates (N-BPs), such as zoledronic acid, induce the formation of a novel ATP analogue (1-adenosin-5'-yl ester 3-(3-methylbut-3-enyl) ester triphosphoric acid; ApppI), as a consequence of the inhibition of farnesyl pyrophosphate synthase and the accumulation of isopentenyl pyrophosphate (IPP). ApppI induces apoptosis, as do comparable metabolites of non-nitrogen-containing bisphosphonates (non-N-BPs). In order to further evaluate a pharmacological role for ApppI, we obtained more detailed data on IPP/ApppI formation in vivo and in vitro. Additionally, zoledronic acid-induced ApppI formation from IPP was compared with the metabolism of clodronate (a non-N-BP) to adenosine 5'(beta,gamma-dichloromethylene) triphosphate (AppCCl2p). EXPERIMENTAL APPROACH: After giving zoledronic acid in vivo to rabbits, IPP/ApppI formation and accumulation was assessed in isolated osteoclasts. The formation of ApppI from IPP was compared with the metabolism of clodronate in MCF-7 cells in vitro. IPP/ApppI and AppCCl2p levels in cell extracts were analysed by mass spectrometry. KEY RESULTS: Isopentenyl pyrophosphate/ApppI were formed in osteoclasts in vivo, after a single, clinically relevant dose of zoledronic acid. Furthermore, exposure of MCF-7 cells in vitro to zoledronic acid at varying times and concentrations induced time- and dose-dependent accumulation of IPP/ApppI. One hour pulse treatment was sufficient to cause IPP accumulation and subsequent ApppI formation, or the metabolism of clodronate into AppCCl2p. CONCLUSIONS AND IMPLICATIONS: This study provided the first conclusive evidence that pro-apoptotic ApppI is a biologically significant molecule, and demonstrated that IPP/ApppI analysis is a sensitive tool for investigating pathways involved in BP action.


Asunto(s)
Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/biosíntesis , Conservadores de la Densidad Ósea/farmacología , Difosfonatos/farmacología , Hemiterpenos/biosíntesis , Imidazoles/farmacología , Osteoclastos/efectos de los fármacos , Adenosina Trifosfato/metabolismo , Animales , Apoptosis , Conservadores de la Densidad Ósea/administración & dosificación , Conservadores de la Densidad Ósea/metabolismo , Línea Celular Tumoral , Ácido Clodrónico/metabolismo , Difosfonatos/administración & dosificación , Humanos , Imidazoles/administración & dosificación , Técnicas In Vitro , Compuestos Organofosforados , Osteoclastos/metabolismo , Conejos , Ácido Zoledrónico
20.
J Biol Chem ; 284(11): 6861-8, 2009 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-19074143

RESUMEN

Rab geranylgeranyl transferase (RGGT) catalyzes the post-translational geranylgeranyl (GG) modification of (usually) two C-terminal cysteines in Rab GTPases. Here we studied the mechanism of the Rab geranylgeranylation reaction by bisphosphonate analogs in which one phosphonate group is replaced by a carboxylate (phosphonocarboxylate, PC). The phosphonocarboxylates used were 3-PEHPC, which was previously reported, and 2-hydroxy-3-imidazo[1,2-a]pyridin-3-yl-2-phosphonopropionic acid ((+)-3-IPEHPC), a >25-fold more potent related compound as measured by both IC50 and Ki.(+)-3-IPEHPC behaves as a mixed-type inhibitor with respect to GG pyrophosphate (GGPP) and an uncompetitive inhibitor with respect to Rab substrates. We propose that phosphonocarboxylates prevent only the second GG transfer onto Rabs based on the following evidence. First, geranylgeranylation of Rab proteins ending with a single cysteine motif such as CAAX, is not affected by the inhibitors, either in vitro or in vivo. Second, the addition of an -AAX sequence onto Rab-CC proteins protects the substrate from inhibition by the inhibitors. Third, we demonstrate directly that in the presence of (+)-3-IPEHPC, Rab-CC and Rab-CXC proteins are modified by only a single GG addition. The presence of (+)-3-IPEHPC resulted in a preference for the Rab N-terminal cysteine to be modified first, suggesting an order of cysteine geranylgeranylation in RGGT catalysis. Our results further suggest that the inhibitor binds to a site distinct from the GGPP-binding site on RGGT. We suggest that phosphonocarboxylate inhibitors bind to a GG-cysteine binding site adjacent to the active site, which is necessary to align the mono-GG-Rab for the second GG addition. These inhibitors may represent a novel therapeutic approach in Rab-mediated diseases.


Asunto(s)
Transferasas Alquil y Aril/antagonistas & inhibidores , Difosfonatos/farmacología , Inhibidores Enzimáticos/farmacología , Fosfatos de Poliisoprenilo/metabolismo , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Piridinas/farmacología , Proteínas de Unión al GTP rab/metabolismo , Transferasas Alquil y Aril/metabolismo , Secuencias de Aminoácidos/fisiología , Animales , Sitios de Unión/fisiología , Línea Celular , Perros , Humanos , Estructura Terciaria de Proteína/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA