Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Sci Signal ; 13(628)2020 04 21.
Artículo en Inglés | MEDLINE | ID: mdl-32317369

RESUMEN

The tricarboxylic acid (TCA) cycle converts the end products of glycolysis and fatty acid ß-oxidation into the reducing equivalents NADH and FADH2 Although mitochondrial matrix uptake of Ca2+ enhances ATP production, it remains unclear whether deprivation of mitochondrial TCA substrates alters mitochondrial Ca2+ flux. We investigated the effect of TCA cycle substrates on MCU-mediated mitochondrial matrix uptake of Ca2+, mitochondrial bioenergetics, and autophagic flux. Inhibition of glycolysis, mitochondrial pyruvate transport, or mitochondrial fatty acid transport triggered expression of the MCU gatekeeper MICU1 but not the MCU core subunit. Knockdown of mitochondrial pyruvate carrier (MPC) isoforms or expression of the dominant negative mutant MPC1R97W resulted in increased MICU1 protein abundance and inhibition of MCU-mediated mitochondrial matrix uptake of Ca2+ We also found that genetic ablation of MPC1 in hepatocytes and mouse embryonic fibroblasts resulted in reduced resting matrix Ca2+, likely because of increased MICU1 expression, but resulted in changes in mitochondrial morphology. TCA cycle substrate-dependent MICU1 expression was mediated by the transcription factor early growth response 1 (EGR1). Blocking mitochondrial pyruvate or fatty acid flux was linked to increased autophagy marker abundance. These studies reveal a mechanism that controls the MCU-mediated Ca2+ flux machinery and that depends on TCA cycle substrate availability. This mechanism generates a metabolic homeostatic circuit that protects cells from bioenergetic crisis and mitochondrial Ca2+ overload during periods of nutrient stress.


Asunto(s)
Canales de Calcio/metabolismo , Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Ácidos Grasos/metabolismo , Mitocondrias Hepáticas/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Proteínas Mitocondriales/metabolismo , Ácido Pirúvico/metabolismo , Animales , Transporte Biológico Activo/genética , Canales de Calcio/genética , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/genética , Células HEK293 , Células HeLa , Células Hep G2 , Humanos , Ratones Noqueados , Mitocondrias Hepáticas/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Proteínas Mitocondriales/genética
2.
Sci Signal ; 12(602)2019 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-31594854

RESUMEN

Ca2+ signals, which facilitate pluripotent changes in cell fate, reflect the balance between cation entry and export. We found that overexpression of either isoform of the Ca2+-extruding plasma membrane calcium ATPase 4 (PMCA4) pump in Jurkat T cells unexpectedly increased activation of the Ca2+-dependent transcription factor nuclear factor of activated T cells (NFAT). Coexpression of the endoplasmic reticulum-resident Ca2+ sensor stromal interaction molecule 1 (STIM1) with the PMCA4b splice variant further enhanced NFAT activity; however, coexpression with PMCA4a depressed NFAT. No PMCA4 splice variant dependence in STIM1 association was observed, whereas partner of STIM1 (POST) preferentially associated with PMCA4b over PMCA4a, which enhanced, rather than inhibited, PMCA4 function. A comparison of global and near-membrane cytosolic Ca2+ abundances during store-operated Ca2+ entry revealed that PMCA4 markedly depressed near-membrane Ca2+ concentrations, particularly when PMCA4b was coexpressed with STIM1. PMCA4b closely associated with both POST and the store-operated Ca2+ channel Orai1. Furthermore, POST knockdown increased the near-membrane Ca2+ concentration, inhibiting the global cytosolic Ca2+ increase. These observations reveal an unexpected role for POST in coupling PMCA4 to Orai1 to promote Ca2+ entry during T cell activation through Ca2+ disinhibition.


Asunto(s)
Señalización del Calcio , Calcio/metabolismo , Membrana Celular/metabolismo , Factores de Transcripción NFATC/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/metabolismo , Retículo Endoplásmico/metabolismo , Células HEK293 , Humanos , Células Jurkat , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Microscopía Fluorescente , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , ATPasas Transportadoras de Calcio de la Membrana Plasmática/genética , Interferencia de ARN , Molécula de Interacción Estromal 1/genética , Molécula de Interacción Estromal 1/metabolismo
3.
Cell Rep ; 26(13): 3709-3725.e7, 2019 03 26.
Artículo en Inglés | MEDLINE | ID: mdl-30917323

RESUMEN

Mitochondrial Ca2+ uniporter (MCU)-mediated Ca2+ uptake promotes the buildup of reducing equivalents that fuel oxidative phosphorylation for cellular metabolism. Although MCU modulates mitochondrial bioenergetics, its function in energy homeostasis in vivo remains elusive. Here we demonstrate that deletion of the Mcu gene in mouse liver (MCUΔhep) and in Danio rerio by CRISPR/Cas9 inhibits mitochondrial Ca2+ (mCa2+) uptake, delays cytosolic Ca2+ (cCa2+) clearance, reduces oxidative phosphorylation, and leads to increased lipid accumulation. Elevated hepatic lipids in MCUΔhep were a direct result of extramitochondrial Ca2+-dependent protein phosphatase-4 (PP4) activity, which dephosphorylates AMPK. Loss of AMPK recapitulates hepatic lipid accumulation without changes in MCU-mediated Ca2+ uptake. Furthermore, reconstitution of active AMPK, or PP4 knockdown, enhances lipid clearance in MCUΔhep hepatocytes. Conversely, gain-of-function MCU promotes rapid mCa2+ uptake, decreases PP4 levels, and reduces hepatic lipid accumulation. Thus, our work uncovers an MCU/PP4/AMPK molecular cascade that links Ca2+ dynamics to hepatic lipid metabolism.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Hepatocitos/metabolismo , Metabolismo de los Lípidos , Proteínas Mitocondriales/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Canales de Calcio/genética , Células Cultivadas , Femenino , Células Hep G2 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias Hepáticas/metabolismo , Proteínas Mitocondriales/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteínas Quinasas/metabolismo , Pez Cebra
4.
ACS Cent Sci ; 5(1): 153-166, 2019 Jan 23.
Artículo en Inglés | MEDLINE | ID: mdl-30693334

RESUMEN

Mitochondrial Ca2+ (mCa2+) uptake mediated by the mitochondrial calcium uniporter (MCU) plays a critical role in signal transduction, bioenergetics, and cell death, and its dysregulation is linked to several human diseases. In this study, we report a new ruthenium complex Ru265 that is cell-permeable, minimally toxic, and highly potent with respect to MCU inhibition. Cells treated with Ru265 show inhibited MCU activity without any effect on cytosolic Ca2+ dynamics and mitochondrial membrane potential (ΔΨm). Dose-dependent studies reveal that Ru265 is more potent than the currently employed MCU inhibitor Ru360. Site-directed mutagenesis of Cys97 in the N-terminal domain of human MCU ablates the inhibitory activity of Ru265, suggesting that this matrix-residing domain is its target site. Additionally, Ru265 prevented hypoxia/reoxygenation injury and subsequent mitochondrial dysfunction, demonstrating that this new inhibitor is a valuable tool for studying the functional role of the MCU in intact biological models.

5.
Nat Commun ; 9(1): 3449, 2018 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-30158529

RESUMEN

Although many factors contribute to cellular differentiation, the role of mitochondria Ca2+ dynamics during development remains unexplored. Because mammalian embryonic epiblasts reside in a hypoxic environment, we intended to understand whether mCa2+ and its transport machineries are regulated during hypoxia. Tissues from multiple organs of developing mouse embryo evidenced a suppression of MICU1 expression with nominal changes on other MCU complex components. As surrogate models, we here utilized human embryonic stem cells (hESCs)/induced pluripotent stem cells (hiPSCs) and primary neonatal myocytes to delineate the mechanisms that control mCa2+ and bioenergetics during development. Analysis of MICU1 expression in hESCs/hiPSCs showed low abundance of MICU1 due to its direct repression by Foxd1. Experimentally, restoration of MICU1 established the periodic cCa2+ oscillations and promoted cellular differentiation and maturation. These findings establish a role of mCa2+ dynamics in regulation of cellular differentiation and reveal a molecular mechanism underlying this contribution through differential regulation of MICU1.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Proteínas de Transporte de Catión/metabolismo , Factores de Transcripción Forkhead/metabolismo , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/metabolismo , Animales , Western Blotting , Calcio/metabolismo , Proteínas de Unión al Calcio/genética , Proteínas de Transporte de Catión/genética , Diferenciación Celular/genética , Diferenciación Celular/fisiología , Línea Celular , Células Cultivadas , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Factores de Transcripción Forkhead/genética , Humanos , Células Madre Pluripotentes Inducidas/citología , Células Madre Pluripotentes Inducidas/metabolismo , Ratones , Ratones Endogámicos C57BL , Mitocondrias/genética , Proteínas de Transporte de Membrana Mitocondrial/genética , Miocitos Cardíacos/citología , Miocitos Cardíacos/metabolismo , Interferencia de ARN
6.
Cell Calcium ; 74: 86-93, 2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-29980025

RESUMEN

Ca2+ flux across the inner mitochondrial membrane (IMM) regulates cellular bioenergetics, intra-cellular cytoplasmic Ca2+ signals, and various cell death pathways. Ca2+ entry into the mitochondria occurs due to the highly negative membrane potential (ΔΨm) through a selective inward rectifying MCU channel. In addition to being regulated by various mitochondrial matrix resident proteins such as MICUs, MCUb, MCUR1 and EMRE, the channel is transcriptionally regulated by upstream Ca2+ cascade, post transnational modification and by divalent cations. The mode of regulation either inhibits or enhances MCU channel activity and thus regulates mitochondrial metabolism and cell fate.


Asunto(s)
Canales de Calcio/metabolismo , Calcio/metabolismo , Fenómenos Fisiológicos Celulares/fisiología , Membranas Mitocondriales/metabolismo , Animales , Canales de Calcio/química , Muerte Celular/fisiología , Citosol/metabolismo , Humanos , Potencial de la Membrana Mitocondrial/fisiología , Mitocondrias/metabolismo , Proteínas de Transporte de Membrana Mitocondrial/química , Proteínas de Transporte de Membrana Mitocondrial/metabolismo
7.
Cell Rep ; 23(4): 1005-1019, 2018 04 24.
Artículo en Inglés | MEDLINE | ID: mdl-29694881

RESUMEN

Mitochondria shape cytosolic calcium ([Ca2+]c) transients and utilize the mitochondrial Ca2+ ([Ca2+]m) in exchange for bioenergetics output. Conversely, dysregulated [Ca2+]c causes [Ca2+]m overload and induces permeability transition pore and cell death. Ablation of MCU-mediated Ca2+ uptake exhibited elevated [Ca2+]c and failed to prevent stress-induced cell death. The mechanisms for these effects remain elusive. Here, we report that mitochondria undergo a cytosolic Ca2+-induced shape change that is distinct from mitochondrial fission and swelling. [Ca2+]c elevation, but not MCU-mediated Ca2+ uptake, appears to be essential for the process we term mitochondrial shape transition (MiST). MiST is mediated by the mitochondrial protein Miro1 through its EF-hand domain 1 in multiple cell types. Moreover, Ca2+-dependent disruption of Miro1/KIF5B/tubulin complex is determined by Miro1 EF1 domain. Functionally, Miro1-dependent MiST is essential for autophagy/mitophagy that is attenuated in Miro1 EF1 mutants. Thus, Miro1 is a cytosolic Ca2+ sensor that decodes metazoan Ca2+ signals as MiST.


Asunto(s)
Calcio/metabolismo , Mitocondrias/metabolismo , Dinámicas Mitocondriales , Receptores Acoplados a Proteínas G/metabolismo , Estrés Fisiológico , Proteínas de Unión al GTP rho/metabolismo , Animales , Células HeLa , Humanos , Ratones , Ratones Mutantes , Mitocondrias/genética , Receptores Acoplados a Proteínas G/genética , Proteínas de Unión al GTP rho/genética
8.
Mol Cell ; 65(6): 1014-1028.e7, 2017 Mar 16.
Artículo en Inglés | MEDLINE | ID: mdl-28262504

RESUMEN

Ca2+ dynamics and oxidative signaling are fundamental mechanisms for mitochondrial bioenergetics and cell function. The MCU complex is the major pathway by which these signals are integrated in mitochondria. Whether and how these coactive elements interact with MCU have not been established. As an approach toward understanding the regulation of MCU channel by oxidative milieu, we adapted inflammatory and hypoxia models. We identified the conserved cysteine 97 (Cys-97) to be the only reactive thiol in human MCU that undergoes S-glutathionylation. Furthermore, biochemical, structural, and superresolution imaging analysis revealed that MCU oxidation promotes MCU higher order oligomer formation. Both oxidation and mutation of MCU Cys-97 exhibited persistent MCU channel activity with higher [Ca2+]m uptake rate, elevated mROS, and enhanced [Ca2+]m overload-induced cell death. In contrast, these effects were largely independent of MCU interaction with its regulators. These findings reveal a distinct functional role for Cys-97 in ROS sensing and regulation of MCU activity.


Asunto(s)
Canales de Calcio/metabolismo , Señalización del Calcio , Calcio/metabolismo , Células Endoteliales/metabolismo , Activación del Canal Iónico , Mitocondrias/metabolismo , Membranas Mitocondriales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Células COS , Canales de Calcio/química , Canales de Calcio/genética , Señalización del Calcio/efectos de los fármacos , Muerte Celular , Hipoxia de la Célula , Chlorocebus aethiops , Cisteína , Células Endoteliales/efectos de los fármacos , Células Endoteliales/patología , Metabolismo Energético , Glutatión/metabolismo , Células HEK293 , Células HeLa , Humanos , Activación del Canal Iónico/efectos de los fármacos , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/efectos de los fármacos , Mitocondrias/patología , Membranas Mitocondriales/efectos de los fármacos , Membranas Mitocondriales/patología , Mutación , Oxidación-Reducción , Multimerización de Proteína , Procesamiento Proteico-Postraduccional , Estructura Cuaternaria de Proteína , Relación Estructura-Actividad , Trombina/farmacología , Factores de Tiempo , Transfección
9.
Cell Rep ; 15(8): 1673-85, 2016 05 24.
Artículo en Inglés | MEDLINE | ID: mdl-27184846

RESUMEN

Mitochondrial Ca(2+) Uniporter (MCU)-dependent mitochondrial Ca(2+) uptake is the primary mechanism for increasing matrix Ca(2+) in most cell types. However, a limited understanding of the MCU complex assembly impedes the comprehension of the precise mechanisms underlying MCU activity. Here, we report that mouse cardiomyocytes and endothelial cells lacking MCU regulator 1 (MCUR1) have severely impaired [Ca(2+)]m uptake and IMCU current. MCUR1 binds to MCU and EMRE and function as a scaffold factor. Our protein binding analyses identified the minimal, highly conserved regions of coiled-coil domain of both MCU and MCUR1 that are necessary for heterooligomeric complex formation. Loss of MCUR1 perturbed MCU heterooligomeric complex and functions as a scaffold factor for the assembly of MCU complex. Vascular endothelial deletion of MCU and MCUR1 impaired mitochondrial bioenergetics, cell proliferation, and migration but elicited autophagy. These studies establish the existence of a MCU complex that assembles at the mitochondrial integral membrane and regulates Ca(2+)-dependent mitochondrial metabolism.


Asunto(s)
Canales de Calcio/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Autofagia , Calcio/metabolismo , Canales de Calcio/química , Movimiento Celular , Células Endoteliales/metabolismo , Eliminación de Gen , Células HEK293 , Células HeLa , Corazón/fisiología , Humanos , Ratones Noqueados , Proteínas Mitocondriales/química , Neovascularización Fisiológica , Unión Proteica , Dominios Proteicos
10.
J Bone Miner Res ; 31(6): 1225-34, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-26763740

RESUMEN

Sclerostin is a potent inhibitor of osteoblastogenesis. Interestingly, newly diagnosed multiple myeloma (MM) patients have high levels of circulating sclerostin that correlate with disease stage and fractures. However, the source and impact of sclerostin in MM remains to be defined. Our goal was to determine the role of sclerostin in the biology of MM and its bone microenvironment as well as investigate the effect of targeting sclerostin with a neutralizing antibody (scl-Ab) in MM bone disease. Here we confirm increased sclerostin levels in MM compared with precursor disease states like monoclonal gammopathy of undetermined significance (MGUS) and smoldering MM. Furthermore, we found that a humanized MM xenograft mouse model bearing human MM cells (NOD-SCID.CB17 male mice injected intravenously with 2.5 million of MM1.S-Luc-GFP cells) demonstrated significantly higher concentrations of mouse-derived sclerostin, suggesting a microenvironmental source of sclerostin. Associated with the increased sclerostin levels, activated ß-catenin expression levels were lower than normal in MM mouse bone marrow. Importantly, a high-affinity grade scl-Ab reversed osteolytic bone disease in this animal model. Because scl-Ab did not demonstrate significant in vitro anti-MM activity, we combined it with the proteasome inhibitor carfilzomib. Our data demonstrated that this combination therapy significantly inhibited tumor burden and improved bone disease in our in vivo MM mouse model. In agreement with our in vivo data, sclerostin expression was noted in marrow stromal cells and osteoblasts of MM patient bone marrow samples. Moreover, MM cells stimulated sclerostin expression in immature osteoblasts while inhibiting osteoblast differentiation in vitro. This was in part regulated by Dkk-1 secreted by MM cells and is a potential mechanism contributing to the osteoblast dysfunction noted in MM. Our data confirm the role of sclerostin as a potential therapeutic target in MM bone disease and provides the rationale for studying scl-Ab combined with proteasome inhibitors in MM. © 2016 American Society for Bone and Mineral Research.


Asunto(s)
Enfermedades Óseas/metabolismo , Glicoproteínas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Mieloma Múltiple/metabolismo , Proteínas de Neoplasias/metabolismo , Osteoblastos/metabolismo , Microambiente Tumoral , Proteínas Adaptadoras Transductoras de Señales , Animales , Enfermedades Óseas/genética , Enfermedades Óseas/patología , Femenino , Glicoproteínas/genética , Xenoinjertos , Humanos , Péptidos y Proteínas de Señalización Intercelular/genética , Masculino , Ratones , Ratones Endogámicos NOD , Ratones SCID , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Proteínas de Neoplasias/genética , Trasplante de Neoplasias , Osteoblastos/patología
11.
Mol Cell ; 60(1): 47-62, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26387735

RESUMEN

Mitochondrial permeability transition is a phenomenon in which the mitochondrial permeability transition pore (PTP) abruptly opens, resulting in mitochondrial membrane potential (ΔΨm) dissipation, loss of ATP production, and cell death. Several genetic candidates have been proposed to form the PTP complex, however, the core component is unknown. We identified a necessary and conserved role for spastic paraplegia 7 (SPG7) in Ca(2+)- and ROS-induced PTP opening using RNAi-based screening. Loss of SPG7 resulted in higher mitochondrial Ca(2+) retention, similar to cyclophilin D (CypD, PPIF) knockdown with sustained ΔΨm during both Ca(2+) and ROS stress. Biochemical analyses revealed that the PTP is a heterooligomeric complex composed of VDAC, SPG7, and CypD. Silencing or disruption of SPG7-CypD binding prevented Ca(2+)- and ROS-induced ΔΨm depolarization and cell death. This study identifies an ubiquitously expressed IMM integral protein, SPG7, as a core component of the PTP at the OMM and IMM contact site.


Asunto(s)
Ciclofilinas/metabolismo , Metaloendopeptidasas/genética , Metaloendopeptidasas/metabolismo , Mitocondrias/metabolismo , Canal Aniónico 1 Dependiente del Voltaje/metabolismo , ATPasas Asociadas con Actividades Celulares Diversas , Sitios de Unión , Calcio/metabolismo , Muerte Celular , Ciclofilinas/química , Células HEK293 , Células HeLa , Humanos , Potencial de la Membrana Mitocondrial , Metaloendopeptidasas/química , Membranas Mitocondriales/metabolismo , Interferencia de ARN , Especies Reactivas de Oxígeno/metabolismo
12.
Br J Haematol ; 169(3): 423-34, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25709080

RESUMEN

Proteasome inhibition induces the accumulation of aggregated misfolded/ubiquitinated proteins in the aggresome; conversely, histone deacetylase 6 (HDAC6) inhibition blocks aggresome formation. Although this rationale has been the basis of proteasome inhibitor (PI) and HDAC6 inhibitor combination studies, the role of disruption of aggresome formation by HDAC6 inhibition has not yet been studied in multiple myeloma (MM). The present study aimed to evaluate the impact of carfilzomib (CFZ) in combination with a selective HDAC6 inhibitor (ricolinostat) in MM cells with respect to the aggresome-proteolysis pathway. We observed that combination treatment of CFZ with ricolinostat triggered synergistic anti-MM effects, even in bortezomib-resistant cells. Immunofluorescent staining showed that CFZ increased the accumulation of ubiquitinated proteins and protein aggregates in the cytoplasm, as well as the engulfment of aggregated ubiquitinated proteins by autophagosomes, which was blocked by ricolinostat. Electron microscopy imaging showed increased autophagy triggered by CFZ, which was inhibited by the addition of ACY-1215. Finally, an in vivo mouse xenograft study confirmed a decrease in tumour volume, associated with apoptosis, following treatment with CFZ in combination with ricolinostat. Our results suggest that ricolinostat inhibits aggresome formation, caused by CFZ-induced inhibition of the proteasome pathway, resulting in enhanced apoptosis in MM cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Ácidos Hidroxámicos/farmacología , Mieloma Múltiple/metabolismo , Oligopéptidos/farmacología , Pirimidinas/farmacología , Animales , Antineoplásicos/farmacología , Autofagia/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Sinergismo Farmacológico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Femenino , Xenoinjertos , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Ratones , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Fagosomas/metabolismo , Inhibidores de Proteasoma/farmacología
13.
J Bone Miner Res ; 30(3): 465-70, 2015 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-25407518

RESUMEN

Decorin is a small, leucine-rich proteoglycan found in the extracellular matrix of various connective tissues with potential effective tumor suppressive properties. Recent data suggest low levels of decorin in multiple myeloma (MM) patients compared to healthy volunteers, as well as in patients with osteolytic bone lesions compared to non-osteolytic lesions. In the present report, we investigated the role of decorin in the MM microenvironment or niche. Our data suggests that decorin is produced by osteoblasts (OBs) but not by MM cells. Furthermore, MM cells decrease OB-induced decorin secretion and this effect is mediated by CCL3. Importantly, neutralizing CCL3 from MM cells restores decorin levels in OBs as does proteasome inhibitors such as carfilzomib. These findings indicate that decorin may indirectly act as an antagonist to MM cell survival and that the interplay between MM and decorin may be an important target to explore in manipulating the tumor niche to inhibit tumorigenesis.


Asunto(s)
Médula Ósea/patología , Decorina/fisiología , Mieloma Múltiple/patología , Microambiente Tumoral , Animales , Línea Celular Tumoral , Humanos , Ratones
14.
Mol Cancer Ther ; 13(11): 2489-500, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25172964

RESUMEN

Despite promising preclinical results with mTOR kinase inhibitors in multiple myeloma, resistance to these drugs may arise via feedback activation loops. This concern is especially true for insulin-like growth factor 1 receptor (IGF1R), because IGF1R signaling is downregulated by multiple AKT and mTOR feedback mechanisms. We have tested this hypothesis in multiple myeloma using the novel selective mTOR kinase inhibitor AZD8055. We evaluated p-mTOR S(2481) as the readout for mTORC2/Akt activity in multiple myeloma cells in the context of mTOR inhibition via AZD8055 or rapamycin. We next validated AZD8055 inhibition of mTORC1 and mTORC2 functions in multiple myeloma cells alone or in culture with bone marrow stroma cells and growth factors. Unlike rapamycin, AZD8055 resulted in apoptosis of multiple myeloma cells. AZD8055 treatment, however, induced upregulation of IGF1R phosphorylation in p-Akt S(473)-expressing multiple myeloma cell lines. Furthermore, exposure of AZD8055-treated cells to IGF1 induced p-Akt S(473) and rescued multiple myeloma cells from apoptosis despite mTOR kinase inhibition and TORC2/Akt blockage. The addition of blocking IGF1R antibody resulted in reversing this effect and increased AZD8055-induced apoptosis. Our study suggests that combination treatment with AZD8055 and IGF1R-blocking agents is a promising strategy in multiple myeloma with potential IGF1R/Akt signaling-mediated survival.


Asunto(s)
Morfolinas/farmacología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/enzimología , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/metabolismo , Animales , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Humanos , Ratones , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Clin Cancer Res ; 20(15): 3955-61, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24958808

RESUMEN

BACKGROUND: Patients with multiple myeloma may be susceptible to osteonecrosis of the jaw (ONJ) and stress fractures due to long-term aminobisphosphonate (aBP) therapy. However, it is unknown whether urinary N-telopeptide (NTX) or other bone biomarkers are predictive of skeletal-related events (SRE) or the impact of cessation of aBP therapy on bone remodeling. METHODS: We studied markers of bone turnover over a 6-month period after a single dose of zoledronic acid in 29 patients with multiple myeloma in remission who previously received 8 to 12 doses of pamidronate or zoledronate (NCT00577642). Our primary objective was to determine the duration of time urinary NTX levels remain suppressed after a single dose of zoledronate. A secondary objective was to identify and correlate other markers of bone remodeling with NTX changes. Thirty cytokines, based on their possible role in bone remodeling, were tested using cytokine arrays. Candidates were confirmed by ELISA. RESULTS: All patients had continued suppression of NTX levels, except 1 patient who had an increase in NTX levels associated with an SRE. GDF-15 and decorin were found to decrease, whereas bone-specific alkaline phosphatase (BSALP) increased. Although not significant in aggregate, osteopontin and osteoprotegerin levels increased in at least half of the patients. CONCLUSION: Our data show that NTX levels continue to be suppressed after aBP therapy, and suggest that suppressed NTX levels may be predictive of freedom from SRE in this patient population. Furthermore, osteoblast suppression by aBP may be reversible in myeloma. These data provide the basis for less frequent dosing of aBPs.


Asunto(s)
Biomarcadores de Tumor/orina , Conservadores de la Densidad Ósea/administración & dosificación , Remodelación Ósea/efectos de los fármacos , Difosfonatos/administración & dosificación , Imidazoles/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Osteonecrosis/diagnóstico , Anciano , Anciano de 80 o más Años , Remodelación Ósea/fisiología , Resorción Ósea/inducido químicamente , Resorción Ósea/diagnóstico , Resorción Ósea/orina , Colágeno Tipo I/orina , Citocinas/orina , Ensayo de Inmunoadsorción Enzimática , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Mieloma Múltiple/metabolismo , Estadificación de Neoplasias , Osteonecrosis/inducido químicamente , Osteonecrosis/orina , Pamidronato , Péptidos/orina , Pronóstico , Ácido Zoledrónico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...