Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Blood ; 132(10): 1050-1063, 2018 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-29997223

RESUMEN

The microRNA (miRNA) cluster miR-17-92 is oncogenic and represents a valuable therapeutic target in c-MYC (MYC)-driven malignancies. Here, we developed novel LNA gapmeR antisense oligonucleotides (ASOs) to induce ribonuclease H-mediated degradation of MIR17HG primary transcripts and consequently prevent biogenesis of miR-17-92 miRNAs (miR-17-92s). The leading LNA ASO, MIR17PTi, impaired proliferation of several cancer cell lines (n = 48) established from both solid and hematologic tumors by on-target antisense activity, more effectively as compared with miR-17-92 inhibitors. By focusing on multiple myeloma (MM), we found that MIR17PTi triggers apoptosis via impairment of homeostatic MYC/miR-17-92 feed-forward loops (FFLs) in patient-derived MM cells and induces MYC-dependent synthetic lethality. We show that alteration of a BIM-centered FFL is instrumental for MIR17PTi to induce cytotoxicity in MM cells. MIR17PTi exerts strong in vivo antitumor activity in nonobese diabetic severe combined immunodeficient mice bearing clinically relevant models of MM, with advantageous safety and pharmacokinetic profiles in nonhuman primates. Altogether, MIR17PTi is a novel pharmacological tool to be tested in early-phase clinical trials against MM and other MYC-driven malignancies.


Asunto(s)
Apoptosis/efectos de los fármacos , MicroARNs/antagonistas & inhibidores , Mieloma Múltiple/tratamiento farmacológico , Oligonucleótidos/farmacología , ARN Neoplásico/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Endogámicos NOD , Ratones SCID , MicroARNs/genética , MicroARNs/metabolismo , Mieloma Múltiple/genética , Mieloma Múltiple/metabolismo , Oligonucleótidos/genética , ARN Largo no Codificante , ARN Neoplásico/genética , ARN Neoplásico/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
2.
J Cell Physiol ; 229(12): 2106-16, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24819167

RESUMEN

The analysis of deregulated microRNAs (miRNAs) is emerging as a novel approach to disclose the regulation of tumor suppressor or tumor promoting pathways in tumor cells. Targeting aberrantly expressed miRNAs is therefore a promising strategy for cancer treatment. By miRNA profiling of primary plasma cells from multiple myeloma (MM) patients, we previously reported increased miR-125a-5p levels associated to specific molecular subgroups. On these premises, we aimed at investigating the biological effects triggered by miR-125a-5p modulation in MM cells. Expression of p53 pathway-related genes was down-regulated in MM cells transfected with miR-125a-5p mimics. Luciferase reporter assays confirmed specific p53 targeting at 3'UTR level by miR-125a-5p mimics. Interestingly, bone marrow stromal cells (BMSCs) affected the miR-125a-5p/p53 axis, since adhesion of MM cells to BMSCs strongly up-regulated miR-125a-5p levels, while reduced p53 expression. Moreover, ectopic miR-125a-5p reduced, while miR-125-5p inhibitors promoted, the expression of tumor suppressor miR-192 and miR-194, transcriptionally regulated by p53. Lentiviral-mediated stable inhibition of miR-125a-5p expression in wild-type p53 MM cells dampened cell growth, increased apoptosis and reduced cell migration. Importantly, inhibition of in vitro MM cell proliferation and migration was also achieved by synthetic miR-125a-5p inhibitors and was potentiated by the co-expression of miR-192 or miR-194. Taken together, our data indicate that miR-125a-5p antagonism results in the activation of p53 pathway in MM cells, underlying the crucial role of this miRNA in the biopathology of MM and providing the molecular rationale for the combinatory use of miR-125a inhibitors and miR-192 or miR-194 mimics for MM treatment.


Asunto(s)
Redes Reguladoras de Genes , MicroARNs/genética , Mieloma Múltiple/genética , Proteína p53 Supresora de Tumor/genética , Regiones no Traducidas 3'/genética , Apoptosis/genética , Movimiento Celular/genética , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Genes Supresores de Tumor , Humanos , MicroARNs/antagonistas & inhibidores , Mieloma Múltiple/patología , Proteína p53 Supresora de Tumor/metabolismo
3.
J Cell Physiol ; 228(7): 1506-15, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23254643

RESUMEN

Skeletal homeostasis relies upon a fine tuning of osteoclast (OCL)-mediated bone resorption and osteoblast (OBL)-dependent bone formation. This balance is unsettled by multiple myeloma (MM) cells, which impair OBL function and stimulate OCLs to generate lytic lesions. Emerging experimental evidence is disclosing a key regulatory role of microRNAs (miRNAs) in the regulation of bone homeostasis suggesting the miRNA network as potential novel target for the treatment of MM-related bone disease (BD). Here, we report that miR-29b expression decreases progressively during human OCL differentiation in vitro. We found that lentiviral transduction of miR-29b into OCLs, even in the presence of MM cells, significantly impairs tartrate acid phosphatase (TRAcP) expression, lacunae generation, and collagen degradation, which are relevant hallmarks of OCL activity. Accordingly, expression of cathepsin K and metalloproteinase 9 (MMP9) as well as actin ring rearrangement were impaired in the presence of miR-29b. Moreover, we found that canonical targets C-FOS and metalloproteinase 2 are suppressed by constitutive miR-29b expression which also downregulated the master OCL transcription factor, NAFTc-1. Overall, these data indicate that enforced expression of miR-29b impairs OCL differentiation and overcomes OCL activation triggered by MM cells, providing a rationale for miR-29b-based treatment of MM-related BD.


Asunto(s)
MicroARNs/genética , MicroARNs/metabolismo , Mieloma Múltiple/terapia , Osteoclastos/metabolismo , Osteoclastos/patología , Osteólisis/terapia , Fosfatasa Ácida/metabolismo , Actinas/metabolismo , Resorción Ósea/etiología , Resorción Ósea/genética , Resorción Ósea/prevención & control , Catepsina K/metabolismo , Diferenciación Celular , Línea Celular Tumoral , Células Cultivadas , Colágeno Tipo I/metabolismo , Expresión Génica , Genes fos , Humanos , Isoenzimas/metabolismo , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , MicroARNs/uso terapéutico , Mieloma Múltiple/complicaciones , Mieloma Múltiple/metabolismo , Factores de Transcripción NFATC/metabolismo , Osteoblastos/metabolismo , Osteoblastos/patología , Osteólisis/etiología , Osteólisis/metabolismo , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Fosfatasa Ácida Tartratorresistente
5.
Clin Cancer Res ; 22(5): 1222-33, 2016 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-26527748

RESUMEN

PURPOSE: The onset of drug resistance is a major cause of treatment failure in multiple myeloma. Although increasing evidence is defining the role of miRNAs in mediating drug resistance, their potential activity as drug-sensitizing agents has not yet been investigated in multiple myeloma. EXPERIMENTAL DESIGN: Here we studied the potential utility of miR-221/222 inhibition in sensitizing refractory multiple myeloma cells to melphalan. RESULTS: miR-221/222 expression inversely correlated with melphalan sensitivity of multiple myeloma cells. Inhibition of miR-221/222 overcame melphalan resistance and triggered apoptosis of multiple myeloma cells in vitro, in the presence or absence of human bone marrow (BM) stromal cells. Decreased multiple myeloma cell growth induced by inhibition of miR-221/222 plus melphalan was associated with a marked upregulation of pro-apoptotic BBC3/PUMA protein, a miR-221/222 target, as well as with modulation of drug influx-efflux transporters SLC7A5/LAT1 and the ABC transporter ABCC1/MRP1. Finally, in vivo treatment of SCID/NOD mice bearing human melphalan-refractory multiple myeloma xenografts with systemic locked nucleic acid (LNA) inhibitors of miR-221 (LNA-i-miR-221) plus melphalan overcame drug resistance, evidenced by growth inhibition with significant antitumor effects together with modulation of PUMA and ABCC1 in tumors retrieved from treated mice. CONCLUSIONS: Taken together, our findings provide the proof of concept that LNA-i-miR-221 can reverse melphalan resistance in preclinical models of multiple myeloma, providing the framework for clinical trials to overcome drug resistance, and improve patient outcome in multiple myeloma.


Asunto(s)
Melfalán/administración & dosificación , MicroARNs/administración & dosificación , MicroARNs/biosíntesis , Mieloma Múltiple/tratamiento farmacológico , Animales , Apoptosis/efectos de los fármacos , Proteínas Reguladoras de la Apoptosis/biosíntesis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , MicroARNs/antagonistas & inhibidores , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/biosíntesis , Mieloma Múltiple/genética , Mieloma Múltiple/patología , Proteínas Proto-Oncogénicas/biosíntesis , Ensayos Antitumor por Modelo de Xenoinjerto
6.
Mol Cancer Ther ; 15(6): 1364-75, 2016 06.
Artículo en Inglés | MEDLINE | ID: mdl-27196750

RESUMEN

Epigenetic abnormalities are common in hematologic malignancies, including multiple myeloma, and their effects can be efficiently counteracted by a class of tumor suppressor miRNAs, named epi-miRNAs. Given the oncogenic role of histone deacetylases (HDAC) in multiple myeloma, we investigated whether their activity could be antagonized by miR-29b, a well-established epi-miRNA. We demonstrated here that miR-29b specifically targets HDAC4 and highlighted that both molecules are involved in a functional loop. In fact, silencing of HDAC4 by shRNAs inhibited multiple myeloma cell survival and migration and triggered apoptosis and autophagy, along with the induction of miR-29b expression by promoter hyperacetylation, leading to the downregulation of prosurvival miR-29b targets (SP1, MCL-1). Moreover, treatment with the pan-HDAC inhibitor SAHA upregulated miR-29b, overcoming the negative control exerted by HDAC4. Importantly, overexpression or inhibition of miR-29b, respectively, potentiated or antagonized SAHA activity on multiple myeloma cells, as also shown in vivo by a strong synergism between miR-29b synthetic mimics and SAHA in a murine xenograft model of human multiple myeloma. Altogether, our results shed light on a novel epigenetic circuitry regulating multiple myeloma cell growth and survival and open new avenues for miR-29b-based epi-therapeutic approaches in the treatment of this malignancy. Mol Cancer Ther; 15(6); 1364-75. ©2016 AACR.


Asunto(s)
Histona Desacetilasas/genética , Ácidos Hidroxámicos/administración & dosificación , MicroARNs/genética , Mieloma Múltiple/genética , ARN Interferente Pequeño/farmacología , Proteínas Represoras/genética , Acetilación/efectos de los fármacos , Animales , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Sinergismo Farmacológico , Epigénesis Genética/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Vectores Genéticos/administración & dosificación , Humanos , Ácidos Hidroxámicos/farmacología , Ratones , MicroARNs/administración & dosificación , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Trasplante de Neoplasias , Regiones Promotoras Genéticas , Proteínas Represoras/antagonistas & inhibidores , Vorinostat
7.
Oncotarget ; 6(15): 12837-61, 2015 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-25968566

RESUMEN

A wealth of studies has highlighted the biological complexity of hematologic malignancies and the role of dysregulated signal transduction pathways. Along with the crucial role of genetic abnormalities, epigenetic aberrations are nowadays emerging as relevant players in cancer development, and significant research efforts are currently focusing on mechanisms by which histone post-translational modifications, DNA methylation and noncoding RNAs contribute to the pathobiology of cancer. As a consequence, these studies have provided the rationale for the development of epigenetic drugs, such as histone deacetylase inhibitors and demethylating compounds, some of which are currently in advanced phase of pre-clinical investigation or in clinical trials. In addition, a more recent body of evidence indicates that microRNAs (miRNAs) might target effectors of the epigenetic machinery, which are aberrantly expressed or active in cancers, thus reverting those epigenetic abnormalities driving tumor initiation and progression. This review will focus on the broad epigenetic activity triggered by members of the miR-29 family, which underlines the potential of miR-29s as candidate epi-therapeutics for the treatment of hematologic malignancies.


Asunto(s)
Neoplasias Hematológicas/genética , Neoplasias Hematológicas/terapia , MicroARNs/genética , Animales , Metilación de ADN , Epigénesis Genética , Humanos , Terapia Molecular Dirigida
8.
Oncotarget ; 6(29): 27343-58, 2015 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-26160841

RESUMEN

miR-21 is an oncogenic microRNA (miRNA) with an emerging role as therapeutic target in human malignancies, including multiple myeloma (MM). Here we investigated whether miR-21 is involved in MM-related bone disease (BD). We found that miR-21 expression is dramatically enhanced, while osteoprotegerin (OPG) is strongly reduced, in bone marrow stromal cells (BMSCs) adherent to MM cells. On this basis, we validated the 3'UTR of OPG mRNA as miR-21 target. Constitutive miR-21 inhibition in lentiviral-transduced BMSCs adherent to MM cells restored OPG expression and secretion. Interestingly, miR-21 inhibition reduced RANKL production by BMSCs. Overexpression of protein inhibitor of activated STAT3 (PIAS3), which is a direct and validated target of miR-21, antagonized STAT3-mediated RANKL gene activation. Finally, we demonstrate that constitutive expression of miR-21 inhibitors in BMSCs restores RANKL/OPG balance and dramatically impairs the resorbing activity of mature osteoclasts. Taken together, our data provide proof-of-concept that miR-21 overexpression within MM-microenviroment plays a crucial role in bone resorption/apposition balance, supporting the design of innovative miR-21 inhibition-based strategies for MM-related BD.


Asunto(s)
MicroARNs/antagonistas & inhibidores , Mieloma Múltiple/metabolismo , Osteoclastos/citología , Osteoprotegerina/metabolismo , Ligando RANK/metabolismo , Células del Estroma/citología , Regiones no Traducidas 3' , Células de la Médula Ósea/citología , Resorción Ósea , Adhesión Celular , Línea Celular Tumoral , Técnicas de Cocultivo , Células HEK293 , Humanos , Interleucina-6/metabolismo , Lentivirus/genética , Células Madre Mesenquimatosas/metabolismo , MicroARNs/metabolismo , Chaperonas Moleculares/metabolismo , Osteoclastos/metabolismo , Proteínas Inhibidoras de STAT Activados/metabolismo , ARN Mensajero/metabolismo , Factor de Transcripción STAT3/metabolismo , Microambiente Tumoral , Regulación hacia Arriba
9.
Clin Cancer Res ; 18(7): 1888-900, 2012 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-22351691

RESUMEN

PURPOSE: Protein kinase CK2 promotes multiple myeloma cell growth by regulating critical signaling pathways. CK2 also modulates proper HSP90-dependent client protein folding and maturation by phosphorylating its co-chaperone CDC37. Because the endoplasmic reticulum (ER) stress/unfolded protein response (UPR) is central in myeloma pathogenesis, we tested the hypothesis that the CK2/CDC37/HSP90 axis could be involved in UPR in myeloma cells. EXPERIMENTAL DESIGN: We analyzed CK2 activity upon ER stress, the effects of its inactivation on the UPR pathways and on ER stress-induced apoptosis. The consequences of CK2 plus HSP90 inhibition on myeloma cell growth in vitro and in vivo and CK2 regulation of HSP90-triggered UPR were determined. RESULTS: CK2 partly localized to the ER and ER stress triggered its kinase activity. CK2 inhibition reduced the levels of the ER stress sensors IRE1α and BIP/GRP78, increased phosphorylation of PERK and EIF2α, and enhanced ER stress-induced apoptosis. Simultaneous inactivation of CK2 and HSP90 resulted in a synergic anti-myeloma effect (combination index = 0.291) and in much stronger alterations of the UPR pathways as compared with the single inhibition of the two molecules. Cytotoxicity from HSP90 and CK2 targeting was present in a myeloma microenvironment model, on plasma cells from patients with myeloma and in an in vivo mouse xenograft model. Mechanistically, CK2 inhibition led to a reduction of IRE1α/HSP90/CDC37 complexes in multiple myeloma cells. CONCLUSIONS: Our results place CK2 as a novel regulator of the ER stress/UPR cascades and HSP90 function in myeloma cells and offer the groundwork to design novel combination treatments for this disease.


Asunto(s)
Apoptosis/fisiología , Quinasa de la Caseína II/metabolismo , Estrés del Retículo Endoplásmico/fisiología , Proteínas HSP90 de Choque Térmico/metabolismo , Mieloma Múltiple/fisiopatología , Respuesta de Proteína Desplegada/fisiología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Benzoquinonas/farmacología , Western Blotting , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/genética , Línea Celular Tumoral , Células Cultivadas , Chaperón BiP del Retículo Endoplásmico , Estrés del Retículo Endoplásmico/genética , Inhibidores Enzimáticos/farmacología , Regulación Neoplásica de la Expresión Génica , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Proteínas HSP90 de Choque Térmico/genética , Humanos , Lactamas Macrocíclicas/farmacología , Ratones , Ratones SCID , Microscopía Confocal , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Transducción de Señal/fisiología , Tapsigargina/farmacología , Respuesta de Proteína Desplegada/genética , Ensayos Antitumor por Modelo de Xenoinjerto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA