Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Brain ; 147(9): 2998-3008, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-38606777

RESUMEN

Apilimod dimesylate is a first-in-class phosphoinositide kinase, FYVE-type zinc finger-containing (PIKfyve) inhibitor with a favourable clinical safety profile and has demonstrated activity in preclinical C9orf72 and TDP-43 amyotrophic lateral sclerosis (ALS) models. In this ALS clinical trial, the safety, tolerability, CNS penetrance and modulation of pharmacodynamic target engagement biomarkers were evaluated. This phase 2a, randomized, double-blind, placebo-controlled, biomarker-end-point clinical trial was conducted in four US centres (ClinicalTrials.gov NCT05163886). Participants with C9orf72 repeat expansions were randomly assigned (2:1) to receive twice-daily oral treatment with 125 mg apilimod dimesylate capsules or matching placebo for 12 weeks, followed by a 12-week open-label extension. Safety was measured as the occurrence of treatment-emergent or serious adverse events attributable to the study drug and tolerability at trial completion or treatment over 12 weeks. Changes from baseline in plasma and CSF and concentrations of apilimod dimesylate and its active metabolites and of pharmacodynamic biomarkers of PIKfyve inhibition [soluble glycoprotein nonmetastatic melanoma protein B (sGPNMB) upregulation] and disease-specific CNS target engagement [poly(GP)] were measured. Between 16 December 2021 and 7 July 2022, 15 eligible participants were enrolled. There were no drug-related serious adverse events reported in the trial. Fourteen (93%) participants completed the double-blind period with 99% dose compliance [n = 9 (90%) apilimod dimesylate; n = 5 (100%) placebo]. At Week 12, apilimod dimesylate was measurable in CSF at 1.63 ng/ml [standard deviation (SD): 0.937]. At Week 12, apilimod dimesylate increased plasma sGPNMB by >2.5-fold (P < 0.001), indicating PIKfyve inhibition, and lowered CSF poly(GP) protein levels by 73% (P < 0.001), indicating CNS tissue-level proof of mechanism. Apilimod dimesylate met prespecified key safety and biomarker end-points in this phase 2a trial and demonstrated CNS penetrance and pharmacodynamic target engagement. Apilimod dimesylate was observed to result in the greatest reduction in CSF poly(GP) levels observed to date in C9orf72 clinical trials.


Asunto(s)
Esclerosis Amiotrófica Lateral , Proteína C9orf72 , Humanos , Masculino , Femenino , Persona de Mediana Edad , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Método Doble Ciego , Adulto , Anciano , Proteína C9orf72/genética , Pirazoles/uso terapéutico , Pirazoles/farmacocinética , Resultado del Tratamiento , Biomarcadores/sangre , Hidrazonas , Morfolinas , Pirimidinas
2.
Proc Natl Acad Sci U S A ; 116(37): 18507-18516, 2019 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-31451639

RESUMEN

Genome-wide phenotypic screens provide an unbiased way to identify genes involved in particular biological traits, and have been widely used in lower model organisms. However, cost and time have limited the utility of such screens to address biological and disease questions in mammals. Here we report a highly efficient piggyBac (PB) transposon-based first-generation (F1) dominant screening system in mice that enables an individual investigator to conduct a genome-wide phenotypic screen within a year with fewer than 300 cages. The PB screening system uses visually trackable transposons to induce both gain- and loss-of-function mutations and generates genome-wide distributed new insertions in more than 55% of F1 progeny. Using this system, we successfully conducted a pilot F1 screen and identified 5 growth retardation mutations. One of these mutants, a Six1/4 PB/+ mutant, revealed a role in milk intake behavior. The mutant animals exhibit abnormalities in nipple recognition and milk ingestion, as well as developmental defects in cranial nerves V, IX, and X. This PB F1 screening system offers individual laboratories unprecedented opportunities to conduct affordable genome-wide phenotypic screens for deciphering the genetic basis of mammalian biology and disease pathogenesis.


Asunto(s)
Mapeo Cromosómico/métodos , Elementos Transponibles de ADN/genética , Genoma , Técnicas de Genotipaje/métodos , Mutagénesis Insercional/métodos , Animales , Animales Recién Nacidos , Mapeo Cromosómico/economía , Modelos Animales de Enfermedad , Embrión de Mamíferos , Estudios de Factibilidad , Femenino , Retardo del Crecimiento Fetal/genética , Fibroblastos , Técnicas de Genotipaje/economía , Humanos , Masculino , Ratones/genética , Ratones Transgénicos , Mutagénesis Insercional/economía , Mutación , Fenotipo , Cultivo Primario de Células
3.
Blood ; 129(13): 1768-1778, 2017 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-28104689

RESUMEN

We identified apilimod as an antiproliferative compound by high-throughput screening of clinical-stage drugs. Apilimod exhibits exquisite specificity for phosphatidylinositol-3-phosphate 5-kinase (PIKfyve) lipid kinase and has selective cytotoxic activity in B-cell non-Hodgkin lymphoma (B-NHL) compared with normal cells. Apilimod displays nanomolar activity in vitro, and in vivo studies demonstrate single-agent efficacy as well as synergy with approved B-NHL drugs. Using biochemical and knockdown approaches, and discovery of a kinase domain mutation conferring resistance, we demonstrate that apilimod-mediated cytotoxicity is driven by PIKfyve inhibition. Furthermore, a critical role for lysosome dysfunction as a major factor contributing to apilimod's cytotoxicity is supported by a genome-wide CRISPR screen. In the screen, TFEB (master transcriptional regulator of lysosomal biogenesis) and endosomal/lysosomal genes CLCN7, OSTM1, and SNX10 were identified as important determinants of apilimod sensitivity. These findings thus suggest that disruption of lysosomal homeostasis with apilimod represents a novel approach to treat B-NHL.


Asunto(s)
Linfoma de Células B/tratamiento farmacológico , Morfolinas/uso terapéutico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/uso terapéutico , Triazinas/uso terapéutico , Antineoplásicos , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Evaluación Preclínica de Medicamentos/métodos , Endosomas/efectos de los fármacos , Endosomas/genética , Ensayos Analíticos de Alto Rendimiento , Humanos , Hidrazonas , Lisosomas/efectos de los fármacos , Lisosomas/genética , Fosfatidilinositol 3-Quinasas , Pirimidinas
4.
Proc Natl Acad Sci U S A ; 110(38): E3640-9, 2013 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-24003131

RESUMEN

Despite considerable efforts to sequence hypermutated cancers such as melanoma, distinguishing cancer-driving genes from thousands of recurrently mutated genes remains a significant challenge. To circumvent the problematic background mutation rates and identify new melanoma driver genes, we carried out a low-copy piggyBac transposon mutagenesis screen in mice. We induced eleven melanomas with mutation burdens that were 100-fold lower relative to human melanomas. Thirty-eight implicated genes, including two known drivers of human melanoma, were classified into three groups based on high, low, or background-level mutation frequencies in human melanomas, and we further explored the functional significance of genes in each group. For two genes overlooked by prevailing discovery methods, we found that loss of membrane associated guanylate kinase, WW and PDZ domain containing 2 and protein tyrosine phosphatase, receptor type, O cooperated with the v-raf murine sarcoma viral oncogene homolog B (BRAF) recurrent V600E mutation to promote cellular transformation. Moreover, for infrequently mutated genes often disregarded by current methods, we discovered recurrent mitogen-activated protein kinase kinase kinase 1 (Map3k1)-activating insertions in our screen, mirroring recurrent MAP3K1 up-regulation in human melanomas. Aberrant expression of Map3k1 enabled growth factor-autonomous proliferation and drove BRAF-independent ERK signaling, thus shedding light on alternative means of activating this prominent signaling pathway in melanoma. In summary, our study contributes several previously undescribed genes involved in melanoma and establishes an important proof-of-principle for the utility of the low-copy transposon mutagenesis approach for identifying cancer-driving genes, especially those masked by hypermutation.


Asunto(s)
Elementos Transponibles de ADN/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Quinasa 1 de Quinasa de Quinasa MAP/metabolismo , Melanoma/genética , Mutagénesis Insercional/genética , Transducción de Señal/fisiología , Animales , Western Blotting , Cartilla de ADN/genética , Regulación Neoplásica de la Expresión Génica/genética , Pruebas Genéticas , Células HEK293 , Humanos , Inmunohistoquímica , Ratones , Ratones Transgénicos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Especificidad de la Especie
5.
Blood ; 120(4): 868-79, 2012 Jul 26.
Artículo en Inglés | MEDLINE | ID: mdl-22613795

RESUMEN

Oncogenic mutations in components of cytokine signaling pathways elicit ligand-independent activation of downstream signaling, enhancing proliferation and survival in acute myeloid leukemia (AML). The myeloproliferative leukemia virus oncogene, MPL, a homodimeric receptor activated by thrombopoietin (THPO), is mutated in myeloproliferative disorders but rarely in AML. Here we show that wild-type MPL expression is increased in a fraction of human AML samples expressing RUNX1-ETO, a fusion protein created by chromosome translocation t(8;21), and that up-regulation of Mpl expression in mice induces AML when coexpressed with RUNX1-ETO. The leukemic cells are sensitive to THPO, activating survival and proliferative responses. Mpl expression is not regulated by RUNX1-ETO in mouse hematopoietic progenitors or leukemic cells. Moreover, we find that activation of PI3K/AKT but not ERK/MEK pathway is a critical mediator of the MPL-directed antiapoptotic function in leukemic cells. Hence, this study provides evidence that up-regulation of wild-type MPL levels promotes leukemia development and maintenance through activation of the PI3K/AKT axis, and suggests that inhibitors of this axis could be effective for treatment of MPL-positive AML.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Proteínas de Fusión Oncogénica/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Receptores de Trombopoyetina/metabolismo , Trombopoyetina/metabolismo , Secuencia de Aminoácidos , Animales , Western Blotting , Médula Ósea/metabolismo , Médula Ósea/patología , Trasplante de Médula Ósea , Ciclo Celular , Proliferación Celular , Cromosomas Humanos Par 21/genética , Cromosomas Humanos Par 8/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Humanos , Técnicas para Inmunoenzimas , Leucemia Mieloide Aguda/genética , Ratones , Datos de Secuencia Molecular , Proteínas de Fusión Oncogénica/genética , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , ARN Mensajero/genética , Proteína 1 Compañera de Translocación de RUNX1 , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptores de Trombopoyetina/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal , Tasa de Supervivencia , Trombopoyetina/genética , Translocación Genética , Células Tumorales Cultivadas
6.
Cancer Cell ; 9(1): 57-68, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16413472

RESUMEN

The acute myeloid leukemia (AML)-associated CBF beta-SMMHC fusion protein impairs hematopoietic differentiation and predisposes to leukemic transformation. The mechanism of leukemia progression, however, is poorly understood. In this study, we report a conditional Cbfb-MYH11 knockin mouse model that develops AML with a median latency of 5 months. Cbf beta-SMMHC expression reduced the multilineage repopulation capacity of hematopoietic stem cells (HSCs) while maintaining their numbers under competitive conditions. The fusion protein induced abnormal myeloid progenitors (AMPs) with limited proliferative potential but leukemic predisposition similar to that of HSCs in transplanted mice. In addition, Cbf beta-SMMHC blocked megakaryocytic maturation at the CFU-Meg to megakaryocyte transition. These data show that a leukemia oncoprotein can inhibit differentiation and proliferation while not affecting the maintenance of long-term HSCs.


Asunto(s)
Leucemia Mieloide/patología , Células Progenitoras Mieloides/patología , Proteínas de Fusión Oncogénica/metabolismo , Preleucemia/patología , Enfermedad Aguda , Animales , Linfocitos B/patología , Plaquetas/patología , Proliferación Celular , Hematopoyesis , Leucemia Mieloide/metabolismo , Megacariocitos/metabolismo , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Células Progenitoras Mieloides/metabolismo , Proteínas de Fusión Oncogénica/genética , Preleucemia/metabolismo
7.
PLoS Genet ; 7(7): e1002110, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21814514

RESUMEN

With recent advances in genomic technologies, candidate human disease genes are being mapped at an accelerated pace. There is a clear need to move forward with genetic tools that can efficiently validate these mutations in vivo. Murine somatic mutagenesis is evolving to fulfill these needs with tools such as somatic transgenesis, humanized rodents, and forward genetics. By combining these resources one is not only able to model disease for in vivo verification, but also to screen for mutations and pathways integral to disease progression and therapeutic intervention. In this review, we briefly outline the current advances in somatic mutagenesis and discuss how these new tools, especially the piggyBac transposon system, can be applied to decipher human biology and disease.


Asunto(s)
Enfermedad/genética , Genómica , Crecimiento y Desarrollo/genética , Mutación/genética , Animales , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Pruebas Genéticas , Humanos , Ratones , Mutagénesis Insercional , Mutágenos/farmacología , Mutación/efectos de los fármacos
8.
Blood Adv ; 3(22): 3661-3673, 2019 11 26.
Artículo en Inglés | MEDLINE | ID: mdl-31751472

RESUMEN

Acute myeloid leukemias (AML) harboring a constitutively active internal tandem duplication (ITD) mutation in the FMS-like kinase tyrosine kinase (FLT3) receptor are associated with poor patient prognosis. Despite initial clinical responses to FLT3 kinase inhibitors, patients eventually relapse. Mechanisms of resistance include the acquisition of secondary FLT3 mutations and protective stromal signaling within the bone marrow niche. Here we show that LAM-003, a prodrug of the heat shock protein 90 inhibitor LAM-003A, has cytotoxic activity against AML cell lines and primary samples harboring FLT3-ITD. LAM-003 regressed tumors in an MV-4-11 xenograft mouse model and extended survival in a MOLM-13 systemic model. LAM-003 displayed synergistic activity with chemotherapeutic drugs and FLT3 inhibitors, with the most robust synergy being obtained with venetoclax, a BCL-2 inhibitor. This finding was verified in a MOLM-13 systemic survival model in which the combination significantly prolonged survival compared with the single agents. Importantly, LAM-003 exhibited equipotent activity against FLT3 inhibitor-resistant mutants of FLT3, such as D835 or F691, in cytotoxic and FLT3 degradation assays. LAM-003 also retained potency in AML cells grown in stromal-conditioned media that were resistant to FLT3 inhibitors. Lastly, a genome-wide CRISPR screen revealed epigenetic regulators, including KDM6A, as determinants of LAM-003 sensitivity in AML cell lines, leading to the discovery of synergy with an EZH2 inhibitor. Collectively, these preclinical findings support the use of LAM-003 in FLT3-ITD patients with AML who no longer respond to FLT3 inhibitor therapy either as a single agent or in combination with drugs known to be active in AML.


Asunto(s)
Antineoplásicos/farmacología , Resistencia a Antineoplásicos/genética , Duplicación de Gen , Leucemia Mieloide Aguda/genética , Inhibidores de Proteínas Quinasas/farmacología , Tirosina Quinasa 3 Similar a fms/genética , Animales , Antineoplásicos/uso terapéutico , Línea Celular Tumoral , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Epigénesis Genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Ratones , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico
9.
Autophagy ; 13(6): 1082-1083, 2017 Jun 03.
Artículo en Inglés | MEDLINE | ID: mdl-28350209

RESUMEN

We identified the PIKFYVE inhibitor apilimod as a potent and selective cytotoxic agent against B-cell non-Hodgkin lymphoma (B-NHL). Our data robustly establish PIKFYVE as the target through which apilimod kills B-NHL cells and show that apilimod-induced death in B-NHL is mediated by broad disruption of lysosome homeostasis characterized by lysosomal swelling, TFEB nuclear translocation, impaired maturation of lysosomal enzymes and incomplete autophagosome clearance. Furthermore, through genome-wide CRISPR knockout screening, we identified specific lysosomal genes (TFEB, CLCN7, OSTM1 and SNX10) as critical determinants of apilimod-induced cytotoxicity. Together these data highlight disruption of lysosome homeostasis through PIKFYVE inhibition as a novel anticancer mechanism in B-NHL and potentially other cancers.


Asunto(s)
Linfocitos B/patología , Linfoma no Hodgkin/tratamiento farmacológico , Inhibidores de las Quinasa Fosfoinosítidos-3 , Inhibidores de Proteínas Quinasas/uso terapéutico , Linfocitos B/efectos de los fármacos , Linfocitos B/enzimología , Endosomas/metabolismo , Humanos , Linfoma no Hodgkin/enzimología , Linfoma no Hodgkin/patología , Lisosomas/metabolismo , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología
10.
Stem Cell Reports ; 4(5): 926-38, 2015 May 12.
Artículo en Inglés | MEDLINE | ID: mdl-25866159

RESUMEN

The mechanisms regulating human embryonic stem (ES) cell self-renewal and differentiation are not well defined in part due to the lack of tools for forward genetic analysis. We present a piggyBac transposon gain of function screen in human ES cells that identifies DENND2C, which genetically cooperates with NANOG to maintain self-renewal in the presence of retinoic acid. We show that DENND2C negatively regulates RHOA activity, which cooperates with NANOG to block differentiation. It has been recently shown that RHOA exists in the nucleus and is activated by DNA damage; however, its nuclear function remains unknown. We discovered that RHOA associates with DNA and that DENND2C affects nuclear RHOA localization, activity, and DNA association. Our study illustrates the power of piggyBac as a cost-effective, efficient, and easy to use tool for forward genetic screens in human ES cells and provides insight into the role of RHOA in the nucleus.


Asunto(s)
Elementos Transponibles de ADN/genética , Vectores Genéticos/metabolismo , Células Madre Embrionarias Humanas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Diferenciación Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Mapeo Cromosómico , Vectores Genéticos/genética , Factores de Intercambio de Guanina Nucleótido/genética , Factores de Intercambio de Guanina Nucleótido/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Células Madre Embrionarias Humanas/citología , Humanos , Mutagénesis Insercional , Proteína Homeótica Nanog , Tretinoina/farmacología , Proteína de Unión al GTP rac1/metabolismo
11.
Pigment Cell Melanoma Res ; 27(2): 253-62, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24283590

RESUMEN

BRAF inhibitors improve melanoma patient survival, but resistance invariably develops. Here we report the discovery of a novel BRAF mutation that confers resistance to PLX4032 employing whole-exome sequencing of drug-resistant BRAF(V600K) melanoma cells. We further describe a new screening approach, a genome-wide piggyBac mutagenesis screen that revealed clinically relevant aberrations (N-terminal BRAF truncations and CRAF overexpression). The novel BRAF mutation, a Leu505 to His substitution (BRAF(L505H) ), is the first resistance-conferring second-site mutation identified in BRAF mutant cells. The mutation replaces a small nonpolar amino acid at the BRAF-PLX4032 interface with a larger polar residue. Moreover, we show that BRAF(L505H) , found in human prostate cancer, is itself a MAPK-activating, PLX4032-resistant oncogenic mutation. Lastly, we demonstrate that the PLX4032-resistant melanoma cells are sensitive to novel, next-generation BRAF inhibitors, especially the 'paradox-blocker' PLX8394, supporting its use in clinical trials for treatment of melanoma patients with BRAF-mutations.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Indoles/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/farmacología , Secuencia de Aminoácidos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Elementos Transponibles de ADN/genética , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Melanoma/enzimología , Melanoma/patología , Modelos Moleculares , Datos de Secuencia Molecular , Mutagénesis Insercional/genética , Proteínas Mutantes/metabolismo , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Vemurafenib
12.
PLoS One ; 6(12): e29486, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22216292

RESUMEN

Genotyping mice by DNA based methods is both laborious and costly. As an alternative, we systematically examined fluorescent proteins expressed in the lens as transgenic markers for mice. A set of eye markers has been selected such that double and triple transgenic animals can be visually identified and that fluorescence intensity in the eyes can be used to distinguish heterozygous from homozygous mice. Taken together, these eye markers dramatically reduce the time and cost of genotyping transgenics and empower analysis of genetic interaction.


Asunto(s)
Biomarcadores/metabolismo , Ojo/metabolismo , Proteínas Luminiscentes/metabolismo , Animales , Ratones , Ratones Transgénicos
13.
PLoS One ; 6(10): e26650, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22039523

RESUMEN

Somatic forward genetic screens have the power to interrogate thousands of genes in a single animal. Retroviral and transposon mutagenesis systems in mice have been designed and deployed in somatic tissues for surveying hematopoietic and solid tumor formation. In the context of cancer, the ability to visually mark mutant cells would present tremendous advantages for identifying tumor formation, monitoring tumor growth over time, and tracking tumor infiltrations and metastases into wild-type tissues. Furthermore, locating mutant clones is a prerequisite for screening and analyzing most other somatic phenotypes. For this purpose, we developed a system using the piggyBac (PB) transposon for somatic mutagenesis with an activated reporter and tracker, called PB-SMART. The PB-SMART mouse genetic screening system can simultaneously induce somatic mutations and mark mutated cells using bioluminescence or fluorescence. The marking of mutant cells enable analyses that are not possible with current somatic mutagenesis systems, such as tracking cell proliferation and tumor growth, detecting tumor cell infiltrations, and reporting tissue mutagenesis levels by a simple ex vivo visual readout. We demonstrate that PB-SMART is highly mutagenic, capable of tumor induction with low copy transposons, which facilitates the mapping and identification of causative insertions. We further integrated a conditional transposase with the PB-SMART system, permitting tissue-specific mutagenesis with a single cross to any available Cre line. Targeting the germline, the system could also be used to conduct F1 screens. With these features, PB-SMART provides an integrated platform for individual investigators to harness the power of somatic mutagenesis and phenotypic screens to decipher the genetic basis of mammalian biology and disease.


Asunto(s)
Elementos Transponibles de ADN , Genes Reporteros , Mutagénesis , Animales , Ratones
14.
Blood ; 105(7): 2900-7, 2005 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-15585652

RESUMEN

Recurrent chromosomal rearrangements are associated with the development of acute myeloid leukemia (AML). The frequent inversion of chromosome 16 creates the CBFB-MYH11 fusion gene that encodes the fusion protein CBFbeta-SMMHC. This fusion protein inhibits the core-binding factor (CBF), resulting in a block of hematopoietic differentiation, and induces leukemia upon the acquisition of additional mutations. A recent genetic screen identified Plag1 and Plagl2 as CBF beta-SMMHC candidate cooperating proteins. In this study, we demonstrate that Plag1 and Plagl2 independently cooperate with CBF beta-SMMHC in vivo to efficiently trigger leukemia with short latency in the mouse. In addition, Plag1 and Plagl2 increased proliferation by inducing G1 to S transition that resulted in the expansion of hematopoietic progenitors and increased cell renewal in vitro. Finally, PLAG1 and PLAGL2 expression was increased in 20% of human AML samples. Interestingly, PLAGL2 was preferentially increased in samples with chromosome 16 inversion, suggesting that PLAG1 and PLAGL2 may also contribute to human AML. Overall, this study shows that Plag1 and Plagl2 are novel leukemia oncogenes that act by expanding hematopoietic progenitors expressing CbF beta-SMMHC.


Asunto(s)
Proteínas de Unión al ADN/genética , Leucemia Mieloide/genética , Proteínas de Fusión Oncogénica/genética , Proteínas de Unión al ARN/genética , Factores de Transcripción/genética , Enfermedad Aguda , Adolescente , Adulto , Animales , Proteínas de Unión al ADN/metabolismo , Femenino , Fase G1/inmunología , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/citología , Humanos , Leucemia Mieloide/fisiopatología , Masculino , Ratones , Ratones Mutantes , Persona de Mediana Edad , Mutagénesis Insercional , Proteínas de Fusión Oncogénica/metabolismo , Proteínas de Unión al ARN/metabolismo , Retroviridae/genética , Fase S/inmunología , Factores de Transcripción/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA