Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Nature ; 586(7831): 769-775, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-33057200

RESUMEN

Myeloproliferative neoplasms (MPNs) are blood cancers that are characterized by the excessive production of mature myeloid cells and arise from the acquisition of somatic driver mutations in haematopoietic stem cells (HSCs). Epidemiological studies indicate a substantial heritable component of MPNs that is among the highest known for cancers1. However, only a limited number of genetic risk loci have been identified, and the underlying biological mechanisms that lead to the acquisition of MPNs remain unclear. Here, by conducting a large-scale genome-wide association study (3,797 cases and 1,152,977 controls), we identify 17 MPN risk loci (P < 5.0 × 10-8), 7 of which have not been previously reported. We find that there is a shared genetic architecture between MPN risk and several haematopoietic traits from distinct lineages; that there is an enrichment for MPN risk variants within accessible chromatin of HSCs; and that increased MPN risk is associated with longer telomere length in leukocytes and other clonal haematopoietic states-collectively suggesting that MPN risk is associated with the function and self-renewal of HSCs. We use gene mapping to identify modulators of HSC biology linked to MPN risk, and show through targeted variant-to-function assays that CHEK2 and GFI1B have roles in altering the function of HSCs to confer disease risk. Overall, our results reveal a previously unappreciated mechanism for inherited MPN risk through the modulation of HSC function.


Asunto(s)
Predisposición Genética a la Enfermedad/genética , Células Madre Hematopoyéticas/patología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/patología , Neoplasias/genética , Neoplasias/patología , Linaje de la Célula/genética , Autorrenovación de las Células , Quinasa de Punto de Control 2/genética , Femenino , Humanos , Leucocitos/patología , Masculino , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Riesgo , Homeostasis del Telómero
2.
FASEB J ; : fj201701568R, 2018 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-29856660

RESUMEN

Glioblastoma is an aggressive and invasive brain malignancy with high mortality rates despite current treatment modalities. In this study, we show that a 7-gene signature, previously found to govern the switch of glioblastomas from dormancy to aggressive tumor growth, correlates with improved overall survival of patients with glioblastoma. Using glioblastoma dormancy models, we validated the role of 2 genes from the signature, thrombospondin-1 ( TSP-1) and epidermal growth factor receptor ( EGFR), as regulators of glioblastoma dormancy and explored their therapeutic potential. EGFR up-regulation was reversed using EGFR small interfering RNA polyplex, antibody, or small-molecule inhibitor. The diminished function of TSP-1 was augmented via a peptidomimetic. The combination of EGFR inhibition and TSP-1 restoration led to enhanced therapeutic efficacy in vitro, in 3-dimensional patient-derived spheroids, and in a subcutaneous human glioblastoma model in vivo. Systemic administration of the combination therapy to mice bearing intracranial murine glioblastoma resulted in marginal therapeutic outcomes, probably due to brain delivery challenges, p53 mutation status, and the aggressive nature of the selected cell line. Nevertheless, this study provides a proof of concept for exploiting regulators of tumor dormancy for glioblastoma therapy. This therapeutic strategy can be exploited for future investigations using a variety of therapeutic entities that manipulate the expression of dormancy-associated genes in glioblastoma as well as in other cancer types.-Tiram, G., Ferber, S., Ofek, P., Eldar-Boock, A., Ben-Shushan, D., Yeini, E., Krivitsky, A., Blatt, R., Almog, N., Henkin, J., Amsalem, O., Yavin, E., Cohen, G., Lazarovici, P., Lee, J. S., Ruppin, E., Milyavsky, M., Grossman, R., Ram, Z., Calderón, M., Haag, R., Satchi-Fainaro, R. Reverting the molecular fingerprint of tumor dormancy as a therapeutic strategy for glioblastoma.

3.
Carcinogenesis ; 38(4): 367-377, 2017 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-28334174

RESUMEN

Self-renewing and multipotent hematopoietic stem cells (HSCs) maintain lifelong hematopoiesis. Their enormous regenerative potential coupled with lifetime persistence in the body, in contrast with the Progenitors, demand tight control of HSCs genome stability. Indeed, failure to accurately repair DNA damage in HSCs is associated with bone marrow failure and accelerated leukemogenesis. Recent observations exposed remarkable differences in several DNA-damage response (DDR) aspects between HSCs and Progenitors, especially in their DNA-repair capacities and susceptibility to apoptosis. Human HSCs in comparison with Progenitors exhibit delayed DNA double-strand break rejoining, persistent DDR signaling activation, higher sensitivity to the cytotoxic effects of ionizing radiation and attenuated expression of DNA-repair genes. Importantly, the distinct DDR of HSCs was also documented in mouse models. Nevertheless, physiological significance and the molecular basis of the HSCs-specific DDR features are only partially understood. Taking radiation-induced DDR as a paradigm, this review will focus on the current advances in understanding the role of cell-intrinsic DDR regulators and the cellular microenvironment in balancing stemness with genome stability. Pre-leukemia HSCs and clonal hematopoiesis evolvement will be discussed as an evolutionary compromise between the need for lifelong blood regeneration and DDR. Uniquely for this review, we outline the differences in HSCs-related DDR as highlighted by various experimental systems and attempt to provide their critical analysis.


Asunto(s)
Sangre/metabolismo , Daño del ADN/genética , ADN/genética , Células Madre Hematopoyéticas/metabolismo , Regeneración/genética , Animales , Reparación del ADN/genética , Células Madre Hematopoyéticas/fisiología , Humanos , Leucemia/genética , Leucemia/patología , Regeneración/fisiología
4.
Int J Cancer ; 140(4): 864-876, 2017 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-27813122

RESUMEN

Quercetin (Que) is an abundant flavonoid in the human diet and high-concentration food supplement with reported pro- and anti-carcinogenic activities. Topoisomerase II (TopoII) inhibition and subsequent DNA damage induction by Que was implicated in the mixed lineage leukemia gene (MLL) rearrangements that can induce infant and adult leukemias. This notion raised concerns regarding possible genotoxicities of Que in hematopoietic stem and progenitor cells (HSPCs). However, molecular targets mediating Que effects on DNA repair relevant to MLL translocations have not been defined. In this study we describe novel and potentially genotoxic Que activities in suppressing non-homologous end joining and homologous recombination pathways downstream of MLL cleavage. Using pharmacological dissection of DNA-PK, ATM and PI3K signalling we defined PI3K inhibition by Que with a concomitant decrease in the abundance of key DNA repair genes to be responsible for DNA repair inhibition. Evidence for the downstream TopoII-independent mutagenic potential of Que was obtained by documenting further increased frequencies of MLL rearrangements in human HSPCs concomitantly treated with Etoposide and Que versus single treatments. Importantly, by engaging a tissue engineered placental barrier, we have established the extent of Que transplacental transfer and hence provided the evidence for Que reaching fetal HSPCs. Thus, Que exhibits genotoxic effects in human HSPCs via different mechanisms when applied continuously and at high concentrations. In light of the demonstrated Que transfer to the fetal compartment our findings are key to understanding the mechanisms underlying infant leukemia and provide molecular markers for the development of safety values.


Asunto(s)
Transformación Celular Neoplásica/efectos de los fármacos , Daño del ADN , Reparación del ADN/efectos de los fármacos , ADN-Topoisomerasas de Tipo II/fisiología , Células Madre Hematopoyéticas/efectos de los fármacos , N-Metiltransferasa de Histona-Lisina/genética , Leucemia/inducido químicamente , Proteína de la Leucemia Mieloide-Linfoide/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Quercetina/toxicidad , Transducción de Señal/efectos de los fármacos , Inhibidores de Topoisomerasa II/toxicidad , Adulto , Ácido Ascórbico/farmacología , Técnicas de Cultivo de Célula , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Relación Dosis-Respuesta a Droga , Etopósido/farmacología , Femenino , Genisteína/farmacología , Histonas/análisis , Humanos , Lactante , Leucemia/genética , Intercambio Materno-Fetal , Fosfatidilinositol 3-Quinasas/fisiología , Embarazo
5.
Proc Natl Acad Sci U S A ; 110(51): 20599-604, 2013 Dec 17.
Artículo en Inglés | MEDLINE | ID: mdl-24297922

RESUMEN

The Lnk (Sh2b3) adaptor protein dampens the response of hematopoietic stem cells and progenitors (HSPCs) to a variety of cytokines by inhibiting JAK2 signaling. As a consequence, Lnk(-/-) mice develop hematopoietic hyperplasia, which progresses to a phenotype resembling the nonacute phase of myeloproliferative neoplasm. In addition, Lnk mutations have been identified in human myeloproliferative neoplasms and acute leukemia. We find that Lnk suppresses the development of radiation-induced acute B-cell malignancies in mice. Lnk-deficient HSPCs recover more effectively from irradiation than their wild-type counterparts, and this resistance of Lnk(-/-) HSPCs to radiation underlies the subsequent emergence of leukemia. A search for the mechanism responsible for radiation resistance identified the cytokine IL-11 as being critical for the ability of Lnk(-/-) HSPCs to recover from irradiation and subsequently become leukemic. In IL-11 signaling, wild-type Lnk suppresses tyrosine phosphorylation of the Src homology region 2 domain-containing phosphatase-2/protein tyrosine phosphatase nonreceptor type 11 and its association with the growth factor receptor-bound protein 2, as well as activation of the Erk MAP kinase pathway. Indeed, Src homology region 2 domain-containing phosphatase-2 has a binding motif for the Lnk Src Homology 2 domain that is phosphorylated in response to IL-11 stimulation. IL-11 therefore drives a pathway that enhances HSPC radioresistance and radiation-induced B-cell malignancies, but is normally attenuated by the inhibitory adaptor Lnk.


Asunto(s)
Rayos gamma/efectos adversos , Interleucina-11/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Leucemia de Células B/metabolismo , Sistema de Señalización de MAP Quinasas/efectos de la radiación , Proteínas de Neoplasias/metabolismo , Neoplasias Inducidas por Radiación/metabolismo , Proteínas/metabolismo , Tolerancia a Radiación/efectos de la radiación , Proteínas Adaptadoras Transductoras de Señales , Secuencias de Aminoácidos , Animales , Proteína Adaptadora GRB2/genética , Proteína Adaptadora GRB2/metabolismo , Humanos , Interleucina-11/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Leucemia de Células B/genética , Leucemia de Células B/patología , Sistema de Señalización de MAP Quinasas/genética , Proteínas de la Membrana , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Neoplasias Inducidas por Radiación/genética , Neoplasias Inducidas por Radiación/patología , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas/genética , Tolerancia a Radiación/genética
6.
Cancer Cell ; 11(2): 133-46, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17292825

RESUMEN

Myocardin is known as an important transcriptional regulator in smooth and cardiac muscle development. Here we found that myocardin is frequently repressed during human malignant transformation, contributing to a differentiation defect. We demonstrate that myocardin is a transcriptional target of TGFbeta required for TGFbeta-mediated differentiation of human fibroblasts. Serum deprivation, intact contact inhibition response, and the p16ink4a/Rb pathway contribute to myocardin induction and differentiation. Restoration of myocardin expression in sarcoma cells results in differentiation and inhibition of malignant growth, whereas inactivation of myocardin in normal fibroblasts increases their proliferative potential. Myocardin expression is reduced in multiple types of human tumors. Collectively, our results demonstrate that myocardin is an important suppressive modifier of the malignant transformation process.


Asunto(s)
Diferenciación Celular , Transformación Celular Neoplásica , Inhibidor p16 de la Quinasa Dependiente de Ciclina/antagonistas & inhibidores , Fibroblastos/citología , Proteínas Nucleares/antagonistas & inhibidores , Transactivadores/antagonistas & inhibidores , Western Blotting , Adhesión Celular , Proliferación Celular , Células Cultivadas , Ensayo de Unidades Formadoras de Colonias , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Metilación de ADN , Fibroblastos/metabolismo , Técnica del Anticuerpo Fluorescente , Regulación de la Expresión Génica , Humanos , Pulmón/embriología , Mesodermo/citología , Mesodermo/metabolismo , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Plásmidos , Regiones Promotoras Genéticas , ARN Interferente Pequeño/farmacología , Transactivadores/genética , Transactivadores/metabolismo , Factor de Crecimiento Transformador beta/farmacología
7.
Blood ; 117(16): 4253-61, 2011 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-21398220

RESUMEN

Src homology 2 domain-containing phosphatase 2 (Shp2), encoded by Ptpn11, is a member of the nonreceptor protein-tyrosine phosphatase family, and functions in cell survival, proliferation, migration, and differentiation in many tissues. Here we report that loss of Ptpn11 in murine hematopoietic cells leads to bone marrow aplasia and lethality. Mutant mice show rapid loss of hematopoietic stem cells (HSCs) and immature progenitors of all hematopoietic lineages in a gene dosage-dependent and cell-autonomous manner. Ptpn11-deficient HSCs and progenitors undergo apoptosis concomitant with increased Noxa expression. Mutant HSCs/progenitors also show defective Erk and Akt activation in response to stem cell factor and diminished thrombopoietin-evoked Erk activation. Activated Kras alleviates the Ptpn11 requirement for colony formation by progenitors and cytokine/growth factor responsiveness of HSCs, indicating that Ras is functionally downstream of Shp2 in these cells. Thus, Shp2 plays a critical role in controlling the survival and maintenance of HSCs and immature progenitors in vivo.


Asunto(s)
Médula Ósea/patología , Eliminación de Gen , Células Madre Hematopoyéticas/metabolismo , Proteína Tirosina Fosfatasa no Receptora Tipo 11/genética , Animales , Ciclo Celular , Muerte Celular , Epistasis Genética , Células Madre Hematopoyéticas/citología , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Células Madre/citología , Células Madre/metabolismo
8.
Cancer Immunol Res ; 11(6): 792-809, 2023 06 02.
Artículo en Inglés | MEDLINE | ID: mdl-37070661

RESUMEN

The pioneering design of chimeric antigen receptor (CAR) T-cell therapy demonstrated the potential of reprogramming the immune system. Nonetheless, T-cell exhaustion, toxicity, and suppressive microenvironments limit their efficacy in solid tumors. We previously characterized a subset of tumor-infiltrating CD4+ T cells expressing the FcγRI receptor. Herein, we detail engineering of a receptor, based on the FcγRI structure, allowing T cells to target tumor cells using antibody intermediates. These T cells showed effective and specific cytotoxicity only when an appropriate antibody was added. Only target-bound antibodies activated these cells, while free antibodies were internalized without activation. Their cytotoxic activity was correlated to target protein density, therefore targeting tumor cells with high antigen density while sparing normal cells with low or no expression. This activation mechanism prevented premature exhaustion. Furthermore, during antibody-dependent cytotoxicity these cells secreted attenuated cytokine levels compared with CAR T cells, thereby enhancing their safety profile. These cells eradicated established melanomas, infiltrated the tumor microenvironment, and facilitated host immune cell recruitment in immunocompetent mice. In NOD/SCID gamma mice the cells infiltrate, persist, and eradicate tumors. As opposed to CAR T-cell therapies, which require changing the receptor across different types of cancer, our engineered T cells remain the same across tumor types, while only the injected antibody changes. Overall, we generated a highly flexible T-cell therapy capable of binding a wide range of tumor cells with high affinity, while preserving the cytotoxic specificity only to cells expressing high density of tumor-associated antigens and using a single manufacturing process.


Asunto(s)
Inmunoterapia Adoptiva , Melanoma , Animales , Ratones , Receptores de IgG , Ensayos Antitumor por Modelo de Xenoinjerto , Ratones SCID , Ratones Endogámicos NOD , Melanoma/terapia , Inmunoglobulinas , Línea Celular Tumoral , Microambiente Tumoral
9.
Leukemia ; 37(4): 751-764, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36720973

RESUMEN

Acute myeloid leukemia (AML) is a heterogeneous, aggressive malignancy with dismal prognosis and with limited availability of targeted therapies. Epigenetic deregulation contributes to AML pathogenesis. KDM6 proteins are histone-3-lysine-27-demethylases that play context-dependent roles in AML. We inform that KDM6-demethylase function critically regulates DNA-damage-repair-(DDR) gene expression in AML. Mechanistically, KDM6 expression is regulated by genotoxic stress, with deficiency of KDM6A-(UTX) and KDM6B-(JMJD3) impairing DDR transcriptional activation and compromising repair potential. Acquired KDM6A loss-of-function mutations are implicated in chemoresistance, although a significant percentage of relapsed-AML has upregulated KDM6A. Olaparib treatment reduced engraftment of KDM6A-mutant-AML-patient-derived xenografts, highlighting synthetic lethality using Poly-(ADP-ribose)-polymerase-(PARP)-inhibition. Crucially, a higher KDM6A expression is correlated with venetoclax tolerance. Loss of KDM6A increased mitochondrial activity, BCL2 expression, and sensitized AML cells to venetoclax. Additionally, BCL2A1 associates with venetoclax resistance, and KDM6A loss was accompanied with a downregulated BCL2A1. Corroborating these results, dual targeting of PARP and BCL2 was superior to PARP or BCL2 inhibitor monotherapy in inducing AML apoptosis, and primary AML cells carrying KDM6A-domain mutations were even more sensitive to the combination. Together, our study illustrates a mechanistic rationale in support of a novel combination therapy for AML based on subtype-heterogeneity, and establishes KDM6A as a molecular regulator for determining therapeutic efficacy.


Asunto(s)
Leucemia Mieloide Aguda , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Humanos , Histona Demetilasas/genética , Histona Demetilasas/metabolismo , Histona Demetilasas con Dominio de Jumonji , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas/uso terapéutico , Proteínas Proto-Oncogénicas c-bcl-2/genética
10.
Nat Commun ; 14(1): 5871, 2023 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-37735473

RESUMEN

The ERG (ETS-related gene) transcription factor is linked to various types of cancer, including leukemia. However, the specific ERG domains and co-factors contributing to leukemogenesis are poorly understood. Drug targeting a transcription factor such as ERG is challenging. Our study reveals the critical role of a conserved amino acid, proline, at position 199, located at the 3' end of the PNT (pointed) domain, in ERG's ability to induce leukemia. P199 is necessary for ERG to promote self-renewal, prevent myeloid differentiation in hematopoietic progenitor cells, and initiate leukemia in mouse models. Here we show that P199 facilitates ERG's interaction with the NCoR-HDAC3 co-repressor complex. Inhibiting HDAC3 reduces the growth of ERG-dependent leukemic and prostate cancer cells, indicating that the interaction between ERG and the NCoR-HDAC3 co-repressor complex is crucial for its oncogenic activity. Thus, targeting this interaction may offer a potential therapeutic intervention.


Asunto(s)
Leucemia , Factores de Transcripción , Animales , Masculino , Ratones , Proteínas Co-Represoras , Regulación de la Expresión Génica , Genes Reguladores
11.
Aging (Albany NY) ; 13(17): 21066-21089, 2021 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-34506302

RESUMEN

The gender gap in life expectancy and cancer incidence suggests differences in the aging process between the sexes. Genomic instability has been recognized as a key factor in aging, but little is known about sex-specific differences. Therefore, we analyzed DNA double-strand break (DSB) repair in cycling human peripheral blood lymphocytes (PBL) from male and female donors of different age. Reporter-based DSB repair analyses revealed differential regulation of pathway usage in PBL from male and female donors with age: Non-homologous end joining (NHEJ) was inversely regulated in men and women; the activity of pathways requiring end processing and strand annealing steps such as microhomology-mediated end joining (MMEJ) declined with age in women but not in men. Screening candidate proteins identified the NHEJ protein KU70 as well as the end resection regulatory factors ATM and BLM showing reduced expression during aging in women. Consistently, the regulatory factor BLM contributed to the MMEJ proficiency in young but not in old women as demonstrated by knockdown analysis. In conclusion, we show that DSB repair is subject to changes upon aging and age-related changes in DSB repair are distinct in men and women.


Asunto(s)
Envejecimiento/fisiología , Roturas del ADN de Doble Cadena , Reparación del ADN/fisiología , Linfocitos/fisiología , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Humanos , Masculino , Persona de Mediana Edad , Factores Sexuales , Adulto Joven
12.
Nat Commun ; 12(1): 2455, 2021 04 28.
Artículo en Inglés | MEDLINE | ID: mdl-33911081

RESUMEN

The mutational mechanisms underlying recurrent deletions in clonal hematopoiesis are not entirely clear. In the current study we inspect the genomic regions around recurrent deletions in myeloid malignancies, and identify microhomology-based signatures in CALR, ASXL1 and SRSF2 loci. We demonstrate that these deletions are the result of double stand break repair by a PARP1 dependent microhomology-mediated end joining (MMEJ) pathway. Importantly, we provide evidence that these recurrent deletions originate in pre-leukemic stem cells. While DNA polymerase theta (POLQ) is considered a key component in MMEJ repair, we provide evidence that pre-leukemic MMEJ (preL-MMEJ) deletions can be generated in POLQ knockout cells. In contrast, aphidicolin (an inhibitor of replicative polymerases and replication) treatment resulted in a significant reduction in preL-MMEJ. Altogether, our data indicate an association between POLQ independent MMEJ and clonal hematopoiesis and elucidate mutational mechanisms involved in the very first steps of leukemia evolution.


Asunto(s)
Hematopoyesis Clonal/genética , Reparación del ADN por Unión de Extremidades/genética , ADN Polimerasa Dirigida por ADN/genética , Leucemia Mieloide/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Afidicolina/farmacología , Calreticulina/genética , Roturas del ADN de Doble Cadena , ADN Polimerasa Dirigida por ADN/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Humanos , Células Progenitoras Mieloides , Proteínas Represoras/genética , Eliminación de Secuencia/genética , Factores de Empalme Serina-Arginina/genética , ADN Polimerasa theta
13.
Biochem Pharmacol ; 168: 412-428, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31325448

RESUMEN

Although having promising anti-myeloma properties, the pan-histone deacetylase inhibitor (HDACi) panobinostat lacks therapeutic activity as a single agent. The aim of the current study was to elucidate the mechanisms underlying multiple myeloma (MM) resistance to panobinostat monotherapy and to define strategies to overcome it. Sensitivity of MM cell lines and primary CD138+ cells from MM patients to panobinostat correlated with reduced expression of the chemokine receptor CXCR4, whereas overexpression of CXCR4 in MM cell lines increased their resistance to panobinostat. Decreased sensitivity to HDACi was associated with reversible G0/G1 cell growth arrest while response was characterized by apoptotic cell death. Analysis of intra-cellular signaling mediators revealed the pro-survival mTOR pathway to be regulated by CXCR4 overexpression. Combining panobinostat with mTOR inhibitor everolimus abrogated the resistance to HDACi and induced synergistic cell death. The combination of panobinostat/everolimus resulted in sustained DNA damage and irreversible suppression of proliferation accompanied by robust apoptosis. Gene expression analysis revealed distinct genetic profiles of single versus combined agent exposure. Whereas panobinostat increased the expression of the cell cycle inhibitor p21, co-treatment with everolimus abrogated the increase in p21 and synergistically downregulated the expression of DNA repair genes and mitotic checkpoint regulators. Importantly, the combination of panobinostat with everolimus effectively targeted CXCR4-expressing resistant MM cells in vivo in the BM niche. In summary, our results uncover the mechanism responsible for the strong synergistic anti-MM activity of dual HDAC and mTOR inhibition and provide the rationale for a novel potential therapeutic approach to treat MM.


Asunto(s)
Antineoplásicos/administración & dosificación , Everolimus/administración & dosificación , Inhibidores de Histona Desacetilasas/administración & dosificación , Mitosis/efectos de los fármacos , Panobinostat/administración & dosificación , Receptores CXCR4/antagonistas & inhibidores , Proteínas de Unión al GTP rho/antagonistas & inhibidores , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Células Cultivadas , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/fisiología , Humanos , Ratones , Mitosis/fisiología , Mieloma Múltiple/tratamiento farmacológico , Mieloma Múltiple/metabolismo , Receptores CXCR4/biosíntesis , Receptores CXCR4/genética , Serina-Treonina Quinasas TOR/antagonistas & inhibidores , Serina-Treonina Quinasas TOR/biosíntesis , Serina-Treonina Quinasas TOR/genética , Ensayos Antitumor por Modelo de Xenoinjerto/métodos , Proteínas de Unión al GTP rho/biosíntesis , Proteínas de Unión al GTP rho/genética
14.
Oncogene ; 38(17): 3103-3118, 2019 04.
Artículo en Inglés | MEDLINE | ID: mdl-30622338

RESUMEN

The cancer stem cell (CSC) model suggests that a subpopulation of cells within the tumor, the CSCs, is responsible for cancer relapse and metastasis formation. CSCs hold unique characteristics, such as self-renewal, differentiation abilities, and resistance to chemotherapy, raising the need for discovering drugs that target CSCs. Previously we have found that the antihypertensive drug spironolactone impairs DNA damage response in cancer cells. Here we show that spironolactone, apart from inhibiting cancerous cell growth, is also highly toxic to CSCs. Notably, we demonstrate that CSCs have high basal levels of DNA double-strand breaks (DSBs). Mechanistically, we reveal that spironolactone does not damage the DNA but impairs DSB repair and induces apoptosis in cancer cells and CSCs while sparing healthy cells. In vivo, spironolactone treatment reduced the size and CSC content of tumors. Overall, we suggest spironolactone as an anticancer reagent, toxic to both cancer cells and, particularly to, CSCs.


Asunto(s)
Antineoplásicos/administración & dosificación , Reparación del ADN/efectos de los fármacos , Neoplasias/tratamiento farmacológico , Células Madre Neoplásicas/efectos de los fármacos , Espironolactona/administración & dosificación , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Proliferación Celular , Supervivencia Celular/efectos de los fármacos , Reposicionamiento de Medicamentos , Células HeLa , Humanos , Ratones , Neoplasias/genética , Espironolactona/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
15.
Leukemia ; 33(8): 2061-2077, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-30705411

RESUMEN

Acute leukemia is an aggressive blood malignancy with low survival rates. A high expression of stem-like programs in leukemias predicts poor prognosis and is assumed to act in an aberrant fashion in the phenotypically heterogeneous leukemia stem cell (LSC) population. A lack of suitable genome engineering tools that can isolate LSCs based on their stemness precludes their comprehensive examination and full characterization. We hypothesized that tagging endogenous stemness-regulatory regions could generate a genome reporter for the putative leukemia stemness-state. Our analysis revealed that the ERG + 85 enhancer region can serve as a marker for stemness-state and a fluorescent lentiviral reporter was developed that can accurately recapitulate the endogenous activity. Using our novel reporter, we revealed cellular heterogeneity in several leukemia cell lines and patient-derived samples. Alterations in reporter activity were associated with transcriptomic and functional changes that were closely related to the hematopoietic stem cell (HSC) identity. Notably, the differentiation potential was skewed towards the erythro-megakaryocytic lineage. Moreover, an ERG + 85High fraction of AML cells could regenerate the original cellular heterogeneity and was enriched for LSCs. RNA-seq analysis coupled with in silico drug-screen analysis identified 4HPR as an effective inhibitor of ERG + 85High leukemia growth. We propose that further utilization of our novel molecular tool will identify crucial determinants of LSCs, thus providing a rationale for their therapeutic targeting.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/fisiología , Elementos de Facilitación Genéticos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Regulador Transcripcional ERG/genética
16.
Cancer Res ; 79(15): 3862-3876, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31175119

RESUMEN

Acute leukemia is a rapidly progressing blood cancer with low survival rates. Unfavorable prognosis is attributed to insufficiently characterized subpopulations of leukemia stem cells (LSC) that drive chemoresistance and leukemia relapse. Here we utilized a genetic reporter that assesses stemness to enrich and functionally characterize LSCs. We observed heterogeneous activity of the ERG+85 enhancer-based fluorescent reporter in human leukemias. Cells with high reporter activity (tagBFPHigh) exhibited elevated expression of stemness and chemoresistance genes and demonstrated increased clonogenicity and resistance to chemo- and radiotherapy as compared with their tagBFPNeg counterparts. The tagBFPHigh fraction was capable of regenerating the original cellular heterogeneity and demonstrated increased invasive ability. Moreover, the tagBFPHigh fraction was enriched for leukemia-initiating cells in a xenograft assay. We identified the ubiquitin hydrolase USP9X as a novel ERG transcriptional target that sustains ERG+85-positive cells by controlling ERG ubiquitination. Therapeutic targeting of USP9X led to preferential inhibition of the ERG-dependent leukemias. Collectively, these results characterize human leukemia cell functional heterogeneity and suggest that targeting ERG via USP9X inhibition may be a potential treatment strategy in patients with leukemia. SIGNIFICANCE: This study couples a novel experimental tool with state-of-the-art approaches to delineate molecular mechanisms underlying stem cell-related characteristics in leukemia cells.


Asunto(s)
Leucemia Mieloide Aguda/genética , Proteínas Oncogénicas/metabolismo , Regulador Transcripcional ERG/metabolismo , Trasplante Heterólogo/métodos , Animales , Línea Celular Tumoral , Modelos Animales de Enfermedad , Expresión Génica , Humanos , Leucemia Mieloide Aguda/mortalidad , Ratones , Análisis de Supervivencia , Transfección
17.
J Immunol Methods ; 336(1): 56-63, 2008 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-18456275

RESUMEN

We have investigated the possibility of using element-tagged antibodies for protein detection and quantification in microplate format using Inductively Coupled Plasma Mass Spectrometry (ICP-MS), and compared the results to conventional immunoassays, such as Enzyme-Linked Immunosorbent Assay (ELISA) and Western blotting. The technique was further employed to detect low levels and measure DNA-binding activity of transcription factor p53 in leukemia cell lysates through its interaction with immobilized oligonucleotides and recognition by element-tagged antibodies. The advantages of ICP-MS detection for routine performance of immunoassays include increased sensitivity, wide dynamic range, minimal interference from complex matrices, and high throughput. Our approach advances the ICP-MS technology and demonstrates its applicability to proteomic studies through the use of antibodies directly labeled with polymer tags bearing multiple atoms of lanthanides. Development of this novel methodology will enable fast and quantitative identification of multiple analytes in a single well.


Asunto(s)
Anticuerpos Monoclonales/química , Inmunoensayo/métodos , Elementos de la Serie de los Lantanoides/química , Espectrometría de Masas/métodos , Proteína p53 Supresora de Tumor/metabolismo , Western Blotting , ADN/metabolismo , Ensayo de Inmunoadsorción Enzimática , Humanos , Células K562 , Factor de Crecimiento Derivado de Plaquetas/análisis , Proteína p53 Supresora de Tumor/análisis
18.
Cancer Res ; 66(7): 3531-40, 2006 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-16585177

RESUMEN

Prostate cancer is the most commonly diagnosed type of cancer in men, and there is no available cure for patients with advanced disease. In vitro model systems are urgently required to permit the study of human prostate cell differentiation and malignant transformation. Unfortunately, human prostate cells are particularly difficult to convert into continuously growing cultures. We report here the successful immortalization without viral oncogenes of prostate epithelial cells and, for the first time, prostate stromal cells. These cells exhibit a significant pattern of authentic prostate-specific features. In particular, the epithelial cell culture is able to differentiate into glandular buds that closely resemble the structures formed by primary prostate epithelial cells. The stromal cells have typical characteristics of prostate smooth muscle cells. These immortalized cultures may serve as a unique experimental platform to permit several research directions, including the study of cell-cell interactions in an authentic prostate microenvironment, prostate cell differentiation, and most significantly, the complex multistep process leading to prostate cell transformation.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Proteínas de Unión al ADN/fisiología , Próstata/citología , Próstata/metabolismo , Neoplasias de la Próstata/metabolismo , Neoplasias de la Próstata/patología , Telomerasa/fisiología , Anciano , Diferenciación Celular/fisiología , Línea Celular Tumoral , Transformación Celular Neoplásica/genética , Células Cultivadas , Inhibidor p16 de la Quinasa Dependiente de Ciclina/biosíntesis , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Células Epiteliales/fisiología , Humanos , Masculino , Próstata/fisiología , Neoplasias de la Próstata/genética , Células del Estroma/citología , Células del Estroma/metabolismo , Células del Estroma/fisiología , Telomerasa/biosíntesis , Telomerasa/genética , Transfección , Proteína p53 Supresora de Tumor/biosíntesis , Proteína p53 Supresora de Tumor/genética
19.
Cancer Res ; 66(22): 10750-9, 2006 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-17108111

RESUMEN

Mutations in p53 are ubiquitous in human tumors. Some p53 mutations not only result in loss of wild-type (WT) activity but also grant additional functions, termed "gain of function." In this study, we explore how the status of p53 affects the immediate response gene activating transcription factor 3 (ATF3) in the 12-O-tetradecanoylphorbol-13-acetate (TPA)-protein kinase C (PKC) pathway. We show that high doses of TPA induce ATF3 in a WT p53-independent manner correlating with PKCs depletion and cell death. We show that cells harboring mutant p53 have attenuated ATF3 induction and are less sensitive to TPA-induced death compared with their p53-null counterparts. Mutagenesis analysis of the ATF3 promoter identified the regulatory motifs cyclic AMP-responsive element binding protein/ATF and MEF2 as being responsible for the TPA-induced activation of ATF3. Moreover, we show that mutant p53 attenuates ATF3 expression by two complementary mechanisms. It interacts with the ATF3 promoter and influences its activity via the MEF2 site, and additionally, it attenuates transcriptional expression of the ATF3 activator MEF2D. These data provide important insights into the molecular mechanisms that underlie mutant p53 gain of function.


Asunto(s)
Factor de Transcripción Activador 3/biosíntesis , Mutación , Acetato de Tetradecanoilforbol/antagonistas & inhibidores , Proteína p53 Supresora de Tumor/genética , Factor de Transcripción Activador 3/antagonistas & inhibidores , Factor de Transcripción Activador 3/genética , Factor de Transcripción Activador 3/metabolismo , Secuencia de Bases , Muerte Celular/efectos de los fármacos , Muerte Celular/genética , Línea Celular Tumoral , Proteína de Unión a Elemento de Respuesta al AMP Cíclico/metabolismo , Humanos , Proteínas de Dominio MADS/genética , Proteínas de Dominio MADS/metabolismo , Factores de Transcripción MEF2 , Datos de Secuencia Molecular , Factores Reguladores Miogénicos/genética , Factores Reguladores Miogénicos/metabolismo , Regiones Promotoras Genéticas , Acetato de Tetradecanoilforbol/farmacología , Transfección , Proteína p53 Supresora de Tumor/metabolismo
20.
Mutat Res ; 808: 74-82, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29079268

RESUMEN

Life-long blood regeneration relies on a rare population of self-renewing hematopoietic stem cells (HSCs). These cells' nearly unlimited self-renewal potential and lifetime persistence in the body signifies the need for tight control of their genome integrity. Their quiescent state, tightly linked with low metabolic activity, is one of the main strategies employed by HSCs to preserve an intact genome. On the other hand, HSCs need to be able to quickly respond to increased blood demands and rapidly increase their cellular output in order to fight infection-associated inflammation or extensive blood loss. This increase in proliferation rate, however, comes at the price of exposing HSCs to DNA damage inevitably associated with the process of DNA replication. Any interference with normal replication fork progression leads to a specialized molecular response termed replication stress (RS). Importantly, increased levels of RS are a hallmark feature of aged HSCs, where an accumulating body of evidence points to causative relationships between RS and the aging-associated impairment of the blood system's functional capacity. In this review, we present an overview of RS in HSCs focusing on its causes and consequences for the blood system of mice and men.


Asunto(s)
Daño del ADN , Reparación del ADN , Replicación del ADN , Células Madre Hematopoyéticas/patología , Animales , Diferenciación Celular , Proliferación Celular , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA