Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 22(11)2021 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-34071360

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive subtype of breast cancer lacking targeted therapy. Here, we evaluated the anti-cancer activity of APR-246, a P53 activator, and CX-5461, a RNA polymerase I inhibitor, in the treatment of TNBC cells. We tested the efficacy of individual and combination therapy of CX-5461 and APR-246 in vitro, using a panel of breast cancer cell lines. Using publicly available breast cancer datasets, we found that components of RNA Pol I are predominately upregulated in basal-like breast cancer, compared to other subtypes, and this upregulation is associated with poor overall and relapse-free survival. Notably, we found that the treatment of breast cancer cells lines with CX-5461 significantly hampered cell proliferation and synergistically enhanced the efficacy of APR-246. The combination treatment significantly induced apoptosis that is associated with cleaved PARP and Caspase 3 along with Annexin V positivity. Likewise, we also found that combination treatment significantly induced DNA damage and replication stress in these cells. Our data provide a novel combination strategy by utilizing APR-246 in combination CX-5461 in killing TNBC cells that can be further developed into more effective therapy in TNBC therapeutic armamentarium.


Asunto(s)
Benzotiazoles/farmacología , Daño del ADN , Replicación del ADN/efectos de los fármacos , Naftiridinas/farmacología , Quinuclidinas/farmacología , Apoptosis/efectos de los fármacos , Apoptosis/genética , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Línea Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Replicación del ADN/genética , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Células MCF-7 , ARN Polimerasa I/antagonistas & inhibidores , ARN Polimerasa I/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología
2.
Commun Biol ; 3(1): 593, 2020 10 21.
Artículo en Inglés | MEDLINE | ID: mdl-33087841

RESUMEN

High expression of centrosomal protein CEP55 has been correlated with clinico-pathological parameters across multiple human cancers. Despite significant in vitro studies and association of aberrantly overexpressed CEP55 with worse prognosis, its causal role in vivo tumorigenesis remains elusive. Here, using a ubiquitously overexpressing transgenic mouse model, we show that Cep55 overexpression causes spontaneous tumorigenesis and accelerates Trp53+/- induced tumours in vivo. At the cellular level, using mouse embryonic fibroblasts (MEFs), we demonstrate that Cep55 overexpression induces proliferation advantage by modulating multiple cellular signalling networks including the hyperactivation of the Pi3k/Akt pathway. Notably, Cep55 overexpressing MEFs have a compromised Chk1-dependent S-phase checkpoint, causing increased replication speed and DNA damage, resulting in a prolonged aberrant mitotic division. Importantly, this phenotype was rescued by pharmacological inhibition of Pi3k/Akt or expression of mutant Chk1 (S280A) protein, which is insensitive to regulation by active Akt, in Cep55 overexpressing MEFs. Moreover, we report that Cep55 overexpression causes stabilized microtubules. Collectively, our data demonstrates causative effects of deregulated Cep55 on genome stability and tumorigenesis which have potential implications for tumour initiation and therapy development.


Asunto(s)
Proteínas de Ciclo Celular/genética , Transformación Celular Neoplásica/genética , Expresión Génica , Inestabilidad Genómica , Animales , Biomarcadores de Tumor , Biopsia , Proteínas de Ciclo Celular/metabolismo , Línea Celular , Transformación Celular Neoplásica/metabolismo , Quinasa 1 Reguladora del Ciclo Celular (Checkpoint 1)/metabolismo , Susceptibilidad a Enfermedades , Fibroblastos/metabolismo , Genotipo , Inmunohistoquímica , Cariotipo , Ganglios Linfáticos/metabolismo , Ganglios Linfáticos/patología , Ratones , Ratones Transgénicos , Microtúbulos/metabolismo , Mitosis , Estabilidad Proteica , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Estrés Fisiológico , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
3.
Trends Mol Med ; 25(7): 595-611, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31078431

RESUMEN

Breast cancer is the most common cancer among women globally. Genomic instability (GI) refers to the increased tendency to accrue genomic alterations. It drives heterogeneity and is a hallmark of cancer. Genomic integrity is closely guarded by several mechanisms, including DNA damage checkpoints, the DNA repair machinery, and the mitotic checkpoint. Alterations in these surveillance mechanisms cause GI. In breast cancer, several pathways maintaining genomic integrity are distinctly altered, including some that have been successfully exploited for therapeutic targeting. In this review, we comprehensively discuss the recent advances on the mechanisms of GI in breast cancer, highlighting DNA repair defects and chromosome segregation errors during mitosis. We further review the clinical implications and therapeutic potential of targeting GI in the era of precision medicine.


Asunto(s)
Neoplasias de la Mama/etiología , Predisposición Genética a la Enfermedad , Variación Genética , Inestabilidad Genómica , Biomarcadores de Tumor , Neoplasias de la Mama/diagnóstico , Neoplasias de la Mama/terapia , Inestabilidad Cromosómica , Daño del ADN , Progresión de la Enfermedad , Femenino , Estudios de Asociación Genética , Humanos , Terapia Molecular Dirigida
4.
Oncogene ; 38(26): 5239-5249, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30971760

RESUMEN

Over 70% of human breast cancers are estrogen receptor-positive (ER+), most of which express MYB. In these and other cell types, the MYB transcription factor regulates the expression of many genes involved in cell proliferation, differentiation, tumorigenesis, and apoptosis. So far, no clear link has been established between MYB and the DNA damage response in breast cancer. Here, we found that silencing MYB in the ER+ breast cancer cell line MCF-7 led to increased DNA damage accumulation, as marked by increased γ-H2AX foci following induction of double-stranded breaks. We further found that this was likely mediated by decreased homologous recombination-mediated repair (HRR), since silencing MYB impaired the formation of RAD51 foci in response to DNA damage. Moreover, cells depleted for MYB exhibited reduced expression of several key genes involved in HRR including BRCA1, PALB2, and TOPBP1. Taken together, these data imply that MYB and its targets play an important role in the response of ER+ breast cancer cells to DNA damage, and suggest that induction of DNA damage along with inhibition of MYB activity could offer therapeutic benefits for ER+ breast cancer and possibly other cancer types.


Asunto(s)
Neoplasias de la Mama/genética , Daño del ADN/genética , Proteínas Proto-Oncogénicas c-myb/fisiología , Receptores de Estrógenos/genética , Reparación del ADN por Recombinación/genética , Proteína BRCA1/genética , Neoplasias de la Mama/patología , Línea Celular Tumoral , Reparación del ADN/genética , Femenino , Humanos , Células MCF-7 , Recombinasa Rad51/genética
5.
J Exp Clin Cancer Res ; 38(1): 85, 2019 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-30777101

RESUMEN

BACKGROUND: Despite the increasing progress in targeted and immune based-directed therapies for other solid organ malignancies, currently there is no targeted therapy available for TNBCs. A number of mechanisms have been reported both in pre-clinical and clinical settings that involve inherent, acquired and adaptive resistance to small molecule inhibitors. Here, we demonstrated a novel resistance mechanism in TNBC cells mediated by PDGFRß in response to JAK2 inhibition. METHODS: Multiple in vitro (subG1, western blotting, immunofluorescence, RT-PCR, Immunoprecipitation), in vivo and publically available datasets were used. RESULTS: We showed that TNBC cells exposed to MEK1/2-JAK2 inhibitors exhibit resistant colonies in anchorage-independent growth assays. Moreover, cells treated with various small molecule inhibitors including JAK2 promote PDGFRß upregulation. Using publically available databases, we showed that patients expressing high PDGFRß or its ligand PDGFB exhibit poor relapse-free survival upon chemotherapeutic treatment. Mechanistically we found that JAK2 expression controls steady state levels of PDGFRß. Thus, co-blockade of PDGFRß with JAK2 and MEK1/2 inhibitors completely eradicated resistant colonies in vitro. We found that triple-combined treatment had a significant impact on CD44+/CD24- stem-cell-like cells. Likewise, we found a significant tumor growth inhibition in vivo through intratumoral CD8+ T cells infiltration in a manner that is reversed by anti-CD8 antibody treatment. CONCLUSION: These findings reveal a novel regulatory role of JAK2-mediated PDGFRß proteolysis and provide an example of a PDGFRß-mediated resistance mechanism upon specific target inhibition in TNBC.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Resistencia a Antineoplásicos/fisiología , Janus Quinasa 2/metabolismo , Linfocitos Infiltrantes de Tumor/inmunología , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Neoplasias de la Mama Triple Negativas/patología , Femenino , Humanos , Inhibidores de las Cinasas Janus/farmacología , MAP Quinasa Quinasa 1/antagonistas & inhibidores , MAP Quinasa Quinasa 2/antagonistas & inhibidores , Neoplasias de la Mama Triple Negativas/inmunología , Neoplasias de la Mama Triple Negativas/metabolismo
6.
EMBO Mol Med ; 10(9)2018 09.
Artículo en Inglés | MEDLINE | ID: mdl-30108112

RESUMEN

The centrosomal protein, CEP55, is a key regulator of cytokinesis, and its overexpression is linked to genomic instability, a hallmark of cancer. However, the mechanism by which it mediates genomic instability remains elusive. Here, we showed that CEP55 overexpression/knockdown impacts survival of aneuploid cells. Loss of CEP55 sensitizes breast cancer cells to anti-mitotic agents through premature CDK1/cyclin B activation and CDK1 caspase-dependent mitotic cell death. Further, we showed that CEP55 is a downstream effector of the MEK1/2-MYC axis. Blocking MEK1/2-PLK1 signaling therefore reduced outgrowth of basal-like syngeneic and human breast tumors in in vivo models. In conclusion, high CEP55 levels dictate cell fate during perturbed mitosis. Forced mitotic cell death by blocking MEK1/2-PLK1 represents a potential therapeutic strategy for MYC-CEP55-dependent basal-like, triple-negative breast cancers.


Asunto(s)
Aneuploidia , Proteínas de Ciclo Celular/metabolismo , Citocinesis , Mitosis , Proteínas Nucleares/metabolismo , Neoplasias de la Mama/patología , Proteína Quinasa CDC2/metabolismo , Caspasas/metabolismo , Proteínas de Ciclo Celular/genética , Muerte Celular , Línea Celular Tumoral , Ciclina B/metabolismo , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Modelos Biológicos , Proteínas Nucleares/genética
7.
Sci Rep ; 7(1): 391, 2017 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-28341829

RESUMEN

USP9X, is highly expressed in neural progenitors and, essential for neural development in mice. In humans, mutations in USP9X are associated with neurodevelopmental disorders. To understand USP9X's role in neural progenitors, we studied the effects of altering its expression in both the human neural progenitor cell line, ReNcell VM, as well as neural stem and progenitor cells derived from Nestin-cre conditionally deleted Usp9x mice. Decreasing USP9X resulted in ReNcell VM cells arresting in G0 cell cycle phase, with a concomitant decrease in mTORC1 signalling, a major regulator of G0/G1 cell cycle progression. Decreased mTORC1 signalling was also observed in Usp9x-null neurospheres and embryonic mouse brains. Further analyses revealed, (i) the canonical mTORC1 protein, RAPTOR, physically associates with Usp9x in embryonic brains, (ii) RAPTOR protein level is directly proportional to USP9X, in both loss- and gain-of-function experiments in cultured cells and, (iii) USP9X deubiquitlyating activity opposes the proteasomal degradation of RAPTOR. EdU incorporation assays confirmed Usp9x maintains the proliferation of neural progenitors similar to Raptor-null and rapamycin-treated neurospheres. Interestingly, loss of Usp9x increased the number of sphere-forming cells consistent with enhanced neural stem cell self-renewal. To our knowledge, USP9X is the first deubiquitylating enzyme shown to stabilize RAPTOR.


Asunto(s)
Autorrenovación de las Células , Diana Mecanicista del Complejo 1 de la Rapamicina/metabolismo , Células-Madre Neurales/metabolismo , Proteína Reguladora Asociada a mTOR/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Puntos de Control del Ciclo Celular , Endopeptidasas/metabolismo , Células HEK293 , Humanos , Ratones , Proteolisis , Transducción de Señal
8.
Mol Oncol ; 11(5): 470-490, 2017 05.
Artículo en Inglés | MEDLINE | ID: mdl-28173629

RESUMEN

Activating KRAS mutations drive colorectal cancer tumorigenesis and influence response to anti-EGFR-targeted therapy. Despite recent advances in understanding Ras signaling biology and the revolution in therapies for melanoma using BRAF inhibitors, no targeted agents have been effective in KRAS-mutant cancers, mainly due to activation of compensatory pathways. Here, by leveraging the largest synthetic lethal genetic interactome in yeast, we identify that KRAS-mutated colorectal cancer cells have augmented homologous recombination repair (HRR) signaling. We found that KRAS mutation resulted in slowing and stalling of the replication fork and accumulation of DNA damage. Moreover, we found that KRAS-mutant HCT116 cells have an increase in MYC-mediated RAD51 expression with a corresponding increase in RAD51 recruitment to irradiation-induced DNA double-strand breaks (DSBs) compared to genetically complemented isogenic cells. MYC depletion using RNA interference significantly reduced IR-induced RAD51 foci formation and HRR. On the contrary, overexpression of either HA-tagged wild-type (WT) MYC or phospho-mutant S62A increased RAD51 protein levels and hence IR-induced RAD51 foci. Likewise, depletion of RAD51 selectively induced apoptosis in HCT116-mutant cells by increasing DSBs. Pharmacological inhibition targeting HRR signaling combined with PARP inhibition selectivity killed KRAS-mutant cells. Interestingly, these differences were not seen in a second isogenic pair of KRAS WT and mutant cells (DLD-1), likely due to their nondependency on the KRAS mutation for survival. Our data thus highlight a possible mechanism by which KRAS-mutant-dependent cells drive HRR in vitro by upregulating MYC-RAD51 expression. These data may offer a promising therapeutic vulnerability in colorectal cancer cells harboring otherwise nondruggable KRAS mutations, which warrants further investigation in vivo.


Asunto(s)
Neoplasias Colorrectales/genética , Recombinación Homóloga , Proteínas Proto-Oncogénicas p21(ras)/genética , Recombinasa Rad51/genética , Saccharomyces cerevisiae/genética , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias Colorrectales/tratamiento farmacológico , Roturas del ADN de Doble Cadena , Daño del ADN , Proteínas de Unión al ADN/genética , Relación Dosis-Respuesta a Droga , Receptores ErbB/genética , Células HCT116 , Humanos , Mutación , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , ARN Interferente Pequeño/genética , Recombinasa Rad51/metabolismo , Factores de Transcripción/genética
9.
Sci Rep ; 7: 39873, 2017 01 04.
Artículo en Inglés | MEDLINE | ID: mdl-28051153

RESUMEN

In breast cancer metastasis, the dynamic continuum involving pro- and anti-inflammatory regulators can become compromised. Over 600 genes have been implicated in metastasis to bone, lung or brain but how these genes might contribute to perturbation of immune function is poorly understood. To gain insight, we adopted a gene co-expression network approach that draws on the functional parallels between naturally occurring bone marrow-derived mesenchymal stem cells (BM-MSCs) and cancer stem cells (CSCs). Our network analyses indicate a key role for metastasis suppressor RARRES3, including potential to regulate the immunoproteasome (IP), a specialized proteasome induced under inflammatory conditions. Knockdown of RARRES3 in near-normal mammary epithelial and breast cancer cell lines increases overall transcript and protein levels of the IP subunits, but not of their constitutively expressed counterparts. RARRES3 mRNA expression is controlled by interferon regulatory factor IRF1, an inducer of the IP, and is sensitive to depletion of the retinoid-related receptor RORA that regulates various physiological processes including immunity through modulation of gene expression. Collectively, these findings identify a novel regulatory role for RARRES3 as an endogenous inhibitor of IP expression, and contribute to our evolving understanding of potential pathways underlying breast cancer driven immune modulation.


Asunto(s)
Complejo de la Endopetidasa Proteasomal/metabolismo , Receptores de Ácido Retinoico/metabolismo , Células de la Médula Ósea/citología , Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Línea Celular Tumoral , Femenino , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Factor 1 Regulador del Interferón/genética , Factor 1 Regulador del Interferón/metabolismo , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Metástasis de la Neoplasia/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/antagonistas & inhibidores , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/inmunología , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Receptores de Ácido Retinoico/antagonistas & inhibidores , Receptores de Ácido Retinoico/genética
10.
Vector Borne Zoonotic Dis ; 13(8): 545-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23930973

RESUMEN

Vector-borne rickettsial infection is a major cause of febrile illnesses throughout the world. Although vertebrates hosting the vectors play a vital role in the natural cycle of rickettsiae, studies have not been conducted on them in Sri Lanka. Therefore, the present study was designed to determine the exposure of dog population in Rajawatta, Thambavita, and areas of the Western Slopes and Unawatuna of Sri Lanka to rickettsial pathogens. A total of 123 dog blood samples were collected from those areas. Samples were tested for antibodies against Rickettsia conorii (RC) of the spotted fever group (SFG), Rickettsia typhi (RT) of the typhus group (TG), and Orientia tsutsugamushi (OT) of the scrub typhus group (ST) of rickettsiae by indirect immunofluorescence antibody test (IFA). Samples with titers ≥ 1:64 were considered as positive in this study. Collectively, 49% dogs were found to have antibodies against the rickettsial agents. Of the dogs, 42%, 24%, and 2% had antibodies against RC, OT, and RT, respectively. The seropositive rate of 100% was observed in areas of the Western Slopes, whereas the lowest rate of 20% was in Unawatuna. Among the positive samples, antibody titers against RC and OT ranged from 1/64 to 1/8192. In contrast, the few dogs that tested positive for RT showed very low titers of 1/64 and 1/128. Results of this study show the extent of exposure to the pathogen and its dispersion in the natural ecology. We suggest that dogs could be acting as reservoirs in the rickettsial transmission cycle or could be effective tracer animals that can be used to detect areas with potential for future outbreaks.


Asunto(s)
Enfermedades de los Perros/microbiología , Orientia tsutsugamushi/inmunología , Infecciones por Rickettsia/veterinaria , Rickettsia conorii/inmunología , Rickettsia typhi/inmunología , Tifus por Ácaros/veterinaria , Animales , Anticuerpos Antibacterianos/sangre , Fiebre Botonosa/epidemiología , Fiebre Botonosa/veterinaria , Chlorocebus aethiops , Reservorios de Enfermedades , Enfermedades de los Perros/epidemiología , Perros , Femenino , Técnica del Anticuerpo Fluorescente Indirecta/veterinaria , Humanos , Masculino , Orientia tsutsugamushi/aislamiento & purificación , Infecciones por Rickettsia/epidemiología , Rickettsia conorii/aislamiento & purificación , Rickettsia typhi/aislamiento & purificación , Tifus por Ácaros/epidemiología , Estudios Seroepidemiológicos , Sri Lanka/epidemiología , Tifus Endémico Transmitido por Pulgas/epidemiología , Tifus Endémico Transmitido por Pulgas/veterinaria , Células Vero
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...