Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
Cell ; 167(1): 260-274.e22, 2016 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-27641504

RESUMEN

The inter- and intra-tumor heterogeneity of breast cancer needs to be adequately captured in pre-clinical models. We have created a large collection of breast cancer patient-derived tumor xenografts (PDTXs), in which the morphological and molecular characteristics of the originating tumor are preserved through passaging in the mouse. An integrated platform combining in vivo maintenance of these PDTXs along with short-term cultures of PDTX-derived tumor cells (PDTCs) was optimized. Remarkably, the intra-tumor genomic clonal architecture present in the originating breast cancers was mostly preserved upon serial passaging in xenografts and in short-term cultured PDTCs. We assessed drug responses in PDTCs on a high-throughput platform and validated several ex vivo responses in vivo. The biobank represents a powerful resource for pre-clinical breast cancer pharmacogenomic studies (http://caldaslab.cruk.cam.ac.uk/bcape), including identification of biomarkers of response or resistance.


Asunto(s)
Bancos de Muestras Biológicas , Neoplasias de la Mama , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Biomarcadores Farmacológicos , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Resistencia a Antineoplásicos/genética , Femenino , Ensayos Analíticos de Alto Rendimiento , Humanos , Ratones , Pruebas de Farmacogenómica , Células Tumorales Cultivadas
2.
Neuroimage ; 280: 120312, 2023 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-37574120

RESUMEN

Activity-dependent myelination is a fundamental mode of brain plasticity which significantly influences network function. We recently discovered that absence seizures, which occur in multiple forms of generalized epilepsy, can induce activity-dependent myelination, which in turn promotes further progression of epilepsy. Structural alterations of myelin are likely to be widespread, given that absence seizures arise from an extensive thalamocortical network involving frontoparietal regions of the bilateral hemispheres. However, the temporal course and spatial extent of myelin plasticity is unknown, due to limitations of gold-standard histological methods such as electron microscopy (EM). In this study, we leveraged magnetization transfer and diffusion MRI for estimation of g-ratios across major white matter tracts in a mouse model of generalized epilepsy with progressive absence seizures. EM was performed on the same brains after MRI. After seizure progression, we found increased myelination (decreased g-ratios) throughout the anterior portion (genu-to-body) of the corpus callosum but not in the posterior portion (body-splenium) nor in the fornix or the internal capsule. Curves obtained from averaging g-ratio values at every longitudinal point of the corpus callosum were statistically different with p<0.001. Seizure-associated myelin differences found in the corpus callosum body with MRI were statistically significant (p = 0.0027) and were concordant with EM in the same region (p = 0.01). Notably, these differences were not detected by diffusion tensor imaging. This study reveals widespread myelin structural change that is specific to the absence seizure network. Furthermore, our findings demonstrate the potential utility and importance of MRI-based g-ratio estimation to non-invasively detect myelin plasticity.


Asunto(s)
Imagen de Difusión Tensora , Epilepsia Generalizada , Animales , Ratones , Imagen por Resonancia Magnética , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Cuerpo Calloso/patología , Convulsiones/diagnóstico por imagen
3.
BMC Genomics ; 19(1): 19, 2018 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-29304755

RESUMEN

BACKGROUND: Patient-Derived Tumour Xenografts (PDTXs) have emerged as the pre-clinical models that best represent clinical tumour diversity and intra-tumour heterogeneity. The molecular characterization of PDTXs using High-Throughput Sequencing (HTS) is essential; however, the presence of mouse stroma is challenging for HTS data analysis. Indeed, the high homology between the two genomes results in a proportion of mouse reads being mapped as human. RESULTS: In this study we generated Whole Exome Sequencing (WES), Reduced Representation Bisulfite Sequencing (RRBS) and RNA sequencing (RNA-seq) data from samples with known mixtures of mouse and human DNA or RNA and from a cohort of human breast cancers and their derived PDTXs. We show that using an In silico Combined human-mouse Reference Genome (ICRG) for alignment discriminates between human and mouse reads with up to 99.9% accuracy and decreases the number of false positive somatic mutations caused by misalignment by >99.9%. We also derived a model to estimate the human DNA content in independent PDTX samples. For RNA-seq and RRBS data analysis, the use of the ICRG allows dissecting computationally the transcriptome and methylome of human tumour cells and mouse stroma. In a direct comparison with previously reported approaches, our method showed similar or higher accuracy while requiring significantly less computing time. CONCLUSIONS: The computational pipeline we describe here is a valuable tool for the molecular analysis of PDTXs as well as any other mixture of DNA or RNA species.


Asunto(s)
Genómica/métodos , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Perfilación de la Expresión Génica , Humanos , Ratones , Mutación , Alineación de Secuencia , Análisis de Secuencia de ADN , Análisis de Secuencia de ARN
5.
J Immunother Cancer ; 11(11)2023 11.
Artículo en Inglés | MEDLINE | ID: mdl-37963637

RESUMEN

BACKGROUND: The metabolism of tryptophan to kynurenines (KYN) by indoleamine-2,3-dioxygenase or tryptophan-2,3-dioxygenase is a key pathway of constitutive and adaptive tumor immune resistance. The immunosuppressive effects of KYN in the tumor microenvironment are predominantly mediated by the aryl hydrocarbon receptor (AhR), a cytosolic transcription factor that broadly suppresses immune cell function. Inhibition of AhR thus offers an antitumor therapy opportunity via restoration of immune system functions. METHODS: The expression of AhR was evaluated in tissue microarrays of head and neck squamous cell carcinoma (HNSCC), non-small cell lung cancer (NSCLC) and colorectal cancer (CRC). A structure class of inhibitors that block AhR activation by exogenous and endogenous ligands was identified, and further optimized, using a cellular screening cascade. The antagonistic properties of the selected AhR inhibitor candidate BAY 2416964 were determined using transactivation assays. Nuclear translocation, target engagement and the effect of BAY 2416964 on agonist-induced AhR activation were assessed in human and mouse cancer cells. The immunostimulatory properties on gene and cytokine expression were examined in human immune cell subsets. The in vivo efficacy of BAY 2416964 was tested in the syngeneic ovalbumin-expressing B16F10 melanoma model in mice. Coculture of human H1299 NSCLC cells, primary peripheral blood mononuclear cells and fibroblasts mimicking the human stromal-tumor microenvironment was used to assess the effects of AhR inhibition on human immune cells. Furthermore, tumor spheroids cocultured with tumor antigen-specific MART-1 T cells were used to study the antigen-specific cytotoxic T cell responses. The data were analyzed statistically using linear models. RESULTS: AhR expression was observed in tumor cells and tumor-infiltrating immune cells in HNSCC, NSCLC and CRC. BAY 2416964 potently and selectively inhibited AhR activation induced by either exogenous or endogenous AhR ligands. In vitro, BAY 2416964 restored immune cell function in human and mouse cells, and furthermore enhanced antigen-specific cytotoxic T cell responses and killing of tumor spheroids. In vivo, oral application with BAY 2416964 was well tolerated, induced a proinflammatory tumor microenvironment, and demonstrated antitumor efficacy in a syngeneic cancer model in mice. CONCLUSIONS: These findings identify AhR inhibition as a novel therapeutic approach to overcome immune resistance in various types of cancers.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas , Dioxigenasas , Neoplasias de Cabeza y Cuello , Neoplasias Pulmonares , Humanos , Ratones , Animales , Triptófano , Receptores de Hidrocarburo de Aril/genética , Carcinoma de Pulmón de Células no Pequeñas/tratamiento farmacológico , Leucocitos Mononucleares/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Neoplasias Pulmonares/tratamiento farmacológico , Quinurenina/metabolismo , Inmunoterapia , Factores Inmunológicos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Microambiente Tumoral
6.
Nat Rev Neurol ; 18(12): 735-746, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36376595

RESUMEN

Within the past decade, multiple lines of evidence have converged to identify a critical role for activity-regulated myelination in tuning the function of neural networks. In this Review, we provide an overview of accumulating evidence that activity-regulated myelination is required for brain adaptation and learning across multiple domains. We then discuss dysregulation of activity-dependent myelination in the context of neurological disease, a novel frontier with the potential to uncover new mechanisms of disease pathogenesis and to develop new therapeutic strategies. Alterations in myelination and neural network function can result from deficient myelin plasticity that impairs neurological function or from maladaptive myelination, in which intact activity-dependent myelination contributes to the disease process by promoting pathological patterns of neuronal activity. These emerging mechanisms suggest new avenues for therapeutic intervention that could more fully address the complex interactions between neurons and oligodendroglia.


Asunto(s)
Vaina de Mielina , Oligodendroglía , Humanos , Vaina de Mielina/fisiología , Neuronas/fisiología , Encéfalo/fisiología
7.
Nat Neurosci ; 25(5): 596-606, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35501379

RESUMEN

Activity-dependent myelination can fine-tune neural network dynamics. Conversely, aberrant neuronal activity, as occurs in disorders of recurrent seizures (epilepsy), could promote maladaptive myelination, contributing to pathogenesis. In this study, we tested the hypothesis that activity-dependent myelination resulting from absence seizures, which manifest as frequent behavioral arrests with generalized electroencephalography (EEG) spike-wave discharges, promote thalamocortical network hypersynchrony and contribute to epilepsy progression. We found increased oligodendrogenesis and myelination specifically within the seizure network in two models of generalized epilepsy with absence seizures (Wag/Rij rats and Scn8a+/mut mice), evident only after epilepsy onset. Aberrant myelination was prevented by pharmacological seizure inhibition in Wag/Rij rats. Blocking activity-dependent myelination decreased seizure burden over time and reduced ictal synchrony as assessed by EEG coherence. These findings indicate that activity-dependent myelination driven by absence seizures contributes to epilepsy progression; maladaptive myelination may be pathogenic in some forms of epilepsy and other neurological diseases.


Asunto(s)
Epilepsia Tipo Ausencia , Epilepsia Generalizada , Animales , Modelos Animales de Enfermedad , Electroencefalografía , Epilepsia Generalizada/genética , Ratones , Canal de Sodio Activado por Voltaje NAV1.6 , Ratas , Ratas Wistar , Convulsiones
8.
Nat Commun ; 12(1): 5406, 2021 09 13.
Artículo en Inglés | MEDLINE | ID: mdl-34518533

RESUMEN

DNA methylation is aberrant in cancer, but the dynamics, regulatory role and clinical implications of such epigenetic changes are still poorly understood. Here, reduced representation bisulfite sequencing (RRBS) profiles of 1538 breast tumors and 244 normal breast tissues from the METABRIC cohort are reported, facilitating detailed analysis of DNA methylation within a rich context of genomic, transcriptional, and clinical data. Tumor methylation from immune and stromal signatures are deconvoluted leading to the discovery of a tumor replication-linked clock with genome-wide methylation loss in non-CpG island sites. Unexpectedly, methylation in most tumor CpG islands follows two replication-independent processes of gain (MG) or loss (ML) that we term epigenomic instability. Epigenomic instability is correlated with tumor grade and stage, TP53 mutations and poorer prognosis. After controlling for these global trans-acting trends, as well as for X-linked dosage compensation effects, cis-specific methylation and expression correlations are uncovered at hundreds of promoters and over a thousand distal elements. Some of these targeted known tumor suppressors and oncogenes. In conclusion, this study demonstrates that global epigenetic instability can erode cancer methylomes and expose them to localized methylation aberrations in-cis resulting in transcriptional changes seen in tumors.


Asunto(s)
Neoplasias de la Mama/genética , Metilación de ADN , Epigénesis Genética , Epigenómica/métodos , Regulación Neoplásica de la Expresión Génica , Estudios de Cohortes , Islas de CpG/genética , Replicación del ADN/genética , Femenino , Genoma Humano/genética , Inestabilidad Genómica/genética , Genómica/métodos , Humanos , Células MCF-7 , Mutación , Regiones Promotoras Genéticas/genética , Análisis de Supervivencia
9.
Eur J Pharmacol ; 883: 173377, 2020 Sep 15.
Artículo en Inglés | MEDLINE | ID: mdl-32687920

RESUMEN

Lithium, commonly used to treat bipolar disorder, potentiates the ability of the muscarinic agonist pilocarpine to induce seizures in rodents. As this potentiation by lithium is reversed by the administration of myo-inositol, the potentiation may be mediated by inhibition of inositol monophosphatase (IMPase), a known target of lithium. Recently, we demonstrated that ebselen is a 'lithium mimetic' in regard to behaviours in both mice and man. Ebselen inhibits IMPase in vitro and lowers myo-inositol in vivo in the brains of mice and men, making ebselen the only known inhibitor of IMPase, other than lithium, that penetrates the blood-brain barrier. Our objective was to determine the effects of ebselen on sensitization to pilocarpine-induced seizures and neural activity. We administered ebselen at different doses and time intervals to mice, followed by injection of a sub-seizure dose of pilocarpine. We assessed seizure and neural activity by a subjective seizure rating scale, by monitoring tremors, and by induction of the immediate early gene c-fos. In contrast to lithium, ebselen did not potentiate the ability of pilocarpine to induce seizures. Unexpectedly, ebselen inhibited pilocarpine-induced tremor as well as pilocarpine-induced increases in c-fos mRNA levels. Both lithium and ebselen inhibit a common target, IMPase, but only lithium potentiates pilocarpine-induced seizures, consistent with their polypharmacology at diverse molecular targets. We conclude that ebselen does not potentiate pilocarpine-induced seizures and instead, reduces pilocarpine-mediated neural activation. This lack of potentiation of muscarinic sensitization may be one reason for the lack of side-effects observed with ebselen treatment clinically.


Asunto(s)
Anticonvulsivantes/farmacología , Azoles/farmacología , Encéfalo/efectos de los fármacos , Cloruro de Litio/toxicidad , Neuronas/efectos de los fármacos , Compuestos de Organoselenio/farmacología , Pilocarpina , Convulsiones/prevención & control , Animales , Anticonvulsivantes/toxicidad , Azoles/toxicidad , Encéfalo/metabolismo , Encéfalo/fisiopatología , Células CHO , Señalización del Calcio/efectos de los fármacos , Cricetulus , Modelos Animales de Enfermedad , Fosfatos de Inositol/metabolismo , Isoindoles , Masculino , Ratones , Neuronas/metabolismo , Compuestos de Organoselenio/toxicidad , Monoéster Fosfórico Hidrolasas/antagonistas & inhibidores , Monoéster Fosfórico Hidrolasas/metabolismo , Proteínas Proto-Oncogénicas c-fos/genética , Proteínas Proto-Oncogénicas c-fos/metabolismo , Receptores Muscarínicos/efectos de los fármacos , Receptores Muscarínicos/genética , Receptores Muscarínicos/metabolismo , Convulsiones/inducido químicamente , Convulsiones/metabolismo , Convulsiones/fisiopatología
10.
Cancer Cell ; 38(4): 516-533.e9, 2020 10 12.
Artículo en Inglés | MEDLINE | ID: mdl-32976773

RESUMEN

PIK3CA, encoding the PI3Kα isoform, is the most frequently mutated oncogene in estrogen receptor (ER)-positive breast cancer. Isoform-selective PI3K inhibitors are used clinically but intrinsic and acquired resistance limits their utility. Improved selection of patients that will benefit from these drugs requires predictive biomarkers. We show here that persistent FOXM1 expression following drug treatment is a biomarker of resistance to PI3Kα inhibition in ER+ breast cancer. FOXM1 drives expression of lactate dehydrogenase (LDH) but not hexokinase 2 (HK-II). The downstream metabolic changes can therefore be detected using MRI of LDH-catalyzed hyperpolarized 13C label exchange between pyruvate and lactate but not by positron emission tomography measurements of HK-II-mediated trapping of the glucose analog 2-deoxy-2-[18F]fluorodeoxyglucose. Rapid assessment of treatment response in breast cancer using this imaging method could help identify patients that benefit from PI3Kα inhibition and design drug combinations to counteract the emergence of resistance.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Fosfatidilinositol 3-Quinasa Clase I/antagonistas & inhibidores , Proteína Forkhead Box M1/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Resistencia a Antineoplásicos/genética , Femenino , Proteína Forkhead Box M1/genética , Fulvestrant/administración & dosificación , Humanos , Imidazoles/administración & dosificación , Células MCF-7 , Imagen por Resonancia Magnética/métodos , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Oxazepinas/administración & dosificación , Receptores de Estrógenos/metabolismo , Tamoxifeno/administración & dosificación , Ensayos Antitumor por Modelo de Xenoinjerto/métodos
11.
Anaesth Crit Care Pain Med ; 38(1): 41-45, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-29605459

RESUMEN

PURPOSE: Analgesic efficacy of ultrasound-guided transverse abdominis plane block, administered a little more medially, just close to the origin of the transverse abdominis muscle has not yet been investigated in patients undergoing unilateral inguinal hernia repair. We hypothesised that medial transverse abdominis plane block would provide comparable postoperative analgesia to ilioinguinal-iliohypogastric nerve block in inguinal hernia repair patients. METHODS: This prospective, randomised trial was conducted in 50 ASA I and II male patients≥18 years of age. Patients were randomised into two groups to receive either pre-incisional ipsilateral ultrasound-guided ilioinguinal-iliohypogastric nerve block or medial transverse abdominis plane block, with 0.3ml/kg of 0.25% bupivacaine. Our primary objective was postoperative 24-hour analgesic consumption and secondary outcomes included pain scores, time to first request for rescue analgesic and side effects, if any, in the postoperative period. RESULTS: There was no significant difference in the total postoperative analgesic consumption [group I: 66.04mg; group II: 68.33mg (P value 0.908)]. Time to first request for rescue analgesic was delayed, though statistically non-significant (P value 0.326), following medial transverse abdominis plane block, with excellent pain relief seen in 58.3% patients as opposed to 45.8% patients in ilioinguinal-iliohypogastric nerve block group. CONCLUSION: Medial transverse abdominis plane block being a novel, simple and easily performed procedure can serve as an useful alternative to ilioinguinal-iliohypogastric nerve block for providing postoperative pain relief in inguinal hernia repair patients.


Asunto(s)
Hernia Inguinal/cirugía , Herniorrafia/métodos , Bloqueo Nervioso/métodos , Dolor Postoperatorio/terapia , Ultrasonografía Intervencional/métodos , Músculos Abdominales/diagnóstico por imagen , Adulto , Anestésicos Locales/administración & dosificación , Bupivacaína/administración & dosificación , Herniorrafia/efectos adversos , Humanos , Masculino , Persona de Mediana Edad , Bloqueo Nervioso/estadística & datos numéricos , Dimensión del Dolor , Estudios Prospectivos , Factores de Tiempo
12.
EMBO Mol Med ; 11(7): e9982, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31273933

RESUMEN

Due to compromised homologous recombination (HR) repair, BRCA1- and BRCA2-mutated tumours accumulate DNA damage and genomic rearrangements conducive of tumour progression. To identify drugs that target specifically BRCA2-deficient cells, we screened a chemical library containing compounds in clinical use. The top hit was chlorambucil, a bifunctional alkylating agent used for the treatment of chronic lymphocytic leukaemia (CLL). We establish that chlorambucil is specifically toxic to BRCA1/2-deficient cells, including olaparib-resistant and cisplatin-resistant ones, suggesting the potential clinical use of chlorambucil against disease which has become resistant to these drugs. Additionally, chlorambucil eradicates BRCA2-deficient xenografts and inhibits growth of olaparib-resistant patient-derived tumour xenografts (PDTXs). We demonstrate that chlorambucil inflicts replication-associated DNA double-strand breaks (DSBs), similarly to cisplatin, and we identify ATR, FANCD2 and the SNM1A nuclease as determinants of sensitivity to both drugs. Importantly, chlorambucil is substantially less toxic to normal cells and tissues in vitro and in vivo relative to cisplatin. Because chlorambucil and cisplatin are equally effective inhibitors of BRCA2-compromised tumours, our results indicate that chlorambucil has a higher therapeutic index than cisplatin in targeting BRCA-deficient tumours.


Asunto(s)
Proteína BRCA1/deficiencia , Proteína BRCA2/deficiencia , Clorambucilo/farmacología , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Receptores Activados del Proliferador del Peroxisoma/antagonistas & inhibidores , Ftalazinas/farmacología , Piperazinas/farmacología , Animales , Línea Celular Tumoral , Cricetinae , Resistencia a Antineoplásicos/genética , Humanos , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/metabolismo , Masculino , Ratones , Ratones SCID , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Sci Rep ; 9(1): 1957, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30760795

RESUMEN

Chimeric Antigen Receptor-T (CAR-T) cell immunotherapy has produced dramatic responses in hematologic malignancies. One of the challenges in the field is the lack of a simple assay for the detection of CARs on the surface of immune effector cells. In this study, we describe a novel luciferase-based assay, termed Topanga Assay, for the detection of CAR expression. The assay utilizes a recombinant fusion protein, called Topanga reagent, generated by joining the extra-cellular domain of a CAR-target in frame with one of the marine luciferases or their engineered derivatives. The assay involves incubation of CAR expressing cells with the Topanga reagent, a few washes and measurement of luminescence. The assay can detect CARs comprising either immunoglobulin- or non-immunoglobulin-based antigen binding domains. We further demonstrate that addition of epitope tags to the Topanga reagent not only allows its convenient one step purification but also extends its use for detection of CAR cells using flow cytometry. However, crude supernatant containing the secreted Topanga reagent can be directly used in both luminescence and flow-cytometry based assays without prior protein purification. Our results demonstrate that the Topanga assay is a highly sensitive, specific, convenient, economical and versatile assay for the detection of CARs.


Asunto(s)
Inmunoterapia Adoptiva/métodos , Luciferasas/metabolismo , Receptores Quiméricos de Antígenos/análisis , Línea Celular , Citometría de Flujo/métodos , Humanos , Linfocitos/metabolismo , Receptores de Antígenos/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Quiméricos de Antígenos/metabolismo , Proteínas Recombinantes de Fusión/metabolismo , Linfocitos T/inmunología
14.
Cell Res ; 28(7): 719-729, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29795445

RESUMEN

Activation of the mitogen-activated protein kinase (MAPK) pathway is frequent in cancer. Drug development efforts have been focused on kinases in this pathway, most notably on RAF and MEK. We show here that MEK inhibition activates JNK-JUN signaling through suppression of DUSP4, leading to activation of HER Receptor Tyrosine Kinases. This stimulates the MAPK pathway in the presence of drug, thereby blunting the effect of MEK inhibition. Cancers that have lost MAP3K1 or MAP2K4 fail to activate JNK-JUN. Consequently, loss-of-function mutations in either MAP3K1 or MAP2K4 confer sensitivity to MEK inhibition by disabling JNK-JUN-mediated feedback loop upon MEK inhibition. In a panel of 168 Patient Derived Xenograft (PDX) tumors, MAP3K1 and MAP2K4 mutation status is a strong predictor of response to MEK inhibition. Our findings suggest that cancers having mutations in MAP3K1 or MAP2K4, which are frequent in tumors of breast, prostate and colon, may respond to MEK inhibitors. Our findings also suggest that MAP3K1 and MAP2K4 are potential drug targets in combination with MEK inhibitors, in spite of the fact that they are encoded by tumor suppressor genes.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Neoplasias del Colon/tratamiento farmacológico , Resistencia a Antineoplásicos/genética , MAP Quinasa Quinasa 4/genética , Quinasa 1 de Quinasa de Quinasa MAP/genética , Neoplasias de la Próstata/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/uso terapéutico , Animales , Bencimidazoles/farmacología , Bencimidazoles/uso terapéutico , Neoplasias de la Mama/genética , Línea Celular Tumoral , Neoplasias del Colon/genética , Femenino , Xenoinjertos , Humanos , Mutación con Pérdida de Función , MAP Quinasa Quinasa 4/antagonistas & inhibidores , Quinasa 1 de Quinasa de Quinasa MAP/antagonistas & inhibidores , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Ratones Endogámicos BALB C , Ratones Desnudos , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neoplasias de la Próstata/genética , Inhibidores de Proteínas Quinasas/farmacología
15.
Exp Biol Med (Maywood) ; 242(4): 436-440, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27895094

RESUMEN

MicroRNA-122 (miR-122) is liver specific and plays an important role in physiology as well as diseases including hepatocellular carcinoma (HCC). Downregulation of miR-122 in HCC modulates apoptosis. Similarly, the putative targets of miR-122, the forkhead box (FOX) family genes also play an important role in the regulation of apoptosis. Hence, an interplay between miR-122 and FOX family genes has been explored in this study. Initially, an augmentation of apoptosis was noticed in HepG2 cells after transfection with miR-122. Further, the predicted miR-122 targets, the FOX family genes ( FOXM1b, FOXP1, and FOXO4) were selected via in silico analysis based on their role in apoptosis. We checked the expression of all these genes at transcript level after the transfection of miR-122 and found that the relative expression of FOXP1 and FOXM1b was significantly downregulated (p < 0.005) and that of FOXO4 was upregulated (p < 0.005). Thus, the finding indicates deregulation of these FOX genes as a result of miR-122 augmentation might be involved in the modulation of apoptosis.


Asunto(s)
Apoptosis/genética , Carcinoma Hepatocelular/patología , Proteína Forkhead Box M1/genética , Factores de Transcripción Forkhead/genética , Neoplasias Hepáticas/patología , MicroARNs/genética , Proteínas Represoras/genética , Factores de Transcripción/genética , Carcinoma Hepatocelular/genética , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Proteína Forkhead Box M1/biosíntesis , Factores de Transcripción Forkhead/biosíntesis , Regulación Neoplásica de la Expresión Génica/genética , Células Hep G2 , Humanos , Hígado/patología , Neoplasias Hepáticas/genética , Proteínas Represoras/biosíntesis , Factores de Transcripción/biosíntesis , Transfección/métodos
16.
Anesth Essays Res ; 11(2): 309-313, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28663612

RESUMEN

BACKGROUND: The basic principle of neuroanesthesia is to provide hemodynamic stability, provision of optimal operative conditions, maintenance of cerebral perfusion pressure, and cerebral oxygenation. AIM: This study was undertaken to see the effect of dexmedetomidine infusion on hemodynamics and its ability to act as an anesthetic adjuvant in patients undergoing supratentorial tumor surgery. SETTING AND DESIGN: Prospective randomized control double blind study. SUBJECTS AND METHODS: In this study, we compared two groups with 25 patients in each group. Group C patients received saline infusion during surgery and 4 µg/kg of fentanyl intravenously (i.v.) at the induction and at pin head application. Group D patients received dexmedetomidine infusion during surgery at the rate of 0.4 µg/kg/h and 2 µg/kg of fentanyl i.v. at the induction and at pin head application. STATISTICAL ANALYSES USED: Parametric data were analyzed using Student's t-test. The categorical data were studied using Chi-squared test or Fisher's test as appropriate. RESULTS: The vitals remained within 20% of baseline in both groups during the study period except at the time of extubation where the rise in heart rate was more than 20% in control group. The requirement of thiopentone for induction was significantly less in dexmedetomidine group. In dexmedetomidine group, less number of patients required intraoperative fentanyl (P < 0.05), and the time to rescue analgesic was also more in Group D (P < 0.05). CONCLUSION: Dexmedetomidine infusion started before surgery maintains hemodynamic stability intraoperatively and is effective in attenuating the cardiovascular responses to intubation, skull pin application, and extubation. It decreases the requirement of other anesthetic agents as well.

17.
EMBO Mol Med ; 9(10): 1398-1414, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28729482

RESUMEN

Maintenance of genome integrity requires the functional interplay between Fanconi anemia (FA) and homologous recombination (HR) repair pathways. Endogenous acetaldehyde, a product of cellular metabolism, is a potent source of DNA damage, particularly toxic to cells and mice lacking the FA protein FANCD2. Here, we investigate whether HR-compromised cells are sensitive to acetaldehyde, similarly to FANCD2-deficient cells. We demonstrate that inactivation of HR factors BRCA1, BRCA2, or RAD51 hypersensitizes cells to acetaldehyde treatment, in spite of the FA pathway being functional. Aldehyde dehydrogenases (ALDHs) play key roles in endogenous acetaldehyde detoxification, and their chemical inhibition leads to cellular acetaldehyde accumulation. We find that disulfiram (Antabuse), an ALDH2 inhibitor in widespread clinical use for the treatment of alcoholism, selectively eliminates BRCA1/2-deficient cells. Consistently, Aldh2 gene inactivation suppresses proliferation of HR-deficient mouse embryonic fibroblasts (MEFs) and human fibroblasts. Hypersensitivity of cells lacking BRCA2 to acetaldehyde stems from accumulation of toxic replication-associated DNA damage, leading to checkpoint activation, G2/M arrest, and cell death. Acetaldehyde-arrested replication forks require BRCA2 and FANCD2 for protection against MRE11-dependent degradation. Importantly, acetaldehyde specifically inhibits in vivo the growth of BRCA1/2-deficient tumors and ex vivo in patient-derived tumor xenograft cells (PDTCs), including those that are resistant to poly (ADP-ribose) polymerase (PARP) inhibitors. The work presented here therefore identifies acetaldehyde metabolism as a potential therapeutic target for the selective elimination of BRCA1/2-deficient cells and tumors.


Asunto(s)
Acetaldehído/metabolismo , Proteína BRCA1/metabolismo , Proteína BRCA2/metabolismo , Recombinasa Rad51/metabolismo , Aldehído Deshidrogenasa Mitocondrial/genética , Aldehído Deshidrogenasa Mitocondrial/metabolismo , Animales , Proteína BRCA1/genética , Proteína BRCA2/genética , Línea Celular Tumoral , Daño del ADN , Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación D2 de la Anemia de Fanconi/metabolismo , Fibroblastos , Recombinación Homóloga , Humanos , Ratones , Ratones Desnudos , Recombinasa Rad51/genética , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Endocr Relat Cancer ; 23(12): T259-T270, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27702751

RESUMEN

Despite remarkable advances in our understanding of the drivers of human malignancies, new targeted therapies often fail to show sufficient efficacy in clinical trials. Indeed, the cost of bringing a new agent to market has risen substantially in the last several decades, in part fuelled by extensive reliance on preclinical models that fail to accurately reflect tumour heterogeneity. To halt unsustainable rates of attrition in the drug discovery process, we must develop a new generation of preclinical models capable of reflecting the heterogeneity of varying degrees of complexity found in human cancers. Patient-derived tumour xenograft (PDTX) models prevail as arguably the most powerful in this regard because they capture cancer's heterogeneous nature. Herein, we review current breast cancer models and their use in the drug discovery process, before discussing best practices for developing a highly annotated cohort of PDTX models. We describe the importance of extensive multidimensional molecular and functional characterisation of models and combination drug-drug screens to identify complex biomarkers of drug resistance and response. We reflect on our own experiences and propose the use of a cost-effective intermediate pharmacogenomic platform (the PDTX-PDTC platform) for breast cancer drug and biomarker discovery. We discuss the limitations and unanswered questions of PDTX models; yet, still strongly envision that their use in basic and translational research will dramatically change our understanding of breast cancer biology and how to more effectively treat it.


Asunto(s)
Antineoplásicos/aislamiento & purificación , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Descubrimiento de Drogas/métodos , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Medicina de Precisión/métodos , Ensayos Antitumor por Modelo de Xenoinjerto , Animales , Neoplasias de la Mama/patología , Femenino , Humanos , Neoplasias Mamarias Experimentales/patología , Trasplante de Neoplasias/métodos
19.
Cell Rep ; 13(11): 2480-2490, 2015 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-26686634

RESUMEN

The transforming growth factor beta (TGF-ß) signaling pathway exerts opposing effects on cancer cells, acting as either a tumor promoter or a tumor suppressor. Here, we show that these opposing effects are a result of the synergy between SMAD3, a downstream effector of TGF-ß signaling, and the distinct epigenomes of breast-tumor-initiating cells (BTICs). These effects of TGF-ß are associated with distinct gene expression programs, but genomic SMAD3 binding patterns are highly similar in the BTIC-promoting and BTIC-suppressing contexts. Our data show cell-type-specific patterns of DNA and histone modifications provide a modulatory layer by determining accessibility of genes to regulation by TGF-ß/SMAD3. LBH, one such context-specific target gene, is regulated according to its DNA methylation status and is crucial for TGF-ß-dependent promotion of BTICs. Overall, these results reveal that the epigenome plays a central and previously overlooked role in shaping the context-specific effects of TGF-ß in cancer.


Asunto(s)
Transducción de Señal/efectos de los fármacos , Proteína smad3/metabolismo , Factor de Crecimiento Transformador beta/farmacología , Sitios de Unión , Línea Celular Tumoral , ADN/química , ADN/metabolismo , Metilación de ADN , Epigénesis Genética , Histonas/metabolismo , Humanos , Células Madre Neoplásicas/citología , Células Madre Neoplásicas/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Proteína Smad2/metabolismo , Transactivadores/antagonistas & inhibidores , Transactivadores/genética , Transactivadores/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda