Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cells Tissues Organs ; 2022 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-35970135

RESUMEN

High-grade serous ovarian carcinoma (HGSC) is associated with late-stage disease presentation and poor prognosis, with limited understanding of early transformation events. Our study presents a comprehensive analysis of tumor progression and organ-specific metastatic dissemination to identify hypoxia-associated molecular, cellular, and histological alterations during HGSC tumor growth. H&E staining and subsequent histological assessment of tumor volume-based categories revealed recapitulation of numerous clinical features, including the prevalence of >0.0625≤0.5cm3 volume tumors and metastatic spread by orthotopic xenografts. The constant evolution of the tissue architecture concerning increased hyaluronic acid deposition, tumor vasculature, necrosis, altered proliferative potential, and gland forming ability of the tumor cells was identified. Flow cytometry and label chase-based molecular profiling across the tumor regenerative hierarchy identified the hypoxia-vasculogenic niche and the hybrid epithelial-mesenchymal tumor-cell state as determinants of self-renewal capabilities of progenitors and cancer stem cells (CSCs). A regulatory network and mathematical model based on tumor histology and molecular signatures predicted hypoxia-inducible factor 1-alpha (HIF1A) as a central node connecting epithelial-mesenchymal transition, metabolic and necrotic pathways in HGSC tumors. Thus, our findings provide a temporal resolution of hypoxia-associated events that sculpt HGSC tumor growth, and an in-depth understanding of it may aid in the early detection and treatment of HGSC.

2.
J Neurooncol ; 157(3): 575-591, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35430703

RESUMEN

BACKGROUND: Glioblastoma initiation and progression is believed to be driven by Glioma stem cells (GSCs). Activation of NOTCH1 and WNT, and more recently, non-canonical WNT5A signaling, has been demonstrated to regulate self-renewal and differentiation of the GSCs crucially. High expression levels of NOTCH1 and WNT in GBM tumors contribute to the sustenance of GSCs and mediate characteristic phenotypic plasticity, which is reflected by the different subtypes and tremendous intra-tumor heterogeneity. However, the coregulation of NOTCH1 and WNT5A is not well understood. Here, we studied the role of these molecules in regulating the characteristics of different GSC subtypes. METHODS: We established a novel GSC-enriched cell model, referred to as NSG-70, from a patient with recurrent GBM. NSG-70 cells harbor a unique cytogenetic feature, viz. isochromosome 9q. At the same time, its expression profiles indicate that it is a mixed lineage comprising proneural and mesenchymal subtypes. We examined the relevance of NOTCH1 and WNT5A signaling and their coordinated action in GBM using these cells and other patient-derived models representing different GSC subtypes. RESULTS: Our data revealed that the downregulation of NOTCH1 resulted in the suppression of stem cell and mesenchymal markers and significantly reduced the levels of WNT5A. NOTCH1 knockdown also led to a notable reduction in the vasculogenic mimicry of GSCs. Interestingly, knockdown of WNT5A exhibited similar effects and drove quiescent GSC towards proliferation. In a complementary manner, ectopic expression of WNT5A or rhWNT5A treatment rescued the effects of NOTCH1 knockdown. CONCLUSION: The resistance of GSCs towards conventional therapies in part due to subtype interconversion demands therapies targeting specific GSC subtype. Our study suggests the need for a combinatorial approach that could effectively target the NOTCH1-WNT5A signaling axis toward eliminating GSCs.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Neoplasias Encefálicas/patología , Línea Celular , Línea Celular Tumoral , Glioblastoma/patología , Glioma/patología , Humanos , Células Madre Neoplásicas/patología , Neovascularización Patológica/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Proteína Wnt-5a/metabolismo
3.
Med J Armed Forces India ; 78(Suppl 1): S7-S13, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-36147383

RESUMEN

The rapid development of advanced high throughput technologies and introduction of high resolution "omics" data through analysis of biological molecules has revamped medical research. Single-cell sequencing in recent years, is in fact revolutionising the field by providing a deeper, spatio-temporal analyses of individual cells within tissues and their relevance to disease. Like conventional sequencing, the single-cell approach deciphers the sequence of nucleotides in a given Deoxyribose Nucleic Acid (DNA), Ribose Nucleic Acid (RNA), Micro Ribose Nucleic Acid (miRNA), epigenetically modified DNA or chromatin DNA; however, the unit of analyses is changed to single cells rather than the entire tissue. Further, a large number of single cells analysed from a single tissue generate a unique holistic perception capturing all kinds of perturbations across different cells in the tissue that increases the precision of data. Inherently, execution of the technique generates a large amount of data, which is required to be processed in a specific manner followed by customised bioinformatic analysis to produce meaningful results. The most crucial role of single-cell sequencing technique is in elucidating the inter-cell genetic, epigenetic, transcriptomic and proteomic heterogeneity in health and disease. The current review presents a brief overview of this cutting-edge technology and its applications in medical research.

4.
Mol Cell Biochem ; 476(12): 4493-4505, 2021 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-34499322

RESUMEN

RNA-binding proteins (RBPs) are critical players in the post-transcriptional regulation of gene expression and are associated with each event in RNA metabolism. The term 'RNA-binding motif' (RBM) is assigned to novel RBPs with one or more RNA recognition motif (RRM) domains that are mainly involved in the nuclear processing of RNAs. RBM47 is a novel RBP conserved in vertebrates with three RRM domains whose contributions to various aspects of cellular functions are as yet emerging. Loss of RBM47 function affects head morphogenesis in zebrafish embryos and leads to perinatal lethality in mouse embryos, thereby assigning it to be an essential gene in early development of vertebrates. Its function as an essential cofactor for APOBEC1 in C to U RNA editing of several targets through substitution for A1CF in the A1CF-APOBEC1 editosome, established a new paradigm in the field. Recent advances in the understanding of its involvement in cancer progression assigned RBM47 to be a tumor suppressor that acts by inhibiting EMT and Wnt/[Formula: see text]-catenin signaling through post-transcriptional regulation. RBM47 is also required to maintain immune homeostasis, which adds another facet to its regulatory role in cellular functions. Here, we review the emerging roles of RBM47 in various biological contexts and discuss the current gaps in our knowledge alongside future perspectives for the field.


Asunto(s)
Desaminasas APOBEC-1/metabolismo , Neoplasias/patología , Edición de ARN , Proteínas de Unión al ARN/metabolismo , Vertebrados/crecimiento & desarrollo , Desaminasas APOBEC-1/genética , Animales , Humanos , Neoplasias/genética , Neoplasias/metabolismo , Proteínas de Unión al ARN/genética
5.
Carcinogenesis ; 41(4): 515-526, 2020 06 17.
Artículo en Inglés | MEDLINE | ID: mdl-31241128

RESUMEN

Cellular plasticity and transitional phenotypes add to complexities of cancer metastasis that can be initiated by single cell epithelial to mesenchymal transition (EMT) or cooperative cell migration (CCM). Our study identifies novel regulatory cross-talks between Tcf21 and Slug in mediating phenotypic and migration plasticity in high-grade serous ovarian adenocarcinoma (HGSC). Differential expression and subcellular localization associate Tcf21, Slug with epithelial, mesenchymal phenotypes, respectively; however, gene manipulation approaches identify their association with additional intermediate phenotypic states, implying the existence of a multistep epithelial-mesenchymal transition program. Live imaging further associated distinct migratory modalities with the Tcf21/Slug status of cell systems and discerned proliferative/passive CCM, active CCM and EMT modes of migration. Tcf21-Slug balance identified across a phenotypic spectrum in HGSC cell lines, associated with microenvironment-induced transitions and the emergence of an epithelial phenotype following drug exposure. Phenotypic transitions and associated functionalities following drug exposure were affirmed to ensue from occupancy of Slug promoter E-box sequences by Tcf21. Our study effectively provides a framework for understanding the relevance of ovarian cancer plasticity as a function of two transcription factors.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Movimiento Celular , Plasticidad de la Célula , Cistadenocarcinoma Seroso/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias Ováricas/patología , Factores de Transcripción de la Familia Snail/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Cistadenocarcinoma Seroso/genética , Cistadenocarcinoma Seroso/metabolismo , Transición Epitelial-Mesenquimal , Femenino , Humanos , Clasificación del Tumor , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Factores de Transcripción de la Familia Snail/genética , Células Tumorales Cultivadas , Microambiente Tumoral , Cicatrización de Heridas
6.
Biochim Biophys Acta ; 1849(9): 1209-18, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26170144

RESUMEN

Slug, a five C2H2 zinc finger (ZF) motif transcription factor mediates cell migration in development, adult tissue repair and regeneration, as well as during tumor metastases through epithelial to mesenchymal transition. At the molecular level, this involves interactions with E-box (CACC/GGTG) consensus elements within target gene promoters to achieve transcriptional repression. However, precise elucidation of events involved in this DNA recognition and binding of specific promoters to regulate target genes have not been achieved. In the present study, we show that besides transcriptional repression, Slug can also directly activate its own expression by preferential binding to specific E-box elements in the distal binding region of its promoter. Our findings suggest that while the first ZF does not contribute to the transcription-associated functions of Slug, all the remaining four ZFs are involved in regulating the expression of target genes with ZF3 and ZF4 being more crucial than ZF2 or ZF5. We also report that recognition and binding preferences of ZFs are defined through intrinsic differences in the E-box core base pairs and/or flanking sequences, with the S2 E-box element being most critical during autoregulation. However, specific target E-box recognition and binding are also defined by the cellular context, which implies that in silico and/or biochemical DNA binding preferences may not necessarily be able to accurately predict in situ events. Our studies thus constitute a novel understanding of transcriptional regulation.


Asunto(s)
Transición Epitelial-Mesenquimal/fisiología , Factores de Transcripción/fisiología , Línea Celular Tumoral , Femenino , Regulación de la Expresión Génica/genética , Humanos , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Regiones Promotoras Genéticas , Unión Proteica , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Transcripción Genética/genética
7.
Pharmacol Res ; 111: 815-819, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27431330

RESUMEN

A major focus of contemporary drug screening strategies is the identification of novel anticancer compounds, which often results in underutilization of resources. Current drug evaluation involves in vivo tumor (xenograft) regression as proof-of-principle for cytotoxicity (POC). However, this end-point lacks any assessment of drug resistance of the residual tumor and its capability to establish refractory and/or recurrent disease, which would represent more appropriate indicators of therapeutic failure. We have recently developed a flow cytometry-based approach for the analyses of intra-tumor cellular heterogeneity across stem cell hierarchies, genetic instability and differential cell cycling fractions, which can potentially be predictive of refractory disease and tumor relapse. Iterating this approach after initial POC screening in the drug discovery pipeline would have a great impact in terms of precision of drug evaluation, design of optimal drug combinations and/or drug repositioning. In this perspective, we highlight how through embracing of a comprehensive, informative and analytical assessment of the cellular content of residual tumors, the fidelity and statistical robustness of preclinical drug discovery can be greatly improved.


Asunto(s)
Antineoplásicos/uso terapéutico , Descubrimiento de Drogas/métodos , Reposicionamiento de Medicamentos/métodos , Citometría de Flujo , Ensayos Analíticos de Alto Rendimiento , Neoplasias/tratamiento farmacológico , Animales , Resistencia a Antineoplásicos , Humanos , Modelos Biológicos , Recurrencia Local de Neoplasia , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Subcell Biochem ; 61: 419-34, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23150261

RESUMEN

The concept of cancer as a stem cell disease has slowly gained ground over the last decade. A 'stem-like' state essentially necessitates that some cells in the developing tumor express the properties of remaining quiescent, self-renewing and regenerating tumors through establishment of aberrant cellular hierarchies. Alternatively, such capacities may also be reacquired through a de-differentiation process. The abnormal cellular differentiation patterns involved during either process during carcinogenesis are likely to be driven through a combination of genetic events and epigenetic regulation. The role(s) of the latter is increasingly being appreciated in acquiring the requisite genomic specificity and flexibility required for phenotypic plasticity, specifically in a context wherein genome sequences are not altered for differentiation to ensue. In this chapter, the recent advances in elucidating epigenetic mechanisms that govern the self-renewal, differentiation and regenerative potentials of cancer stem cells will be presented.


Asunto(s)
Epigénesis Genética , Regulación Neoplásica de la Expresión Génica , Neoplasias/genética , Células Madre Neoplásicas/metabolismo , Animales , Diferenciación Celular , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cromatina/metabolismo , Ensamble y Desensamble de Cromatina , Metilación de ADN , Predisposición Genética a la Enfermedad , Histonas/metabolismo , Humanos , Neoplasias/metabolismo , Neoplasias/patología , Células Madre Neoplásicas/patología , Fenotipo
9.
Stem Cell Rev Rep ; 19(2): 475-490, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35986129

RESUMEN

RNA-binding proteins (RBPs) are pivotal for regulating gene expression as they are involved in each step of RNA metabolism. Several RBPs are essential for viable growth and development in mammals. RNA-binding motif 47 (RBM47) is an RRM-containing RBP whose role in mammalian embryonic development is poorly understood yet deemed to be essential since its loss in mouse embryos leads to perinatal lethality. In this study, we attempted to elucidate the significance of RBM47 in cell-fate decisions of mouse embryonic stem cells (mESCs). Downregulation of Rbm47 did not affect mESC maintenance and the cell cycle but perturbed the expression of primitive endoderm (PrE) markers and increased GATA4 + PrE-like cells. However, the PrE misregulation could be reversed by either overexpressing Rbm47 or treating the knockdown mESCs with the inhibitors of FGFR or MEK, suggesting an implication of RBM47 in regulating FGF-ERK signaling. Rbm47 knockdown affected the multi-lineage differentiation potential of mESCs as it regressed teratoma in NSG mice and led to a skewed expression of differentiation markers in serum-induced monolayer differentiation. Further, lineage-specific differentiation revealed that Rbm47 is essential for proper differentiation of mESCs towards neuroectodermal and endodermal fate. Taken together, we assign a hitherto unknown role(s) to RBM47 in a subtle regulation of mESC differentiation.


Asunto(s)
Endodermo , Células Madre Embrionarias de Ratones , Proteínas de Unión al ARN , Animales , Ratones , Diferenciación Celular/genética , Desarrollo Embrionario , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo
10.
Comput Struct Biotechnol J ; 21: 5153-5164, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37920814

RESUMEN

The molecular and functional diversity generated by chimeric transcripts (CTs) that are derived from two genes is indicated to contribute to tumor cell survival. Several gaps yet exist. The present research is a systematic study of the spectrum of CTs identified in RNA sequencing datasets of 160 ovarian cancer samples in the The Cancer Genome Atlas (TCGA) (https://portal.gdc.cancer.gov). Structural annotation revealed complexities emerging from chromosomal localization of partner genes, differential splicing and inclusion of regulatory, untranslated regions. Identification of phenotype-specific associations further resolved a dynamically modulated mesenchymal signature during transformation. On an evolutionary background, protein-coding CTs were indicated to be highly conserved, while non-coding CTs may have evolved more recently. We also realized that the current premise postulating structural alterations or neighbouring gene readthrough generating CTs is not valid in instances wherein the parental genes are genomically distanced. In addressing this lacuna, we identified the essentiality of specific spatiotemporal arrangements mediated gene proximities in 3D space for the generation of CTs. All these features together suggest non-random mechanisms towards increasing the molecular diversity in a cell through chimera formation either in parallel or with cross-talks with the indigenous regulatory network.

11.
Transl Oncol ; 15(1): 101257, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34715620

RESUMEN

The involvement of cancer stem cells (CSCs) in driving tumor dormancy and drug resistance is well established. Most therapeutic regimens however are ineffective in targeting these regenerative populations. We report the development and evaluation of a monoclonal antibody, mAb150, which targets the metastasis associated antigen, Annexin A2 (AnxA2) through recognition of a N-terminal epitope. Treatment with mAb150 potentiated re-entry of CSCs into the cell cycle that perturbed tumor dormancy and facilitated targeting of CSCs as was validated by in vitro and in vivo assays. Epigenetic potentiation further improved mAb150 efficacy in achieving total tumor regression by targeting regenerative populations to achieve tumor regression, specifically in high-grade serous ovarian adenocarcinoma.

12.
Differentiation ; 79(4-5): 218-31, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20207472

RESUMEN

Biallelic inactivation of the CREB-binding protein (CREBBP) a transcriptional co-activator produces an embryonic lethal phenotype in mice. In humans, re-arrangements in CREBBP are associated with the Rubinstein-Taybi Syndrome (RSTS) that is characterised by craniofacial, skeletal and neuronal symptoms. Neuronal defects in RSTS can be attributed to genetic re-arrangements in CREBBP, which has been implicated in synaptic plasticity and long-term memory. The present study was designed to investigate the role of CREBBP re-arrangements during neuronal differentiation. Towards this, deletion constructs of pCREBBP, viz. pDeltaCB-HAT and pDeltaHAT-CT were generated and transfected into NT2 cells. Expression profiling of the components of Notch, Wnt, SHH and Retinoid signaling along with screening of the neuronal markers was carried out in the NT2 cells and their mutant derivatives. ChIP-PCRs along with co-immunoprecipitations were also performed in these cells to investigate defects due to inappropriate interaction of mutated CREEBP with the corresponding transcription factor and other transcription regulatory proteins both at steady state as well as during differentiation. Mutant NT2 cells lacking the CREB, BROMO and HAT domains (CB-HAT) were highly proliferative and showed limited differentiation; while mutant NT2 cells expressing CREBBP lacking the HAT and CTAD domains (HAT-CT) are proliferation deficient and differentiate rapidly albeit generating an insufficient number of neurons. Altered CREBBP structure resulted in changes in HAT activity, cell cycle profiles and expression of basal levels of components of Notch, SHH, Wnt and retinoid pathways known to be critical in the proliferation and differentiation of neuronal progenitors. At the chromatin level, aberrant signaling correlated with altered binding affinities of the (CREBBP-transcription factor) complexes to promoter regions of components of these pathways. Thus, differentiation defects are manifested early at the genomic level leading to aberrant transcription of the genes involved in differentiation along the neuronal lineage.


Asunto(s)
Proteína de Unión a CREB/genética , Diferenciación Celular/fisiología , Reordenamiento Génico , Neuronas/fisiología , Animales , Biomarcadores/metabolismo , Proteína de Unión a CREB/metabolismo , Ciclo Celular/fisiología , Línea Celular , Proteínas Hedgehog/metabolismo , Humanos , Ratones , Neuronas/citología , Regiones Promotoras Genéticas , Receptores Notch/metabolismo , Retinoides/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt/metabolismo
13.
Reproduction ; 140(1): 33-41, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20368192

RESUMEN

The isolation and identification of stem-like cells in solid tumors or cancer stem cells (CSCs) have been exciting developments of the last decade, although these rare populations had been earlier identified in leukemia. CSC biology necessitates a detailed delineation of normal stem cell functioning and maintenance of homeostasis within the organ. Ovarian CSC biology has unfortunately not benefited from a pre-established knowledge of stem cell lineage demarcation and functioning in the normal organ. In the absence of such information, some of the classical parameters such as long-term culture-initiating assays to isolate stem cell clones from tumors, screening and evaluation of other epithelial stem cell surface markers, dye efflux, and label retention have been applied toward the putative isolation of CSCs from ovarian tumors. The present review presents an outline of the various approaches developed so far and the various perspectives revealed that are now required to be dealt with toward better disease management.


Asunto(s)
Células Madre Neoplásicas/fisiología , Neoplasias Ováricas/patología , Adenocarcinoma/patología , Animales , Apoptosis , Biomarcadores de Tumor , Separación Celular , Femenino , Humanos , Receptores de Hialuranos/metabolismo , Ratones
14.
Stem Cells ; 27(3): 498-508, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19253934

RESUMEN

Recruitment and localization of endothelial precursors within tumors is a potential area for the development of therapeutics, because their functional contribution to tumor vasculature is realized to be important for cancer cell survival. However, the exact nature of the recruited cell type and cellular events orchestrating the entire phenomenon remains obscure. We report that human ovarian cancer is frequently associated with cells expressing the stem cell surface marker CD133. We further show that these CD133-expressing cells are nontumorigenic in nature, and they augment tumor development through their vasculogenic potential. This cell population is attracted by cancer stem cells (CSCs) and retains a direct physical association within the CSC-derived spheroids. Our study further delineates the contribution of these vasculogenic CD133(+) stem cells, termed by us as endothelial stem cells (EnSCs) to the developing tumor vasculature during disease progression. In support of their being stem cells, the EnSCs have a capability of establishing an entire endothelial cell hierarchy. We conclude that such EnSCs play a crucial role in ensuring the development of long-term tumor vasculature to complement CSC-driven tumor development and disease progression.


Asunto(s)
Antígenos CD/metabolismo , Glicoproteínas/metabolismo , Metástasis de la Neoplasia/fisiopatología , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Péptidos/metabolismo , Células Madre/metabolismo , Células Madre/fisiología , Antígeno AC133 , Adulto , Anciano , Anciano de 80 o más Años , Diferenciación Celular , Células Cultivadas , Endotelio Vascular/citología , Femenino , Citometría de Flujo , Humanos , Persona de Mediana Edad , Modelos Biológicos , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Células Madre Neoplásicas/fisiología , Células Madre/citología
15.
Stem Cells ; 27(9): 2059-68, 2009 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-19544473

RESUMEN

The transcriptional repressors Snail and Slug contribute to cancer progression by mediating epithelial-mesenchymal transition (EMT), which results in tumor cell invasion and metastases. We extend this current understanding to demonstrate their involvement in the development of resistance to radiation and paclitaxel. The process is orchestrated through the acquisition of a novel subset of gene targets that is repressed under conditions of stress, effectively inactivating p53-mediated apoptosis, while another subset of targets continues to mediate EMT. Repressive activities are complemented by a concurrent derepression of specific genes resulting in the acquisition of stem cell-like characteristics. Such cells are bestowed with three critical capabilities, namely EMT, resistance to p53-mediated apoptosis, and a self-renewal program, that together define the functionality and survival of metastatic cancer stem cells. EMT provides a mechanism of escape to a new, less adverse niche; resistance to apoptosis ensures cell survival in conditions of stress in the primary tumor; whereas acquisition of "stemness" ensures generation of the critical tumor mass required for progression of micrometastases to macrometastases. Our findings, besides achieving considerable expansion of the inventory of direct genes targets, more importantly demonstrate that such elegant cooperative modulation of gene regulation mediated by Snail and Slug is critical for a cancer cell to acquire stem cell characteristics toward resisting radiotherapy- or chemotherapy-mediated cellular stress, and this may be a determinative aspect of aggressive cancer metastases.


Asunto(s)
Apoptosis/fisiología , Resistencia a Antineoplásicos/fisiología , Neoplasias Ováricas/metabolismo , Factores de Transcripción/fisiología , Proteína p53 Supresora de Tumor/metabolismo , Apoptosis/genética , Sitios de Unión , Línea Celular Tumoral , Resistencia a Antineoplásicos/genética , Femenino , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica/genética , Regulación Neoplásica de la Expresión Génica/fisiología , Genoma Humano/genética , Humanos , Immunoblotting , Etiquetado Corte-Fin in Situ , Neoplasias Ováricas/tratamiento farmacológico , Neoplasias Ováricas/radioterapia , Regiones Promotoras Genéticas/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factores de Transcripción de la Familia Snail , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteína p53 Supresora de Tumor/genética
16.
Methods Mol Biol ; 2019: 1-14, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31359385

RESUMEN

Retinoid and rexinoid receptors are known to regulate key processes during development, differentiation, and cell death in vertebrates. However, their contributions to progression of malignant disease remain largely elusive although it is realized that transformed cancer cells, which essentially evade apoptosis, may display altered molecular expressions or functions associated with retinoid signaling. Here, using a progression model of ovarian cancer, we describe a proteomics-based approach including experimental procedures toward identification and validation of altered protein profiles during transformation. Effectively, this specifies loss of RXR-γ during progression of epithelial ovarian cancer.


Asunto(s)
Carcinoma Epitelial de Ovario/patología , Neoplasias Ováricas/patología , Proteómica/métodos , Receptor gamma X Retinoide/deficiencia , Animales , Carcinoma Epitelial de Ovario/metabolismo , Línea Celular Tumoral , Progresión de la Enfermedad , Electroforesis en Gel Bidimensional , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Ratones , Trasplante de Neoplasias , Neoplasias Ováricas/metabolismo , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción
17.
J Clin Med ; 8(7)2019 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-31261795

RESUMEN

The experimental evaluation of metastasis overly focuses on the gain of migratory and invasive properties, while disregarding the contributions of cellular plasticity, extra-cellular matrix heterogeneity, niche interactions, and tissue architecture. Traditional cell-based assays often restrict the inclusion of these processes and warrant the implementation of approaches that provide an enhanced spatiotemporal resolution of the metastatic cascade. Time lapse imaging represents such an underutilized approach in cancer biology, especially in the context of disease progression. The inclusion of time lapse microscopy and microfluidic devices in routine assays has recently discerned several nuances of the metastatic cascade. Our review emphasizes that a complete comprehension of metastasis in view of evolving ideologies necessitates (i) the use of appropriate, context-specific assays and understanding their inherent limitations; (ii) cautious derivation of inferences to avoid erroneous/overestimated clinical extrapolations; (iii) corroboration between multiple assay outputs to gauge metastatic potential; and (iv) the development of protocols with improved in situ implications. We further believe that the adoption of improved quantitative approaches in these assays can generate predictive algorithms that may expedite therapeutic strategies targeting metastasis via the development of disease relevant model systems. Such approaches could potentiate the restructuring of the cancer metastasis paradigm through an emphasis on the development of next-generation real-time assays.

18.
J Clin Med ; 8(3)2019 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-30857227

RESUMEN

Molecular stratification of high-grade serous ovarian carcinoma (HGSC) for targeted therapy is a pertinent approach in improving prognosis of this highly heterogeneous disease. Enabling the same necessitates identification of class-specific biomarkers and their robust detection in the clinic. We have earlier resolved three discrete molecular HGSC classes associated with distinct functional behavior based on their gene expression patterns, biological networks, and pathways. An important difference revealed was that Class 1 is likely to exhibit cooperative cell migration (CCM), Class 2 undergoes epithelial to mesenchymal transition (EMT), while Class 3 is possibly capable of both modes of migration. In the present study, we define clinical stratification of HGSC tumors through the establishment of standard operating procedures for immunohistochemistry and histochemistry based detection of a panel of biomarkers including TCF21, E-cadherin, PARP1, Slug, AnnexinA2, and hyaluronan. Further development and application of scoring guidelines based on expression of this panel in cell line-derived xenografts, commercial tissue microarrays, and patient tumors led to definitive stratification of samples. Biomarker expression was observed to vary significantly between primary and metastatic tumors suggesting class switching during disease progression. Another interesting feature in the study was of enhanced CCM-marker expression in tumors following disease progression and chemotherapy. These stratification principles and the new information thus generated is the first step towards class-specific personalized therapies in the disease.

19.
Pharmacol Ther ; 194: 161-184, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30268772

RESUMEN

Cancer metastasis and therapy resistance are the major unsolved clinical challenges, and account for nearly all cancer-related deaths. Both metastasis and therapy resistance are fueled by epithelial plasticity, the reversible phenotypic transitions between epithelial and mesenchymal phenotypes, including epithelial-mesenchymal transition (EMT) and mesenchymal-epithelial transition (MET). EMT and MET have been largely considered as binary processes, where cells detach from the primary tumor as individual units with many, if not all, traits of a mesenchymal cell (EMT) and then convert back to being epithelial (MET). However, recent studies have demonstrated that cells can metastasize in ways alternative to traditional EMT paradigm; for example, they can detach as clusters, and/or occupy one or more stable hybrid epithelial/mesenchymal (E/M) phenotypes that can be the end point of a transition. Such hybrid E/M cells can integrate various epithelial and mesenchymal traits and markers, facilitating collective cell migration. Furthermore, these hybrid E/M cells may possess higher tumor-initiation and metastatic potential as compared to cells on either end of the EMT spectrum. Here, we review in silico, in vitro, in vivo and clinical evidence for the existence of one or more hybrid E/M phenotype(s) in multiple carcinomas, and discuss their implications in tumor-initiation, tumor relapse, therapy resistance, and metastasis. Together, these studies drive the emerging notion that cells in a hybrid E/M phenotype may occupy 'metastatic sweet spot' in multiple subtypes of carcinomas, and pathways linked to this (these) hybrid E/M state(s) may be relevant as prognostic biomarkers as well as a promising therapeutic targets.


Asunto(s)
Resistencia a Antineoplásicos , Células Epiteliales , Células Madre Mesenquimatosas , Metástasis de la Neoplasia , Animales , Plasticidad de la Célula , Transición Epitelial-Mesenquimal , Humanos , Neoplasias/tratamiento farmacológico , Neoplasias/patología , Fenotipo
20.
Front Oncol ; 8: 633, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30619767

RESUMEN

Metastatic dissemination generates an aggressive disease facilitated by enhanced migratory and invasive properties. Experimental approaches employ several in vitro and in vivo assays toward quantification of these functionalities. In vitro assessments of cell motility often employ endpoint assays that rely on the global efficacy of wound closure and thwart quantification of migratory phenotypes observed during metastatic dissemination. Recent studies highlight the distinct signatures associated with individual vs. collective cell migration and necessitate the incorporation of these modalities into routine analyses. Advances in live cell imaging that permit real-time visualization of pathophysiological processes can be employed toward elucidating phenotypic plasticity associated with cell migration to overcome caveats inherent to end-point assays. Herein, we corroborate live cell imaging with the in vitro scratch assay toward quantification of migratory modalities in transformed cells. Our protocol describes a step-by-step approach for live cell setup of the scratch assay, and details analyses employed toward definition of three quantitative metrics viz., displacement, velocity and number of nearest neighbors. The current protocol (from scratch induction to data acquisition) is implemented for ~30 h and provides global/single-cell resolution of migratory phenotypes as opposed to the endpoint assays. Routine application of this protocol in cancer biology can aid the design of therapeutic regimes targeting specific migratory modalities and significantly contribute to the dissection of associated molecular networks.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA