Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 174(3): 716-729.e27, 2018 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-29961576

RESUMEN

Single-cell RNA sequencing technologies suffer from many sources of technical noise, including under-sampling of mRNA molecules, often termed "dropout," which can severely obscure important gene-gene relationships. To address this, we developed MAGIC (Markov affinity-based graph imputation of cells), a method that shares information across similar cells, via data diffusion, to denoise the cell count matrix and fill in missing transcripts. We validate MAGIC on several biological systems and find it effective at recovering gene-gene relationships and additional structures. Applied to the epithilial to mesenchymal transition, MAGIC reveals a phenotypic continuum, with the majority of cells residing in intermediate states that display stem-like signatures, and infers known and previously uncharacterized regulatory interactions, demonstrating that our approach can successfully uncover regulatory relations without perturbations.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Algoritmos , Línea Celular , Epistasis Genética/genética , Redes Reguladoras de Genes/genética , Humanos , Cadenas de Markov , MicroARNs/genética , ARN Mensajero/genética , Programas Informáticos
2.
Cell ; 168(4): 670-691, 2017 02 09.
Artículo en Inglés | MEDLINE | ID: mdl-28187288

RESUMEN

Metastases account for the great majority of cancer-associated deaths, yet this complex process remains the least understood aspect of cancer biology. As the body of research concerning metastasis continues to grow at a rapid rate, the biological programs that underlie the dissemination and metastatic outgrowth of cancer cells are beginning to come into view. In this review we summarize the cellular and molecular mechanisms involved in metastasis, with a focus on carcinomas where the most is known, and we highlight the general principles of metastasis that have begun to emerge.


Asunto(s)
Carcinoma/patología , Metástasis de la Neoplasia/patología , Animales , Plaquetas/metabolismo , Carcinoma/genética , Carcinoma/metabolismo , Movimiento Celular , Transición Epitelial-Mesenquimal , Humanos , Invasividad Neoplásica , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/patología , Células Neoplásicas Circulantes/metabolismo , Células Neoplásicas Circulantes/patología , Neutrófilos/metabolismo , Linfocitos T/inmunología , Microambiente Tumoral
3.
Breast Cancer Res ; 25(1): 23, 2023 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-36859337

RESUMEN

Stratifying breast cancer into specific molecular or histologic subtypes aids in therapeutic decision-making and predicting outcomes; however, these subtypes may not be as distinct as previously thought. Patients with luminal-like, estrogen receptor (ER)-expressing tumors have better prognosis than patients with more aggressive, triple-negative or basal-like tumors. There is, however, a subset of luminal-like tumors that express lower levels of ER, which exhibit more basal-like features. We have found that breast tumors expressing lower levels of ER, traditionally considered to be luminal-like, represent a distinct subset of breast cancer characterized by the emergence of basal-like features. Lineage tracing of low-ER tumors in the MMTV-PyMT mouse mammary tumor model revealed that basal marker-expressing cells arose from normal luminal epithelial cells, suggesting that luminal-to-basal plasticity is responsible for the evolution and emergence of basal-like characteristics. This plasticity allows tumor cells to gain a new lumino-basal phenotype, thus leading to intratumoral lumino-basal heterogeneity. Single-cell RNA sequencing revealed SOX10 as a potential driver for this plasticity, which is known among breast tumors to be almost exclusively expressed in triple-negative breast cancer (TNBC) and was also found to be highly expressed in low-ER tumors. These findings suggest that basal-like tumors may result from the evolutionary progression of luminal tumors with low ER expression.


Asunto(s)
Neoplasias Mamarias Animales , Receptores de Estrógenos , Animales , Ratones , Fenotipo , Expresión Génica , Modelos Animales de Enfermedad
4.
Proc Natl Acad Sci U S A ; 114(12): E2337-E2346, 2017 03 21.
Artículo en Inglés | MEDLINE | ID: mdl-28270621

RESUMEN

Neoplastic cells within individual carcinomas often exhibit considerable phenotypic heterogeneity in their epithelial versus mesenchymal-like cell states. Because carcinoma cells with mesenchymal features are often more resistant to therapy and may serve as a source of relapse, we sought to determine whether such cells could be further stratified into functionally distinct subtypes. Indeed, we find that a basal epithelial marker, integrin-ß4 (ITGB4), can be used to enable stratification of mesenchymal-like triple-negative breast cancer (TNBC) cells that differ from one another in their relative tumorigenic abilities. Notably, we demonstrate that ITGB4+ cancer stem cell (CSC)-enriched mesenchymal cells reside in an intermediate epithelial/mesenchymal phenotypic state. Among patients with TNBC who received chemotherapy, elevated ITGB4 expression was associated with a worse 5-year probability of relapse-free survival. Mechanistically, we find that the ZEB1 (zinc finger E-box binding homeobox 1) transcription factor activity in highly mesenchymal SUM159 TNBC cells can repress expression of the epithelial transcription factor TAp63α (tumor protein 63 isoform 1), a protein that promotes ITGB4 expression. In addition, we demonstrate that ZEB1 and ITGB4 are important in modulating the histopathological phenotypes of tumors derived from mesenchymal TNBC cells. Hence, mesenchymal carcinoma cell populations are internally heterogeneous, and ITGB4 is a mechanistically driven prognostic biomarker that can be used to identify the more aggressive subtypes of mesenchymal carcinoma cells in TNBC. The ability to rapidly isolate and mechanistically interrogate the CSC-enriched, partially mesenchymal carcinoma cells should further enable identification of novel therapeutic opportunities to improve the prognosis for high-risk patients with TNBC.


Asunto(s)
Carcinoma/metabolismo , Integrina beta4/metabolismo , Mesodermo/metabolismo , Células Madre Neoplásicas/metabolismo , Neoplasias de la Mama Triple Negativas/metabolismo , Carcinoma/genética , Carcinoma/mortalidad , Línea Celular Tumoral , Transición Epitelial-Mesenquimal , Femenino , Humanos , Integrina beta4/genética , Mesodermo/citología , Pronóstico , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/mortalidad , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo
5.
Blood ; 123(17): 2682-90, 2014 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-24596419

RESUMEN

The MYB oncogene is widely expressed in acute leukemias and is important for the continued proliferation of leukemia cells, suggesting that MYB may be a therapeutic target in these diseases. However, realization of this potential requires a significant therapeutic window for MYB inhibition, given its essential role in normal hematopoiesis, and an approach for developing an effective therapeutic. We previously showed that the interaction of c-Myb with the coactivator CBP/p300 is essential for its transforming activity. Here, by using cells from Booreana mice which carry a mutant allele of c-Myb, we show that this interaction is essential for in vitro transformation by the myeloid leukemia oncogenes AML1-ETO, AML1-ETO9a, MLL-ENL, and MLL-AF9. We further show that unlike cells from wild-type mice, Booreana cells transduced with AML1-ETO9a or MLL-AF9 retroviruses fail to generate leukemia upon transplantation into irradiated recipients. Finally, we have begun to explore the molecular mechanisms underlying these observations by gene expression profiling. This identified several genes previously implicated in myeloid leukemogenesis and HSC function as being regulated in a c-Myb-p300-dependent manner. These data highlight the importance of the c-Myb-p300 interaction in myeloid leukemogenesis and suggest disruption of this interaction as a potential therapeutic strategy for acute myeloid leukemia.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Leucemia Mieloide Aguda/metabolismo , Proteínas Proto-Oncogénicas c-myb/metabolismo , Factores de Transcripción p300-CBP/metabolismo , Alelos , Animales , Transformación Celular Neoplásica , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Proteínas de Unión al ADN/metabolismo , Perfilación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Ratones Mutantes , Mutación , Proteínas de Fusión Oncogénica/metabolismo , Oncogenes , Proteínas Proto-Oncogénicas/metabolismo , Factores de Transcripción/metabolismo
6.
RNA ; 19(12): 1767-80, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24158791

RESUMEN

Metastasis is a complex, multistep process involved in the progression of cancer from a localized primary tissue to distant sites, often characteristic of the more aggressive forms of this disease. Despite being studied in great detail in recent years, the mechanisms that govern this process remain poorly understood. In this study, we identify a novel role for miR-139-5p in the inhibition of breast cancer progression. We highlight its clinical relevance by reviewing miR-139-5p expression across a wide variety of breast cancer subtypes using in-house generated and online data sets to show that it is most frequently lost in invasive tumors. A biotin pull-down approach was then used to identify the mRNA targets of miR-139-5p in the breast cancer cell line MCF7. Functional enrichment analysis of the pulled-down targets showed significant enrichment of genes in pathways previously implicated in breast cancer metastasis (P < 0.05). Further bioinformatic analysis revealed a predicted disruption to the TGFß, Wnt, Rho, and MAPK/PI3K signaling cascades, implying a potential role for miR-139-5p in regulating the ability of cells to invade and migrate. To corroborate this finding, using the MDA-MB-231 breast cancer cell line, we show that overexpression of miR-139-5p results in suppression of these cellular phenotypes. Furthermore, we validate the interaction between miR-139-5p and predicted targets involved in these pathways. Collectively, these results suggest a significant functional role for miR-139-5p in breast cancer cell motility and invasion and its potential to be used as a prognostic marker for the aggressive forms of breast cancer.


Asunto(s)
Carcinoma Ductal de Mama/genética , Regulación Neoplásica de la Expresión Génica , MicroARNs/fisiología , Neoplasias de la Mama Triple Negativas/genética , Secuencia de Bases , Sitios de Unión , Carcinoma Ductal de Mama/metabolismo , Carcinoma Ductal de Mama/secundario , Línea Celular Tumoral , Movimiento Celular , Proliferación Celular , Replicación del ADN , Femenino , Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Invasividad Neoplásica , Interferencia de ARN , Transducción de Señal , Neoplasias de la Mama Triple Negativas/metabolismo , Neoplasias de la Mama Triple Negativas/patología
7.
Epigenomics ; 16(5): 293-308, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38356412

RESUMEN

Background: Triple-negative breast cancer (TNBC) is an aggressive disease with limited treatment options. Eribulin, a chemotherapeutic drug, induces epigenetic changes in cancer cells, suggesting a unique mechanism of action. Materials & methods: MDA-MB 231 cells were treated with eribulin and paclitaxel, and the samples from 53 patients treated with neoadjuvant eribulin were compared with those from 14 patients who received the standard-of-care treatment using immunohistochemistry. Results: Eribulin treatment caused significant DNA methylation changes in drug-tolerant persister TNBC cells, and it also elicited changes in the expression levels of epigenetic modifiers (DNMT1, TET1, DNMT3A/B) in vitro and in primary TNBC tumors. Conclusion: These findings provide new insights into eribulin's mechanism of action and potential biomarkers for predicting TNBC treatment response.


Asunto(s)
Metilación de ADN , Furanos , Policétidos Poliéteres , Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Cetonas/farmacología , Cetonas/uso terapéutico , ADN/metabolismo , Línea Celular Tumoral , Oxigenasas de Función Mixta/genética , Proteínas Proto-Oncogénicas/genética
8.
Cell Rep Med ; 5(4): 101504, 2024 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-38593809

RESUMEN

Targeted therapies have improved outcomes for certain cancer subtypes, but cytotoxic chemotherapy remains a mainstay for triple-negative breast cancer (TNBC). The epithelial-to-mesenchymal transition (EMT) is a developmental program co-opted by cancer cells that promotes metastasis and chemoresistance. There are no therapeutic strategies specifically targeting mesenchymal-like cancer cells. We report that the US Food and Drug Administration (FDA)-approved chemotherapeutic eribulin induces ZEB1-SWI/SNF-directed chromatin remodeling to reverse EMT that curtails the metastatic propensity of TNBC preclinical models. Eribulin induces mesenchymal-to-epithelial transition (MET) in primary TNBC in patients, but conventional chemotherapy does not. In the treatment-naive setting, but not after acquired resistance to other agents, eribulin sensitizes TNBC cells to subsequent treatment with other chemotherapeutics. These findings provide an epigenetic mechanism of action of eribulin, supporting its use early in the disease process for MET induction to prevent metastatic progression and chemoresistance. These findings warrant prospective clinical evaluation of the chemosensitizing effects of eribulin in the treatment-naive setting.


Asunto(s)
Antineoplásicos , Furanos , Cetonas , Policétidos Poliéteres , Neoplasias de la Mama Triple Negativas , Estados Unidos , Humanos , Neoplasias de la Mama Triple Negativas/patología , Ensamble y Desensamble de Cromatina , Estudios Prospectivos , Antineoplásicos/uso terapéutico
9.
Nucleic Acids Res ; 39(11): 4664-79, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21317192

RESUMEN

To gain insight into the mechanisms by which the Myb transcription factor controls normal hematopoiesis and particularly, how it contributes to leukemogenesis, we mapped the genome-wide occupancy of Myb by chromatin immunoprecipitation followed by massively parallel sequencing (ChIP-Seq) in ERMYB myeloid progenitor cells. By integrating the genome occupancy data with whole genome expression profiling data, we identified a Myb-regulated transcriptional program. Gene signatures for leukemia stem cells, normal hematopoietic stem/progenitor cells and myeloid development were overrepresented in 2368 Myb regulated genes. Of these, Myb bound directly near or within 793 genes. Myb directly activates some genes known critical in maintaining hematopoietic stem cells, such as Gfi1 and Cited2. Importantly, we also show that, despite being usually considered as a transactivator, Myb also functions to repress approximately half of its direct targets, including several key regulators of myeloid differentiation, such as Sfpi1 (also known as Pu.1), Runx1, Junb and Cebpb. Furthermore, our results demonstrate that interaction with p300, an established coactivator for Myb, is unexpectedly required for Myb-mediated transcriptional repression. We propose that the repression of the above mentioned key pro-differentiation factors may contribute essentially to Myb's ability to suppress differentiation and promote self-renewal, thus maintaining progenitor cells in an undifferentiated state and promoting leukemic transformation.


Asunto(s)
Regulación de la Expresión Génica , Células Progenitoras Mieloides/metabolismo , Mielopoyesis/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/genética , Transcripción Genética , Animales , Sitios de Unión , Células Cultivadas , Cromatina/metabolismo , Inmunoprecipitación de Cromatina , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Genómica , Histonas/metabolismo , Leucemia/genética , Ratones , Ratones Endogámicos C57BL , Células Progenitoras Mieloides/citología , Factores de Transcripción p300-CBP/metabolismo
10.
bioRxiv ; 2023 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-37333096

RESUMEN

Triple-negative breast cancer (TNBC) is an aggressive disease subtype with limited treatment options. Eribulin is a chemotherapeutic approved for the treatment of advanced breast cancer that has been shown to elicit epigenetic changes. We investigated the effect of eribulin treatment on genome-scale DNA methylation patterns in TNBC cells. Following repeated treatment, The results showed that eribulin-induced changes in DNA methylation patterns evident in persister cells. Eribulin also affected the binding of transcription factors to genomic ZEB1 binding sites and regulated several cellular pathways, including ERBB and VEGF signaling and cell adhesion. Eribulin also altered the expression of epigenetic modifiers including DNMT1, TET1, and DNMT3A/B in persister cells. Data from primary human TNBC tumors supported these findings: DNMT1 and DNMT3A levels were altered by eribulin treatment in human primary TNBC tumors. Our results suggest that eribulin modulates DNA methylation patterns in TNBC cells by altering the expression of epigenetic modifiers. These findings have clinical implications for using eribulin as a therapeutic agent.

11.
bioRxiv ; 2023 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-37131809

RESUMEN

The epithelial-mesenchymal transition (EMT) is a developmental program co-opted by tumor cells that aids the initiation of the metastatic cascade. Tumor cells that undergo EMT are relatively chemoresistant, and there are currently no therapeutic avenues specifically targeting cells that have acquired mesenchymal traits. We show that treatment of mesenchymal-like triple-negative breast cancer (TNBC) cells with the microtubule-destabilizing chemotherapeutic eribulin, which is FDA-approved for the treatment of advanced breast cancer, leads to a mesenchymal-epithelial transition (MET). This MET is accompanied by loss of metastatic propensity and sensitization to subsequent treatment with other FDA-approved chemotherapeutics. We uncover a novel epigenetic mechanism of action that supports eribulin pretreatment as a path to MET induction that curtails metastatic progression and the evolution of therapy resistance.

12.
Blood ; 116(26): 5849-58, 2010 Dec 23.
Artículo en Inglés | MEDLINE | ID: mdl-20610815

RESUMEN

Identification of genes that regulate the development, self-renewal, and differentiation of stem cells is of vital importance for understanding normal organogenesis and cancer; such knowledge also underpins regenerative medicine. Here we demonstrate that chemical mutagenesis of mice combined with advances in hematopoietic stem cell reagents and genome resources can efficiently recover recessive mutations and identify genes essential for generation and proliferation of definitive hematopoietic stem cells and/or their progeny. We used high-throughput fluorescence-activated cell sorter to analyze 9 subsets of blood stem cells, progenitor cells, circulating red cells, and platelets in more than 1300 mouse embryos at embryonic day (E) 14.5. From 45 pedigrees, we recovered 6 strains with defects in definitive hematopoiesis. We demonstrate rapid identification of a novel mutation in the c-Myb transcription factor that results in thrombocythemia and myelofibrosis as proof of principal of the utility of our fluorescence-activated cell sorter-based screen. Such phenotype-driven approaches will provide new knowledge of the genes, protein interactions, and regulatory networks that underpin stem cell biology.


Asunto(s)
Embrión de Mamíferos/citología , Embrión de Mamíferos/fisiología , Perfilación de la Expresión Génica , Genes Recesivos , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/fisiología , Animales , Biomarcadores/metabolismo , Diferenciación Celular , Células Cultivadas , Femenino , Citometría de Flujo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C3H , Ratones Endogámicos C57BL , Mutación/genética , Fenotipo , Proteínas Proto-Oncogénicas c-myb/genética , Proteínas Proto-Oncogénicas c-myb/metabolismo
13.
Cancers (Basel) ; 14(5)2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-35267444

RESUMEN

The epithelial-to-mesenchymal transition (EMT) and its reversal, the mesenchymal-to-epithelial transition (MET) are critical components of the metastatic cascade in breast cancer and many other solid tumor types. Recent work has uncovered the presence of a variety of states encompassed within the EMT spectrum, each of which may play unique roles or work collectively to impact tumor progression. However, defining EMT status is not routinely carried out to determine patient prognosis or dictate therapeutic decision-making in the clinic. Identifying and quantifying the presence of various EMT states within a tumor is a critical first step to scoring patient tumors to aid in determining prognosis. Here, we review the major strides taken towards translating our understanding of EMT biology from bench to bedside. We review previously used approaches including basic immunofluorescence staining, flow cytometry, single-cell sequencing, and multiplexed tumor mapping. Future studies will benefit from the consideration of multiple methods and combinations of markers in designing a diagnostic tool for detecting and measuring EMT in patient tumors.

14.
Methods Cell Biol ; 171: 149-161, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35953198

RESUMEN

Tumor heterogeneity presents an ongoing challenge to disease progression and treatment in many solid tumor types. Understanding the roots of intra-tumoral heterogeneity and how it may relate to the high incidence of metastasis is critical in overcoming disease relapse and chemoresistance. The epithelial-to-mesenchymal transition is a dynamic cellular program that is co-opted by cancer cells to enhance, among others, migratory and invasive cell traits. It is a key contributor to heterogeneity, chemo-resistance, and metastasis in many carcinoma-types, with the intermediate or hybrid EMT state playing a critical role due to its increased tumor-initiating potential. A critical component in utilizing this knowledge in patient treatment is to first detect and score the impact of EMT in a patient sample. Here, we provide a detailed protocol to detect EMT states and quantify the resulting epithelial-mesenchymal heterogeneity within tumors using a novel multiplexed immunostaining approach and analysis method. This protocol and concept can easily be adapted using custom panels of markers to explore other sources of tumoral heterogeneity in addition to EMT.


Asunto(s)
Transición Epitelial-Mesenquimal , Humanos , Benzopiranos , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Fenoles , Fenotipo
15.
Epigenomics ; 14(9): 519-535, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35382559

RESUMEN

Background: Epithelial-to-mesenchymal transition (EMT) is an early step in the invasion-metastasis cascade, involving progression through intermediate cell states. Due to challenges with isolating intermediate cell states, genome-wide cytosine modifications that define transition are not completely understood. Methods: The authors measured multiple DNA cytosine modification marks and chromatin accessibility across clonal populations residing in specific EMT states. Results: Clones exhibiting more intermediate EMT phenotypes demonstrated increased 5-hydroxymethylcytosine and decreased 5-methylcytosine. Open chromatin regions containing increased 5-hydroxymethylcytosine CpG loci were enriched in EMT transcription factor motifs and were associated with Rho GTPases. Conclusion: The results indicate the importance of both distinct and shared epigenetic profiles associated with EMT processes that may be targeted to prevent EMT progression.


Asunto(s)
Citosina , Metilación de ADN , Cromatina/genética , Epigénesis Genética , Transición Epitelial-Mesenquimal/genética , Humanos , Factores de Transcripción/genética
16.
Sci Adv ; 8(31): eabj8002, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35921406

RESUMEN

The epithelial-to-mesenchymal transition (EMT) is frequently co-opted by cancer cells to enhance migratory and invasive cell traits. It is a key contributor to heterogeneity, chemoresistance, and metastasis in many carcinoma types, where the intermediate EMT state plays a critical tumor-initiating role. We isolate multiple distinct single-cell clones from the SUM149PT human breast cell line spanning the EMT spectrum having diverse migratory, tumor-initiating, and metastatic qualities, including three unique intermediates. Using a multiomics approach, we identify CBFß as a key regulator of metastatic ability in the intermediate state. To quantify epithelial-mesenchymal heterogeneity within tumors, we develop an advanced multiplexed immunostaining approach using SUM149-derived orthotopic tumors and find that the EMT state and epithelial-mesenchymal heterogeneity are predictive of overall survival in a cohort of stage III breast cancer. Our model reveals previously unidentified insights into the complex EMT spectrum and its regulatory networks, as well as the contributions of epithelial-mesenchymal plasticity (EMP) in tumor heterogeneity in breast cancer.


Asunto(s)
Neoplasias de la Mama , Neoplasias de la Mama/patología , Línea Celular Tumoral , Progresión de la Enfermedad , Transición Epitelial-Mesenquimal/genética , Femenino , Humanos , Metástasis de la Neoplasia
17.
Dev Cell ; 55(5): 544-557.e6, 2020 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-33120014

RESUMEN

Differentiation therapy utilizes our understanding of the hierarchy of cellular systems to pharmacologically induce a shift toward terminal commitment. While this approach has been a paradigm in treating certain hematological malignancies, efforts to translate this success to solid tumors have met with limited success. Mammary-specific activation of PKA in mouse models leads to aberrant differentiation and diminished self-renewing potential of the basal compartment, which harbors mammary repopulating cells. PKA activation results in tumors that are more benign, exhibiting reduced metastatic propensity, loss of tumor-initiating potential, and increased sensitivity to chemotherapy. Analysis of tumor histopathology revealed features of overt differentiation with papillary characteristics. Longitudinal single-cell profiling at the hyperplasia and tumor stages uncovered an altered path of tumor evolution whereby PKA curtails the emergence of aggressive subpopulations. Acting through the repression of SOX4, PKA activation promotes tumor differentiation and represents a possible adjuvant to chemotherapy for certain breast cancers.


Asunto(s)
Diferenciación Celular , Autorrenovación de las Células , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Neoplasias Mamarias Animales/enzimología , Neoplasias Mamarias Animales/patología , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Linaje de la Célula , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Activación Enzimática , Femenino , Amplificación de Genes , Sitios Genéticos , Genoma Humano , Humanos , Neoplasias Mamarias Animales/genética , Ratones , Metástasis de la Neoplasia , Factores de Transcripción SOXC/metabolismo , Transducción de Señal
18.
Nat Commun ; 11(1): 2042, 2020 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-32341362

RESUMEN

The endothelial cell adhesion molecule E-selectin is a key component of the bone marrow hematopoietic stem cell (HSC) vascular niche regulating balance between HSC self-renewal and commitment. We now report in contrast, E-selectin directly triggers signaling pathways that promote malignant cell survival and regeneration. Using acute myeloid leukemia (AML) mouse models, we show AML blasts release inflammatory mediators that upregulate endothelial niche E-selectin expression. Alterations in cell-surface glycosylation associated with oncogenesis enhances AML blast binding to E-selectin and enable promotion of pro-survival signaling through AKT/NF-κB pathways. In vivo AML blasts with highest E-selectin binding potential are 12-fold more likely to survive chemotherapy and main contributors to disease relapse. Absence (in Sele-/- hosts) or therapeutic blockade of E-selectin using small molecule mimetic GMI-1271/Uproleselan effectively inhibits this niche-mediated pro-survival signaling, dampens AML blast regeneration, and strongly synergizes with chemotherapy, doubling the duration of mouse survival over chemotherapy alone, whilst protecting endogenous HSC.


Asunto(s)
Resistencia a Antineoplásicos , Selectina E/antagonistas & inhibidores , Selectina E/metabolismo , Leucemia Mieloide Aguda/tratamiento farmacológico , Animales , Antineoplásicos/uso terapéutico , Médula Ósea , Técnicas de Cocultivo , Modelos Animales de Enfermedad , Femenino , Glucolípidos/uso terapéutico , Glicosilación , Células Madre Hematopoyéticas/citología , Humanos , Inflamación , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal
19.
Sci Transl Med ; 10(436)2018 04 11.
Artículo en Inglés | MEDLINE | ID: mdl-29643230

RESUMEN

Patients undergoing surgical resection of primary breast tumors confront a risk for metastatic recurrence that peaks sharply 12 to 18 months after surgery. The cause of early metastatic relapse in breast cancer has long been debated, with many ascribing these relapses to the natural progression of the disease. Others have proposed that some aspect of surgical tumor resection triggers the outgrowth of otherwise-dormant metastases, leading to the synchronous pattern of relapse. Clinical data cannot distinguish between these hypotheses, and previous experimental approaches have not provided clear answers. Such uncertainty hinders the development and application of therapeutic approaches that could potentially reduce early metastatic relapse. We describe an experimental model system that definitively links surgery and the subsequent wound-healing response to the outgrowth of tumor cells at distant anatomical sites. Specifically, we find that the systemic inflammatory response induced after surgery promotes the emergence of tumors whose growth was otherwise restricted by a tumor-specific T cell response. Furthermore, we demonstrate that perioperative anti-inflammatory treatment markedly reduces tumor outgrowth in this model, suggesting that similar approaches might substantially reduce early metastatic recurrence in breast cancer patients.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/metabolismo , Animales , Biomarcadores de Tumor/inmunología , Neoplasias de la Mama/inmunología , Linfocitos T CD8-positivos/metabolismo , Línea Celular Tumoral , Femenino , Ratones , Metástasis de la Neoplasia/inmunología , Recurrencia Local de Neoplasia/inmunología , Recurrencia Local de Neoplasia/metabolismo
20.
Artículo en Inglés | MEDLINE | ID: mdl-28057845

RESUMEN

Although important strides have been made in targeted therapy for certain leukemias and subtypes of breast cancer, the standard of care for most carcinomas still involves chemotherapy, radiotherapy, surgery, or a combination of these. Two processes serve as obstacles to the successful treatment of carcinomas. First, a majority of deaths from these types of cancers occurs as a result of distant metastases and not the primary tumors themselves. Second, subsets of cells that are able to survive conventional therapy drive the aggressive relapse of the tumors, often in forms that are resistant to treatment. A frequently observed feature of malignant carcinomas is the loss of epithelial traits and the gain of certain mesenchymal ones that are programmed by the cell-biological program termed the epithelial-to-mesenchymal transition (EMT). The EMT program can confer (i) an ability to disseminate, (ii) an ability to become stem-like tumor-initiating cells, (iii) an ability to found new tumor colonies at distant anatomical sites, and (iv) an elevated resistance to therapy. These multiple powers of the EMT program explain why it has become an attractive target for therapeutic intervention. Recent work has revealed the variable nature of the EMT, with multiple versions of the program being observed depending on the tissue context and the stage of tumor progression. In this review, we attempt to crystallize emerging concepts in the research on EMT and stemness and discuss the benefits of using a differentiation-based therapeutic strategy for the eradication of stem-like populations that have adopted various versions of the EMT program.


Asunto(s)
Diferenciación Celular/fisiología , Transformación Celular Neoplásica/patología , Transición Epitelial-Mesenquimal/fisiología , Leucemia/tratamiento farmacológico , Células Madre Neoplásicas/citología , Animales , Humanos , Leucemia/metabolismo , Neoplasias/tratamiento farmacológico , Neoplasias/metabolismo , Neoplasias/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA