Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 30
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
J Mammary Gland Biol Neoplasia ; 29(1): 3, 2024 Jan 30.
Artículo en Inglés | MEDLINE | ID: mdl-38289401

RESUMEN

During female adolescence and pregnancy, rising levels of hormones result in a cyclic source of signals that control the development of mammary tissue. While such alterations are well understood from a whole-gland perspective, the alterations that such hormones bring to organoid cultures derived from mammary glands have yet to be fully mapped. This is of special importance given that organoids are considered suitable systems to understand cross species breast development. Here we utilized single-cell transcriptional profiling to delineate responses of murine and human normal breast organoid systems to female hormones across evolutionary distinct species. Collectively, our study represents a molecular atlas of epithelial dynamics in response to estrogen and pregnancy hormones.


Asunto(s)
Mama , Estrógenos , Adolescente , Embarazo , Humanos , Animales , Ratones , Femenino , Organoides
2.
bioRxiv ; 2023 Sep 29.
Artículo en Inglés | MEDLINE | ID: mdl-37808747

RESUMEN

During female adolescence and pregnancy, rising levels of hormones result in a cyclic source of signals that control the development of mammary tissue. While such alterations are well understood from a whole-gland perspective, the alterations that such hormones bring to organoid cultures derived from mammary glands have yet to be fully mapped. This is of special importance given that organoids are considered suitable systems to understand cross species breast development. Here we utilized single-cell transcriptional profiling to delineate responses of murine and human normal breast organoid systems to female hormones across evolutionary distinct species. Collectively, our study represents a molecular atlas of epithelial dynamics in response to estrogen and pregnancy hormones.

3.
Int J Mol Sci ; 24(13)2023 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-37446393

RESUMEN

L-Asparaginase (ASNase) is a biopharmaceutical used as an essential drug in the treatment of acute lymphoblastic leukemia (ALL). Yet, some cases of ALL are naturally resistant to ASNase treatment, which results in poor prognosis. The REH ALL cell line, used as a model for studying the most common subtype of ALL, is considered resistant to treatment with ASNase. Cathepsin B (CTSB) is one of the proteases involved in the regulation of in vivo ASNase serum half-life and it has also been associated with the progression and resistance to treatment of several solid tumors. Previous works have shown that, in vitro, ASNase is degraded when incubated with REH cell lysate, which is prevented by a specific CTSB inhibitor, suggesting a function of this protease in the ASNase resistance of REH cells. In this work, we utilized a combination of CRISPR/Cas9 gene targeting and enzymatic measurements to investigate the relevance of CTSB on ASNase treatment resistance in the ALL model cell line. We found that deletion of CTSB in REH ALL cells did not confer ASNase treatment sensitivity, thus suggesting that intrinsic expression of CTSB is not a mechanism that drives the resistant nature of these ALL cells to enzymes used as the first-line treatment against leukemia.


Asunto(s)
Antineoplásicos , Leucemia-Linfoma Linfoblástico de Células Precursoras , Humanos , Asparaginasa/farmacología , Asparaginasa/metabolismo , Factor Intrinseco/uso terapéutico , Catepsina B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Línea Celular , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico
4.
J Mammary Gland Biol Neoplasia ; 28(1): 16, 2023 07 14.
Artículo en Inglés | MEDLINE | ID: mdl-37450228

RESUMEN

The onset of pregnancy marks the start of offspring development, and represents the key physiological event that induces re-organization and specialization of breast tissue. Such drastic tissue remodeling has also been linked to epithelial cell transformation and the establishment of breast cancer (BC). While patient outcomes for BC overall continue to improve across subtypes, prognosis remains dismal for patients with gestational breast cancer (GBC) and post-partum breast cancer (PPBC), as pregnancy and lactation pose additional complications and barriers to several gold standard clinical approaches. Moreover, delayed diagnosis and treatment, coupled with the aggressive time-scale in which GBC metastasizes, inevitably contributes to the higher incidence of disease recurrence and patient mortality. Therefore, there is an urgent and evident need to better understand the factors contributing to the establishment and spreading of BC during pregnancy. In this review, we provide a literature-based overview of the diagnostics and treatments available to patients with BC more broadly, and highlight the treatment deficit patients face due to gestational status. Further, we review the current understanding of the molecular and cellular mechanisms driving GBC, and discuss recent advances in model systems that may support the identification of targetable approaches to block BC development and dissemination during pregnancy. Our goal is to provide an updated perspective on GBC, and to inform critical areas needing further exploration to improve disease outcome.


Asunto(s)
Neoplasias de la Mama , Embarazo , Femenino , Humanos , Neoplasias de la Mama/patología , Periodo Posparto , Pronóstico , Lactancia , Modelos Biológicos
5.
bioRxiv ; 2023 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-37292919

RESUMEN

Despite the progress made in identifying cellular factors and mechanisms that predict progression and metastasis, breast cancer remains the second leading cause of death for women in the US. Using The Cancer Genome Atlas and mouse models of spontaneous and invasive mammary tumorigenesis, we identified that loss of function of interferon regulatory factor 5 (IRF5) is a predictor of metastasis and survival. Histologic analysis of Irf5 -/- mammary glands revealed expansion of luminal and myoepithelial cells, loss of organized glandular structure, and altered terminal end budding and migration. RNA-seq and ChIP-seq analyses of primary mammary epithelial cells from Irf5 +/+ and Irf5 -/- littermate mice revealed IRF5-mediated transcriptional regulation of proteins involved in ribosomal biogenesis. Using an invasive model of breast cancer lacking Irf5 , we demonstrate that IRF5 re-expression inhibits tumor growth and metastasis via increased trafficking of tumor infiltrating lymphocytes and altered tumor cell protein synthesis. These findings uncover a new function for IRF5 in the regulation of mammary tumorigenesis and metastasis. Highlights: Loss of IRF5 is a predictor of metastasis and survival in breast cancer.IRF5 contributes to the regulation of ribosome biogenesis in mammary epithelial cells.Loss of IRF5 function in mammary epithelial cells leads to increased protein translation.

6.
Sci Transl Med ; 15(686): eabl4414, 2023 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-36888695

RESUMEN

Natural killer (NK) cells are cytotoxic lymphocytes that accumulate within the tumor microenvironment and are generally considered to be antitumorigenic. Using single-cell RNA sequencing and functional analysis of multiple triple-negative breast cancer (TNBC) and basal tumor samples, we observed a unique subcluster of Socs3highCD11b-CD27- immature NK cells that were present only in TNBC samples. These tumor-infiltrating NK cells expressed a reduced cytotoxic granzyme signature and, in mice, were responsible for activating cancer stem cells through Wnt signaling. NK cell-mediated activation of these cancer stem cells subsequently enhanced tumor progression in mice, whereas depletion of NK cells or Wnt ligand secretion from NK cells by LGK-974 decreased tumor progression. In addition, NK cell depletion or inhibition of their function improved anti-programmed cell death ligand 1 (PD-L1) antibody or chemotherapy response in mice with TNBC. Furthermore, tumor samples from patients with TNBC and non-TNBC revealed that increased numbers of CD56bright NK cells were present in TNBC tumors and were correlated to poor overall survival in patients with TNBC. Together, our findings identify a population of protumorigenic NK cells that may be exploited for both diagnostic and therapeutic strategies to improve outcomes for patients with TNBC.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama Triple Negativas , Humanos , Animales , Ratones , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Antineoplásicos/uso terapéutico , Células Asesinas Naturales , Antígeno B7-H1/metabolismo , Microambiente Tumoral
7.
Exp Biol Med (Maywood) ; 247(24): 2176-2183, 2022 12.
Artículo en Inglés | MEDLINE | ID: mdl-36408534

RESUMEN

While mouse models and two-dimensional (2D) cell culture systems have dominated as research tools for cancer biology, three-dimensional (3D) cultures have gained traction as a new approach that retains features of in vivo biology within an in vitro system. Over time, 3D culture systems have evolved from spheroids and tumorspheres to organoids, and by doing so, they have become more complex and representative of original tissue. Such technological improvements have mostly benefited the study of heterogeneous solid tumors, like those found in breast cancer (BC), by providing an attractive avenue for scalable drug testing and biobank generation. Experimentally, organoids have been used in the BC field to dissect mechanisms related to cellular invasion and metastasis-and through co-culture methods-epithelial interactions with stromal and immune cells. In addition, organoid studies of wild-type mouse models and healthy donor samples have provided insight into the basic developmental cellular and molecular biology of the mammary gland, which may inform one's understanding of the initial stages of cancer development and progression.


Asunto(s)
Neoplasias , Esferoides Celulares , Ratones , Animales , Técnicas de Cocultivo , Células Tumorales Cultivadas , Organoides , Modelos Animales de Enfermedad , Neoplasias/patología
8.
Cell Rep ; 37(10): 110099, 2021 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-34879282

RESUMEN

Pregnancy reprograms mammary epithelial cells (MECs) to control their responses to pregnancy hormone re-exposure and carcinoma progression. However, the influence of pregnancy on the mammary microenvironment is less clear. Here, we used single-cell RNA sequencing to profile the composition of epithelial and non-epithelial cells in mammary tissue from nulliparous and parous female mice. Our analysis indicates an expansion of γδ natural killer T-like immune cells (NKTs) following pregnancy and upregulation of immune signaling molecules in post-pregnancy MECs. We show that expansion of NKTs following pregnancy is due to elevated expression of the antigen-presenting molecule CD1d on MECs. Loss of CD1d expression on post-pregnancy MECs, or overall lack of activated NKTs, results in mammary oncogenesis. Collectively, our findings illustrate how pregnancy-induced changes modulate the communication between MECs and the immune microenvironment and establish a causal link between pregnancy, the immune microenvironment, and mammary oncogenesis.


Asunto(s)
Proliferación Celular , Transformación Celular Neoplásica/inmunología , Células Epiteliales/inmunología , Activación de Linfocitos , Glándulas Mamarias Animales/inmunología , Neoplasias Mamarias Experimentales/inmunología , Células T Asesinas Naturales/inmunología , Paridad , Animales , Antígenos CD1d/metabolismo , Comunicación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Regulación Neoplásica de la Expresión Génica , Genes BRCA1 , Genes myc , Glándulas Mamarias Animales/metabolismo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Neoplasias Mamarias Experimentales/patología , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Células T Asesinas Naturales/metabolismo , Embarazo , Receptores de Antígenos de Linfocitos T gamma-delta/metabolismo , Transducción de Señal , Microambiente Tumoral
9.
J Med Chem ; 64(18): 13902-13917, 2021 09 23.
Artículo en Inglés | MEDLINE | ID: mdl-34515477

RESUMEN

The nucleosome remodeling factor (NURF) alters chromatin accessibility through interactions with its largest subunit,the bromodomain PHD finger transcription factor BPTF. BPTF is overexpressed in several cancers and is an emerging anticancer target. Targeting the BPTF bromodomain presents a potential strategy for its inhibition and the evaluation of its functional significance; however, inhibitor development for BPTF has lagged behind those of other bromodomains. Here we describe the development of pyridazinone-based BPTF inhibitors. The lead compound, BZ1, possesses a high potency (Kd = 6.3 nM) and >350-fold selectivity over BET bromodomains. We identify an acidic triad in the binding pocket to guide future designs. We show that our inhibitors sensitize 4T1 breast cancer cells to doxorubicin but not BPTF knockdown cells, suggesting a specificity to BPTF. Given the high potency and good physicochemical properties of these inhibitors, we anticipate that they will be useful starting points for chemical tool development to explore the biological roles of BPTF.


Asunto(s)
Antineoplásicos/farmacología , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Piridazinas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Secuencia de Aminoácidos , Animales , Antígenos Nucleares/química , Antineoplásicos/química , Antineoplásicos/toxicidad , Línea Celular Tumoral , Diseño de Fármacos , Ratones , Estructura Molecular , Proteínas del Tejido Nervioso/química , Dominios Proteicos , Piridazinas/química , Piridazinas/toxicidad , Relación Estructura-Actividad , Factores de Transcripción/química
10.
Oncogene ; 40(25): 4384-4397, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-34103681

RESUMEN

Recurrence of metastatic breast cancer stemming from acquired endocrine and chemotherapy resistance remains a health burden for women with luminal (ER+) breast cancer. Disseminated ER+ tumor cells can remain viable but quiescent for years to decades. Contributing factors to metastatic spread include the maintenance and expansion of breast cancer stem cells (CSCs). Breast CSCs frequently exist as a minority population in therapy resistant tumors. In this study, we show that cytoplasmic complexes composed of steroid receptor (SR) co-activators, PELP1 and SRC-3, modulate breast CSC expansion through upregulation of the HIF-activated metabolic target genes PFKFB3 and PFKFB4. Seahorse metabolic assays demonstrated that cytoplasmic PELP1 influences cellular metabolism by increasing both glycolysis and mitochondrial respiration. PELP1 interacts with PFKFB3 and PFKFB4 proteins, and inhibition of PFKFB3 and PFKFB4 kinase activity blocks PELP1-induced tumorspheres and protein-protein interactions with SRC-3. PFKFB4 knockdown inhibited in vivo emergence of circulating tumor cell (CTC) populations in mammary intraductal (MIND) models. Application of PFKFB inhibitors in combination with ER targeted therapies blocked tumorsphere formation in multiple models of advanced breast cancer including tamoxifen (TamR) and paclitaxel (TaxR) resistant models, murine tumor cells, and ER+ patient-derived organoids (PDxO). Together, our data suggest that PELP1, SRC-3, and PFKFBs cooperate to drive ER+ tumor cell populations that include CSCs and CTCs. Identifying non-ER pharmacological targets offers a useful approach to blocking metastatic escape from standard of care ER/estrogen (E2)-targeted strategies to overcome endocrine and chemotherapy resistance.


Asunto(s)
Neoplasias de la Mama/genética , Proteínas Co-Represoras/genética , Resistencia a Antineoplásicos/genética , Coactivador 3 de Receptor Nuclear/genética , Fosfofructoquinasa-2/genética , Receptores de Estrógenos/genética , Factores de Transcripción/genética , Animales , Mama/patología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/patología , Línea Celular Tumoral , Estrógenos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Humanos , Células MCF-7 , Ratones , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Paclitaxel/farmacología , Fosforilación/genética , Tamoxifeno/farmacología , Regulación hacia Arriba/genética
11.
J Mammary Gland Biol Neoplasia ; 26(1): 43-66, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33988830

RESUMEN

The developing mammary gland depends on several transcription-dependent networks to define cellular identities and differentiation trajectories. Recent technological advancements that allow for single-cell profiling of gene expression have provided an initial picture into the epithelial cellular heterogeneity across the diverse stages of gland maturation. Still, a deeper dive into expanded molecular signatures would improve our understanding of the diversity of mammary epithelial and non-epithelial cellular populations across different tissue developmental stages, mouse strains and mammalian species. Here, we combined differential mammary gland fractionation approaches and transcriptional profiles obtained from FACS-isolated mammary cells to improve our definitions of mammary-resident, cellular identities at the single-cell level. Our approach yielded a series of expression signatures that illustrate the heterogeneity of mammary epithelial cells, specifically those of the luminal fate, and uncovered transcriptional changes to their lineage-defined, cellular states that are induced during gland development. Our analysis also provided molecular signatures that identified non-epithelial mammary cells, including adipocytes, fibroblasts and rare immune cells. Lastly, we extended our study to elucidate expression signatures of human, breast-resident cells, a strategy that allowed for the cross-species comparison of mammary epithelial identities. Collectively, our approach improved the existing signatures of normal mammary epithelial cells, as well as elucidated the diversity of non-epithelial cells in murine and human breast tissue. Our study provides a useful resource for future studies that use single-cell molecular profiling strategies to understand normal and malignant breast development.


Asunto(s)
Células Epiteliales/fisiología , Perfilación de la Expresión Génica/métodos , Glándulas Mamarias Animales/fisiología , Glándulas Mamarias Humanas/fisiología , Análisis de Secuencia de ARN/métodos , Análisis de la Célula Individual/métodos , Transcriptoma , Animales , Linaje de la Célula/fisiología , Células Epiteliales/citología , Femenino , Humanos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Humanas/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL
12.
Semin Cell Dev Biol ; 114: 93-112, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33082117

RESUMEN

Our understanding of the molecular events underpinning the development of mammalian organ systems has been increasing rapidly in recent years. With the advent of new and improved next-generation sequencing methods, we are now able to dig deeper than ever before into the genomic and epigenomic events that play critical roles in determining the fates of stem and progenitor cells during the development of an embryo into an adult. In this review, we detail and discuss the genes and pathways that are involved in mammary gland development, from embryogenesis, through maturation into an adult gland, to the role of pregnancy signals in directing the terminal maturation of the mammary gland into a milk producing organ that can nurture the offspring. We also provide an overview of the latest research in the single-cell genomics of mammary gland development, which may help us to understand the lineage commitment of mammary stem cells (MaSCs) into luminal or basal epithelial cells that constitute the mammary gland. Finally, we summarize the use of 3D organoid cultures as a model system to study the molecular events during mammary gland development. Our increased investigation of the molecular requirements for normal mammary gland development will advance the discovery of targets to predict breast cancer risk and the development of new breast cancer therapies.


Asunto(s)
Células Epiteliales/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Humanas/crecimiento & desarrollo , Animales , Diferenciación Celular , Femenino , Humanos
13.
J Mammary Gland Biol Neoplasia ; 25(4): 351-366, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33131024

RESUMEN

The use of mouse derived mammary organoids can provide a unique strategy to study mammary gland development across a normal life cycle, as well as offering insights into how malignancies form and progress. Substantial cellular and epigenomic changes are triggered in response to pregnancy hormones, a reaction that engages molecular and cellular changes that transform the mammary epithelial cells into "milk producing machines". Such epigenomic alterations remain stable in post-involution mammary epithelial cells and control the reactivation of gene transcription in response to re-exposure to pregnancy hormones. Thus, a system that tightly controls exposure to pregnancy hormones, epigenomic alterations, and activation of transcription will allow for a better understanding of such molecular switches. Here, we describe the characterization of ex vivo cultures to mimic the response of mammary organoid cultures to pregnancy hormones and to understand gene regulation and epigenomic reprogramming on consecutive hormone exposure. Our findings suggest that this system yields similar epigenetic modifications to those reported in vivo, thus representing a suitable model to closely track epigenomic rearrangement and define unknown players of pregnancy-induced development.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Estradiol/metabolismo , Glándulas Mamarias Animales/crecimiento & desarrollo , Progesterona/metabolismo , Prolactina/metabolismo , Animales , Diferenciación Celular/genética , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Epigénesis Genética , Células Epiteliales/fisiología , Femenino , Código de Histonas , Histonas/genética , Lactancia/genética , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Modelos Animales , Organoides , Embarazo
14.
Front Oncol ; 10: 569985, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33072601

RESUMEN

Myeloid cell heterogeneity remains poorly studied in breast cancer, and particularly in premalignancy. Here, we used single cell RNA sequencing to characterize macrophage diversity in mouse pre-invasive lesions as compared to lesions undergoing localized invasion. Several subpopulations of macrophages with transcriptionally distinct profiles were identified, two of which resembled macrophages in the steady state. While all subpopulations expressed tumor-promoting genes, many of the populations expressed pro-inflammatory genes, differing from reports in tumor-associated macrophages. Gene profiles of the myeloid cells were similar between early and late stages of premalignancy, although expansion of some subpopulations occurred. These results unravel macrophage heterogeneity in early progression and may provide insight into early intervention strategies that target macrophages.

15.
J Exp Med ; 217(10)2020 10 05.
Artículo en Inglés | MEDLINE | ID: mdl-32667673

RESUMEN

C-C chemokine receptor type 2 (CCR2) is expressed on monocytes and facilitates their recruitment to tumors. Though breast cancer cells also express CCR2, its functions in these cells are unclear. We found that Ccr2 deletion in cancer cells led to reduced tumor growth and approximately twofold longer survival in an orthotopic, isograft breast cancer mouse model. Deletion of Ccr2 in cancer cells resulted in multiple alterations associated with better immune control: increased infiltration and activation of cytotoxic T lymphocytes (CTLs) and CD103+ cross-presenting dendritic cells (DCs), as well as up-regulation of MHC class I and down-regulation of checkpoint regulator PD-L1 on the cancer cells. Pharmacological or genetic targeting of CCR2 increased cancer cell sensitivity to CTLs and enabled the cancer cells to induce DC maturation toward the CD103+ subtype. Consistently, Ccr2-/- cancer cells did not induce immune suppression in Batf3-/- mice lacking CD103+ DCs. Our results establish that CCR2 signaling in cancer cells can orchestrate suppression of the immune response.


Asunto(s)
Inmunidad Adaptativa/inmunología , Tolerancia Inmunológica , Neoplasias Mamarias Experimentales/inmunología , Receptores CCR2/fisiología , Inmunidad Adaptativa/fisiología , Animales , Apoptosis , Antígeno B7-H1/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/fisiología , Femenino , Antígenos de Histocompatibilidad Clase I/metabolismo , Tolerancia Inmunológica/inmunología , Tolerancia Inmunológica/fisiología , Interferones/metabolismo , Ratones , Ratones Endogámicos C57BL , Receptores CCR2/inmunología , Linfocitos T Citotóxicos/inmunología , Linfocitos T Citotóxicos/fisiología
16.
Nat Commun ; 11(1): 2649, 2020 05 27.
Artículo en Inglés | MEDLINE | ID: mdl-32461571

RESUMEN

Pregnancy causes a series of cellular and molecular changes in mammary epithelial cells (MECs) of female adults. In addition, pregnancy can also modify the predisposition of rodent and human MECs to initiate oncogenesis. Here, we investigate how pregnancy reprograms enhancer chromatin in the mammary epithelium of mice and influences the transcriptional output of the oncogenic transcription factor cMYC. We find that pregnancy induces an expansion of the active cis-regulatory landscape of MECs, which influences the activation of pregnancy-related programs during re-exposure to pregnancy hormones in vivo and in vitro. Using inducible cMYC overexpression, we demonstrate that post-pregnancy MECs are resistant to the downstream molecular programs induced by cMYC, a response that blunts carcinoma initiation, but does not perturb the normal pregnancy-induced epigenomic landscape. cMYC overexpression drives post-pregnancy MECs into a senescence-like state, and perturbations of this state increase malignant phenotypic changes. Taken together, our findings provide further insight into the cell-autonomous signals in post-pregnancy MECs that underpin the regulation of gene expression, cellular activation, and resistance to malignant development.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , Animales , Carcinogénesis/genética , Transformación Celular Neoplásica/patología , Epigénesis Genética , Epigenoma , Células Epiteliales/metabolismo , Femenino , Regulación de la Expresión Génica , Glándulas Mamarias Animales/citología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Oncogenes/genética , Embarazo , Complicaciones del Embarazo/etiología , Complicaciones del Embarazo/genética , Transducción de Señal , Factores de Transcripción/genética , Factores de Transcripción/metabolismo
17.
Trends Mol Med ; 25(10): 866-881, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31383623

RESUMEN

Several studies have made strong efforts to understand how age and parity modulate the risk of breast cancer. A holistic understanding of the dynamic regulation of the morphological, cellular, and molecular milieu of the mammary gland offers insights into the drivers of breast cancer development as well as into potential prophylactic interventions, the latter being a longstanding ambition of the research and clinical community aspiring to eradicate the disease. In this review we discuss mechanisms that react to pregnancy signals, and we delineate the nuances of pregnancy-associated dynamism that contribute towards either breast cancer development or prevention. Further definition of the molecular basis of parity and breast cancer risk may allow the elaboration of tools to predict and survey those who are at risk of breast cancer development.


Asunto(s)
Neoplasias de la Mama/prevención & control , Complicaciones del Embarazo/prevención & control , Animales , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Epigenómica , Femenino , Humanos , Embarazo , Complicaciones del Embarazo/genética , Complicaciones del Embarazo/metabolismo , Factores de Riesgo
18.
Org Biomol Chem ; 17(7): 2020-2027, 2019 02 13.
Artículo en Inglés | MEDLINE | ID: mdl-30706071

RESUMEN

Bromodomain and PHD finger containing protein transcription factor (BPTF) is an epigenetic protein involved in chromatin remodelling and is a potential anticancer target. The BPTF bromodomain has one reported small molecule inhibitor (AU1, rac-1). Here, advances made on the structure-activity relationship of a BPTF bromodomain ligand are reported using a combination of experimental and molecular dynamics simulations leading to the active enatiomer (S)-1. Additionally, a ligand deconstruction analysis was conducted to characterize important pharmacophores for engaging the BPTF bromodomain. These studies have been enabled by a protein-based fluorine NMR approach, highlighting the versatility of the method for selectivity, ligand deconstruction, and ligand binding. To enable future analysis of biological activity, cell growth analyses in a panel of cancer cell lines were carried out using CRISPR-Cas9 and (S)-1 to identify cell-based model systems that are sensitive to BPTF inhibition.


Asunto(s)
Proteínas del Tejido Nervioso/antagonistas & inhibidores , Pirazoles/farmacología , Piridinas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Antígenos Nucleares , Proliferación Celular , Cristalografía por Rayos X , Humanos , Ligandos , Espectroscopía de Resonancia Magnética , Simulación de Dinámica Molecular , Estructura Molecular , Pirazoles/síntesis química , Pirazoles/química , Piridinas/síntesis química , Piridinas/química , Bibliotecas de Moléculas Pequeñas/síntesis química , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
19.
Stem Cell Reports ; 9(1): 23-31, 2017 07 11.
Artículo en Inglés | MEDLINE | ID: mdl-28579392

RESUMEN

Chromatin remodeling is a key requirement for transcriptional control of cellular differentiation. However, the factors that alter chromatin architecture in mammary stem cells (MaSCs) are poorly understood. Here, we show that BPTF, the largest subunit of the NURF chromatin remodeling complex, is essential for MaSC self-renewal and differentiation of mammary epithelial cells (MECs). BPTF depletion arrests cells at a previously undefined stage of epithelial differentiation that is associated with an incapacity to achieve the luminal cell fate. Moreover, genome-wide analysis of DNA accessibility following genetic or chemical inhibition, suggests a role for BPTF in maintaining the open chromatin landscape at enhancers regions in MECs. Collectively, our study implicates BPTF in maintaining the unique epigenetic state of MaSCs.


Asunto(s)
Antígenos Nucleares/metabolismo , Proliferación Celular , Cromatina/metabolismo , Células Epiteliales/citología , Glándulas Mamarias Animales/citología , Proteínas del Tejido Nervioso/metabolismo , Células Madre/citología , Factores de Transcripción/metabolismo , Animales , Antígenos Nucleares/genética , Diferenciación Celular , Células Cultivadas , Cromatina/genética , Ensamble y Desensamble de Cromatina , Epigénesis Genética , Células Epiteliales/metabolismo , Femenino , Regulación del Desarrollo de la Expresión Génica , Glándulas Mamarias Animales/metabolismo , Proteínas del Tejido Nervioso/genética , Células Madre/metabolismo , Factores de Transcripción/genética
20.
ACS Chem Biol ; 10(10): 2246-56, 2015 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-26158404

RESUMEN

Bromodomain-containing protein dysregulation is linked to cancer, diabetes, and inflammation. Selective inhibition of bromodomain function is a newly proposed therapeutic strategy. We describe a (19)F NMR dual screening method for small molecule discovery using fluorinated tryptophan resonances on two bromodomain-containing proteins. The chemical shift dispersion of (19)F resonances within fluorine-labeled proteins enables the simultaneous analysis of two fluorinated bromodomains by NMR. A library of 229 small molecules was screened against the first bromodomain of Brd4 and the BPTF bromodomain. We report the first small molecule selective for BPTF over Brd4, termed AU1. The Kd = 2.8 µM for AU1, which is active in a cell-based reporter assay. No binding is detected with Brd4. Three new Brd4 inhibitors with submicromolar affinity were also discovered. Brd4 hits were validated in a thermal stability assay and potency determined via fluorescence anisotropy. The speed, ease of interpretation, and low protein concentration needed for protein-observed (19)F NMR experiments in a multiprotein format offers a new method to discover and characterize selective ligands for bromodomain-containing proteins.


Asunto(s)
Antígenos Nucleares/química , Proteínas del Tejido Nervioso/química , Proteínas Nucleares/química , Factores de Transcripción/química , Unión Competitiva , Proteínas de Ciclo Celular , Línea Celular , Sistemas de Liberación de Medicamentos , Evaluación Preclínica de Medicamentos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Flúor/química , Humanos , Concentración 50 Inhibidora , Imagen por Resonancia Magnética , Estructura Molecular , Estructura Terciaria de Proteína , Relación Estructura-Actividad , Temperatura
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA