Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
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.

2.
Int Urogynecol J ; 34(8): 1915-1921, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36795112

RESUMEN

INTRODUCTION AND HYPOTHESIS: The incidence of trocar bladder puncture during midurethral sling (MUS) surgery varies widely. We aim to further characterize risk factors for bladder puncture and examine its long-term impact on storage and emptying. METHODS: This is an Institutional Review Board-approved, retrospective chart review of women who underwent MUS surgery at our institution from 2004 to 2018 with ≥12 months of follow-up. Unless prolonged catheterization was necessary, a voiding trial was performed prior to discharge, or the next morning in outpatients, regardless of puncture. Preoperative and postoperative details were obtained from office charts and operative records. RESULTS: Of 1,500 women, 1,063 (71%) had retropubic (RP) and 437 (29%) had transobturator MUS surgery. Mean follow-up was 34 months. Thirty-five women (2.3%) sustained a bladder puncture. RP approach and lower BMI were significantly associated with puncture. No statistical association was found between bladder puncture and age, previous pelvic surgery, or concomitant surgery. Mean day of discharge and day of successful voiding trial were not statistically different between the puncture and nonpuncture groups. There was no statistically significant difference in de novo storage and emptying symptoms between the two groups. Fifteen women in the puncture group had cystoscopy during follow-up and none had bladder exposure. Level of the resident performing trocar passage was not associated with bladder puncture. CONCLUSIONS: Lower BMI and RP approach are associated with bladder puncture during MUS surgery. Bladder puncture is not associated with additional perioperative complications, long-term urinary storage/voiding sequelae, or delayed bladder sling exposure. Standardized training minimizes bladder punctures in trainees of all levels.


Asunto(s)
Cabestrillo Suburetral , Incontinencia Urinaria de Esfuerzo , Femenino , Humanos , Vejiga Urinaria , Estudios Retrospectivos , Incontinencia Urinaria de Esfuerzo/cirugía , Incontinencia Urinaria de Esfuerzo/complicaciones , Cabestrillo Suburetral/efectos adversos , Instrumentos Quirúrgicos/efectos adversos , Resultado del Tratamiento
3.
J Perinat Med ; 50(2): 207-218, 2022 Feb 23.
Artículo en Inglés | MEDLINE | ID: mdl-34717055

RESUMEN

OBJECTIVES: Oxytocin (OXT) is widely used to facilitate labor. However, little is known about the effects of perinatal OXT exposure on the developing brain. We investigated the effects of maternal OXT administration on gene expression in perinatal mouse brains. METHODS: Pregnant C57BL/6 mice were treated with saline or OXT at term (n=6-7/group). Dams and pups were euthanized on gestational day (GD) 18.5 after delivery by C-section. Another set of dams was treated with saline or OXT (n=6-7/group) and allowed to deliver naturally; pups were euthanized on postnatal day 9 (PND9). Perinatal/neonatal brain gene expression was determined using Illumina BeadChip Arrays and real time quantitative PCR. Differential gene expression analyses were performed. In addition, the effect of OXT on neurite outgrowth was assessed using PC12 cells. RESULTS: Distinct and sex-specific gene expression patterns were identified in offspring brains following maternal OXT administration at term. The microarray data showed that female GD18.5 brains exhibited more differential changes in gene expression compared to male GD18.5 brains. Specifically, Cnot4 and Frmd4a were significantly reduced by OXT exposure in male and female GD18.5 brains, whereas Mtap1b, Srsf11, and Syn2 were significantly reduced only in female GD18.5 brains. No significant microarray differences were observed in PND9 brains. By quantitative PCR, OXT exposure reduced Oxtr expression in female and male brains on GD18.5 and PND9, respectively. PC12 cell differentiation assays revealed that OXT induced neurite outgrowth. CONCLUSIONS: Prenatal OXT exposure induces sex-specific differential regulation of several nervous system-related genes and pathways with important neural functions in perinatal brains.


Asunto(s)
Oxitocina , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Femenino , Expresión Génica/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , Oxitocina/farmacología , Embarazo
4.
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
5.
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.

6.
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
7.
Mol Biol Rep ; 47(4): 3241-3248, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-32219771

RESUMEN

In mammary gland development, normal stem cell activity occurs in the embryonic stage and postnatally. Research supports that certain breast cancers contain a small sub-population of cells that mimic stem-like activity. It is believed stem cell activation in the mutated mature human mammary tissue is what drives quiescent epithelial cells to convert to mesenchymal states initiating migration, invasion, and metastasis in breast cancer. The goal of the work reported herein was to investigate early mammary development gene expression in the postnatal pig using fine needle biopsy methods in order to establish a reliable model for human breast cancer detection. Tissue samples were collected from pig mammary glands beginning at Day 11 of age through Day 39 in order to capture early postnatal-growth gene expression. Based on the initial clustering analysis, two distinct clusters of gene expression profiles occurred before and after Day 25 of mammary development. Gene set enrichment analysis (GSEA) ontology indicated the cellular processes that changed after Day 25, and many of these processes were implicated in epithelial-mesenchymal transition (EMT) signaling events. Gene expression in the postnatal pig was compared with the Epithelial-Mesenchymal Transition gene database (dbEMT) confirming the presence of EMT activity in this early developmental program. Information from this study will provide insight into early postnatal mammary gland development. In addition, mechanisms exploited by mutated mammary epithelial cells leading to cancer initiation and growth may be detected considering that mutated mammary epithelial cells can reactivate early developmental signals.


Asunto(s)
Transición Epitelial-Mesenquimal/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/metabolismo , Animales , Neoplasias de la Mama/metabolismo , Células Epiteliales/metabolismo , Femenino , Expresión Génica/genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica/genética , Organogénesis/genética , Transducción de Señal/fisiología , Células Madre/metabolismo , Porcinos/genética , Transcriptoma/genética
8.
Curr Urol Rep ; 18(1): 1, 2017 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-28092069

RESUMEN

The etiology of men's lower urinary tract storage and voiding symptoms involves a contribution from both detrusor and outlet. As such, treatment of benign prostatic enlargement (BPE) ± benign prostatic obstruction (BPO) with standard alpha-adrenergic blockade and 5-alpha reductase inhibitor therapy may leave a population of men with persistent and bothersome urinary storage symptoms. An abundance of adequately powered, randomized, placebo-controlled trials indicate that the use of antimuscarinics and beta-3 adrenergic agonists, either alone or in combination with standard BPE/BPO therapy, leads to improvement in storage symptoms. At the same time, metrics associated with urinary emptying, such as maximum flow rate, post-void residual urinary volume, and incidence of treatment-associated urinary retention, appear to be stable and not significantly impacted by the addition of antimuscarinics.


Asunto(s)
Hiperplasia Prostática/terapia , Vejiga Urinaria Hiperactiva/terapia , Humanos , Síntomas del Sistema Urinario Inferior/etiología , Síntomas del Sistema Urinario Inferior/terapia , Masculino , Hiperplasia Prostática/complicaciones , Ensayos Clínicos Controlados Aleatorios como Asunto , Vejiga Urinaria Hiperactiva/etiología , Retención Urinaria
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA