Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Mamm Genome ; 29(9-10): 632-655, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30073618

RESUMEN

The breast-feeding neonate depends on mother's milk for both macronutrients and micronutrients including minerals. The goals of the present study were to document the effects of genetic background in mice on milk concentrations of select minerals and to use genome-wide association study (GWAS) to identify quantitative trait loci (QTL) regulating milk mineral concentrations. Milk samples from lactating mice in each of 31 different inbred strains of the mouse diversity panel (MDP) were analyzed by inductively coupled plasma-optical emission spectroscopy to determine the concentrations of calcium (Ca), copper (Cu), iron (Fe), potassium (K), magnesium (Mg), sodium (Na), phosphorus (P), sulfur (S), and zinc (Zn). GWAS identified a single pleiotropic milk mineral concentration QTL (Mmcq) on chromosome 3 for Ca, Mg, and P. For the remaining minerals, six QTL were detected for Fe, four for K, three for Zn, and one for S. Intersecting the Mmcq with published chromatin immunoprecipitation sequence data identified 15 out of 4633 high-linkage disequilibrium single-nucleotide polymorphisms that resided in signal transducer and activation of transcription 5 (STAT5) binding regions. A milk Fe-associated locus (Mmcq9) on chromosome 1 contained an SNP that localized to a STAT5 binding region and intersected with a HOMER motif predicted to bind the transcriptional regulator E74-Like ETS transcription factor 5. This locus also contained the genes for solute carrier family (Slc) members Slc9a2, Slc9a4, Slc39a10, and Slc40a1. Expression analysis of these transporters supports the conclusion that Slc9a2 and Slc40a1 within the mammary gland could mediate the effect of Mmcq9 on milk Fe concentration.


Asunto(s)
Proteínas de Transporte de Catión/genética , Mapeo Cromosómico , Hierro/metabolismo , Lactancia/genética , Leche/química , Sitios de Carácter Cuantitativo/genética , Intercambiadores de Sodio-Hidrógeno/genética , Animales , Sitios de Unión/genética , Simulación por Computador , Femenino , Expresión Génica , Estudio de Asociación del Genoma Completo , Hierro/análisis , Desequilibrio de Ligamiento , Ratones , Leche/metabolismo , Minerales/análisis , Minerales/metabolismo , Polimorfismo de Nucleótido Simple , Factores de Transcripción/metabolismo
2.
Mamm Genome ; 29(1-2): 24-37, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29487996

RESUMEN

Breast cancer risk is intimately intertwined with exposure to estrogens. While more than 160 breast cancer risk loci have been identified in humans, genetic interactions with estrogen exposure remain to be established. Strains of rodents exhibit striking differences in their responses to endogenous ovarian estrogens (primarily 17ß-estradiol). Similar genetic variation has been observed for synthetic estrogen agonists (ethinyl estradiol) and environmental chemicals that mimic the actions of estrogens (xenoestrogens). This review of literature highlights the extent of variation in responses to estrogens among strains of rodents and compiles the genetic loci underlying pathogenic effects of excessive estrogen signaling. Genetic linkage studies have identified a total of the 35 quantitative trait loci (QTL) affecting responses to 17ß-estradiol or diethylstilbestrol in five different tissues. However, the QTL appear to act in a tissue-specific manner with 9 QTL affecting the incidence or latency of mammary tumors induced by 17ß-estradiol or diethylstilbestrol. Mammary gland development during puberty is also exquisitely sensitive to the actions of endogenous estrogens. Analysis of mammary ductal growth and branching in 43 strains of inbred mice identified 20 QTL. Regions in the human genome orthologous to the mammary development QTL harbor loci associated with breast cancer risk or mammographic density. The data demonstrate extensive genetic variation in regulation of estrogen signaling in rodent mammary tissues that alters susceptibility to tumors. Genetic variants in these pathways may identify a subset of women who are especially sensitive to either endogenous estrogens or environmental xenoestrogens and render them at increased risk of breast cancer.


Asunto(s)
Neoplasias de la Mama/genética , Estrógenos/genética , Neoplasias Mamarias Animales/genética , Sitios de Carácter Cuantitativo/genética , Animales , Neoplasias de la Mama/patología , Estradiol/genética , Estradiol/metabolismo , Femenino , Predisposición Genética a la Enfermedad , Variación Genética , Humanos , Glándulas Mamarias Humanas/metabolismo , Neoplasias Mamarias Animales/patología , Ratones , Factores de Riesgo
3.
Am J Physiol Endocrinol Metab ; 312(3): E136-E149, 2017 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-27894063

RESUMEN

Mammalian glutaredoxin 3 (Grx3) has been shown to be important for regulating cellular redox homeostasis in the cell. Our previous studies indicate that Grx3 is significantly overexpressed in various human cancers including breast cancer and demonstrate that Grx3 controls cancer cell growth and invasion by regulating reactive oxygen species (ROS) and NF-κB signaling pathways. However, it remains to be determined whether Grx3 is required for normal mammary gland development and how it contributes to epithelial cell proliferation and differentiation in vivo. In the present study, we examined Grx3 expression in different cell types within the developing mouse mammary gland (MG) and found enhanced expression of Grx3 at pregnancy and lactation stages. To assess the physiological role of Grx3 in MG, we generated the mutant mice in which Grx3 was deleted specifically in mammary epithelial cells (MECs). Although the reduction of Grx3 expression had only minimal effects on mammary ductal development in virgin mice, it did reduce alveolar density during pregnancy and lactation. The impairment of lobuloalveolar development was associated with high levels of ROS accumulation and reduced expression of milk protein genes. In addition, proliferative gene expression was significantly suppressed with proliferation defects occurring in knockout MECs during alveolar development compared with wild-type controls. Therefore, our findings suggest that Grx3 is a key regulator of ROS in vivo and is involved in pregnancy-dependent mammary gland development and secretory activation through modulating cellular ROS.


Asunto(s)
Células Epiteliales/metabolismo , Glutarredoxinas/genética , Lactancia/genética , Glándulas Mamarias Animales/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Western Blotting , Proliferación Celular/genética , Ciclina D1/genética , Femenino , Regulación del Desarrollo de la Expresión Génica , Inmunohistoquímica , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Ratones Noqueados , Proteínas de la Leche/genética , FN-kappa B/metabolismo , Embarazo , Preñez , Ligando RANK/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Receptor Activador del Factor Nuclear kappa-B/metabolismo , Receptores de Progesterona/metabolismo , Transducción de Señal
4.
J Lipid Res ; 57(9): 1696-711, 2016 09.
Artículo en Inglés | MEDLINE | ID: mdl-27474222

RESUMEN

We aimed to characterize the lipidomic, metabolomic, and transcriptomic profiles in preterm piglets administered enteral (ENT) formula or three parenteral lipid emulsions [parenteral nutrition (PN)], Intralipid (IL), Omegaven (OV), or SMOFlipid (SL), for 14 days. Piglets in all parenteral lipid groups showed differential organ growth versus ENT piglets; whole body growth rate was lowest in IL piglets, yet there were no differences in either energy expenditure or (13)C-palmitate oxidation. Plasma homeostatic model assessment of insulin resistance demonstrated insulin resistance in IL, but not OV or SL, compared with ENT. The fatty acid and acyl-CoA content of the liver, muscle, brain, and plasma fatty acids reflected the composition of the dietary lipids administered. Free carnitine and acylcarnitine (ACT) levels were markedly reduced in the PN groups compared with ENT piglets. Genes associated with oxidative stress and inflammation were increased, whereas those associated with alternative pathways of fatty acid oxidation were decreased in all PN groups. Our results show that new generation lipid emulsions directly enrich tissue fatty acids, especially in the brain, and lead to improved growth and insulin sensitivity compared with a soybean lipid emulsion. In all total PN groups, carnitine levels are limiting to the formation of ACTs and gene expression reflects the stress of excess lipid on liver function.


Asunto(s)
Aceites de Pescado/administración & dosificación , Metabolismo de los Lípidos/genética , Fosfolípidos/administración & dosificación , Aceite de Soja/administración & dosificación , Triglicéridos/metabolismo , Animales , Animales Recién Nacidos/crecimiento & desarrollo , Animales Recién Nacidos/metabolismo , Grasas Insaturadas en la Dieta/administración & dosificación , Emulsiones/administración & dosificación , Ácidos Grasos/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ácido Palmítico/administración & dosificación , Ácido Palmítico/metabolismo , Nutrición Parenteral , Porcinos/crecimiento & desarrollo , Porcinos/metabolismo
5.
Mamm Genome ; 26(1-2): 57-79, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25552398

RESUMEN

Genetic background plays a dominant role in mammary gland development and breast cancer (BrCa). Despite this, the role of genetics is only partially understood. This study used strain-dependent variation in an inbred mouse mapping panel, to identify quantitative trait loci (QTL) underlying structural variation in mammary ductal development, and determined if these QTL correlated with genomic intervals conferring BrCa susceptibility in humans. For about half of the traits, developmental variation among the complete set of strains in this study was greater (P < 0.05) than that of previously studied strains, or strains in current common use for mammary gland biology. Correlations were also detected with previously reported variation in mammary tumor latency and metastasis. In-silico genome-wide association identified 20 mammary development QTL (Mdq). Of these, five were syntenic with previously reported human BrCa loci. The most significant (P = 1 × 10(-11)) association of the study was on MMU6 and contained the genes Plxna4, Plxna4os1, and Chchd3. On MMU5, a QTL was detected (P = 8 × 10(-7)) that was syntenic to a human BrCa locus on h12q24.5 containing the genes Tbx3 and Tbx5. Intersection of linked SNP (r(2) > 0.8) with genomic and epigenomic features, and intersection of candidate genes with gene expression and survival data from human BrCa highlighted several for further study. These results support the conclusion that mammary tumorigenesis and normal ductal development are influenced by common genetic factors and that further studies of genetically diverse mice can improve our understanding of BrCa in humans.


Asunto(s)
Neoplasias de la Mama/genética , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones Endogámicos/genética , Sitios de Carácter Cuantitativo/genética , Animales , Neoplasias de la Mama/fisiopatología , Mapeo Cromosómico , Simulación por Computador , Femenino , Estudio de Asociación del Genoma Completo , Técnicas Histológicas , Humanos , Ratones , Polimorfismo de Nucleótido Simple/genética , Especificidad de la Especie , Sintenía/genética , Tomografía Óptica
6.
J Mammary Gland Biol Neoplasia ; 17(2): 167-88, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22752723

RESUMEN

This paper resulted from a conference entitled "Lactation and Milk: Defining and refining the critical questions" held at the University of Colorado School of Medicine from January 18-20, 2012. The mission of the conference was to identify unresolved questions and set future goals for research into human milk composition, mammary development and lactation. We first outline the unanswered questions regarding the composition of human milk (Section I) and the mechanisms by which milk components affect neonatal development, growth and health and recommend models for future research. Emerging questions about how milk components affect cognitive development and behavioral phenotype of the offspring are presented in Section II. In Section III we outline the important unanswered questions about regulation of mammary gland development, the heritability of defects, the effects of maternal nutrition, disease, metabolic status, and therapeutic drugs upon the subsequent lactation. Questions surrounding breastfeeding practice are also highlighted. In Section IV we describe the specific nutritional challenges faced by three different populations, namely preterm infants, infants born to obese mothers who may or may not have gestational diabetes, and infants born to undernourished mothers. The recognition that multidisciplinary training is critical to advancing the field led us to formulate specific training recommendations in Section V. Our recommendations for research emphasis are summarized in Section VI. In sum, we present a roadmap for multidisciplinary research into all aspects of human lactation, milk and its role in infant nutrition for the next decade and beyond.


Asunto(s)
Lactancia Materna , Desarrollo Infantil , Lactancia , Glándulas Mamarias Humanas/crecimiento & desarrollo , Glándulas Mamarias Humanas/metabolismo , Leche Humana/metabolismo , Morfogénesis , Adulto , Animales , Animales Recién Nacidos , Investigación Biomédica/tendencias , Susceptibilidad a Enfermedades , Femenino , Humanos , Lactante , Recién Nacido , Intestinos/crecimiento & desarrollo , Intestinos/microbiología , Glándulas Mamarias Animales , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/prevención & control , Leche/metabolismo
7.
Am J Physiol Endocrinol Metab ; 303(3): E365-76, 2012 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-22649065

RESUMEN

Lactose synthesis is believed to be rate limiting for milk production. However, understanding the molecular events controlling lactose synthesis in humans is still rudimentary. We have utilized our established model of the RNA isolated from breast milk fat globule from seven healthy, exclusively breastfeeding women from 6 h to 42 days following delivery to determine the temporal coordination of changes in gene expression in the carbohydrate metabolic processes emphasizing the lactose synthesis pathway in human mammary epithelial cell. We showed that milk lactose concentrations increased from 75 to 200 mM from 6 to 96 h. Milk progesterone concentrations fell by 65% at 24 h and were undetectable by day 3. Milk prolactin peaked at 36 h and then declined progressively afterward. In concordance with lactose synthesis, gene expression of galactose kinase 2, UDP-glucose pyrophosphorylase 2 (UGP2), and phosphoglucomutase 1 increased 18-, 10-, and threefold, respectively, between 6 and 72 h. Between 6 and 96 h, gene expression of UDP-galactose transporter 2 (SLC35A2) increased threefold, whereas glucose transporter 1 was unchanged. Gene expression of lactose synthase no. 3 increased 1.7-fold by 96 h, whereas α-lactalbumin did not change over the entire study duration. Gene expression of prolactin receptor (PRLR) and its downstream signal transducer and activator of transcription complex 5 (STAT5) were increased 10- and 2.5-fold, respectively, by 72 h. In summary, lactose synthesis paralleled the induction of gene expression of proteins involved in UDP-galactose synthesis and transport, suggesting that they are potentially rate limiting in lactose synthesis and thus milk production. Progesterone withdrawal may be the signal that triggers PRLR signaling via STAT5, which may in turn induce UGP2 and SLC35A2 expression.


Asunto(s)
Lactancia/genética , Redes y Vías Metabólicas/genética , Leche Humana/metabolismo , Uridina Difosfato Galactosa/biosíntesis , Uridina Difosfato Galactosa/metabolismo , Adolescente , Adulto , Transporte Biológico/genética , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica/fisiología , Hormonas/sangre , Humanos , Lactancia/sangre , Lactancia/metabolismo , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/fisiología , Metabolismo/genética , Análisis por Micromatrices , Modelos Biológicos , Embarazo , Adulto Joven
8.
Biology (Basel) ; 11(7)2022 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-36101404

RESUMEN

Breastfeeding offers a broad spectrum of health benefits for infants. However, overnutrition and a steady increase in maternal obesity in the U.S. have made it harder for many mothers to produce and express breastmilk, and the quality of milk from obese mothers is also frequently compromised. Adipocytes, the primary cell type in the non-lactating breast, display a drastic morphological and functional change during lactation in mice. Lipid-filled adipocytes undergo lipolysis, and lipid droplets disappear to provide fatty acids and energy for breastmilk production. Once the animal stops lactation, these lipid-depleted adipocytes return as lipid-laden cells. This dynamic remodeling of the tissue is likely the result of active intercellular communications. Connexin43 (Cx43) is the most abundant connexin in the mammary adipose tissue that makes up the gap junctions for direct intercellular communications. Its expression is increased during lactation and reduced in obese mammary adipose tissue, which is resistant to lactation-induced remodeling. However, whether Cx43 is required for adipocyte remodeling and breastmilk production to support neonates' growth has not been established. In this study, we used doxycycline-inducible adipocyte-specific Cx43-deleted mice and demonstrated that adipocyte Cx43 played a vital role in determining the carbohydrate levels in breastmilk, which may subsequently affect neonates' growth.

9.
Physiol Genomics ; 43(8): 381-91, 2011 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-21205870

RESUMEN

Growth hormone is one of few pharmacologic agents known to augment milk production in humans. We hypothesized that recombinant human GH (rhGH) increases the expression of cell proliferation and milk protein synthesis genes. Sequential milk and blood samples collected over four days were obtained from five normal lactating women. Following 24 h of baseline milk and blood sampling, rhGH (0.1 mg/kg/day) was administered subcutaneously once daily for 3 days. Gene expression changes were determined by microarray studies utilizing milk fat globule RNA isolated from each milk sample. Following rhGH administration, DNA synthesis and cell cycle genes were induced, while no significant changes were observed in the expression of milk synthesis genes. Expression of glycolysis and citric acid cycle genes were increased by day 4 compared with day 1, while lipid synthesis genes displayed a circadian-like pattern. Cell cycle gene upregulation occurred after a lag of ∼2 days, likely explaining the failure to increase milk production after only 3 days of rhGH treatment. We conclude that rhGH induces expression of cellular proliferation and metabolism genes but does not induce milk protein gene expression, as potential mechanisms for increasing milk production and could account for the known effect of rhGH to increase milk production following 7-10 days.


Asunto(s)
Glucolípidos/análisis , Glicoproteínas/análisis , Hormona de Crecimiento Humana/administración & dosificación , Lactancia/efectos de los fármacos , Lactancia/genética , Proteínas de la Leche/efectos de los fármacos , Proteínas de la Leche/genética , Adulto , Proteínas de Ciclo Celular/sangre , Proteínas de Ciclo Celular/efectos de los fármacos , Proteínas de Ciclo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Femenino , Expresión Génica/efectos de los fármacos , Humanos , Factor I del Crecimiento Similar a la Insulina/efectos de los fármacos , Factor I del Crecimiento Similar a la Insulina/metabolismo , Gotas Lipídicas , Análisis por Micromatrices/métodos , Proteínas Recombinantes/administración & dosificación
10.
Physiol Genomics ; 43(6): 271-85, 2011 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-21189371

RESUMEN

The regulation of mitochondrial biogenesis and function in the lactating mammary cell is poorly understood. The goal of this study was to use proteomics to relate temporal changes in mammary cell mitochondrial function during lactation to changes in the proteins that make up this organelle. The hypothesis tested was that changes in mammary cell mitochondrial biogenesis and function during lactation would be accounted for by coordinated changes in the proteins of the electron transport chain and that some of these proteins might be linked by their expression patterns to PPARGC1α and AMP kinase. The mitochondrial proteome was studied along with markers of mitochondrial biogenesis and function in mammary tissue collected from mice over the course of a single prolonged lactation cycle. Mammary tissue concentrations of AMP and ADP were increased (P < 0.05) during early lactation and then declined with prolonged lactation. Similar changes were also observed for mitochondrial ATP synthesis activity, mitochondrial mass and DNA copy number. Analysis of the mammary cell mitochondrial proteome identified 244 unique proteins. Of these, only two proteins of the electron transport chain were found to increase during early lactation. In contrast, coordinated changes in numerous electron transport chain proteins were observed both during mid- and late lactation. There were six proteins that could be directly linked to PPARGC1α through network analysis. Abundance of PPARGC-1α and phosphorylation of AMP kinase was highest on day 2 postpartum. The results suggest that the increases in mammary mitochondria ATP synthesis activity during early lactation results from changes in only a limited number proteins. In addition, decreases in a handful of proteins linked to lipid oxidation could be temporally linked to decreases in PPARGC1α and phospho-AMP kinase suggesting potential roles for these proteins in coordinating mammary gland metabolism during early lactation.


Asunto(s)
Lactancia/fisiología , Glándulas Mamarias Animales/metabolismo , Mitocondrias/metabolismo , Adenosina Difosfato/metabolismo , Adenosina Monofosfato/metabolismo , Animales , Variaciones en el Número de Copia de ADN , Femenino , Regulación del Desarrollo de la Expresión Génica , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Fosforilación Oxidativa , Proteómica
11.
J Nutr ; 140(12): 2193-200, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20980637

RESUMEN

Prematurity and overfeeding in infants are associated with insulin resistance in childhood and may increase the risk of adult disease. Total parenteral nutrition (TPN) is a major source of infant nutritional support and may influence neonatal metabolic function. Our aim was to test the hypothesis that TPN induces increased adiposity and insulin resistance compared with enteral nutrition (EN) in neonatal pigs. Neonatal pigs were either fed enteral formula orally or i.v. administered a TPN mixture for 17 d; macronutrient intake was similar in both groups. During the 17-d period, we measured body composition by dual-energy X-ray absorptiometry scanning; fasting i.v. glucose tolerance tests (IVGTT) and hyperinsulinemic-euglycemic clamps (CLAMP) were performed to quantify insulin resistance. On d 17, tissue was collected after 1-h, low-dose CLAMP for tissue insulin signaling assays. TPN pigs gained less lean and more body fat and developed hepatic steatosis compared with EN pigs. After 7 and 13 d, IVGTT showed evidence of insulin resistance in the TPN compared with the EN group. Fasting plasma glucose and insulin also were higher in TPN pigs. CLAMP showed that insulin sensitivity was markedly lower in TPN pigs than in EN pigs. TPN also reduced the abundance of the insulin receptor, insulin receptor substrate 1, and phosphatidylinositol 3 kinase in skeletal muscle and liver and the proliferation of total pancreatic cells and ß-cells. Hepatic proinflammatory genes as well as c-Jun-N-terminal kinase 1 phosphorylation, plasma interleukin 6, and tumor necrosis factor-α were all higher in TPN pigs than in EN pigs. The results demonstrate that chronic TPN induces a hepatic inflammatory response that is associated with significant insulin resistance, hepatic steatosis, and fat deposition compared with EN in neonatal pigs. Further studies are warranted to establish the mechanism of TPN-induced insulin resistance and hepatic metabolic dysfunction and whether there are persistent metabolic consequences of this lifesaving form of infant nutritional support.


Asunto(s)
Animales Recién Nacidos , Hígado Graso/etiología , Hepatitis/etiología , Resistencia a la Insulina , Nutrición Parenteral , Animales , Porcinos
12.
Animals (Basel) ; 10(6)2020 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-32481546

RESUMEN

EGF acts as a ligand of the EGF receptor (EGFR) to activate the EGFR-mediated signaling pathways and is involved in the regulation of cell physiology. However, the roles of EGFR mediated signaling pathways in the regulation of lipid metabolism in goat mammary epithelial cells (GMECs) are poorly understood. To evaluate the impact of EGF on GMECs, the triglyceride (TG) content and lipid droplet were detected, using TG assay and immunofluorescence. Further, expression of lipogenic genes, the protein kinase B (Akt), phospholipase C-γ1 (PLC-γ1) and extracellular signal-regulated kinases (ERK)1/2 signaling pathways were measured by real-time polymerase chain reaction and Western blot, respectively. The results showed that the mRNA expression of EGFR gene was significantly upregulated in lactating goat mammary gland tissues compared to non-lactation period (p < 0.05). TG contents in EGF-treated GMECs were significantly increased (p < 0.05), and an increase of lipid droplets was also detected. In vitro studies demonstrated that the mRNA levels of lipogenesis-related FASN, ACC, SCD1, LXRa, LXRb and SP1 genes were positively correlated to the mRNA level of EGFR gene shown by gene overexpression and silencing (p < 0.05). The phosphorylations of Akt, ERK1/2 and PLC-γ1 in GMECs were greatly upregulated in the presence of EGF, and specific inhibitors were capable of blocking the phosphorylation of Akt, ERK1/2 and PLC-γ1. Compared with EGF-treated GMECs, the mRNA levels of FASN, ACC and SCD1 were significantly decreased in GMECs co-treated with PLC-γ1 and Akt inhibitor and EGF (p < 0.05), and TG content was also dropped significantly. These observations implied that EGFR plays an important role in regulating de novo fatty acid synthesis in GMECs, mainly mediated by Akt and PLC-γ1 signaling pathways.

13.
Physiol Genomics ; 37(1): 12-22, 2009 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-19018045

RESUMEN

The molecular physiology underlying human milk production is largely unknown because of limitations in obtaining tissue samples. Determining gene expression in normal lactating women would be a potential step toward understanding why some women struggle with or fail at breastfeeding their infants. Recently, we demonstrated the utility of RNA obtained from breast milk fat globule (MFG) to detect mammary epithelial cell (MEC)-specific gene expression. We used MFG RNA to determine the gene expression profile of human MEC during lactation. Microarray studies were performed using Human Ref-8 BeadChip arrays (Illumina). MFG RNA was collected every 3 h for 24 h from five healthy, exclusively breastfeeding women. We determined that 14,070 transcripts were expressed and represented the MFG transcriptome. According to GeneSpring GX 9, 156 ontology terms were enriched (corrected P < 0.05), which include cellular (n = 3,379 genes) and metabolic (n = 2,656) processes as the most significantly enriched biological process terms. The top networks and pathways were associated primarily with cellular activities most likely involved with milk synthesis. Multiple sampling over 24 h enabled us to demonstrate core circadian clock gene expression and the periodicity of 1,029 genes (7%) enriched for molecular functions involved in cell development, growth, proliferation, and cell morphology. In addition, we found that the MFG transcriptome was comparable to the metabolic gene expression profile described for the lactating mouse mammary gland. This paper is the first to describe the MFG transcriptome in sequential human samples over a 24 h period, providing valuable insights into gene expression in the human MEC.


Asunto(s)
Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Glucolípidos/genética , Glicoproteínas/genética , Lactancia/genética , Glándulas Mamarias Humanas/metabolismo , Adolescente , Adulto , Animales , Lactancia Materna , Análisis por Conglomerados , Femenino , Redes Reguladoras de Genes , Humanos , Gotas Lipídicas , Ratones , Prolactina/sangre , Programas Informáticos , Factores de Tiempo
14.
Mol Cell Biol ; 26(24): 9302-14, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17030631

RESUMEN

Insulin receptor substrates (IRSs) are signaling adaptors that play a major role in the metabolic and mitogenic actions of insulin and insulin-like growth factors. Reports have recently noted increased levels, or activity, of IRSs in many human cancers, and some have linked this to poor patient prognosis. We found that overexpressed IRS-1 was constitutively phosphorylated in vitro and in vivo and that transgenic mice overexpressing IRS-1 or IRS-2 in the mammary gland showed progressive mammary hyperplasia, tumorigenesis, and metastasis. Tumors showed extensive squamous differentiation, a phenotype commonly seen with activation of the canonical beta-catenin signaling pathway. Consistent with this, IRSs were found to bind beta-catenin in vitro and in vivo. IRS-induced tumorigenesis is unique, given that the IRSs are signaling adaptors with no intrinsic kinase activity, and this supports a growing literature indicating a role for IRSs in cancer. This study defines IRSs as oncogene proteins in vivo and provides new models to develop inhibitors against IRSs for anticancer therapy.


Asunto(s)
Péptidos y Proteínas de Señalización Intracelular/genética , Neoplasias Pulmonares/secundario , Neoplasias Mamarias Experimentales/genética , Fosfoproteínas/biosíntesis , Fosfoproteínas/genética , Proteínas Adaptadoras Transductoras de Señales/biosíntesis , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Línea Celular Transformada , Femenino , Humanos , Hiperplasia , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular/fisiología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Transgénicos , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Fosfoproteínas/fisiología , Transducción de Señal/fisiología
15.
J Endocrinol ; 194(2): 327-36, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17641282

RESUMEN

Expression of insulin receptor substrates (IRS)-1 and -2 within the mammary gland was found to be high at mid-lactation and dramatically decreased with mammary involution. This observation supports the hypothesis that these proteins are induced in the mammary gland with lactogenesis and involved in normal milk synthesis. To test this hypothesis, lactation capacity, along with indices of mammary secretory cell glucose metabolism and cell signaling were compared in normal mice and mice carrying targeted mutations in either the Irs1 or Irs2 genes. Mammary IRS-1 and IRS-2 protein levels were increased within 1 day of parturition and reached maximal levels by 5 days post partum. Dams carrying germline mutations of Irs1 or Irs2 displayed reduced lactation capacity as assessed by weight gain of pup litters. The reduction was more dramatic in Irs1(-/-) versus Irs2(-/-) dams. Maternal body weight was also reduced in Irs1(-/-) dams as well as in Irs1(+/-) Irs2(+/-) dams. The loss of IRS-1 had little impact on mammary gland expression of milk protein mRNAs, glucose transport, or on the abundance and subcellular localization of hexokinases I and II. The loss of IRS-1 was associated with a compensatory increase in insulin-induced IRS-2 phosphorylation; however, the loss of IRS-1 did also cause a reduction in insulin-dependent mammary gland-specific activation of Akt phosphorylation. These results support the conclusion that IRS-1 is important for insulin-dependent activation of Akt signaling within the lactating mammary gland, but that loss of this protein has only modest impact on normal milk synthesis, since related signaling proteins such as IRS-2 may act in compensatory fashion.


Asunto(s)
Lactancia/fisiología , Glándulas Mamarias Animales/metabolismo , Leche/química , Fosfoproteínas/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Pérdida de Peso , Animales , Transporte Biológico , Northern Blotting , Western Blotting , Femenino , Glucosa/metabolismo , Insulina/metabolismo , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Ratones , Ratones Noqueados , Fosfoproteínas/metabolismo , Fosforilación , Embarazo
16.
Cancer Res ; 65(9): 3781-7, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15867374

RESUMEN

The insulin-like growth factor I receptor (IGF-IR) is a transmembrane tyrosine kinase that is essential to growth and development and also thought to provide a survival signal for the maintenance of the transformed phenotype. There has been increasing interest in further understanding the role of IGF-I signaling in cancer and in developing receptor antagonists for therapeutic application. We describe herein a novel animal model that involves transgenic expression of a fusion receptor that is constitutively activated by homodimerization. Transgenic mice that expressed the activated receptor showed aberrant development of the mammary glands and developed salivary and mammary adenocarcinomas as early as 8 weeks of age. Xenograft tumors and a cell line were derived from the transgenic animals and are sensitive to inhibition by a novel small-molecule inhibitor of the IGF-IR kinase. This new model should provide new opportunities for further understanding how aberrant IGF-IR signaling leads to tumorigenesis and for optimizing novel antagonists of the receptor kinase.


Asunto(s)
Adenocarcinoma/genética , Transformación Celular Neoplásica/genética , Neoplasias Mamarias Experimentales/genética , Receptor IGF Tipo 1/biosíntesis , Neoplasias de las Glándulas Salivales/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Animales , Antígenos CD8/biosíntesis , Antígenos CD8/genética , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Femenino , Humanos , Neoplasias Mamarias Experimentales/enzimología , Neoplasias Mamarias Experimentales/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos ICR , Ratones Desnudos , Ratones Transgénicos , Trasplante de Neoplasias , Embarazo , Receptor IGF Tipo 1/antagonistas & inhibidores , Receptor IGF Tipo 1/genética , Proteínas Recombinantes de Fusión/biosíntesis , Proteínas Recombinantes de Fusión/genética , Neoplasias de las Glándulas Salivales/enzimología , Neoplasias de las Glándulas Salivales/patología , Transfección , Transgenes/genética , Trasplante Heterólogo
17.
Exp Gerontol ; 41(3): 271-81, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16442254

RESUMEN

Milk synthesis by the mammary gland declines during prolonged lactation despite the continued suckling stimulus and complete removal of mammary secretions. Although this process has been hypothesized to result from cellular aging there has been no reported analysis of aging markers in the lactating mammary gland. The goal of these studies was to relate lactation performance in the mouse during a single prolonged lactation cycle to changes in mammary development and mitochondrial oxidative damage. During an artificially prolonged lactation cycle, the capacity of the dams to support litter growth decreased over time. This decrease was associated with decreased mammary epithelial content. Cell proliferation, along with the percentage of mammary progenitor cells, was high during early lactation, but low during prolonged lactation. Apoptosis increased during prolonged lactation. Oxidative damage to mitochondrial DNA increased during the early postpartum period and remained elevated through the end of the cycle. In contrast oxidative damage to mitochondrial protein was high during early lactation and decreased through mid lactation to increase again with prolonged lactation. The results suggest that a single prolonged lactation cycle may replicate on an accelerated basis some of the changes that occur with a lifetime of aging in organs possessing more stable cell populations.


Asunto(s)
Senescencia Celular/fisiología , Lactancia/fisiología , Glándulas Mamarias Animales/fisiología , Mitocondrias/fisiología , Estrés Oxidativo/fisiología , Animales , Apoptosis/fisiología , División Celular/fisiología , ADN Mitocondrial/fisiología , Células Epiteliales/fisiología , Femenino , Peroxidación de Lípido/fisiología , Glándulas Mamarias Animales/crecimiento & desarrollo , Ratones , Ratones Endogámicos , Proteínas Mitocondriales/metabolismo , Oxidación-Reducción , Periodo Posparto , Células Madre/fisiología
18.
Cancer Res ; 63(17): 5203-8, 2003 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-14500345

RESUMEN

E-cadherin is an important mediator of cell-cell interactions, and has been shown to play a crucial role in breast tumor suppression. Its inactivation occurs through instability at its chromosomal locus and mutations, but also through epigenetic mechanisms such as promoter hypermethylation and transcriptional silencing. We show here that the potent mitogen estrogen causes down-regulation of E-cadherin levels in both normal and tumorigenic breast epithelial cells, and that this down-regulation is reversed by antiestrogens. The reduction in E-cadherin levels is via a decrease in promoter activity and subsequent mRNA levels. Chromatin immunoprecipitation assays revealed that estrogen receptor and corepressors were bound to the E-cadherin promoter, and that overexpression of corepressors such as scaffold attachment factor B resulted in enhanced repression of E-cadherin. We propose that estrogen-mediated down-regulation of E-cadherin is a novel way of reducing E-cadherin levels in estrogen receptor-positive breast cancer.


Asunto(s)
Neoplasias de la Mama/metabolismo , Cadherinas/biosíntesis , Estradiol/farmacología , Regulación Neoplásica de la Expresión Génica/fisiología , Tamoxifeno/análogos & derivados , Neoplasias de la Mama/genética , Cadherinas/genética , Línea Celular Transformada , Medio de Cultivo Libre de Suero , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/fisiología , Moduladores de los Receptores de Estrógeno/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Proteínas Nucleares , Co-Represor 1 de Receptor Nuclear , Regiones Promotoras Genéticas , ARN Mensajero/biosíntesis , ARN Mensajero/genética , Receptor Cross-Talk/fisiología , Receptores de Estrógenos/biosíntesis , Receptores de Estrógenos/fisiología , Proteínas Represoras/biosíntesis , Proteínas Represoras/genética , Tamoxifeno/farmacología , Células Tumorales Cultivadas
19.
Clin Cancer Res ; 9(1 Pt 2): 516S-23S, 2003 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-12538509

RESUMEN

The explosion of signal transduction research over the last 10 years has provided a unique insight into the complexity of these intricate pathways. Whereas intermediates of multiple signaling pathways have been identified, understanding their function and, in particular, the interactions between them has become a daunting task. The increasing evidence that many of these pathways can cross-talk with each other via signal transactivation inevitably raises the question of how cells determine specificity in signaling. Despite the mind-numbing complexity of these pathways, progress has been made in developing highly specific and potent signal transduction inhibitors (STIs). STIs show promise in the treatment of cancer in preclinical studies and are currently in a number of clinical trials. Whereas many of these agents were "rationally designed," we barely understand their mechanisms of action. This review will highlight how recent studies using these STIs have elucidated novel mechanisms of STI action that may be used in the development of new therapeutic strategies for the treatment of cancer.


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Receptor Cross-Talk/fisiología , Proteínas Tirosina Quinasas Receptoras/metabolismo , Transducción de Señal/efectos de los fármacos , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/enzimología , Neoplasias de la Mama/genética , Regulación hacia Abajo , Inhibidores Enzimáticos/uso terapéutico , Femenino , Humanos
20.
Mol Endocrinol ; 17(6): 1054-65, 2003 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12637587

RESUMEN

Previous studies from our laboratory have demonstrated that p190-B RhoGAP (p190-B) is differentially expressed in the Cap cells of terminal end buds (TEBs) and poorly differentiated rodent mammary tumors. Based on these observations we hypothesized that p190-B might play an essential role in invasion of the TEBs into the surrounding fat pad during ductal morphogenesis. To test this hypothesis, mammary development was studied in p190-B-deficient mice. A haploinsufficiency phenotype was observed in p190-B heterozygous mice as indicated by decreased number and rate of ductal outgrowth(s) at 3, 4, and 5 wk of age when compared with their wild-type littermates. This appeared to result from decreased proliferation in the Cap cells of the TEBs, a phenotype remarkably similar to that observed previously in IGF-I receptor null mammary epithelium. Furthermore, decreased expression of insulin receptor substrates 1 and 2 were observed in TEBs of p190-B heterozygous mice. These findings are consistent with decreased IGF signaling observed previously in p190-B-/- mouse embryo fibroblasts. To further assess if this defect was cell autonomous or due to systemic endocrine effects, the mammary anlagen from p190-B+/+, p190-B+/-, and p190-B-/- mice was rescued by transplantation into the cleared fat pad of recipient Rag1-/- mice. Surprisingly, as opposed to 75-80% outgrowths observed using wild-type donor epithelium, only 40% of the heterozygous and none of the p190-B-/- epithelial transplants displayed any outgrowths. Together, these results suggest that p190-B regulates ductal morphogenesis, at least in part, by modulating the IGF signaling axis.


Asunto(s)
Factores de Intercambio de Guanina Nucleótido/fisiología , Glándulas Mamarias Animales/crecimiento & desarrollo , Proteínas Nucleares/fisiología , Fosfoproteínas/metabolismo , Maduración Sexual/fisiología , Somatomedinas/metabolismo , Análisis de Varianza , Animales , División Celular/fisiología , Proteínas de Unión al ADN , Femenino , Proteínas Activadoras de GTPasa , Factores de Intercambio de Guanina Nucleótido/genética , Heterocigoto , Proteínas Sustrato del Receptor de Insulina , Péptidos y Proteínas de Señalización Intracelular , Glándulas Mamarias Animales/citología , Glándulas Mamarias Animales/metabolismo , Ratones , Morfogénesis , Proteínas Nucleares/genética , Proteínas Represoras , Maduración Sexual/genética , Transducción de Señal
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA