Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
1.
Biology (Basel) ; 11(7)2022 Jul 07.
Article in English | MEDLINE | ID: mdl-36101404

ABSTRACT

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.

2.
Animals (Basel) ; 10(6)2020 May 28.
Article in English | MEDLINE | ID: mdl-32481546

ABSTRACT

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.

3.
Mamm Genome ; 29(9-10): 632-655, 2018 10.
Article in English | MEDLINE | ID: mdl-30073618

ABSTRACT

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.


Subject(s)
Cation Transport Proteins/genetics , Chromosome Mapping , Iron/metabolism , Lactation/genetics , Milk/chemistry , Quantitative Trait Loci/genetics , Sodium-Hydrogen Exchangers/genetics , Animals , Binding Sites/genetics , Computer Simulation , Female , Gene Expression , Genome-Wide Association Study , Iron/analysis , Linkage Disequilibrium , Mice , Milk/metabolism , Minerals/analysis , Minerals/metabolism , Polymorphism, Single Nucleotide , Transcription Factors/metabolism
4.
Mamm Genome ; 29(1-2): 24-37, 2018 02.
Article in English | MEDLINE | ID: mdl-29487996

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Estrogens/genetics , Mammary Neoplasms, Animal/genetics , Quantitative Trait Loci/genetics , Animals , Breast Neoplasms/pathology , Estradiol/genetics , Estradiol/metabolism , Female , Genetic Predisposition to Disease , Genetic Variation , Humans , Mammary Glands, Human/metabolism , Mammary Neoplasms, Animal/pathology , Mice , Risk Factors
6.
Am J Physiol Endocrinol Metab ; 312(3): E136-E149, 2017 03 01.
Article in English | MEDLINE | ID: mdl-27894063

ABSTRACT

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.


Subject(s)
Epithelial Cells/metabolism , Glutaredoxins/genetics , Lactation/genetics , Mammary Glands, Animal/metabolism , Reactive Oxygen Species/metabolism , Animals , Blotting, Western , Cell Proliferation/genetics , Cyclin D1/genetics , Female , Gene Expression Regulation, Developmental , Immunohistochemistry , Mammary Glands, Animal/growth & development , Mice , Mice, Knockout , Milk Proteins/genetics , NF-kappa B/metabolism , Pregnancy , Pregnancy, Animal , RANK Ligand/metabolism , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Progesterone/metabolism , Signal Transduction
7.
J Lipid Res ; 57(9): 1696-711, 2016 09.
Article in English | MEDLINE | ID: mdl-27474222

ABSTRACT

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.


Subject(s)
Fish Oils/administration & dosage , Lipid Metabolism/genetics , Phospholipids/administration & dosage , Soybean Oil/administration & dosage , Triglycerides/metabolism , Animals , Animals, Newborn/growth & development , Animals, Newborn/metabolism , Dietary Fats, Unsaturated/administration & dosage , Emulsions/administration & dosage , Fatty Acids/metabolism , Liver/drug effects , Liver/metabolism , Male , Palmitic Acid/administration & dosage , Palmitic Acid/metabolism , Parenteral Nutrition , Swine/growth & development , Swine/metabolism
8.
Mamm Genome ; 26(1-2): 57-79, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25552398

ABSTRACT

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.


Subject(s)
Breast Neoplasms/genetics , Mammary Glands, Animal/growth & development , Mice, Inbred Strains/genetics , Quantitative Trait Loci/genetics , Animals , Breast Neoplasms/physiopathology , Chromosome Mapping , Computer Simulation , Female , Genome-Wide Association Study , Histological Techniques , Humans , Mice , Polymorphism, Single Nucleotide/genetics , Species Specificity , Synteny/genetics , Tomography, Optical
9.
J Mammary Gland Biol Neoplasia ; 17(2): 167-88, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22752723

ABSTRACT

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.


Subject(s)
Breast Feeding , Child Development , Lactation , Mammary Glands, Human/growth & development , Mammary Glands, Human/metabolism , Milk, Human/metabolism , Morphogenesis , Adult , Animals , Animals, Newborn , Biomedical Research/trends , Disease Susceptibility , Female , Humans , Infant , Infant, Newborn , Intestines/growth & development , Intestines/microbiology , Mammary Glands, Animal , Metabolic Diseases/etiology , Metabolic Diseases/prevention & control , Milk/metabolism
10.
Am J Physiol Endocrinol Metab ; 303(3): E365-76, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-22649065

ABSTRACT

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.


Subject(s)
Lactation/genetics , Metabolic Networks and Pathways/genetics , Milk, Human/metabolism , Uridine Diphosphate Galactose/biosynthesis , Uridine Diphosphate Galactose/metabolism , Adolescent , Adult , Biological Transport/genetics , Female , Gene Expression Profiling , Gene Expression Regulation/physiology , Hormones/blood , Humans , Lactation/blood , Lactation/metabolism , Mammary Glands, Human/metabolism , Mammary Glands, Human/physiology , Metabolism/genetics , Microarray Analysis , Models, Biological , Pregnancy , Young Adult
11.
Physiol Genomics ; 43(8): 381-91, 2011 Apr 27.
Article in English | MEDLINE | ID: mdl-21205870

ABSTRACT

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.


Subject(s)
Glycolipids/analysis , Glycoproteins/analysis , Human Growth Hormone/administration & dosage , Lactation/drug effects , Lactation/genetics , Milk Proteins/drug effects , Milk Proteins/genetics , Adult , Cell Cycle Proteins/blood , Cell Cycle Proteins/drug effects , Cell Cycle Proteins/metabolism , Cell Proliferation/drug effects , Female , Gene Expression/drug effects , Humans , Insulin-Like Growth Factor I/drug effects , Insulin-Like Growth Factor I/metabolism , Lipid Droplets , Microarray Analysis/methods , Recombinant Proteins/administration & dosage
12.
Physiol Genomics ; 43(6): 271-85, 2011 Mar 29.
Article in English | MEDLINE | ID: mdl-21189371

ABSTRACT

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.


Subject(s)
Lactation/physiology , Mammary Glands, Animal/metabolism , Mitochondria/metabolism , Adenosine Diphosphate/metabolism , Adenosine Monophosphate/metabolism , Animals , DNA Copy Number Variations , Female , Gene Expression Regulation, Developmental , Mammary Glands, Animal/growth & development , Mice , Oxidative Phosphorylation , Proteomics
13.
J Nutr ; 140(12): 2193-200, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20980637

ABSTRACT

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.


Subject(s)
Animals, Newborn , Fatty Liver/etiology , Hepatitis/etiology , Insulin Resistance , Parenteral Nutrition , Animals , Swine
14.
Physiol Genomics ; 37(1): 12-22, 2009 Mar 03.
Article in English | MEDLINE | ID: mdl-19018045

ABSTRACT

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.


Subject(s)
Gene Expression Profiling , Gene Expression Regulation , Glycolipids/genetics , Glycoproteins/genetics , Lactation/genetics , Mammary Glands, Human/metabolism , Adolescent , Adult , Animals , Breast Feeding , Cluster Analysis , Female , Gene Regulatory Networks , Humans , Lipid Droplets , Mice , Prolactin/blood , Software , Time Factors
15.
J Endocrinol ; 198(1): 61-70, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18577570

ABSTRACT

GH, prolactin (PRL), and IGF-I stimulate lactation-related metabolic processes in mammary epithelial cells. However, the ability of these factors to stimulate milk production in animals varies depending on species and experimental variables. Previous work in our laboratory demonstrated that transgenic overexpression of des(1-3)IGF-I within the mammary glands of lactating mouse dams increased lactation capacity during prolonged lactation. This work also suggested that some of the effects of the overexpressed IGF-I may have been mediated through elevated concentrations of IGF-I or PRL in the systemic circulation. In the present study, murine GH and PRL, and a human IGF-I analog, long-R3-IGF-I (LR3), were administered as s.c. injections to compare their ability to enhance milk production, and alter mammary gland signaling and gene expression. Lactation capacity, as measured by litter gain, was increased (P<0.05) by GH, but not by PRL. LR3 increased (P<0.05) mammary phospho-Akt and suppressors of cytokines signaling 3 (SOCS3) gene expression, and had a modest ability to increase (P<0.05) lactation capacity. GH both increased (P<0.05) mammary SOCS1 expression and decreased (P<0.05) mammary expression of tryptophan hydroxylase 1, the rate-limiting enzyme in the synthesis of serotonin and a potential feedback inhibitor of lactation. These results suggest that while both GH and IGF-I stimulate milk production in the lactating mouse, the effect of GH may be additionally mediated through IGF-I-independent effects associated with repression of mammary serotonin synthesis.


Subject(s)
Growth Hormone/pharmacology , Insulin-Like Growth Factor I/analogs & derivatives , Lactation/drug effects , Mammary Glands, Animal/drug effects , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Insulin-Like Growth Factor I/pharmacology , Mammary Glands, Animal/metabolism , Mice , Pregnancy , Prolactin/pharmacology , Signal Transduction , Suppressor of Cytokine Signaling Proteins/genetics
16.
J Endocrinol ; 195(3): 503-11, 2007 Dec.
Article in English | MEDLINE | ID: mdl-18000312

ABSTRACT

Little is known of the molecular regulation of human milk production because of limitations in obtaining mammary tissue from lactating women. Our objectives were to evaluate whether RNA isolated from breast milk fat globules (MFGs) could be an alternative to mammary biopsies and to determine whether intense breast pumping, which increases prolactin (PRL) secretion, will upregulate alpha-lactalbumin (alpha-LA, a major determinant of lactose synthesis) transcription. RNA was isolated from MFG and transcripts of interest were identified and quantitated by real-time RT-PCR using an external standard for normalization. In addition, we performed microarray studies to determine MFG RNA gene expression profile. Ten lactating women were studied using two protocols: protocol A with intense pumping from 0800 to 0814 h followed by short pumping and protocol B with intense pumping from 1200 to 1214 h preceded by short pumping. Plasma PRL and MFG alpha-LA mRNA expression were measured. During protocol A, plasma PRL (61+/-7-248+/-43 mug/l by 14 min) and alpha-LA (3.5+/-0.9 fold by 6 h; P<0.03) increased. During protocol B, PRL gradually increased over 4 h from 69+/-14 to 205+/-28 mug/l, and further to 329+/-23 mug/l by 12 min of intense pumping; alpha-LA mRNA expression did not increase significantly. We conclude that MFGs provide a unique source to study the in vivo regulation of gene expression in mammary epithelial cells. alpha-LA mRNA is abundant in the MFG and its expression may be regulated by hormonal and temporal factors.


Subject(s)
Breast Feeding , Breast/metabolism , Gene Expression , Suction/methods , Actins/metabolism , Adult , Animals , Epithelium/metabolism , Female , Glycolipids/genetics , Glycoproteins/genetics , Humans , Lactalbumin/genetics , Lipid Droplets , Mice , Microarray Analysis , Prolactin/blood , RNA/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Time Factors , Transcription, Genetic
17.
Dev Biol ; 309(1): 137-49, 2007 Sep 01.
Article in English | MEDLINE | ID: mdl-17662267

ABSTRACT

P190-B RhoGAP (p190-B, also known as ARHGAP5) has been shown to play an essential role in invasion of the terminal end buds (TEBs) into the surrounding fat pad during mammary gland ductal morphogenesis. Here we report that embryos with a homozygous p190-B gene deletion exhibit major defects in embryonic mammary bud development. Overall, p190-B-deficient buds were smaller in size, contained fewer cells, and displayed characteristics of impaired mesenchymal proliferation and differentiation. Consistent with the reported effects of p190-B deletion on IGF-1R signaling, IGF-1R-deficient embryos also displayed a similar small mammary bud phenotype. However, unlike the p190-B-deficient embryos, the IGF-1R-deficient embryos exhibited decreased epithelial proliferation and did not display mesenchymal defects. Because both IGF and p190-B signaling affect IRS-1/2, we examined IRS-1/2 double knockout embryonic mammary buds. These embryos displayed major defects similar to the p190-B-deficient embryos including smaller bud size. Importantly, like the p190-B-deficient buds, proliferation of the IRS-1/2-deficient mesenchyme was impaired. These results indicate that IGF signaling through p190-B and IRS proteins is critical for mammary bud formation and ensuing epithelial-mesenchymal interactions necessary to sustain mammary bud morphogenesis.


Subject(s)
GTPase-Activating Proteins/metabolism , Mammary Glands, Animal/embryology , Receptor, IGF Type 1/metabolism , Signal Transduction , Animals , Epithelium/embryology , Epithelium/physiology , Female , GTPase-Activating Proteins/genetics , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Male , Mammary Glands, Animal/metabolism , Mesoderm/physiology , Mice , Mice, Knockout , Phosphoproteins/genetics , Phosphoproteins/metabolism , Receptor, IGF Type 1/genetics
18.
J Endocrinol ; 194(2): 327-36, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17641282

ABSTRACT

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.


Subject(s)
Lactation/physiology , Mammary Glands, Animal/metabolism , Milk/chemistry , Phosphoproteins/genetics , Proto-Oncogene Proteins c-akt/metabolism , Weight Loss , Animals , Biological Transport , Blotting, Northern , Blotting, Western , Female , Glucose/metabolism , Insulin/metabolism , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Phosphoproteins/metabolism , Phosphorylation , Pregnancy
19.
Cell Cycle ; 6(6): 705-13, 2007 Mar 15.
Article in English | MEDLINE | ID: mdl-17374994

ABSTRACT

Insulin receptor substrates (IRSs) are adaptor proteins that link signaling from upstream activators to multiple downstream effectors to modulate normal growth, metabolism, survival, and differentiation. Recent cell culture studies have shown that IRSs can interact with, and are functionally required for, the transforming ability of many oncogenes. Consistent with this, IRSs are elevated and hyperactive in many human tumors. IRSs respond to many extracellular signals that are critical for mammary gland development, and we have shown that IRSs disrupt normal mammary acini formation in vitro, and cause mammary tumorigenesis and metastasis in vivo. In this review we will discuss the role of IRSs in both transformation and cancer progression.


Subject(s)
Cell Transformation, Neoplastic/metabolism , Cell Transformation, Neoplastic/pathology , Intracellular Signaling Peptides and Proteins/physiology , Phosphoproteins/physiology , Proto-Oncogene Proteins/physiology , Animals , Cell Transformation, Neoplastic/genetics , Disease Progression , Humans , Insulin Receptor Substrate Proteins
20.
J Mammary Gland Biol Neoplasia ; 12(1): 59-70, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17286209

ABSTRACT

In most species the functional activity of the mammary gland during lactation follows a biphasic developmental pattern. This pattern starts with a rapid increase in milk output that occurs with secretory activation and continues with a more gradual increase until the point of peak lactation is reached. Following this gain-of-function phase, the ability of the gland to produce milk decreases. This decrease occurs even if the lactation is prolonged by the presence of continued suckling stimulus and complete milk removal. This review describes the current state of our knowledge concerning the factors that regulate milk synthesis capacity by the mammary gland during the lactation cycle. The review describes four potential alternatives as mechanisms governing the process, which we refer to as secretory diminution. These alternatives are not presented as mutually exclusive of each other or other possible mechanisms, but are proposed as potential contributing mechanisms.


Subject(s)
Lactation/physiology , Mammary Glands, Animal/physiology , Mammary Glands, Human/physiology , Animals , Apoptosis/physiology , Female , Humans , Mice
SELECTION OF CITATIONS
SEARCH DETAIL
...