Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 96
Filter
1.
J Mol Endocrinol ; 50(1): 79-90, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23048206

ABSTRACT

Prolactin is essential for normal mammary gland development and differentiation, and has been shown to promote tumor cell proliferation and chemotherapeutic resistance. Soluble isoforms of the prolactin receptor (PrlR) have been reported to regulate prolactin bioavailability by functioning as 'prolactin-binding proteins'. Included in this category is Δ7/11, a product of alternate splicing of the PrlR primary transcript. However, the direct interactions of prolactin with Δ7/11, and the resulting effect on cell behavior, have not been investigated. Herein, we demonstrate the ability of Δ7/11 to bind prolactin using a novel proximity ligation assay and traditional immunoprecipitation techniques. Biochemical analyses demonstrated that Δ7/11 was heavily glycosylated, similar to the extracellular domain of the primary PrlR, and that glycosylation regulated the cellular localization and secretion of Δ7/11. Low levels of Δ7/11 were detected in serum samples of healthy volunteers, but were undetectable in human milk samples. Expression of Δ7/11 was also detected in six of the 62 primary breast tumor biopsies analyzed; however, no correlation was found with Δ7/11 expression and tumor histotype or other patient demographics. Functional analysis demonstrated the ability of Δ7/11 to inhibit prolactin-induced cell proliferation as well as alter prolactin-induced rescue of cell cycle arrest/early senescence events in breast epithelial cells. Collectively, these data demonstrate that Δ7/11 is a novel regulatory mechanism of prolactin bioavailability and signaling.


Subject(s)
Carrier Proteins/metabolism , Animals , Biopsy , Blotting, Western , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CHO Cells , Carrier Proteins/blood , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Cricetinae , Cricetulus , Female , Glycosylation , Humans , Immunoprecipitation , Milk, Human/metabolism , Polymerase Chain Reaction , Prolactin/metabolism , Protein Binding , Recombinant Proteins/blood , Recombinant Proteins/metabolism
2.
Horm Cancer ; 1(2): 71-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-21113329

ABSTRACT

Prolactin (PRL) is a peptide hormone necessary for normal growth and development of the human breast. In addition, high levels of PRL in plasma correlate with increased risk of breast cancer, especially among postmenopausal women. Several isoforms of PRL exist in human circulation, including a 16 kDa isoform that is an N-terminal fragment of the full-length 23 kDa PRL. 16 kDa PRL has been shown to be anti-angiogenic in vitro and in vivo, and to reduce formation of tumors from prostate, colon and melanoma cancer cell lines. Here we explore the effect of 16 kDa PRL expression in vitro and in vivo using two breast cancer cell line models (MCF-7 and MDA-MB-231) and also the HCT-116 colon cancer cell line. In all three cell lines, 16 kDa PRL expression inhibited cell proliferation in vitro compared to empty vector controls. In vivo results were markedly different between the two types of cell lines. HCT-116 cells expressing 16 kDa PRL exhibited reduced vascularization and tumor formation, consistent with published results. The breast cancer cell lines expressing 16 kDa PRL also exhibited inhibition of angiogenesis in vivo but no reduction in tumor size or formation. These results suggest that the effects of 16 kDa PRL on tumor formation may vary across tissue types. The unique sensitivity of breast cancer to PRL as a mitogen and/or additional factors in the mammary gland environment (e.g. local hormone/mitogen concentration) may play a dominant role in tumor formation in vivo, thus outweighing the anti-angiogenesis effects and in vitro reduction in cell proliferation induced by 16 kDa PRL.


Subject(s)
Breast Neoplasms/blood supply , Breast Neoplasms/pathology , Neovascularization, Pathologic , Prolactin/metabolism , Protein Isoforms/pharmacology , Angiogenesis Inhibitors/metabolism , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Female , HCT116 Cells , Humans , Mice , Mice, Nude , Neoplasm Transplantation
3.
Br J Cancer ; 103(7): 1097-102, 2010 Sep 28.
Article in English | MEDLINE | ID: mdl-20736944

ABSTRACT

BACKGROUND: Previous prospective studies have found an association between prolactin (PRL) levels and increased risk of breast cancer. Using data from a population-based breast cancer case-control study conducted in two cities in Poland (2000-2003), we examined the association of PRL levels with breast cancer risk factors among controls and with tumour characteristics among the cases. METHODS: We analysed PRL serum levels among 773 controls without breast cancer matched on age and residence to 776 invasive breast cancer cases with available pretreatment serum. Tumours were centrally reviewed and prepared as tissue microarrays for immunohistochemical analysis. Breast cancer risk factors, assessed by interview, were related to serum PRL levels among controls using analysis of variance. Mean serum PRL levels by tumour characteristics are reported. These associations also were evaluated using polytomous logistic regression. RESULTS: Prolactin levels were associated with nulliparity in premenopausal (P=0.05) but not in postmenopausal women. Associations in postmenopausal women included an inverse association with increasing body mass index (P=0.0008) and direct association with use of recent/current hormone therapy (P=0.0006). In case-only analyses, higher PRL levels were more strongly associated with lobular compared with ductal carcinoma among postmenopausal women (P=0.02). Levels were not different by tumour size, grade, node involvement or oestrogen receptor, progesterone receptor, or human epidermal growth factor receptor 2 status. CONCLUSIONS: Our analysis demonstrates that PRL levels are higher among premenopausal nulliparous as compared with parous women. Among postmenopausal women, levels were higher among hormone users and lower among obese women. These results may have value in understanding the mechanisms underlying several breast cancer risk factor associations.


Subject(s)
Breast Neoplasms/blood , Prolactin/blood , Adult , Case-Control Studies , Female , Humans , Middle Aged , Parity , Poland/epidemiology , Postmenopause , Pregnancy , Premenopause , Risk Factors
4.
J Cell Physiol ; 219(3): 659-66, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19170109

ABSTRACT

Epithelial-mesenchymal transition (EMT) is a process occurring during both embryogenesis and early stages of invasive cancer. Epithelial cells that undergo EMT become more migratory and invasive with a mesenchymal morphology. Herein we assess EMT induction in a mouse mammary epithelial cell line driven by Msx2, a homeobox-containing transcription factor important during mammary gland development. NMuMG cells, a normal mouse mammary epithelial cell line, stably transfected with a Msx2 cDNA showed downregulation of an epithelial marker E-cadherin and upregulation of the mesenchymal markers vimentin and N-cadherin. Furthermore, overexpression of Cripto-1, a member of the epidermal growth factor-CFC protein family already known to be involved in EMT, was detected in Msx2-transfected cells. The expression of Cripto-1 was accompanied by activation of the tyrosine kinase c-Src pathway and an increase in the invasive ability of the cells. Functional assays also demonstrated inhibition of the invasive behavior of the Msx2-transfected cells by a c-Src specific inhibitor. Moreover, immunohistochemistry of human infiltrating breast carcinomas showed positive staining for Msx2 only in the infiltrating tumor cells while the non-infiltrating tumor cells were negative. These results suggest that Msx2 may play a significant role in promoting EMT in epithelial cells that acquire properties involved in tumor invasion. J. Cell. Physiol. 219: 659-666, 2009. Published 2009 Wiley-Liss, Inc.


Subject(s)
Epidermal Growth Factor/metabolism , Homeodomain Proteins/metabolism , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CSK Tyrosine-Protein Kinase , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Ductal, Breast/pathology , Cell Line , DNA Primers/genetics , Epidermal Growth Factor/antagonists & inhibitors , Epidermal Growth Factor/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Homeodomain Proteins/genetics , Humans , Mammary Glands, Animal/growth & development , Membrane Glycoproteins/antagonists & inhibitors , Membrane Glycoproteins/genetics , Mesoderm/cytology , Mesoderm/metabolism , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasm Invasiveness/physiopathology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Protein-Tyrosine Kinases/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Signal Transduction , Transfection , Up-Regulation , src-Family Kinases
5.
Oncogene ; 28(2): 219-30, 2009 Jan 15.
Article in English | MEDLINE | ID: mdl-18836481

ABSTRACT

Transgenic mice expressing the Notch 4 intracellular domain (ICD) (Int3) in the mammary gland have two phenotypes: arrest of mammary alveolar/lobular development and mammary tumorigenesis. Notch4 signaling is mediated primarily through the interaction of Int3 with the transcription repressor/activator Rbpj. We have conditionally ablated the Rbpj gene in the mammary glands of mice expressing whey acidic protein (Wap)-Int3. Interestingly, Rbpj knockout mice (Wap-Cre(+)/Rbpj(-/-)/Wap-Int3) have normal mammary gland development, suggesting that the effect of endogenous Notch signaling on mammary gland development is complete by day 15 of pregnancy. RBP-J heterozygous (Wap-Cre(+)/Rbpj(-/+)/Wap-Int3) and Rbpj control (Rbpj(flox/flox)/Wap-Int3) mice are phenotypically the same as Wap-Int3 mice with respect to mammary gland development and tumorigenesis. In addition, the Wap-Cre(+)/Rbpj(-/-)/Wap-Int3-knockout mice also developed mammary tumors at a frequency similar to Rbpj heterozygous and Wap-Int3 control mice but with a slightly longer latency. Thus, the effect on mammary gland development is dependent on the interaction of the Notch ICD with the transcription repressor/activator Rbpj, and Notch-induced mammary tumor development is independent of this interaction.


Subject(s)
Mammary Glands, Animal/embryology , Mammary Neoplasms, Experimental/genetics , Neoplasm Proteins/physiology , Proto-Oncogene Proteins/physiology , Receptors, Notch/physiology , Adenocarcinoma, Papillary/genetics , Adenocarcinoma, Papillary/pathology , Agar , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Cycle Proteins/metabolism , Cell Transformation, Viral/genetics , Female , Homeodomain Proteins/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Knockout , Mice, Nude , Milk Proteins/genetics , Neoplasm Proteins/genetics , Pregnancy , Protein Structure, Tertiary , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Receptor, Notch4 , Receptors, Notch/chemistry , Receptors, Notch/deficiency , Receptors, Notch/genetics , Recombinant Fusion Proteins/physiology , Repressor Proteins/genetics , Terminal Repeat Sequences/genetics , Transcription Factor HES-1 , Tumor Cells, Cultured/cytology
6.
BMC Cell Biol ; 9: 46, 2008 Aug 18.
Article in English | MEDLINE | ID: mdl-18710550

ABSTRACT

BACKGROUND: The normal growth and function of mammary epithelial cells depend on interactions with the supportive stroma. Alterations in this communication can lead to the progression or expansion of malignant growth. The human mammary gland contains two distinctive types of fibroblasts within the stroma. The epithelial cells are surrounded by loosely connected intralobular fibroblasts, which are subsequently surrounded by the more compacted interlobular fibroblasts. The different proximity of these fibroblasts to the epithelial cells suggests distinctive functions for these two subtypes. In this report, we compared the gene expression profiles between the two stromal subtypes. METHODS: Fresh normal breast tissue was collected from reduction mammoplasty patients and immediately placed into embedding medium and frozen on dry ice. Tissue sections were subjected to laser capture microscopy to isolate the interlobular from the intralobular fibroblasts. RNA was prepared and subjected to microarray analysis using the Affymetrix Human Genome U133 GeneChip. Data was analyzed using the Affy and Limma packages available from Bioconductor. Findings from the microarray analysis were validated by RT-PCR and immunohistochemistry. RESULTS: No statistically significant difference was detected between the gene expression profiles of the interlobular and intralobular fibroblasts by microarray analysis and RT-PCR. However, for some of the genes tested, the protein expression patterns between the two subtypes of fibroblasts were significantly different. CONCLUSION: This study is the first to report the gene expression profiles of the two distinct fibroblast populations within the human mammary gland. While there was no significant difference in the gene expression profiles between the groups, there was an obvious difference in the expression pattern of several proteins tested. This report also highlights the importance of studying gene regulation at both the transcriptional and post-translational level.


Subject(s)
Gene Expression Regulation , Mammary Glands, Human/cytology , Mammary Glands, Human/metabolism , Adolescent , Adult , Female , Fibroblasts/metabolism , Gene Expression Profiling , Genotype , Humans , Phenotype , Proteins/genetics , Proteins/metabolism
7.
Oncogene ; 26(54): 7526-34, 2007 Nov 29.
Article in English | MEDLINE | ID: mdl-17546050

ABSTRACT

Branching morphogenesis within the peripubertal mouse mammary gland is directed by progesterone (P). A role for the homeobox-containing transcription factor, Msx2, during branching morphogenesis is suggested from its ontogenic expression profile and hormonal regulation. Herein, we define the spatio-temporal control of Msx2 expression, the regulation of its expression by P and its direct role in ductal branching morphogenesis. P induces Msx2 in the presence of estrogen (E) both in vitro and in vivo while absence of the P receptor (PR) decreased Msx2 expression. Stable transfection of PR into mouse mammary epithelial cells increased the endogenous expression of Msx2 and their ability to undergo branching morphogenesis in vitro. Furthermore, normal mammary cells stably-transfected with Msx2 demonstrated increased branching morphogenesis in vitro while transgenic mice expressing Msx2 in their mammary glands demonstrated enhanced lateral branching during early development. The action of P on branching morphogenesis appears to involve Bmp2/4. Together, these data demonstrate that P, acting through PR-A and the Bmp2/4 pathway, induces Msx2 to enhance ductal branching in the mammary glands.


Subject(s)
DNA-Binding Proteins/genetics , Homeodomain Proteins/genetics , Mammary Glands, Animal/physiology , Morphogenesis/physiology , Progesterone/pharmacology , Animals , Female , Gene Expression Regulation/drug effects , Mammary Glands, Animal/drug effects , Mice , Mice, Inbred BALB C , Morphogenesis/drug effects , Ovariectomy , Polymerase Chain Reaction , RNA/genetics , RNA/isolation & purification , Receptors, Progesterone/physiology , Signal Transduction
8.
Oncogene ; 26(51): 7204-12, 2007 Nov 08.
Article in English | MEDLINE | ID: mdl-17496925

ABSTRACT

Estrogen and its receptor alpha (ERalpha) have been implicated in the tissue-specific tumorigenesis associated with BRCA1 mutations. However, the majority of breast cancers developed in human BRCA1 mutation carriers are ERalpha-negative, challenging the link between BRCA1 and estrogen/ERalpha in breast cancer formation. Using a mouse model lacking the full-length form of BRCA1, here we show that ERalpha is highly expressed in the premalignant mammary gland and initiation stages of tumorigenesis, although its expression is gradually diminished during mammary tumor progression. We demonstrate that the absence of full-length BRCA1 increases sensitivity of cells to estrogen-induced extracellular signal-regulated kinase 1/2 phosphorylation and cyclin D1 expression. The absence of BRCA1 turns the proliferation of ERalpha-positive cells from a paracrine fashion to an autocrine or endocrine fashion. Consequently, BRCA1-mutant cells are sensitized to estrogen-induced cell proliferation in vitro and mammary tumorigenesis in vivo. These findings illustrate a molecular mechanism for estrogen/ERalpha signals in BRCA1-associated tissue-specific tumor formation, and identify several key elements in the estrogen/ERalpha-signaling cascade that can serve as potential therapeutic targets for BRCA1-associated tumorigenesis.


Subject(s)
BRCA1 Protein/physiology , Estrogens/physiology , Signal Transduction/physiology , Animals , BRCA1 Protein/genetics , Cell Line , Cyclin D1/metabolism , Mice , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Phosphorylation
9.
Endocrinology ; 144(6): 2683-94, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12746333

ABSTRACT

Insulin receptor substrates (IRS) are central integrators of hormone, cytokine, and growth factor signaling. IRS proteins can be phosphorylated by a number of signaling pathways critical to normal mammary gland development. Studies in transgenic mice that overexpress IGF-I in the mammary gland suggested that IRS expression is important in the regulation of normal postlactational mammary involution. The goal of these studies was to examine IRS expression in the mouse mammary gland and determine the importance of IRS-1 to mammary development in the virgin mouse. IRS-1 and -2 show distinct patterns of protein expression in the virgin mouse mammary gland, and protein abundance is dramatically increased during pregnancy and lactation, but rapidly lost during involution. Consistent with hormone regulation, IRS-1 protein levels are reduced by ovariectomy, induced by combined treatment with estrogen and progesterone, and vary considerably throughout the estrous cycle. These changes occur without similar changes in mRNA levels, suggesting posttranscriptional control. Mammary glands from IRS-1 null mice have smaller fat pads than wild-type controls, but this reduction is proportional to the overall reduction in body size. Development of the mammary duct (terminal endbuds and branch points) is not altered by the loss of IRS-1, and pregnancy-induced proliferation is not changed. These data indicate that IRS undergo complex developmental and hormonal regulation in the mammary gland, and that IRS-1 is more likely to regulate mammary function in lactating mice than in virgin or pregnant mice.


Subject(s)
Estrogens/pharmacology , Mammary Glands, Animal/physiology , Phosphoproteins/genetics , Progesterone/pharmacology , Signal Transduction/physiology , Adipose Tissue/chemistry , Adipose Tissue/growth & development , Adipose Tissue/physiology , Animals , Estrous Cycle/physiology , Female , Gene Expression/drug effects , Gene Expression/physiology , Insulin Receptor Substrate Proteins , Intracellular Signaling Peptides and Proteins , Mammary Glands, Animal/chemistry , Mammary Glands, Animal/growth & development , Mice , Mice, Inbred Strains , Ovariectomy , Phosphoproteins/analysis , Pregnancy , Signal Transduction/drug effects
10.
Br J Cancer ; 88(8): 1301-9, 2003 Apr 22.
Article in English | MEDLINE | ID: mdl-12698200

ABSTRACT

Prolactin (PRL) has been reported to inhibit apoptosis in various cell types and to serve as a cofactor in the upregulation of CD25 on T cells during activation. We investigated a possible relation between prolactin receptor (PRL-R) or IL-2 receptor alpha (IL-2Ralpha, CD25) expression on circulating T lymphocytes and their apoptosis in patients with breast cancer. Peripheral blood mononuclear cells obtained from 25 patients, 25 normal controls (NC) and three cord blood samples were evaluated for Annexin V binding and expression of CD95, CD25, and PRL-R on CD3(+) T cells by multicolour flow cytometry. Plasma levels of PRL, sCD95L, and sIL-2R were determined in patients and controls and related to T-cell apoptosis. The ability of PRL to protect T cells from apoptosis induced by various agents was also studied. Expression of PRL-R on the surface of T cells was comparable in patients with breast cancer and NC, but PRL plasma levels in patients were significantly lower (P<0.05). In patients, 18+/-11% (mean+/-s.d.) of CD3(+) cells bound Annexin V, compared to 9+/-6% in NC (P<0.0004). Percentages of CD3(+)Fas(+) and CD3(+)CD25(+) cells were higher in the peripheral circulation of patients than NC (P<0.0001 and <0.04, respectively). Levels of sFasL were lowest in plasma of the patients with the highest proportions of CD3(+)Fas(+) T cells. Most T cells undergoing apoptosis were CD3(+)CD25(-) in patients, and the proportion of CD3(+)CD25(-) Annexin V(+) cells was significantly increased in patients compared to NC (P<0.006). Ex vivo PRL protected T cells from starvation-induced or anti-CD3Ab-induced but not from Fas/FasL-dependent apoptosis. These results indicate that expression of CD25 but not of PRL-R on the surface of activated T lymphocytes appears to be involved in modulating Fas/Fas - ligand interactions, which are, in part, responsible for apoptosis of T lymphocytes and excessive turnover of immune cells in the circulation of patients with breast cancer.


Subject(s)
Apoptosis/immunology , Breast Neoplasms/immunology , Receptors, Interleukin-2/immunology , Receptors, Prolactin/physiology , T-Lymphocytes/immunology , Aged , Antigens, CD/immunology , Breast Neoplasms/pathology , Female , Fetal Blood , Humans , Jurkat Cells , Middle Aged , Neoplasm Staging , Reference Values , fas Receptor/immunology
11.
J Mol Endocrinol ; 30(1): 31-47, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12580759

ABSTRACT

Endocrine and autocrine prolactin (PRL) exerts effects on normal breast and breast cancer cells, and high serum PRL is a poor prognostic factor for colorectal cancer. Here we tested the hypothesis that short isoforms of the PRL receptor (PRLR) in human tissue regulate the actions of PRL in cancer. Using 3' RACE we isolated five splice variants of the human PRLR (hPRLR), three of which encode the complete extracellular binding domain. Two of these isoforms, short form 1a (SF1a) and short form 1b (SF1b), possess unique intracellular domains encoded by splicing to exon 11 from exons 10 and 9 respectively. A third novel isoform (delta7/11) reflects alternative splicing from exon 7 to exon 11 and encodes a secreted soluble PRL-binding protein. Additional splice variants of SF1b and delta7/11 that lacked exon 4 (delta4-SF1b and delta4-delta7/11) were also identified. Functional analyses indicated that hPRLR-SF1b is a strong dominant-negative to the differentiative function of the PRLR long form while hPRLR-SF1a is a weaker dominant-negative. Differential abundance of SF1a, SF1b and delta7/11 expression was detected in normal breast, colon, placenta, kidney, liver, ovary and pancreas, and breast and colon tumors. Taken together, these data indicate the presence of multiple isoforms of the hPRLR that may function to modulate the endocrine and autocrine effects of PRL in normal human tissue and cancer.


Subject(s)
Alternative Splicing , Carrier Proteins/genetics , Exons , Protein Isoforms/genetics , Receptors, Prolactin/genetics , Amino Acid Sequence , Animals , Blotting, Northern , CHO Cells , Carrier Proteins/metabolism , Cell Differentiation , Cloning, Molecular , Cricetinae , DNA Primers , DNA, Complementary , Humans , Molecular Sequence Data , Placental Lactogen/metabolism , Protein Isoforms/metabolism , Receptors, Prolactin/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
J Clin Pathol ; 54(12): 956-60, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11729217

ABSTRACT

AIMS: Prolactin plays an important role in the proliferation and differentiation of normal breast epithelium, and possibly in the development of breast carcinoma. The effects of prolactin are mediated by its receptor; thus, alteration in the expression of this receptor could be important in studying the biology of breast cancer. This investigation was aimed at comparing the expression of prolactin receptors in normal, benign, and malignant breast tissue. MATERIAL/METHODS: The expression of prolactin receptors was studied in paraffin wax embedded sections of 102 breast biopsies (93 female and nine male), using the monoclonal antibody B6.2, and the avidin-biotin immunoperoxidase technique. Six biopsies were normal, 34 had benign lesions, and 62 were malignant. RESULTS: In normal cases, prolactin receptor positivity was seen only on the luminal borders of the epithelial cells lining ducts and acini. In most benign lesions, variable degrees of luminal and cytoplasmic staining were seen. Cells showing apocrine metaplasia and florid regular ductal epithelial hyperplasia were mostly negative. In malignant cases, the staining pattern was mostly cytoplasmic and heterogeneous. Forty one of the 59 carcinomas in women showed a degree of positivity involving 10-100% of the tumour cells. A significant direct correlation was found between prolactin receptor and oestrogen receptor staining when only cases that scored more than 100/300 for the latter receptor, using the H scoring system, were considered (p = 0.0207). No correlation was found between prolactin receptors and progesterone receptors, patient's age, tumour size, tumour grade, or axillary lymph node status. CONCLUSIONS: Prolactin receptors seem to be expressed at different cellular sites in normal, benign, and malignant breast epithelial cells. The receptor is expressed in more than two thirds of female breast carcinomas, suggesting that it may play a role in the pathogenesis of the disease. The positivity is correlated with moderate and strong staining for oestrogen receptors in tissue sections, but not with other prognostic factors.


Subject(s)
Breast Diseases/metabolism , Breast/chemistry , Neoplasm Proteins/analysis , Receptors, Progesterone/analysis , Adult , Aged , Aged, 80 and over , Breast Neoplasms/chemistry , Breast Neoplasms, Male/chemistry , Carcinoma, Ductal, Breast/chemistry , Chi-Square Distribution , Female , Fibroadenoma/chemistry , Humans , Immunohistochemistry/methods , Male , Middle Aged
13.
Dev Dyn ; 222(3): 354-67, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11747071

ABSTRACT

Protein tyrosine kinases and phosphatases are signaling molecules involved in all aspects of development, including proliferation, differentiation, and apoptosis. How disruption of protein tyrosine phosphatase affects mammary gland development is not entirely clear. We examined the effects of sodium vanadate, which is known to primarily inhibit tyrosine phosphatases, in mouse mammary gland development in whole organ culture. Mammary epithelial differentiation was effectively inhibited by vanadate in a dose-dependent manner as indicated by lack of epithelial alveoli compared to the contralateral non-treated gland controls. Mammary glands in the differentiation medium after four days in the presence of vanadate did not differentiate into alveoli. Instead, they exhibited prominent terminal end buds and lost the distinctive epithelial structures. The inhibitory effect of vanadate on mammary epithelial cell differentiation was irreversible after one day of treatment. Immunohistochemical staining for PCNA (Proliferating Cell Nuclear Antigen) showed that vanadate-treated glands exhibited elevated proliferation signals in the differentiation medium. Expression of beta-casein protein in the vanadate-treated glands decreased dramatically and progressively. Short-term exposure (up to 72 hours) of mammary glands to vanadate resulted in an increase in mammary epithelial cell density and loss of organization of the mammary structures. TUNEL assay of mammary glands with prolonged exposure to vanadate revealed widespread apoptosis. Furthermore, some cells were still proliferating or expressing beta-casein after prolonged exposure to vanadate. Taken together, these data indicate that vanadate treatment blocks mammary epithelial cell differentiation and promotes abnormal proliferation and apoptosis, likely through the inhibition of protein tyrosine phosphatase-mediated signaling.


Subject(s)
Mammary Glands, Animal/drug effects , Mammary Glands, Animal/growth & development , Vanadates/pharmacology , Animals , Animals, Newborn/growth & development , Apoptosis/physiology , Caseins/antagonists & inhibitors , Cell Differentiation/drug effects , Cell Division/drug effects , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelium/drug effects , Epithelium/growth & development , Female , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred BALB C , Organ Culture Techniques , Reference Values , Time Factors , Vanadates/administration & dosage
14.
Dev Dyn ; 222(2): 192-205, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11668597

ABSTRACT

Ductal branching within the mammary gland is stimulated by prolactin (PRL) and progesterone (P) acting through their receptors (PRLR and PR). Analysis of mammary gland PRLR expression revealed increasing expression of the long form (L-PRLR) and two of the three short forms (S1- and S3-PRLR) during puberty that became maximal late in pubescence and early gestation, then declined during gestation. By contrast, S2-PRLR mRNA levels remained constant. Examination of stromal PRLR revealed the consistent expression of L-PRLR mRNA. By contrast, S1-PRLR was present only in the mammary fat pad of neonates, whereas high neonatal expression of S2-PRLR became undetectable during puberty. Stromal expression of S3-PRLR decreased to low levels during puberty and was undetectable during lactation and involution. Exogenous PRL stimulated DNA synthesis in both epithelial and adjacent stromal cells in vivo. Distribution of PRLR mRNA in mammary epithelium was homogeneous before puberty and heterogeneous during puberty, gestation, and early lactation. A mutual role for PRLR and PR was suggested wherein PR mRNA increased beyond 6 weeks to maximal levels during puberty and gestation then became undetectable during lactation. In situ hybridization revealed that PR mRNA distribution is homogeneous in the ductal epithelium before 6 weeks and heterogenous during puberty and gestation and that PRLR and PR are similarly distributed in the ductal epithelium. Neither hormone stimulated DNA synthesis in mammary glands of ovariectomized females while their effects interacted markedly. These results demonstrate differential PRLR transcription by epithelial and stromal cells and a similar distribution of PRLR and PR that may facilitate the interaction between P and PRL during ductal branching in the mammary gland.


Subject(s)
Mammary Glands, Animal/growth & development , Receptors, Progesterone/metabolism , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , Transcription, Genetic/physiology , Adipose Tissue/physiology , Animals , Cell Division/drug effects , Cell Division/physiology , Drug Synergism , Epithelial Cells/cytology , Epithelial Cells/metabolism , Estrogens/pharmacology , Female , Gene Expression Regulation, Developmental , Mammary Glands, Animal/cytology , Mammary Glands, Animal/physiology , Mice , Mice, Inbred BALB C , Ovariectomy , Progesterone/pharmacology , Prolactin/pharmacology , RNA, Messenger/analysis , Receptors, Progesterone/genetics , Stromal Cells/cytology , Stromal Cells/metabolism
15.
J Clin Endocrinol Metab ; 86(8): 3826-32, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11502819

ABSTRACT

PRL is a major growth and differentiating hormone in the human breast, with activation of the PRL-PRL receptor complex increasingly recognized as an important mechanism in the induction and progression of mammary tumors. Although constitutive activation of various hormone and growth factor receptors is newly recognized as a common cause of tumor development, the PRL receptor gene has not been analyzed for similar aberrations in breast and other tumors. Therefore, using bacterial artificial chromosomes containing the PRL receptor gene and intron-spanning PCR, we determined the exon-surrounding intron sequences providing primers for the first analysis of the entire coding region of the human PRL receptor gene. We examined the presence of PRL receptor in 41 breast tumors by immunohistochemistry and attempted a correlation of its expression to pathological grading of the disease. Then tumor cells were isolated by laser capture microdissection to examine DNA from 30 patients for PRL receptor mutations. The PRL receptor immunoreactive score did not correlate to the tumor size, histopathological grading, age, or family history of patients. PRL receptor immunoreactivity was predominantly found in steroid hormone receptor-positive tumors, but without overall correlation of immunoreactive score. In both PRL receptor-positive and PRL receptor- negative breast cancer cells, direct sequencing of the coding sequence of the PRL receptor gene did not detect any somatic or hereditary gene aberrations. In conclusion, PRL receptor mutations do not appear to be common in human breast cancer, suggesting that constitutive activation of the PRL receptor can be excluded as a major cause of mammary tumor genesis. The molecular structure of the PRL receptor seems to remain intact in tumor tissue, and systemic and local production of PRL may participate in tumor cell growth and proliferation through functional receptors.


Subject(s)
Breast Neoplasms/genetics , Breast Neoplasms/pathology , Receptors, Prolactin/genetics , Adult , Aged , Aged, 80 and over , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Lobular/genetics , Carcinoma, Lobular/pathology , DNA Mutational Analysis , DNA Primers , DNA, Neoplasm/genetics , Exons , Female , Humans , Immunohistochemistry , Introns , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Polymerase Chain Reaction , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis , Receptors, Prolactin/analysis
16.
Mol Cell Endocrinol ; 181(1-2): 47-56, 2001 Jul 05.
Article in English | MEDLINE | ID: mdl-11476940

ABSTRACT

Keratinocyte growth factor (KGF) is a stroma-derived mitogen mediating epithelial-stromal interactions. We investigated the role of KGF during epithelial-stromal interactions accompanying ruminant mammogenesis. Target-specificity of KGF was demonstrated in that KGF-stimulated proliferation of bovine mammary epithelial, but not ovine mammary stromal cells. Consistent with a paracrine function, 4.6, 2.4, 1.5 and 0.9 kb mRNA transcripts were expressed by bovine stromal, but not epithelial cells. Within the ovine mammary gland, 2.4 and 1.5 kb KGF mRNAs were expressed in the fat pad while only the 2.4 kb transcript was transcribed in parenchyma. The abundance of KGF mRNA was greater in the extra-parenchymal fat pad than in the contralateral epithelium-free fat pad prior to puberty, and was less in parenchyma than in the intact or epithelium-free fat pads. Ovariectomy tended to increase KGF transcription while estrogen reduced expression. Of several tissues, mammary parenchyma expressed a 2.4 kb mRNA while adipose tissues expressed a 1.5 kb transcript. These results demonstrate local and systemic regulation of KGF transcription and support a paracrine role for KGF during ruminant mammogenesis.


Subject(s)
Epithelial Cells/metabolism , Fibroblast Growth Factors/genetics , Fibroblast Growth Factors/metabolism , Mammary Glands, Animal/growth & development , Paracrine Communication , Sheep/genetics , Animals , Cattle , Cell Division/drug effects , Cells, Cultured , Epithelial Cells/cytology , Epithelial Cells/drug effects , Estrogens/pharmacology , Fibroblast Growth Factor 7 , Fibroblast Growth Factors/pharmacology , Fibroblasts , Gene Expression Regulation/drug effects , In Situ Hybridization , Mammary Glands, Animal/cytology , Mammary Glands, Animal/drug effects , Mammary Glands, Animal/metabolism , Mitogens/genetics , Mitogens/metabolism , Mitogens/pharmacology , Ovariectomy , Paracrine Communication/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sheep/growth & development , Species Specificity , Stromal Cells/cytology , Stromal Cells/drug effects , Stromal Cells/metabolism , Substrate Specificity
17.
Mol Endocrinol ; 15(5): 819-31, 2001 May.
Article in English | MEDLINE | ID: mdl-11328861

ABSTRACT

Accompanying changes in the development and function of the mammary gland is the establishment of a vascular network of critical importance for lactogenesis and tumorigenesis. A potent angiogenic and permeability factor that regulates vascular development in association with epithelial-stromal interactions is vascular endothelial growth factor (VEGF). Analysis of VEGF transcription by RT-PCR revealed mRNA for all three VEGF isoforms (VEGF120, 164, 188) within the mammary gland of nulliparous females. During pregnancy the level of VEGF188 declined and became undetectable during lactation in association with the increased abundance of VEGF120 and VEGF164 mRNAS: All three isoforms were expressed at consistent levels within the cleared mammary fat pad throughout development. Furthermore, the presence of VEGF188 mRNA in omental adipose tissue at various stages established that VEGF188 is expressed specifically in adipose tissue within the mammary gland. Using 3T3-L1 preadipocytes it was demonstrated that VEGF188 mRNA transcription occurs as a late event during lipogenesis distinct from earlier induction of VEGF120 and VEGF164 mRNA during differentiation. In contrast, HC11 mammary epithelial cells only expressed mRNA for VEGF120 and VEGF164. Localization of VEGF mRNA and protein revealed that VEGF is expressed in stromal cells of the mammary gland in nulliparous females and then undergoes a transition to epithelial expression during lactation. By contrast, mRNA for the VEGF receptors, Flk-1 and Flt-1, localized to stromal cells within the mammary fat pad during virgin and gestational development and was expressed in the interstitial tissue basal to epithelial cells during lactation. Taken together, these results support the conclusion that VEGF is differentially transcribed by specific cell types within the mammary gland, and that under hormonal regulation it functions in an autocrine/paracrine manner.


Subject(s)
Endothelial Growth Factors/biosynthesis , Gene Expression Regulation, Developmental/physiology , Lymphokines/biosynthesis , Mammary Glands, Animal/physiology , Adipocytes/physiology , Animals , Blotting, Northern , Blotting, Western , Endothelial Growth Factors/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Extracellular Matrix Proteins/biosynthesis , Extracellular Matrix Proteins/metabolism , Female , Immunohistochemistry , In Situ Hybridization , Lymphokines/genetics , Male , Mammary Glands, Animal/blood supply , Mammary Glands, Animal/cytology , Mice , Mice, Inbred BALB C , Myosin Heavy Chains , Neovascularization, Physiologic/physiology , Nonmuscle Myosin Type IIB , Pregnancy , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Growth Factor/biosynthesis , Receptors, Growth Factor/metabolism , Receptors, Vascular Endothelial Growth Factor , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/cytology , Stromal Cells/metabolism , Transcriptional Activation/physiology , Vascular Endothelial Growth Factor A , Vascular Endothelial Growth Factor Receptor-1 , Vascular Endothelial Growth Factors
18.
J Endocrinol ; 167(1): 39-52, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11018751

ABSTRACT

Development of the functional secretory epithelium in the mammary gland of the female mouse requires the elongation of the anlage through the mammary fat pad to form the primary/secondary ductal network from which tertiary ductal side-branches and lobuloalveoli develop. In this study we examined the hormonal requirements for the spatial development of the primary/secondary epithelial network and tertiary side-branches by quantifying ductal growth and epithelial cell proliferation in normal and hormone-treated BALB/c mice between 21 and 39 days of age. In normal mice, an allometric increase in ductal length commenced at 31 days of age and resulted in completion of the primary/secondary ductal network by 39 days of age. Concurrent with this allometric growth was a significant increase in cellular proliferation in the terminal end-buds (TEBs) of the ductal epithelium from 29 days of age, as determined by 5-bromo-2'-deoxyuridine (BrdU) incorporation. A level of cellular proliferation similar to that in the TEBs of 33-day-old control mice could be induced in the TEBs of 25-day-old mice following treatment for 1 day with estrogen (E), or progesterone (P) or both (E/P), indicating that both E and P were mitogenic for epithelial cells of the peripubertal TEBs. However, the period of allometric ductal growth in untreated mice did not correspond to an increase in serum E or P (which might have been expected during the estrous cycle). In addition, epithelial growth was not observed in mammary glands from 24-day-old mice that were cultured in vitro with E, P or E/P. In contrast to treatment with E, treatment with P promoted a dramatic increase, relative to control mice, in the number of tertiary branch points upon the primary/secondary ductal network. BrdU labeling of mammary glands from 24- 33-day-old mice pelleted with cholesterol (C), E, P or E/P confirmed the greater mitogenicity of P on the epithelial cells of the secondary/tertiary ducts as compared with C or E. Concurrent with these changes, localized progesterone receptor (PR) expression in clusters of cells in the ductal epithelium was associated with structures that histologically resembled early branch points from ductules. In conclusion, our results suggest that additional endocrine growth factor(s) other than E and P contribute to the development of the primary/secondary ductal network, and that P is responsible for the formation of tertiary side-branches in the mammary glands of mice during puberty.


Subject(s)
Aging/physiology , Mammary Glands, Animal/drug effects , Progesterone/pharmacology , Animals , Cell Division , Epithelium/drug effects , Epithelium/growth & development , Estradiol/pharmacology , Estrogens/blood , Female , Gene Expression , In Situ Hybridization , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mice , Mice, Inbred BALB C , Organ Culture Techniques , Progesterone/blood , RNA, Messenger/genetics , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism
19.
Endocrine ; 12(3): 265-71, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10963047

ABSTRACT

The liver is an essential target tissue for growth hormone (GH) and prolactin (PRL). The aim of this study was to determine the in situ expression of growth hormone receptor (GHR) and prolactin receptor (PRLR) in hepatocellular carcinomas and to compare the results with normal liver. For this purpose, in situ hybridization (ISH) and immunohistochemical techniques were performed and several tests were conducted to validate the results. By radioactive ISH, all the hepatocellular carcinomas studied showed labeling for GHR and PRLR mRNAs. Relative expression levels, determined by computer-assisted microdensity, were higher in hepatocellular carcinomas than in normal liver. Immunohistochemistry led us to confirm the constant expression of both receptor proteins in hepatocellular carcinomas and normal liver and to demonstrate their localization not only in the cytoplasm but also in the nucleus. These results confirm that the liver is a major GH and PRL target tissue and suggest that in hepatocellular carcinomas the proliferative effects of these hormones may be increased by a higher expression of their receptors.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Gene Expression , Liver Neoplasms/metabolism , Receptors, Prolactin/genetics , Receptors, Somatotropin/genetics , Adult , Aged , Aged, 80 and over , Cell Nucleus/chemistry , Cytoplasm/chemistry , Female , Growth Hormone/pharmacology , Humans , Immunohistochemistry , In Situ Hybridization , Male , Middle Aged , Prolactin/pharmacology , RNA, Messenger/analysis
20.
Mol Endocrinol ; 14(3): 359-68, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10707954

ABSTRACT

Deletion of the transcription factor CCAAT/enhancer binding protein (C/EBP)beta results in a severe inhibition of lobuloalveolar development in the mouse mammary gland. Because progesterone receptor (PR) is requisite for alveolar development, the expression of PR was investigated in C/EBPbeta-/- mice. Unexpectedly, the number of PR-positive cells, as well as the levels of PR mRNA, were elevated 3-fold in the mammary glands of C/EBPbeta-/- mice. Furthermore, in contrast to wild-type nulliparous mice, in which PR distribution shifted from a uniform to nonuniform pattern between 8-12 weeks of age, C/EBPbeta-/- mice exhibited uniform PR distribution throughout all stages of mammary development analyzed. No change in C/EBPbeta mRNA levels was observed in the mammary glands of PR-/- mice, suggesting that PR acts in a pathway either in parallel to or downstream of C/EBPbeta. The overexpression and disrupted cellular distribution of PR in C/EBPbeta-/- mice were coincident with a striking 10-fold decrease in cell proliferation after acute steroid hormone treatment, assayed by incorporation of bromodeoxyuridine. In wild-type mice, PR and bromodeoxyuridine-positive cells were adjacent to each other and rarely colocalized. No differences in the level or pattern of PR expression were observed in the uterus, suggesting that C/EBPbeta influences PR in a mammary-specific fashion. Together, these data suggest that C/EBPbeta may control cell fate decisions in the mammary gland through the appropriate temporal and spatial expression of molecular markers, such as PR, that induce the proliferation of alveolar progenitor cells via juxtacrine mechanisms.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , Mammary Glands, Animal/growth & development , Nuclear Proteins/physiology , Receptors, Progesterone/metabolism , Animals , Biomarkers , CCAAT-Enhancer-Binding Proteins , Cell Division/drug effects , Cell Lineage , DNA Replication/drug effects , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Fluorescent Antibody Technique, Indirect , Gene Deletion , In Situ Hybridization , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Nuclear Proteins/deficiency , Nuclear Proteins/genetics , RNA, Messenger/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL
...