Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
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
2.
J Dairy Sci ; 85(2): 365-77, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11913696

ABSTRACT

Insulin-like growth factor I (IGF-I) is known to regulate mammary gland development. This regulation occurs through effects on both cell cycle progression and apoptosis. Our laboratory has studied the IGF-I-dependent regulation of these processes by using transgenic and knockout mouse models that exhibit alterations in the IGF-I axis. Our studies of transgenic mice that overexpress IGF-I during pregnancy and lactation have demonstrated that this growth factor slows the apoptotic loss of mammary epithelial cells during the declining phase of lactation but has minimal effects during early lactation on milk composition or lactational capacity. In contrast, our analysis of early developmental processes in mammary tissue from mice carrying a targeted mutation in the IGF-I receptor gene suggests that IGF-dependent stimulation of cell cycle progression is more important to early mammary gland development than potential anti-apoptotic effects. With both models, the effects of perturbing the IGF-I axis are dependent on the physiological state of the animal. The diminished ductal development that occurs in response to loss of the IGF-I receptor is dramatically restored during pregnancy, whereas the ability of overexpressed IGF-I to protect mammary cells from apoptosis does not occur if the mammary gland is induced to undergo forced involution. Data from our laboratory on the expression of IGF-signaling molecules in the mammary gland suggest that this effect of physiological context may be related to the expression of members of the insulin receptor substrate family.


Subject(s)
Apoptosis/genetics , Insulin-Like Growth Factor I/physiology , Lactation/genetics , Mammary Glands, Animal/growth & development , Animals , Cattle , Female , Insulin-Like Growth Factor I/genetics , Mammary Glands, Animal/physiology , Mice , Mice, Knockout , Mice, Transgenic , Milk/chemistry , Models, Animal , Signal Transduction/genetics
3.
Endocrinology ; 142(11): 4937-45, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11606462

ABSTRACT

IGF-I mediates mammary ductal development through stimulation of terminal end bud (TEB) development; however, no published data exist on the mechanism through which this occurs. The mechanism of IGF-I action on the TEB was studied by determining the requirement for the IGF-I receptor (IGF-IR) in IGF-I-dependent ductal development. We hypothesized that loss of the IGF-IR would disrupt mammary ductal development through a combination of decreased proliferation or increased apoptosis. Because IGF-IR null mice die at birth, embryonic mammary gland transplantation was used to study the effects of a disrupted IGF-IR gene. Analyses of grafts after 4 or 8 wk of development demonstrated a limited growth potential of the null mammary epithelium in virgin hosts. Bromodeoxyuridine labeling and terminal deoxynucleotidyltransferase-mediated deoxy-UTP nick-end labeling showed that cell proliferation was significantly decreased in null TEBs, but apoptosis was not. In addition, both the size and number of TEBs were reduced in null outgrowths. In pregnant hosts, null ductal growth was stimulated beyond the level seen in virgin hosts. These findings directly establish a proliferation-dependent role for the IGF-IR in the cells of the TEB. Additionally, this study indicates that pregnancy-dependent compensatory mechanisms can stimulate mammary development in the absence of an IGF-IR.


Subject(s)
Mammary Glands, Animal/cytology , Receptor, IGF Type 1/physiology , Animals , Apoptosis/physiology , Cell Division/physiology , Epithelial Cells/cytology , Female , Fetal Tissue Transplantation , Gene Targeting , Male , Mammary Glands, Animal/anatomy & histology , Mammary Glands, Animal/embryology , Mammary Glands, Animal/growth & development , Mice , Mice, Knockout/genetics , Pregnancy , Receptor, IGF Type 1/genetics , Time Factors , Transplantation, Heterotopic
4.
J Mammary Gland Biol Neoplasia ; 5(1): 19-30, 2000 Jan.
Article in English | MEDLINE | ID: mdl-10791765

ABSTRACT

Transgenic and knockout mice have become valuable experimental systems with which to study specific molecular events within the mammary gland of an intact animal. These models have provided a wealth of information about the effects of a number of oncogenes and growth factors. This review focuses on results obtained from the application of transgenic and knockout models to determine the roles of insulin and insulin-like growth factors (IGF) in the regulation of mammary gland development, lactation and tumorigenesis. Transgenic models which overexpress IGF-I or -II display specific alterations in mammary gland development and an increased incidence of mammary tumors. Analysis of mammary gland development in knockout mice which are deficient in IGF-I or the IGF-I receptor supports the conclusion that the IGF system is important for normal mammary gland development. This review discusses these observations in detail and attempts to fit them into a larger picture of IGF and insulin action in the mammary gland.


Subject(s)
Breast/physiology , Insulin-Like Growth Factor II/physiology , Insulin-Like Growth Factor I/physiology , Mammary Glands, Animal/physiology , Animals , Breast Neoplasms/physiopathology , Female , Humans , Mammary Neoplasms, Animal/physiopathology , Mice , Mice, Knockout , Mice, Transgenic
5.
Oncogene ; 19(7): 889-98, 2000 Feb 17.
Article in English | MEDLINE | ID: mdl-10702797

ABSTRACT

Mammary tumorigenesis was analysed in transgenic mice which overexpress des(1-3)hIGF-I (WAP-DES) and/or a mutant form of p53 (p53172R-H). Nonlactating, multiparous WAP-DES mice exhibited hyperplastic lesions termed mammary interepithelial neoplasia (MIN) which constitutively expressed WAP-DES. By 23 months of age, 53% of the WAP-DES mice developed mammary adenocarcinomas. A 75% reduction in both apoptosis and proliferation was observed in the normal mammary glands of WAP-DES mice. Mammary tumor incidence in WAP-DES/p53 bitransgenic mice was similar to that of WAP-DES and 2 - 3-fold greater than that of nontransgenic and p53172R-H females. Tumor latency, however, was reduced by 8 months in bitransgenic mice as compared to mice of the other three genotypes. Aneuploidy was frequently observed in tumors from bitransgenic and p53172R-H mice, but not from mice expressing only the WAP-DES transgene. Expression of IGFBP3 was elevated in tumors from WAP-DES, but not bitransgenic mice, indicating an alteration in the p53/IGF-I axis. These studies indicate that overexpression of des(1-3)hIGF-I increases the frequency of MIN and stochastic mammary tumors and that the appearance of tumors displaying genomic instability is accelerated by mutant p53172R-H. Oncogene (2000) 19, 889 - 898.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/genetics , Mutation/genetics , Peptide Fragments/metabolism , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis/genetics , DNA Replication/genetics , Drug Synergism , Female , Male , Mammary Neoplasms, Experimental/metabolism , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred ICR , Mice, Transgenic
6.
Carcinogenesis ; 20(9): 1715-20, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10469615

ABSTRACT

We have developed an in vivo model system of mouse mammary preneoplasias in order to examine the cell and molecular changes that occur during tumorigenesis. Most of these preneoplasias are characterized by an alveolar hyperplasia morphologically similar to that present in normal pregnant mammary gland, but have tumor forming capabilities ranging from very low to high. One of these hyperplasias, the TM3 HOG (transformed mammary hyperplastic outgrowth), forms tumors infrequently and has the unusual characteristic of spontaneous regression. We have observed that 7-8 months post-transplantation into the cleared mammary fat pad of a BALB/c mouse, the TM3 hyperplasia will begin to regress, leaving only a sparse ductal tree with remnant alveolar structures by 10-12 months post-transplantation. We have sought to elucidate the mechanism of this regression by determining the apoptotic and proliferative rates of the alveolar cells during TM3 HOG development. Studies show that apoptotic rates in the TM3 HOG are consistently high (4-7%) at all times after transplantation. This apoptotic rate is higher than the rates found in other preneoplasias in our system and approach the rates observed in the normal involuting gland. An unusual p53 mutation, a serine insertion at codon 233, may be causally related to the high spontaneous apoptotic frequencies as well as elevated inducible apoptotic frequencies in TM3. In addition, a sudden decrease ( approximately 63%) in proliferation occurs around 8 months post-transplantation. Furthermore, transplantation experiments indicate that the ability of the 8-month-old host and/or mammary gland to support growth of preneoplastic mammary tissues is markedly diminished compared with 3- or 6-month-old hosts. The results presented here suggest that the persistent high apoptotic rates, concomitant with decreased proliferation rates, may be responsible for TM3's regression and implicate a unique mutant p53 as a causal factor. Additionally, the results suggest that host determinants can interact with specific molecular changes in the preneoplastic cells to influence growth and progression of the preneoplastic populations.


Subject(s)
Apoptosis/genetics , Genes, p53 , Mammary Glands, Animal/pathology , Precancerous Conditions/genetics , Age Factors , Animals , Apoptosis/radiation effects , Cell Division , Codon/genetics , Female , Gamma Rays , Hyperplasia , Mammary Glands, Animal/transplantation , Mice , Mice, Inbred BALB C , Mutation , Precancerous Conditions/pathology , Remission, Spontaneous , Tumor Suppressor Protein p53/physiology
7.
Cell Prolif ; 29(11): 623-39, 1996 Nov.
Article in English | MEDLINE | ID: mdl-9105418

ABSTRACT

Neoplastic transformation of mouse mammary epithelial cells is the result of several identifiable phenotypic changes which presumably require sequential genetic alterations. In our model system, mammary cells progress from a mortal state (virgin duct) to several morphologically distinct intermediate states. The intermediate states are distinct cell populations that are phenotypically identified as immortal, non-tumourigenic (i.e. EL11), weakly tumourigenic ductal/alveolar hyperplasia (i.e. EL12) and moderately tumourigenic alveolar hyperplasiaa (i.e. TM12) to invasive tumours (i.e. EL12T/TM12T). We have studied the changes in total cyclin A and B1 levels, cyclin A and B1 complexed to cdc2, cyclin B1cdc2 kinase activity and cyclin D proteins in EL11 and EL12 immortalized outgrowth lines. Results revealed increased levels in total cyclin B1 (> 5-fold), cyclin B1/cdc2 (3-4-fold) and cyclin B1/cdc2 kinase activity (2-3.5-fold) in EL11 and EL12 phenotypes when compared to control mammary gland (virgin). No changes in the levels of total cyclin A or cycln A associated to cdc2 were observed. Cyclin D1, D2 and D3 protein levels were low in the EL11 immortal ductal outgrowth. Exposure to hormones via a pituitary isograft stimulated the synthesis of cyclin D1 and D2 but not D3 associated to cdk4 as well as total cdk4 proteins. Bromodeoxyuridine (BrdUrd) labelling indices showed marked increases in immortal ductal outgrowths (EL11 and EL12) when compared to virgin, suggesting that epithelial cells are cycling in these cell populations. Even in the presence of hormone stimulation, EL11 outgrowths were not tumourigenic, suggesting that other events are necessary to drive the cells to a tumourigenic phenotype. The results suggest that increased levels of cyclin B1 and cyclin B1-cdc2 kinase activities are early events and may be an important marker for the immortalized phenotype.


Subject(s)
CDC2 Protein Kinase/metabolism , Cell Transformation, Neoplastic , Cyclin B , Cyclins/biosynthesis , Gene Expression Regulation, Neoplastic , Mammary Glands, Animal/pathology , Mammary Neoplasms, Experimental , Animals , Cell Differentiation , Cell Division , Cyclin B1 , Enzyme Activation , Female , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Mammary Neoplasms, Experimental/pathology , Mice , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...