Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 141
Filter
Add more filters










Publication year range
1.
Nat Commun ; 8: 16069, 2017 07 17.
Article in English | MEDLINE | ID: mdl-28714474

ABSTRACT

Precise spatiotemporal gene regulation is paramount for the establishment and maintenance of cell-specific programmes. Although there is evidence that chromatin neighbourhoods, formed by the zinc-finger protein CTCF, can sequester enhancers and their target genes, there is limited in vivo evidence for CTCF demarcating super-enhancers and preventing cross talk between distinct regulatory elements. Here, we address these questions in the Wap locus with its mammary-specific super-enhancer separated by CTCF sites from widely expressed genes. Mutational analysis demonstrates that the Wap super-enhancer controls Ramp3, despite three separating CTCF sites. Their deletion in mice results in elevated expression of Ramp3 in mammary tissue through augmented promoter-enhancer interactions. Deletion of the distal CTCF-binding site results in loss of Ramp3 expression in non-mammary tissues. This suggests that CTCF sites are porous borders, allowing a super-enhancer to activate a secondary target. Likewise, CTCF sites shield a widely expressed gene from suppressive influences of a silent locus.


Subject(s)
CCCTC-Binding Factor/genetics , Chromatin/metabolism , Gene Expression Regulation/genetics , Mammary Glands, Animal/metabolism , Milk Proteins/genetics , Receptor Activity-Modifying Protein 3/genetics , Regulatory Elements, Transcriptional/genetics , Animals , Binding Sites , CCCTC-Binding Factor/metabolism , DNA Mutational Analysis , Enhancer Elements, Genetic/genetics , Female , Gene Regulatory Networks , Mice , Milk Proteins/metabolism , Promoter Regions, Genetic , Receptor Activity-Modifying Protein 3/metabolism
2.
Leukemia ; 30(8): 1725-33, 2016 08.
Article in English | MEDLINE | ID: mdl-27046463

ABSTRACT

Mutations that activate FMS-like tyrosine kinase 3 (FLT3) are frequent occurrences in acute myeloid leukemia. Two distinct types of mutations have been described: internal duplication of the juxtamembranous domain (ITD) and point mutations of the tyrosine kinase domain (TKD). Although both mutations lead to constitutive FLT3 signaling, only FLT3-ITD strongly activates signal transducer and activator of transcription 5 (STAT5). In a murine transplantation model, FLT3-ITD induces a myeloproliferative neoplasm, whereas FLT3-TKD leads to a lymphoid malignancy with significantly longer latency. Here we report that the presence of STAT5 is critical for the development of a myeloproliferative disease by FLT3-ITD in mice. Deletion of Stat5 in FLT3-ITD-induced leukemogenesis leads not only to a significantly longer survival (82 vs 27 days) of the diseased mice, but also to an immunophenotype switch with expansion of the lymphoid cell compartment. Interestingly, we were able to show differential STAT5 activation in FLT3-ITD(+) myeloid and lymphoid murine progenitors. STAT5 target genes such as Oncostatin M were highly expressed in FLT3-ITD(+) myeloid but not in FLT3-ITD(+) lymphoid progenitor cells. Strikingly, FLT3-TKD expression in combination with Oncostatin M is sufficient to reverse the phenotype to a myeloproliferative disease in FLT3-TKD mice. Thus, lineage-specific STAT5 activation in hematopoietic progenitor cells predicts the FLT3(+)-mediated leukemic phenotype in mice.


Subject(s)
Cell Lineage/genetics , Hematopoietic Stem Cells/metabolism , Leukemia, Myeloid, Acute/pathology , STAT5 Transcription Factor/genetics , Transcriptional Activation/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Carcinogenesis/genetics , Leukemia, Myeloid, Acute/genetics , Lymphoid Progenitor Cells/metabolism , Mice , Mutation , Myeloid Cells/metabolism , Oncostatin M , STAT5 Transcription Factor/metabolism
3.
Cell Death Differ ; 23(1): 41-51, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26045049

ABSTRACT

After cessation of lactation, involution of the mouse mammary gland proceeds in two distinct phases, a reversible and an irreversible one, which leads to the death and removal of alveolar cells. Cell death is preceded by the loss of STAT5 activity, which abrogates cell differentiation and gain of STAT3 activity. Despite early observations implicating BCL2 (B cell lymphoma 2) family proteins in this process, recent evidence suggests that STAT3-controlled cathepsin activity is most critical for cell death at the early stage of involution. Somewhat surprisingly, this cell death associates with but does not depend on the activation of pro-apoptotic effector caspases. However, transgenic overexpression of BCL2, that blocks caspase activation, delays involution while conditional deletion of BclX accelerates this process, suggesting that BCL2 family proteins are needed for the effective execution of involution. Here, we report on the transcriptional induction of multiple pro-apoptotic BCL2 family proteins of the 'BH3-only' subgroup during involution and the rate-limiting role of BIM in this process. Loss of Bim delayed epithelial cell clearance during involution after forced weaning in mice, whereas the absence of related Bmf had minor and loss of Bad or Noxa no impact on this process. Consistent with a contribution of BCL2 family proteins to the second wave of cell death during involution, loss of Bim reduced the number of apoptotic cells in this irreversible phase. Notably, the expression changes observed within the BCL2 family did not depend on STAT3 signalling, in line with its initiating role early in the process, but rather appear to result from relief of repression by STAT5. Our findings support the existence of a signalling circuitry regulating the irreversible phase of involution in mice by engaging BH3-only protein-driven mitochondrial apoptosis.


Subject(s)
Apoptosis Regulatory Proteins/biosynthesis , Cell Death/genetics , Mammary Glands, Animal/metabolism , Membrane Proteins/biosynthesis , Proto-Oncogene Proteins c-bcl-2/biosynthesis , Proto-Oncogene Proteins/biosynthesis , STAT5 Transcription Factor/genetics , Animals , Apoptosis Regulatory Proteins/genetics , Bcl-2-Like Protein 11 , Caspases/biosynthesis , Cell Differentiation/genetics , Female , Gene Expression Regulation, Developmental , Lactation/genetics , Lactation/metabolism , Mammary Glands, Animal/growth & development , Membrane Proteins/genetics , Mice , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins c-bcl-2/genetics , STAT5 Transcription Factor/biosynthesis
4.
Cell Death Differ ; 15(4): 783-92, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18259195

ABSTRACT

The transcription factors signal transducer and activator of transcription 5a and 5b (Stat5) are activated by the neuroprotective and neurotrophic cytokines, erythropoietin (EPO) and growth hormone (GH). Here, we show a dissociation of the intracellular pathway mediating the protective effect of EPO against glutamate toxicity from that needed for its neurotrophic activity using hippocampal neuronal cultures from Stat5a/b-knockout (Stat5(-/-)) mouse fetuses. Both pretreatment and post-treatment with EPO counteracted glutamate-induced cell death in Stat5(+/+) and Stat5(-/-) neurons. Acute pharmacological inhibition of Janus kinase 2 (JAK2)/Stat signalling had no effect on EPO neuroprotection, whereas inhibition of phosphatidylinositol-3' kinase (PI3K)/Akt pathway abolished the protective effect of EPO in both Stat5(+/+) and Stat5(-/-) neurons. GH effectively protected Stat5(+/+) cells against glutamate toxicity but had no effect in Stat5(-/-) neurons or in Stat5(+/+) neurons treated with JAK2/Stat or PI3K inhibitor. EPO and GH stimulated neurite outgrowth and branching of Stat5(+/+) neurons by activating PI3K/Akt signalling but had no trophic effect in Stat5(-/-) cells. We conclude that in hippocampal neurons, Stat5 is not required for neuroprotection by EPO but is together with Akt essential for its neurotrophic activity. Both Stat5 and Akt are needed for neuroprotective and neurotrophic signalling of GH in neurons.


Subject(s)
Erythropoietin/metabolism , Hippocampus/metabolism , Nerve Growth Factors/metabolism , Neurons/metabolism , Neuroprotective Agents/metabolism , STAT5 Transcription Factor/metabolism , Signal Transduction , Animals , Cell Survival , Cells, Cultured , Chromones/pharmacology , Cytoprotection , Glutamic Acid/toxicity , Hippocampus/drug effects , Hippocampus/embryology , Hippocampus/pathology , Human Growth Hormone/metabolism , Humans , Janus Kinase 2/antagonists & inhibitors , Janus Kinase 2/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Morpholines/pharmacology , Neurites/metabolism , Neurons/drug effects , Neurons/pathology , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Phosphorylation , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/metabolism , Recombinant Proteins , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/genetics , Signal Transduction/drug effects , Triterpenes/pharmacology
5.
Genomics ; 78(3): 129-34, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11735219

ABSTRACT

The signal transducers and activators of transcription (STAT) 5 and 3 are critical for mammary alveolar development during pregnancy and remodeling during involution. In the mouse, STAT3, STAT5a, and STAT5b are encoded by adjacent genes on chromosome 11 (60.5 cM). To identify additional genes in the Stat3/5 locus that may participate in normal and neoplastic development of the mammary gland, we have cloned and sequenced 500 kb and searched for genes preferentially expressed in mammary tissue. We identified six known genes and cloned two new genes, termed D11Lgp1 and D11Lgp2. Both genes are most highly expressed in normal mammary tissue and mammary tumors from several transgenic mouse models. LGP1 consists of 532 and 530 amino acids in mouse and human, respectively (88% similarity). A region in the carboxy-terminal half of LGP1 has limited homology with Arabidopsis thaliana GH3-like proteins. Immunofluorescence studies demonstrated that LGP1 is located in the nuclear envelope and the endoplasmic reticulum. LGP2 is a cytoplasmic protein of 678 amino acids.


Subject(s)
DNA Helicases/genetics , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Membrane Proteins/genetics , Milk Proteins , Trans-Activators/genetics , Amino Acid Sequence , Animals , Humans , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT3 Transcription Factor , STAT5 Transcription Factor , Sequence Homology, Amino Acid , Tumor Suppressor Proteins
6.
Mech Dev ; 109(2): 281-93, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11731240

ABSTRACT

In the mammary gland Bcl-x is the most abundant cell survival factor from the Bcl-2 family. Since Bcl-x null mice die around day 12 of embryogenesis, the relevance of this protein in organ development and function is poorly understood. In erythroid cells bcl-x gene expression is controlled by cytokines and the transcription factor Stat5 (signal transducer and activator of transcription). However, we identified that bcl-x RNA levels in mammary tissue from prolactin receptor- and Stat5-null mice were indistinguishable from wild type mice. We have proposed that Bcl-x might control the survival of mammary epithelial cells throughout pregnancy, lactation, and the early stages of involution, and we have now tested this hypothesis through the conditional deletion of the bcl-x gene from mouse mammary epithelium. Conditional (floxed) bcl-x alleles were excised from alveolar cells during pregnancy using a Cre transgene under the control of the whey acidic protein gene promoter. Deletion of the bcl-x gene from the entire epithelial compartment (ducts and alveoli) was achieved by expressing Cre-recombinase under control of the mouse mammary tumor virus long terminal repeat. The absence of Bcl-x did not compromise proliferation and differentiation of mammary ductal and alveolar epithelial cells in virgin mice and during pregnancy and lactation. However, epithelial cell death and tissue remodeling were accelerated in the bcl-x conditional knockout mice during the first stage of involution. Concomitant deletion of the bax gene did not significantly modify the Bcl-x phenotype. Our results suggest that Bcl-x is not essential during mammopoiesis, but is critical for controlled apoptosis during the first phase of involution.


Subject(s)
Apoptosis , Epithelial Cells/pathology , Gene Deletion , Lactation/physiology , Mammary Glands, Animal/pathology , Milk Proteins , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Alleles , Animals , Blotting, Southern , Blotting, Western , Cell Differentiation , DNA-Binding Proteins/metabolism , Female , Genotype , Integrases/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Phenotype , RNA/metabolism , Receptors, Prolactin/metabolism , Recombination, Genetic , Ribonucleases/metabolism , STAT5 Transcription Factor , Spleen/cytology , Trans-Activators/metabolism , Transgenes , Viral Proteins/metabolism , bcl-X Protein
7.
J Cell Biol ; 155(4): 531-42, 2001 Nov 12.
Article in English | MEDLINE | ID: mdl-11706048

ABSTRACT

Functional development of mammary epithelium during pregnancy depends on prolactin signaling. However, the underlying molecular and cellular events are not fully understood. We examined the specific contributions of the prolactin receptor (PrlR) and the signal transducers and activators of transcription 5a and 5b (referred to as Stat5) in the formation and differentiation of mammary alveolar epithelium. PrlR- and Stat5-null mammary epithelia were transplanted into wild-type hosts, and pregnancy-mediated development was investigated at a histological and molecular level. Stat5-null mammary epithelium developed ducts but failed to form alveoli, and no milk protein gene expression was observed. In contrast, PrlR-null epithelium formed alveoli-like structures with small open lumina. Electron microscopy revealed undifferentiated features of organelles and a perturbation of cell-cell contacts in PrlR- and Stat5-null epithelia. Expression of NKCC1, an Na-K-Cl cotransporter characteristic for ductal epithelia, and ZO-1, a protein associated with tight junction, were maintained in the alveoli-like structures of PrlR- and Stat5-null epithelia. In contrast, the Na-Pi cotransporter Npt2b, and the gap junction component connexin 32, usually expressed in secretory epithelia, were undetectable in PrlR- and Stat5-null mice. These data demonstrate that signaling via the PrlR and Stat5 is critical for the proliferation and differentiation of mammary alveoli during pregnancy.


Subject(s)
DNA-Binding Proteins/physiology , Mammary Glands, Animal/cytology , Milk Proteins , Pregnancy, Animal , Trans-Activators/physiology , Animals , Cell Differentiation , Cell Division , Connexins/metabolism , Connexins/physiology , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Epidermal Growth Factor/administration & dosage , Epidermal Growth Factor/metabolism , Epithelial Cells/cytology , Female , Growth Hormone/administration & dosage , Growth Hormone/metabolism , Mammary Glands, Animal/anatomy & histology , Mammary Glands, Animal/embryology , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Receptors, Prolactin/genetics , Receptors, Prolactin/metabolism , Receptors, Prolactin/physiology , STAT5 Transcription Factor , Sodium-Potassium-Chloride Symporters/metabolism , Solute Carrier Family 12, Member 2 , Trans-Activators/genetics , Trans-Activators/metabolism , Gap Junction beta-1 Protein
8.
Oncogene ; 20(48): 7115-9, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11704837

ABSTRACT

Loss of cell cycle regulation in mammary epithelium results in impaired mammary gland development and neoplasia. We investigated the consequences of the absence of pRb in mammary epithelial cells during normal development and in mice that express an oncogene in the mammary epithelium. Since pRb-deficiency results in embryonic lethality, we transplanted pRb-null mammary anlagen into wild hosts. pRb-deficient mammary epithelia were capable of functional differentiation in term animals and they regenerated a differentiated gland even after multiple pregnancies. In serial transplantations no significant differences were found in outgrowth of pRb-deficient and wild type epithelia indicating that the absence of pRb does not lead to transformation. Likewise the effect of a TGFbeta1 transgene was not altered in the absence of pRb. The susceptibility of mammary epithelium to form tumors was assessed in three different models. No differences in tumor incidence were found between wild type and Rb +/- WAP-int3, MMTV-PyMT transgenic and Brcal-/- epithelia. These results demonstrate that the absence of pRb does not affect normal mammary gland development and tumorigenesis in three different mouse models investigated and suggest that loss of more than one member of the pRb pathway is required to induce mammary tumors.


Subject(s)
Genes, Retinoblastoma , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Experimental/genetics , Receptors, Cell Surface , Retinoblastoma Protein/deficiency , Animals , Antigens, Polyomavirus Transforming/genetics , Cell Cycle/genetics , Cell Differentiation , Crosses, Genetic , Female , Genes, BRCA1 , Mammary Glands, Animal/embryology , Mammary Glands, Animal/transplantation , Mammary Neoplasms, Experimental/pathology , Mammary Tumor Virus, Mouse/genetics , Mammary Tumor Virus, Mouse/physiology , Mice , Mice, Knockout , Mice, Transgenic , Milk Proteins/genetics , Oncogenes , Pregnancy , Proto-Oncogene Proteins/genetics , Receptor, Notch4 , Receptors, Notch , Retinoblastoma Protein/physiology , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta1 , Transgenes
9.
Dev Cell ; 1(4): 467-75, 2001 Oct.
Article in English | MEDLINE | ID: mdl-11703938

ABSTRACT

Unlike most other organs, development of the mammary gland occurs predominantly after birth, under the control of steroid and peptide hormones. Once the gland is established, cycles of proliferation, functional differentiation, and death of alveolar epithelium occur repeatedly with each pregnancy. Although it is unique in this respect, the signaling pathways utilized by the gland are shared with other cell types, and have been tailored to meet the needs of this secretory tissue. Here we discuss the signaling pathways that have been adopted by the mammary gland for its own purposes, and the functions they perform.


Subject(s)
Breast/growth & development , Mammary Glands, Animal/growth & development , Signal Transduction/physiology , Animals , Breast/cytology , Female , Humans , Mammary Glands, Animal/cytology
10.
J Immunol ; 167(9): 5018-26, 2001 Nov 01.
Article in English | MEDLINE | ID: mdl-11673510

ABSTRACT

IL-5 stimulation of CD38-activated murine splenic B cells induces mu-gamma1 CSR at the DNA level leading to a high level of IgG1 production. Further addition of IL-4 in the system enhances IL-5-dependent mu-gamma1 CSR. Although some of the postreceptor signaling events initiated by IL-5 in activated B cells have been characterized, the involvement of Stat in IL-5 signaling has not been thoroughly evaluated. In this study, we examined the activation of Stat5 and activation-induced cytidine deaminase (AID) in CD38-activated murine splenic B cells by IL-5. The role of Stat5a and Stat5b in IL-5-induced mu-gamma1 CSR and also IgG1 and IgM production was documented, as IL-5 does not act on CD38-stimulated splenic B cells from Stat5a(-/-) and Stat5b(-/-) mice. Expression levels of CD38-induced germline gamma1 transcripts and AID in Stat5a(-/-) and Stat5b(-/-) B cells upon IL-5 stimulation were comparable to those of wild-type B cells. The impaired mu-gamma1 CSR by Stat5b(-/-) B cells, but not by Stat5a(-/-) B cells, was rescued in part by IL-4, as the addition of IL-4 to the culture of CD38- and IL-5-stimulated B cells induced mu-gamma1 CSR leading to IgG1 production. Analysis of cell division cycle number of wild-type B cells revealed that mu-gamma1 CSR was observed after five or six cell divisions. Stat5a(-/-) and Stat5b(-/-) B cells showed similar cell division cycles, but they did not undergo mu-gamma1 CSR. Our data support the notion that both Stat5a and Stat5b are essential for IL-5-dependent mu;-gamma1 CSR and Ig secretion; however, their major target may not be AID. Stat5a and Stat5b are not redundant, but rather are at least partially distinctive in their function.


Subject(s)
Antigens, CD , B-Lymphocytes/metabolism , DNA-Binding Proteins/physiology , Immunoglobulin Class Switching , Immunoglobulin G/biosynthesis , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin M/biosynthesis , Interleukin-5/pharmacology , Milk Proteins , Repressor Proteins , Trans-Activators/physiology , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Animals , Antigens, Differentiation/pharmacology , Cytidine Deaminase/metabolism , Immunoglobulin G/classification , Immunoglobulin G/genetics , Immunoglobulin M/genetics , Lymphocyte Activation , Membrane Glycoproteins , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , NAD+ Nucleosidase/pharmacology , Positive Regulatory Domain I-Binding Factor 1 , RNA, Messenger/analysis , Recombination, Genetic , STAT5 Transcription Factor , Transcription Factors/biosynthesis
11.
Mol Cell Endocrinol ; 183(1-2): 151-63, 2001 Oct 25.
Article in English | MEDLINE | ID: mdl-11604235

ABSTRACT

Milk production remains suppressed in mammals during late pregnancy despite high levels of lactogenic polypeptide hormones. At parturition, associated with a precipitous fall in circulating progesterone, rising glucocorticoid levels synergize with prolactin to initiate copious milk production. This synergy is mediated at least in part through the coordinated activation of glucocorticoid receptors and transcription factor Stat5, particularly Stat5a. Here we show that two proline-juxtaposed serine residues within the transactivation domain of Stat5a are phosphorylated in the mammary gland during late gestation and lactation, and that these phosphorylation sites inhibit the transcriptional activity of Stat5a in the absence of glucocorticoid receptor costimulation. Specifically, transfection assays revealed that phosphorylation of residues S725 and S779 of Stat5a cooperatively suppressed prolactin-stimulated transcription from the beta-casein promoter in both COS-7 kidney and MCF-7 mammary cells. This suppression was associated with shortened duration and reduced amplitude of nuclear DNA binding activity of wild type Stat5a relative to that of the serine phosphorylation-defective Stat5 mutant. However, costimulation of glucocorticoid receptors completely reversed the suppressive effect of Stat5a serine phosphorylation on beta-casein gene transcription. We propose that serine phosphorylation within the transactivation domain may limit the activity of Stat5a in the absence of proper coactivation by glucocorticoid receptors.


Subject(s)
Caseins/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Mammary Glands, Animal/metabolism , Milk Proteins , Phosphoserine/metabolism , Prolactin/pharmacology , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Caseins/metabolism , Cattle , Cell Line , Culture Media, Serum-Free , DNA-Binding Proteins/genetics , Female , Humans , Immunoblotting , Lactation/physiology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Phosphorylation , Pregnancy , Receptors, Glucocorticoid/metabolism , STAT5 Transcription Factor , Sequence Alignment , Trans-Activators/genetics , Tumor Cells, Cultured , Tumor Suppressor Proteins
12.
Trends Endocrinol Metab ; 12(9): 402-8, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11595542

ABSTRACT

Contemporary gene-targeting techniques now make it possible to alter specific genes in the genome. As a result, a plethora of mouse models have been generated that allow researchers to dissect cell-signaling pathways involved in mammary gland development and in breast cancer. But what have we learned so far? What do these models teach us? This review presents a global picture of how the analyses and comparison of individual knockout mouse models provide important insights into basic mammary gland biology. Particular emphasis is placed upon what is currently known about the signaling pathways involved in the establishment of the mammary ductal tree, and its subsequent proliferation at pregnancy and lactation. In addition to these well-established pathways, we address recent data that describe the role of lesser-known genes in the development of the mammary epithelium.


Subject(s)
Mammary Glands, Animal/physiology , Mice, Knockout , Animals , Epithelial Cells/physiology , Female , Lactation/physiology , Mice , Pregnancy , Signal Transduction/physiology
15.
Genomics ; 71(2): 150-5, 2001 Jan 15.
Article in English | MEDLINE | ID: mdl-11161808

ABSTRACT

Signal transducers and activators of transcription (Stat) are transcription factors that can be activated by many cytokines. While Drosophila contains only one Stat (d-Stat), mammals contain seven, with STATs 3, 5a, and 5b being the closest functional relatives. To understand the evolutionary relationship between d-Stat and vertebrate STATs 3 and 5, we isolated, sequenced, and analyzed the zebrafish Stat3 (z-Stat3) gene and a 500-kb region spanning mouse chromosome 11, 60.5 cM containing three Stat genes (m-Stats). Within this region we identified the genes encoding m-Stats 3, 5a, and 5b, Cnp1, Hcrt/Orexin, Ptrf, GCN5, mDj11, and four new genes. The 5' ends of the m-Stat5a and m-Stat5b genes are juxtaposed to each other, and the 3' ends of the m-Stat3 and Stat5a genes face each other. While the m-Stat5a and m-Stat3 genes have one promoter each, which are active in many tissues, the m-Stat5b gene acquired two distinct promoters. The distal promoter is expressed ubiquitously, and transcription from the proximal promoter is restricted to liver, muscle, and mammary tissue. Through a comparison of exon-intron boundaries from the m-Stat3, m-Stat5a, and m-Stat5b, z-Stat3, and d-Stat genes, we deduced their evolutionary relationship. We propose that the Stat3 and Stat5 lineages are derived from the duplication of a common primordial gene and that d-Stat is a part of the Stat5 lineage.


Subject(s)
DNA-Binding Proteins/genetics , Milk Proteins , Trans-Activators/genetics , Animals , Base Sequence , Conserved Sequence , Drosophila/genetics , Evolution, Molecular , Exons , Introns , Mice , Molecular Sequence Data , Multigene Family , Protein Structure, Tertiary , RNA, Messenger/metabolism , STAT3 Transcription Factor , STAT5 Transcription Factor , Sequence Analysis, DNA , Tissue Distribution , Zebrafish/genetics , Zebrafish Proteins
16.
Dev Biol ; 229(1): 163-75, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11133161

ABSTRACT

Prolactin (Prl)-induced phosphorylation of Stat (signal transducer and activator of transcription) 5 is considered a key event in functional mammary development and differentiation. We now demonstrate that not only Prl, but also growth hormone (GH) and epidermal growth factor (EGF), can activate Stat5 in mammary tissue. We investigated the roles of these hormones in mammary development using mice in which the respective receptors had been inactivated. Although Prl receptor (PrlR)-null mice are infertile, we were able to maintain pregnancies in a few mice by treatment with progesterone. Mammary tissue in these mice was severely underdeveloped and exhibited limited differentiation as assessed by the phosphorylation status of Stat5 and the expression of milk protein genes. PrlR +/- mice showed impaired mammary development and alveolar differentiation during pregnancy, which corresponded with reduced phosphorylation levels of Stat5a and 5b, and impaired expression of milk protein genes. Development of the glands in these mice was arrested at around day 13 of pregnancy. While Prl activated Stat5 only in the epithelium, GH and EGF activated Stat5 preferentially in the stroma. To assess the relevance of the GH receptor (GHR) in the mammary gland, we transplanted GHR-null epithelium into cleared fat pads of wild-type mice. These experiments demonstrated that the GHR in the epithelium is not required for functional mammary development. Similarly, the EGFR in the epithelium is not required for alveolar development. In contrast, epithelial PrlR is required for mammary development and milk protein gene expression during pregnancy. Although GH is not required for alveolar development, we were able to demonstrate its lactogenic function in cultured mammary epithelium from PrlR-null mice. However, ductal development in GHR-null mice was impaired, supporting the notion that GH signals through the stromal compartment. Our findings demonstrate that GH, Prl, and EGF activate Stat5 in separate compartments, which in turn reflects their specific roles in ductal and alveolar development and differentiation.


Subject(s)
DNA-Binding Proteins/metabolism , Epidermal Growth Factor/physiology , Growth Hormone/physiology , Lactation/physiology , Mammary Glands, Animal/growth & development , Prolactin/physiology , Trans-Activators/metabolism , Animals , Caseins/genetics , Epithelial Cells/physiology , Epithelial Cells/transplantation , ErbB Receptors/genetics , Female , Gene Expression Regulation , Mice , Mice, Mutant Strains , Milk Proteins/genetics , Receptors, Prolactin/genetics , STAT5 Transcription Factor , Stromal Cells/physiology
17.
Transgenic Res ; 10(6): 545-53, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11817542

ABSTRACT

Cre-loxP based gene deletion approaches hold great promise to enhance our understanding of molecular pathways controlling mammary development and breast cancer. We reported earlier the generation of transgenic mice that express the Cre recombinase under the control of the mouse mammary tumor virus (MMTV) long terminal repeat (LTR). These mice have become a valuable research tool to delete genes specifically in the mammary gland, other secretory organs, and the female germline. We have now characterized in depth the expression of the MMTV-Cre transgene using the ROSA26-lox-Stop-lox-LacZ reporter strain to determine the temporal and spatial activation of Cre on the level of single cells. Our results show that MMTV-mediated Cre-activation is restricted to specific cell types of various secretory tissues and the hematopoietic system. Secondly, the timing of Cre expression varies between tissues and cell types. Some tissues express Cre during embryonic development, while other selected cell types highly activate Cre around puberty, suggesting a strong influence of steroid hormones on the transcriptional activation of the MMTV-LTR. Thirdly, Cre expression in the female germline is restricted to individual mouse lines and is therefore dependent on the site of integration of the transgene. Information provided by this study will guide the researcher to those cell types and developmental stages at which a phenotype can be expected upon deletion of relevant genes.


Subject(s)
Integrases/biosynthesis , Integrases/genetics , Mammary Tumor Virus, Mouse/genetics , Terminal Repeat Sequences/genetics , Viral Proteins/biosynthesis , Viral Proteins/genetics , Animals , Female , Gene Deletion , Genes, Reporter , Male , Mice , Mice, Knockout , Mice, Transgenic , Oocytes/metabolism , Phenotype , Sex Factors , Time Factors , Transgenes
18.
Nucleic Acids Res ; 28(22): 4452-9, 2000 Nov 15.
Article in English | MEDLINE | ID: mdl-11071932

ABSTRACT

The Microarray Explorer (MAExplorer) is a versatile Java-based data mining bioinformatic tool for analyzing quantitative cDNA expression profiles across multiple microarray platforms and DNA labeling systems. It may be run as either a stand-alone application or as a Web browser applet over the Internet. With this program it is possible to (i) analyze the expression of individual genes, (ii) analyze the expression of gene families and clusters, (iii) compare expression patterns and (iv) directly access other genomic databases for clones of interest. Data may be downloaded as required from a Web server or in the case of the stand-alone version, reside on the user's computer. Analyses are performed in real-time and may be viewed and directly manipulated in images, reports, scatter plots, histograms, expression profile plots and cluster analyses plots. A key feature is the clone data filter for constraining a working set of clones to those passing a variety of user-specified logical and statistical tests. Reports may be generated with hypertext Web access to UniGene, GenBank and other Internet databases for sets of clones found to be of interest. Users may save their explorations on the Web server or local computer and later recall or share them with other scientists in this groupware Web environment. The emphasis on direct manipulation of clones and sets of clones in graphics and tables provides a high level of interaction with the data, making it easier for investigators to test ideas when looking for patterns. We have used the MAExplorer to profile gene expression patterns of 1500 duplicated genes isolated from mouse mammary tissue. We have identified genes that are preferentially expressed during pregnancy and during lactation. One gene we identified, carbonic anhydrase III, is highly expressed in mammary tissue from virgin and pregnant mice and in gene knock-out mice with underdeveloped mammary epithelium. Other genes, which include those encoding milk proteins, are preferentially expressed during lactation.


Subject(s)
DNA, Complementary/genetics , Mammary Glands, Animal/metabolism , Oligonucleotide Array Sequence Analysis , Animals , Blotting, Northern , Carbonic Anhydrases/genetics , Caseins/genetics , Female , Gene Expression Profiling , Gene Expression Regulation, Developmental , Mammary Glands, Animal/growth & development , Mice , Mice, Inbred C57BL , Mice, Knockout , Pregnancy , Procollagen/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism
19.
Development ; 127(22): 4949-58, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11044408

ABSTRACT

Bcl-x is a member of the Bcl2 family and has been suggested to be important for the survival and maturation of various cell types including the erythroid lineage. To define the consequences of Bcl-x loss in erythroid cells and other adult tissues, we have generated mice conditionally deficient in the Bcl-x gene using the Cre-loxP recombination system. The temporal and spatial excision of the floxed Bcl-x locus was achieved by expressing the Cre recombinase gene under control of the MMTV-LTR. By the age of five weeks, Bcl-x conditional mutant mice exhibited hyperproliferation of megakaryocytes and a decline in the number of circulating platelets. Three-month-old animals suffered from severe hemolytic anemia, hyperplasia of immature erythroid cells and profound enlargement of the spleen. We demonstrate that Bcl-x is only required for the survival of erythroid cells at the end of maturation, which includes enucleated reticulocytes in circulation. The extensive proliferation of immature erythroid cells in the spleen and bone marrow might be the result of a fast turnover of late red blood cell precursors and accelerated erythropoiesis in response to tissue hypoxia. The increase in cell death of late erythroid cells is independent from the proapoptotic factor Bax, as demonstrated in conditional double mutant mice for Bcl-x and Bax. Mice conditionally deficient in Bcl-x permitted us for the first time to study the effects of Bcl-x deficiency on cell proliferation, maturation and survival under physiological conditions in an adult animal.


Subject(s)
Anemia, Hemolytic/genetics , Erythrocytes/metabolism , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/physiology , Splenomegaly/genetics , Viral Proteins , Anemia, Hemolytic/pathology , Animals , Apoptosis , Base Sequence , Cell Differentiation , Cell Survival , DNA Primers/genetics , Erythroblasts/pathology , Gene Deletion , Integrases/genetics , Mammary Tumor Virus, Mouse/genetics , Megakaryocytes/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Proto-Oncogene Proteins c-bcl-2/deficiency , Reticulocytes/pathology , Spleen/pathology , Splenomegaly/pathology , Thrombocytopenia/genetics , bcl-X Protein
20.
Mol Endocrinol ; 14(7): 1038-52, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10894153

ABSTRACT

Restricted germ cell loss through apoptosis is initiated in the fetal gonad around embryonic day 13.5 (E13.5) as part of normal germ cell development. The mechanism of this germ cell attrition is unknown. We show that Bcl-x plays a crucial role in maintaining the survival of mouse germ cells during gonadogenesis. A bcl-x hypomorphic mouse was generated through the introduction of a neomycin (neo) gene into the promoter of the bcl-x gene by homologous recombination. Mice that contained two copies of the hypomorphic allele had severe reproductive defects attributed to compromised germ cell development. Males with two mutant alleles lacked spermatogonia and were sterile; females showed a severely reduced population of primordial and primary follicles and exhibited greatly impaired fertility. Primordial germ cells (PGCs) in bcl-x hypomorph mice migrated to the genital ridge by E12.5 but were depleted by E15.5, a time when Bcl-x and Bax were present. Two additional bcl-x transcripts were identified in fetal germ cells more than 300 bp upstream of previously reported start sites. Insertion of a neo cassette led to a down-regulation of the bcl-x gene at E12.5 in the hypomorph. Bax was detected by immunohistochemistry in germ cells from bcl-x hypomorph and control testes at E12.5 and E13.5. Bcl-x function was restored, and animals of both genders were fertile after removal of the neo selection cassette using Cre-mediated recombination. Alternatively, the loss of Bcl-x function in the hypomorph was corrected by the deletion of both copies of the bax gene, resulting in a restoration of germ cell survival. These findings demonstrate that the balance of Bcl-x and Bax control PGC survival and apoptosis.


Subject(s)
Embryo, Mammalian/cytology , Germ Cells/physiology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins/genetics , Animals , Base Sequence , Cell Survival/genetics , Embryonic and Fetal Development/genetics , Female , Gene Expression Regulation, Developmental , Male , Mice , Mice, Inbred Strains , Mice, Mutant Strains , Molecular Sequence Data , Oocytes/pathology , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Spermatogonia/pathology , Testis/embryology , Transcription, Genetic , bcl-2-Associated X Protein , bcl-X Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...