Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Proc Natl Acad Sci U S A ; 118(21)2021 05 25.
Article in English | MEDLINE | ID: mdl-34006643

ABSTRACT

HER2-positive (HER2+) breast cancers (BrCs) contain approximately equal numbers of ERα+HER2+ and ERα-HER2+ cases. An enduring obstacle is the unclear cell lineage-related characteristics of these BrCs. Although ERα+HER2+ BrCs could lose ERα to become ERα-HER2+ BrCs, direct evidence is missing. To investigate ERα dependencies and their implications during BrC growth and metastasis, we generated ERαCreRFP-T mice that produce an RFP-marked ERα+ mammary gland epithelial cell (MGEC) lineage. RCAS virus-mediated expression of Erbb2, a rodent Her2 homolog, first produced comparable numbers of ERα+RFP+Erbb2+ and ERα-RFP-Erbb2+ MGECs. Early hyperplasia developed mostly from ERα+RFP+Erbb2+ cells and ERα-RFP-Erbb2+ cells in these lesions were rare. The subsequently developed ductal carcinomas in situ had 64% slow-proliferating ERα+RFP+Erbb2+ cells, 15% fast-proliferating ERα-RFP+Erbb2+ cells derived from ERα+RFP+Erbb2+ cells, and 20% fast-proliferating ERα-RFP-Erbb2+ cells. The advanced tumors had mostly ERα-RFP+Erbb2+ and ERα-RFP-Erbb2+ cells and only a very small population of ERα+RFP+Erbb2+ cells. In ERα-RFP+Erbb2+ cells, GATA3 and FoxA1 decreased expression and ERα promoter regions became methylated, consistent with the loss of ERα expression. Lung metastases consisted of mostly ERα-RFP+Erbb2+ cells, a few ERα-RFP-Erbb2+ cells, and no ERα+RFP+Erbb2+ cells. The high metastatic capacity of ERα-RFP+Erbb2+ cells was associated with ERK1/2 activation. These results show that the slow-proliferating, nonmetastatic ERα+RFP+Erbb2+ cells progressively lose ERα during tumorigenesis to become fast-proliferating, highly metastatic ERα-RFP+Erbb2+ cells. The ERα-Erbb2+ BrCs with an ERα+ origin are more aggressive than those ERα-Erbb2+ BrCs with an ERα- origin, and thus, they should be distinguished and treated differently in the future.


Subject(s)
Breast Neoplasms/genetics , Carcinoma, Intraductal, Noninfiltrating/genetics , Estrogen Receptor alpha/genetics , Gene Expression Regulation, Neoplastic , Lung Neoplasms/genetics , Receptor, ErbB-2/genetics , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Carcinoma, Intraductal, Noninfiltrating/metabolism , Carcinoma, Intraductal, Noninfiltrating/secondary , Cell Line, Tumor , Cell Lineage/genetics , Cell Lineage/immunology , Cell Proliferation , Cell Transformation, Neoplastic , Estrogen Receptor alpha/metabolism , Female , GATA3 Transcription Factor/genetics , GATA3 Transcription Factor/metabolism , Hepatocyte Nuclear Factor 3-alpha/genetics , Hepatocyte Nuclear Factor 3-alpha/metabolism , Humans , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mitogen-Activated Protein Kinase 1/genetics , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Neoplasm Invasiveness , Promoter Regions, Genetic , Receptor, ErbB-2/metabolism , Signal Transduction , Tumor Burden , Xenograft Model Antitumor Assays
2.
Nature ; 550(7677): 529-533, 2017 10 26.
Article in English | MEDLINE | ID: mdl-29019984

ABSTRACT

In several organ systems, the transitional zone between different types of epithelium is a hotspot for pre-neoplastic metaplasia and malignancy, but the cells of origin for these metaplastic epithelia and subsequent malignancies remain unknown. In the case of Barrett's oesophagus, intestinal metaplasia occurs at the gastro-oesophageal junction, where stratified squamous epithelium transitions into simple columnar cells. On the basis of a number of experimental models, several alternative cell types have been proposed as the source of this metaplasia but in all cases the evidence is inconclusive: no model completely mimics Barrett's oesophagus in terms of the presence of intestinal goblet cells. Here we describe a transitional columnar epithelium with distinct basal progenitor cells (p63+KRT5+KRT7+) at the squamous-columnar junction of the upper gastrointestinal tract in a mouse model. We use multiple models and lineage tracing strategies to show that this squamous-columnar junction basal cell population serves as a source of progenitors for the transitional epithelium. On ectopic expression of CDX2, these transitional basal progenitors differentiate into intestinal-like epithelium (including goblet cells) and thereby reproduce Barrett's metaplasia. A similar transitional columnar epithelium is present at the transitional zones of other mouse tissues (including the anorectal junction) as well as in the gastro-oesophageal junction in the human gut. Acid reflux-induced oesophagitis and the multilayered epithelium (believed to be a precursor of Barrett's oesophagus) are both characterized by the expansion of the transitional basal progenitor cells. Our findings reveal a previously unidentified transitional zone in the epithelium of the upper gastrointestinal tract and provide evidence that the p63+KRT5+KRT7+ basal cells in this zone are the cells of origin for multi-layered epithelium and Barrett's oesophagus.


Subject(s)
Barrett Esophagus/pathology , Cell Lineage , Epithelial Cells/pathology , Epithelium/pathology , Esophagogastric Junction/pathology , Stem Cells/pathology , Animals , Barrett Esophagus/genetics , Barrett Esophagus/metabolism , CDX2 Transcription Factor/genetics , CDX2 Transcription Factor/metabolism , Cell Tracking , Esophagitis/metabolism , Esophagitis/pathology , Esophagogastric Junction/metabolism , Gastroesophageal Reflux , Goblet Cells/metabolism , Goblet Cells/pathology , Humans , Keratin-5/metabolism , Keratin-7/metabolism , Metaplasia/metabolism , Metaplasia/pathology , Mice , Phosphoproteins/metabolism , Stem Cells/metabolism , Trans-Activators/metabolism
3.
Proc Natl Acad Sci U S A ; 114(43): 11494-11499, 2017 10 24.
Article in English | MEDLINE | ID: mdl-29073077

ABSTRACT

Twist1 is an epithelial-mesenchymal transition (EMT)-inducing transcription factor (TF) that promotes cell migration and invasion. To determine the intrinsic role of Twist1 in EMT and breast cancer initiation, growth, and metastasis, we developed mouse models with an oncogene-induced mammary tumor containing wild-type (WT) Twist1 or tumor cell-specific Twist1 knockout (Twist1TKO). Twist1 knockout showed no effects on tumor initiation and growth. In both models with early-stage tumor cells, Twist1, and mesenchymal markers were not expressed, and lung metastasis was absent. Twist1 expression was detected in ∼6% of the advanced WT tumor cells. Most of these Twist1+ cells coexpressed several other EMT-inducing TFs (Snail, Slug, Zeb2), lost ERα and luminal marker K8, acquired basal cell markers (K5, p63), and exhibited a partial EMT plasticity (E-cadherin+/vimentin+). In advanced Twist1TKO tumor cells, Twist1 knockout largely diminished the expression of the aforementioned EMT-inducing TFs and basal and mesenchymal markers, but maintained the expression of the luminal markers. Circulating tumor cells (CTCs) were commonly detected in mice with advanced WT tumors, but not in mice with advanced Twist1TKO tumors. Nearly all WT CTCs coexpressed Twist1 with other EMT-inducing TFs and both epithelial and mesenchymal markers. Mice with advanced WT tumors developed extensive lung metastasis consisting of luminal tumor cells with silenced Twist1 and mesenchymal marker expression. Mice with advanced Twist1TKO tumors developed very little lung metastasis. Therefore, Twist1 is required for the expression of other EMT-inducing TFs in a small subset of tumor cells. Together, they induce partial EMT, basal-like tumor progression, intravasation, and metastasis.


Subject(s)
Cell Plasticity , Lung Neoplasms/secondary , Mammary Neoplasms, Animal/metabolism , Nuclear Proteins/metabolism , Twist-Related Protein 1/metabolism , Animals , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Female , Gene Expression Regulation, Neoplastic , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Knockout , Neoplasm Invasiveness , Nuclear Proteins/genetics , Twist-Related Protein 1/genetics
4.
Sci Adv ; 10(26): eadl2675, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38941473

ABSTRACT

Declined memory is a hallmark of Alzheimer's disease (AD). Experiments in rodents and human postmortem studies suggest that serotonin (5-hydroxytryptamine, 5-HT) plays a role in memory, but the underlying mechanisms are unknown. Here, we investigate the role of 5-HT 2C receptor (5-HT2CR) in regulating memory. Transgenic mice expressing a humanized HTR2C mutation exhibit impaired plasticity of hippocampal ventral CA1 (vCA1) neurons and reduced memory. Further, 5-HT neurons project to and synapse onto vCA1 neurons. Disruption of 5-HT synthesis in vCA1-projecting neurons or deletion of 5-HT2CRs in the vCA1 impairs neural plasticity and memory. We show that a selective 5-HT2CR agonist, lorcaserin, improves synaptic plasticity and memory in an AD mouse model. Cumulatively, we demonstrate that hippocampal 5-HT2CR signaling regulates memory, which may inform the use of 5-HT2CR agonists in the treatment of dementia.


Subject(s)
Alzheimer Disease , Memory , Mice, Transgenic , Neuronal Plasticity , Receptor, Serotonin, 5-HT2C , Animals , Humans , Receptor, Serotonin, 5-HT2C/metabolism , Receptor, Serotonin, 5-HT2C/genetics , Memory/drug effects , Memory/physiology , Mice , Neuronal Plasticity/drug effects , Alzheimer Disease/metabolism , Hippocampus/metabolism , Hippocampus/drug effects , Serotonin/metabolism , Disease Models, Animal , CA1 Region, Hippocampal/metabolism , CA1 Region, Hippocampal/drug effects , Neurons/metabolism , Neurons/drug effects , Serotonin 5-HT2 Receptor Agonists/pharmacology
5.
Proc Natl Acad Sci U S A ; 106(21): 8513-8, 2009 May 26.
Article in English | MEDLINE | ID: mdl-19433796

ABSTRACT

ASC-2, a multifunctional coactivator, forms a steady-state complex, named ASCOM (for ASC-2 COMplex), that contains the histone H3-lysine-4 (H3K4)-methyltransferase MLL3 or its paralogue MLL4. Somewhat surprisingly, given prior indications of redundancy between MLL3 and MLL4, targeted inactivation of the MLL3 H3K4-methylation activity in mice is found to result in ureter epithelial tumors. Interestingly, this phenotype is exacerbated in a p53(+/-) background and the tumorigenic cells are heavily immunostained for gammaH2AX, indicating a contribution of MLL3 to the DNA damage response pathway through p53. Consistent with the in vivo observations, and the demonstration of a direct interaction between p53 and ASCOM, cell-based assays have revealed that ASCOM, through ASC-2 and MLL3/4, acts as a p53 coactivator and is required for H3K4-trimethyation and expression of endogenous p53-target genes in response to the DNA damaging agent doxorubicin. In support of redundant functions for MLL3 and MLL4 for some events, siRNA-mediated down-regulation of both MLL3 and MLL4 is required to suppress doxorubicin-inducible expression of several p53-target genes. Importantly, this study identifies a specific H3K4 methytransferase complex, ASCOM, as a physiologically relevant coactivator for p53 and implicates ASCOM in the p53 tumor suppression pathway in vivo.


Subject(s)
Histone-Lysine N-Methyltransferase/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Transformation, Neoplastic , DNA Damage/genetics , DNA Repair/genetics , Enzyme Activation , Histone-Lysine N-Methyltransferase/genetics , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Transgenic , Nuclear Receptor Coactivators , Tumor Suppressor Protein p53/genetics , Ureteral Neoplasms/enzymology , Ureteral Neoplasms/genetics , Ureteral Neoplasms/pathology
6.
Nat Med ; 28(12): 2537-2546, 2022 12.
Article in English | MEDLINE | ID: mdl-36536256

ABSTRACT

Serotonin reuptake inhibitors and receptor agonists are used to treat obesity, anxiety and depression. Here we studied the role of the serotonin 2C receptor (5-HT2CR) in weight regulation and behavior. Using exome sequencing of 2,548 people with severe obesity and 1,117 control individuals without obesity, we identified 13 rare variants in the gene encoding 5-HT2CR (HTR2C) in 19 unrelated people (3 males and 16 females). Eleven variants caused a loss of function in HEK293 cells. All people who carried variants had hyperphagia and some degree of maladaptive behavior. Knock-in male mice harboring a human loss-of-function HTR2C variant developed obesity and reduced social exploratory behavior; female mice heterozygous for the same variant showed similar deficits with reduced severity. Using the 5-HT2CR agonist lorcaserin, we found that depolarization of appetite-suppressing proopiomelanocortin neurons was impaired in knock-in mice. In conclusion, we demonstrate that 5-HT2CR is involved in the regulation of human appetite, weight and behavior. Our findings suggest that melanocortin receptor agonists might be effective in treating severe obesity in individuals carrying HTR2C variants. We suggest that HTR2C should be included in diagnostic gene panels for severe childhood-onset obesity.


Subject(s)
Obesity, Morbid , Receptor, Serotonin, 5-HT2C , Animals , Child , Female , Humans , Male , Mice , HEK293 Cells , Obesity/genetics , Receptor, Serotonin, 5-HT2C/genetics , Serotonin , Serotonin 5-HT2 Receptor Agonists/pharmacology , Adaptation, Psychological
7.
Biol Reprod ; 82(4): 706-13, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20018910

ABSTRACT

Normal endometrial function requires a balance of progesterone (P4) and estrogen (E2) effects. E2 acts to stimulate the proliferation of uterine epithelial cells, while P4 action inhibits E2-mediated proliferation of the epithelium. P4 through its cognate receptor, the P4 receptor (Pgr), has important roles in the establishment and maintenance of pregnancy. In previous studies, we have identified ERBB receptor feedback inhibitor 1 (Errfi1) as a downstream target of Pgr action in the uterus. Herein, we show that Errfi1 mRNA expression was significantly increased in the uterus after Day 2.5 of gestation. Its expression is also induced in the uterus by acute E2 treatment, and this induction is synergistically induced by chronic E2 and P4 treatment. Although it is known that conditional ablation of Errfi1 in the Pgr-positive cells (Errfi1(d/d)) results in infertility, the function of Errfi1 in reproductive biology has remained elusive. Using Errfi1(d/d) mice, we have identified Errfi1 as an important mediator of uterine implantation. Epithelial ESR1 and target genes were significantly increased in the uteri of Errfi1(d/d) mice. Our results identify a new signaling paradigm of steroid hormone regulation in female reproductive biology that adds insight into the underlying dysregulation of hormonal signaling in human reproductive disorders such as endometriosis and endometrial cancer.


Subject(s)
Embryo Implantation/genetics , Estrogen Receptor alpha/physiology , Intracellular Signaling Peptides and Proteins/physiology , Animals , Embryo Implantation/drug effects , Embryo Implantation/physiology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Female , Gene Expression Regulation/drug effects , Gestational Age , Gonadal Steroid Hormones/pharmacology , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Uterus/drug effects , Uterus/metabolism , Uterus/physiology
8.
PLoS Genet ; 3(6): e102, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17590085

ABSTRACT

Progesterone and estrogen are critical regulators of uterine receptivity. To facilitate uterine remodeling for embryo attachment, estrogen activity in the uterine epithelia is attenuated by progesterone; however, the molecular mechanism by which this occurs is poorly defined. COUP-TFII (chicken ovalbumin upstream promoter transcription factor II; also known as NR2F2), a member of the nuclear receptor superfamily, is highly expressed in the uterine stroma and its expression is regulated by the progesterone-Indian hedgehog-Patched signaling axis that emanates from the epithelium. To further assess COUP-TFII uterine function, a conditional COUP-TFII knockout mouse was generated. This mutant mouse is infertile due to implantation failure, in which both embryo attachment and uterine decidualization are impaired. Using this animal model, we have identified a novel genetic pathway in which BMP2 lies downstream of COUP-TFII. Epithelial progesterone-induced Indian hedgehog regulates stromal COUP-TFII, which in turn controls BMP2 to allow decidualization to manifest in vivo. Interestingly, enhanced epithelial estrogen activity, which impedes maturation of the receptive uterus, was clearly observed in the absence of stromal-derived COUP-TFII. This finding is consistent with the notion that progesterone exerts its control of implantation through uterine epithelial-stromal cross-talk and reveals that stromal-derived COUP-TFII is an essential mediator of this complex cross-communication pathway. This finding also provides a new signaling paradigm for steroid hormone regulation in female reproductive biology, with attendant implications for furthering our understanding of the molecular mechanisms that underlie dysregulation of hormonal signaling in such human reproductive disorders as endometriosis and endometrial cancer.


Subject(s)
COUP Transcription Factor II/physiology , Embryo Implantation/physiology , Progesterone/physiology , Receptors, Estrogen/antagonists & inhibitors , Uterus/metabolism , Animals , COUP Transcription Factor II/deficiency , COUP Transcription Factor II/genetics , Cell Communication/genetics , Epithelium/metabolism , Epithelium/physiology , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Pregnancy , Receptor Cross-Talk/physiology , Receptors, Estrogen/metabolism , Stromal Cells/metabolism , Stromal Cells/physiology
9.
Nat Commun ; 11(1): 997, 2020 02 21.
Article in English | MEDLINE | ID: mdl-32081850

ABSTRACT

Prostate development depends on balanced cell proliferation and differentiation, and acetylated KLF5 is known to alter epithelial proliferation. It remains elusive whether post-translational modifications of transcription factors can differentially determine adult stem/progenitor cell fate. Here we report that, in human and mouse prostates, Klf5 is expressed in both basal and luminal cells, with basal cells preferentially expressing acetylated Klf5. Functionally, Klf5 is indispensable for maintaining basal progenitors, their luminal differentiation, and the proliferation of their basal and luminal progenies. Acetylated Klf5 is also essential for basal progenitors' maintenance and proper luminal differentiation, as deacetylation of Klf5 causes excess basal-to-luminal differentiation; attenuates androgen-mediated organoid organization; and retards postnatal prostate development. In basal progenitor-derived luminal cells, Klf5 deacetylation increases their proliferation and attenuates their survival and regeneration following castration and subsequent androgen restoration. Mechanistically, Klf5 deacetylation activates Notch signaling. Klf5 and its acetylation thus contribute to postnatal prostate development and regeneration by controlling basal progenitor cell fate.


Subject(s)
Kruppel-Like Transcription Factors/metabolism , Prostate/growth & development , Prostate/metabolism , Acetylation , Androgens/metabolism , Animals , Cell Differentiation , Cell Proliferation , Humans , Kruppel-Like Transcription Factors/deficiency , Kruppel-Like Transcription Factors/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Orchiectomy , Organoids/cytology , Organoids/metabolism , Prostate/cytology , Regeneration , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism
10.
Dev Cell ; 44(6): 752-761.e4, 2018 03 26.
Article in English | MEDLINE | ID: mdl-29587145

ABSTRACT

Basal cells (BCs) are p63-expressing multipotent progenitors of skin, tracheoesophageal and urinary tracts. p63 is abundant in developing airways; however, it remains largely unclear how embryonic p63+ cells contribute to the developing and postnatal respiratory tract epithelium, and ultimately how they relate to adult BCs. Using lineage-tracing and functional approaches in vivo, we show that p63+ cells arising from the lung primordium are initially multipotent progenitors of airway and alveolar lineages but later become restricted proximally to generate the tracheal adult stem cell pool. In intrapulmonary airways, these cells are maintained immature to adulthood in bronchi, establishing a rare p63+Krt5- progenitor cell population that responds to H1N1 virus-induced severe injury. Intriguingly, this pool includes a CC10 lineage-labeled p63+Krt5- cell subpopulation required for a full H1N1-response. These data elucidate key aspects in the establishment of regionally distinct adult stem cell pools in the respiratory system, potentially with relevance to other organs.


Subject(s)
Cell Lineage , Embryonic Stem Cells/cytology , Influenza A Virus, H1N1 Subtype/physiology , Lung/cytology , Phosphoproteins/physiology , Respiratory Mucosa/cytology , Stem Cells/cytology , Trans-Activators/physiology , Animals , Cell Differentiation , Cells, Cultured , Embryonic Stem Cells/metabolism , Embryonic Stem Cells/virology , Female , Lung/metabolism , Lung/virology , Male , Mice , Mice, Knockout , Respiratory Mucosa/metabolism , Respiratory Mucosa/virology , Stem Cells/metabolism , Stem Cells/virology , Trachea/cytology , Trachea/metabolism , Trachea/virology
11.
Nat Cell Biol ; 19(8): 904-914, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28737769

ABSTRACT

After influenza infection, lineage-negative epithelial progenitors (LNEPs) exhibit a binary response to reconstitute epithelial barriers: activating a Notch-dependent ΔNp63/cytokeratin 5 (Krt5) remodelling program or differentiating into alveolar type II cells (AEC2s). Here we show that local lung hypoxia, through hypoxia-inducible factor (HIF1α), drives Notch signalling and Krt5pos basal-like cell expansion. Single-cell transcriptional profiling of human AEC2s from fibrotic lungs revealed a hypoxic subpopulation with activated Notch, suppressed surfactant protein C (SPC), and transdifferentiation toward a Krt5pos basal-like state. Activated murine Krt5pos LNEPs and diseased human AEC2s upregulate strikingly similar core pathways underlying migration and squamous metaplasia. While robust, HIF1α-driven metaplasia is ultimately inferior to AEC2 reconstitution in restoring normal lung function. HIF1α deletion or enhanced Wnt/ß-catenin activity in Sox2pos LNEPs blocks Notch and Krt5 activation, instead promoting rapid AEC2 differentiation and migration and improving the quality of alveolar repair.


Subject(s)
Cell Lineage , Cell Proliferation , Cell Transdifferentiation , Epithelial Cells/metabolism , Hypoxia/metabolism , Influenza, Human/metabolism , Orthomyxoviridae Infections/metabolism , Oxygen/metabolism , Pulmonary Alveoli/metabolism , Regeneration , Animals , Cell Movement , Cells, Cultured , Disease Models, Animal , Epithelial Cells/pathology , Epithelial Cells/virology , Female , Gene Expression Profiling , Genotype , Humans , Hypoxia/genetics , Hypoxia/pathology , Hypoxia/virology , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Influenza A Virus, H1N1 Subtype/pathogenicity , Influenza, Human/genetics , Influenza, Human/pathology , Influenza, Human/virology , Keratin-5/genetics , Keratin-5/metabolism , Male , Mice, Transgenic , Orthomyxoviridae Infections/genetics , Orthomyxoviridae Infections/pathology , Orthomyxoviridae Infections/virology , Phenotype , Phosphoproteins/genetics , Phosphoproteins/metabolism , Pulmonary Alveoli/pathology , Pulmonary Alveoli/virology , Receptors, Notch/metabolism , SOXB1 Transcription Factors/genetics , SOXB1 Transcription Factors/metabolism , Single-Cell Analysis , Time Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism , Wnt Signaling Pathway
12.
Dev Cell ; 43(4): 436-448.e6, 2017 11 20.
Article in English | MEDLINE | ID: mdl-29103953

ABSTRACT

Puberty is characterized by dynamic tissue remodeling in the mammary gland involving ductal elongation, resolution into the mature epithelial bilayer, and lumen formation. To decipher the cellular mechanisms underlying these processes, we studied the fate of putative stem cells, termed cap cells, present in terminal end buds of pubertal mice. Employing a p63CreERT2-based lineage-tracing strategy, we identified a unipotent fate for proliferative cap cells that only generated cells with basal features. Furthermore, we observed that dislocated "cap-in-body" cells underwent apoptosis, which aided lumen formation during ductal development. Basal lineage-specific profiling and genetic loss-of-function experiments revealed a critical role for FOXO transcription factors in mediating these proliferative versus apoptotic fates. Importantly, these studies revealed a mode of WNT signaling-mediated FOXO1 inhibition, potentially mediated through AKT. Together, these data suggest that the WNT pathway confers proliferative and survival advantages on cap cells via regulation of FOXO1 localization.


Subject(s)
Apoptosis/physiology , Breast/metabolism , Forkhead Box Protein O1/metabolism , Homeostasis/physiology , Stem Cells/cytology , Wnt Signaling Pathway/physiology , Animals , Breast/cytology , Cell Lineage/physiology , Cell Proliferation/physiology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mice, Transgenic
13.
Mol Endocrinol ; 19(2): 399-408, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15528275

ABSTRACT

Insulin-induced gene-1 (Insig-1) and its homolog Insig-2 encode closely related proteins of the endoplasmic reticulum that block proteolytic activation of sterol regulatory element binding proteins, membrane-bound transcription factors that activate synthesis of cholesterol and fatty acids in animal cells. These proteins also restrict lipogenesis in mature adipocytes and block differentiation of preadipocytes. Herein, we identified a novel 1alpha,25-dihydroxyvitamin D3 [1,25-(OH)2D3] response element in the promoter region of Insig-2 gene, which specifically binds to the heterodimer of retinoid X receptor and vitamin D receptor (VDR) and directs VDR-mediated transcriptional activation in a 1,25-(OH)2D3-dependent manner. Interestingly, 1,25-(OH)2D3 is known to directly suppress the expression of peroxisome proliferator-activated receptor gamma2 protein and inhibits adipocyte differentiation of 3T3-L1 preadipocytes and murine bone marrow stromal cells. Consistent with an idea that the antiadipogenic action of 1,25-(OH)2D3 may also involve up-regulation of Insig-2, we found that 1,25-(OH)2D3 transiently but strongly induces Insig-2 expression in 3T3-L1 cells. This novel regulatory circuit may also play important roles in other lipogenic cell types that express VDR, and collectively our results suggest an intriguing, new linkage between 1,25-(OH)2D3 and lipogenesis.


Subject(s)
Adipocytes/cytology , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/genetics , Promoter Regions, Genetic , Response Elements , Vitamin D/metabolism , 3T3-L1 Cells , Adipocytes/metabolism , Animals , Base Sequence , Bone Marrow Cells/cytology , Calcitriol/metabolism , Cell Differentiation , DNA Primers/chemistry , Dimerization , Dose-Response Relationship, Drug , Ligands , Mice , Models, Biological , Molecular Sequence Data , PPAR gamma/metabolism , Plasmids/metabolism , Protein Binding , Receptors, Calcitriol/metabolism , Retinoid X Receptors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation , Transfection
14.
Int J Biol Sci ; 10(9): 1007-17, 2014.
Article in English | MEDLINE | ID: mdl-25210499

ABSTRACT

The prostate epithelium is composed of basal (BC), luminal (LEC), and neuroendocrine (NEC) cells. It is unclear how many subtypes of BCs in the prostate and which subtype of BCs contains the main stem cell niche in the adult prostate. Here we report seven BC subpopulations according to their p63, cytokeratin 14 (K14) and K5 expression patterns, including p63-positive/K14-negative/K5-negative (p63+/K14-/K5-), p63-/K14+/K5-, p63-/K14-/K5+, p63+/K14+/K5-, p63+/K14-/K5+, p63-/K14+/K5+, and p63+/K14+/K5+ BCs. We generated a p63-CreERT2 knock-in mouse line that expresses tamoxifen-inducible Cre activity in the p63-expressing cells, including the prostate BCs. We then crossbred this line with ROSA26R mice, and generated p63-CreERT2×ROSA26R bi-genic mice harboring the Cre-activated ß-galactosidase reporter gene. We treated these bi-genic mice with tamoxifen to mark the p63+ BCs at different ages or under different hormonal conditions, and then traced the lineage differentiation of these genetically labeled BCs. We discovered that these p63+ BCs contain self-renewable stem cells in culture and efficiently differentiated into LECs, NECs and BCs in the postnatal, adult and re-generating mouse prostates. Therefore, BC population contains heterogeneous BCs that express different combinations of the p63, K14 and K5 differentiation markers. Because K14+ and K5+ BCs were previously shown to be extremely inefficient to produce LECs in adulthood, we propose that the p63+/K5-/K14- subpopulation of BCs contains most stem-like cells, especially in adult animals.


Subject(s)
Epithelial Cells/classification , Gene Expression Regulation, Neoplastic/physiology , Phosphoproteins/metabolism , Prostatic Neoplasms/pathology , Trans-Activators/metabolism , Androgens/pharmacology , Animals , Cell Differentiation , Epithelial Cells/cytology , Epithelium/physiology , Estrogen Antagonists/pharmacology , Homeostasis , Male , Mice , Multipotent Stem Cells/classification , Multipotent Stem Cells/cytology , Multipotent Stem Cells/physiology , Phosphoproteins/genetics , Tamoxifen/pharmacology , Trans-Activators/genetics
16.
Int J Biol Sci ; 10(10): 1116-27, 2014.
Article in English | MEDLINE | ID: mdl-25332686

ABSTRACT

Neuroendocrine tumor cells (NETCs) are commonly observed in prostate cancer. Their presence is associated with castration resistance, metastasis and poor prognosis. Cellular and molecular mechanisms for NETC initiation and growth are unknown. TRAMP mice develop heterogeneous adenocarcinomas induced by expression of the SV40-T/t oncogene in prostate epithelial cells. Here, we demonstrate prostate tumors in TRAMP mice with a mixed genetic background are characterized mostly by atypical hyperplasia (AH) containing steroid receptor coactiator-3-positive, androgen receptor-positive and synaptophysin-negative (SRC-3+/AR+/Syp-) cells. Few SRC-3+/AR-/Syp+ NETCs are present in their prostates. We generated TRAMP mice in which SRC-3 was specifically ablated in AR+/Syp- prostatic epithelial cells (termed PE3KOT mice). In these animals, we observed a substantial reduction in SRC-3-/AR+/Syp- AH tumor growth. There was a corresponding increase in SRC-3-/AR+/Syp- phyllodes lesions, suggesting SRC-3 knockout can convert aggressive AH tumors with mostly epithelial tumor cells into less aggressive phyllodes lesions with mostly stromal tissue. Surprisingly, PE3KOT mice developed many more SRC-3+/AR-/Syp+ NETCs versus control TRAMP mice, indicating SRC-3 expression was retained in NETCs. In contrast, TRAMP mice with global SRC-3 knockout did not develop any NETC, indicating SRC-3 is required for developing NETC. Analysis of cell-differentiating markers revealed that these NETCs might not be derived from the mature AR-/Syp+ neuroendocrine cells or the AR+/Syp- luminal epithelial tumor cells. Instead, these NETCs might originate from the SV40-T/t-transformed intermediate/progenitor epithelial cells. In summary, SRC-3 is required for both AR+/Syp- AH tumor growth and AR-/Syp+ NETC development, suggesting SRC-3 is a target for inhibiting aggressive prostate cancer containing NETCs.


Subject(s)
Neuroendocrine Tumors/metabolism , Nuclear Receptor Coactivator 3/metabolism , Prostatic Neoplasms/metabolism , Animals , Gene Knockout Techniques , Histological Techniques , Immunohistochemistry , Male , Mice , Nuclear Receptor Coactivator 3/genetics , Synaptophysin/metabolism
17.
J Clin Endocrinol Metab ; 99(3): E427-37, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24423359

ABSTRACT

CONTEXT: Endometriosis is one of the most common gynecological diseases in women with a prevalence rate of approximately 10%. Chronic pelvic inflammation has been observed in patients with endometriosis and is associated with disease severity. However, how pelvic inflammation promotes endometriosis progression remains unknown. OBJECTIVE: The objective of the study was to investigate the regulatory network of proinflammatory cytokines in endometriosis progression. DESIGN, SETTINGS, AND PATIENTS: Immunostaining of human endometrial (n = 21) and endometriotic (n = 36) sections, quantitative RT-PCR, Western blotting, chromatin immunoprecipitation, and luciferase reporter assays in primary culture human endometrial stromal cells were performed. Autologous transplantation of uterine endometrium from control chicken ovalbumin upstream promoter-transcription factor II [(COUP-TFII) flox/flox] and uterus-specific COUP-TFII knockout mice was performed. RESULTS: Expression of COUP-TFII was significantly reduced in endometriotic stroma. Reduction of COUP-TFII in endometriotic stromal cells was mediated by proinflammatory cytokines including IL-1ß, TNF-α, and TGF-ß1 via a common effector, microRNA-302a. Treatment with these proinflammatory cytokines increased the expression of microRNA-302a, which targets the 3'untranslated region of COUP-TFII to cause its down-regulation. Intriguingly, down-regulation of COUP-TFII in endometrial stromal cells resulted in de-repression of cyclooxygenase-2 (COX-2). Further investigation demonstrated that COUP-TFII directly binds to COX-2 promoter to inhibit its transcription. Forced expression of COUP-TFII inhibited IL-1ß-induced COX-2 up-regulation, whereas the knockdown of COUP-TFII augmented this effect. CONCLUSION: Because overexpression of COX-2 has been demonstrated to be a master regulator in endometriosis progression, our data demonstrate the critical function of proinflammatory cytokines and the COUP-TFII regulatory gene network in the progression of endometriosis.


Subject(s)
COUP Transcription Factor II/genetics , Cytokines/physiology , Endometriosis/genetics , Inflammation Mediators/physiology , Uterine Diseases/genetics , Animals , COUP Transcription Factor II/metabolism , Cells, Cultured , Cytokines/pharmacology , Down-Regulation/drug effects , Down-Regulation/genetics , Endometriosis/metabolism , Endometrium/metabolism , Endometrium/pathology , Female , Gene Regulatory Networks/drug effects , Humans , Inflammation Mediators/pharmacology , Mice , Mice, Knockout , Stromal Cells/metabolism , Stromal Cells/pathology , Uterine Diseases/metabolism
18.
Cancer Res ; 73(16): 5090-9, 2013 Aug 15.
Article in English | MEDLINE | ID: mdl-23811943

ABSTRACT

Endometrial cancer is preceded by endometrial hyperplasia, unopposed estrogen exposure, and genetic alterations, but the precise causes of endometrial cancer remain uncertain. Mig-6, mainly known as a negative regulator of the EGF receptor, is an important mediator of progesterone signaling in the uterus, where it mediates tumor suppression by modulating endometrial stromal-epithelial communications. In this study, we investigated the function of Mig-6 in the uterine epithelium using a tissue-specific gene knockout strategy, in which floxed Mig-6 (Mig-6(f/f)) mice were crossed to Wnt7a-Cre mice (Wnt7a(cre+)Mig-6(f/f)). Wnt7a(cre+)Mig-6(f/f) mice developed endometrial hyperplasia and estrogen-dependent endometrial cancer, exhibiting increased proliferation in epithelial cells as well as apoptosis in subepithelial stromal cells. We documented increased expression of NOTCH1 and BIRC3 in epithelial cells of Wnt7a(cre+)Mig-6(f/f) mice and decreased expression of the progesterone receptor (PR) in stromal cells. Progesterone therapy controls endometrial growth and prevents endometrial cancer, but the effectiveness of progesterone as a treatment for women with endometrial cancer is less clear. We noted that the hyperplasic phenotype of Wnt7a(cre+)Mig-6(f/f) mice was prevented by progesterone treatment, whereas this treatment had no effect in PR(cre/+)Mig-6(f/f) mice where Mig-6 was deleted in both the epithelial and stromal compartments of the uterus. In contrast, activation of progesterone signaling in the stroma regulated proliferation and apoptosis in the epithelium via suppression of ERα signaling. In summary, our results establish that epithelial Mig-6 functions as a critical tumor suppressor that mediates the ability of progesterone to prevent the development of endometrial cancer.


Subject(s)
Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Epithelial Cells/metabolism , Epithelium/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Tumor Suppressor Proteins/genetics , Animals , Apoptosis/physiology , Baculoviral IAP Repeat-Containing 3 Protein , Cell Growth Processes/physiology , Endometrial Hyperplasia/genetics , Endometrial Hyperplasia/metabolism , Endometrial Hyperplasia/pathology , Endometrial Neoplasms/pathology , Endometrial Neoplasms/prevention & control , Epithelial Cells/pathology , Epithelium/pathology , Estrogen Receptor alpha/genetics , Estrogen Receptor alpha/metabolism , Estrogens/genetics , Estrogens/metabolism , Female , Inhibitor of Apoptosis Proteins/genetics , Inhibitor of Apoptosis Proteins/metabolism , Mice , Progesterone/genetics , Progesterone/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, Progesterone/genetics , Receptors, Progesterone/metabolism , Stromal Cells/metabolism , Stromal Cells/pathology , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases , Uterus/metabolism , Uterus/pathology , Wnt Proteins/genetics , Wnt Proteins/metabolism
19.
Mol Endocrinol ; 24(6): 1297-304, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20382891

ABSTRACT

Nuclear receptors and coregulators orchestrate diverse aspects of biological functions and inappropriate expression of these factors often associates with human diseases. The present study describes a conditional overexpression system consisting of a minigene located at the Rosa26 locus in the genome of mouse embryonic stem (ES) cells. Before activation, the minigene is silent due to a floxed STOP cassette inserted between the promoter and the transgene. Upon cre-mediated excision of the STOP cassette, the minigene constitutively expresses the tagged transgene driven by the ubiquitous CAGGS promoter. Thus, this system can be used to express target gene in any tissue in a spatial and/or temporal manner if respective cre mouse lines are available. Serving as proof of principle, the CAG-S-hCOUP-TFI allele was generated in ES cells and subsequently in mice. This allele was capable of conditionally overexpressing human chicken ovalbumin upstream promoter-transcription factor I (COUP-TFI) in all tissues tested upon activation by cre drivers. This allele was further subjected to address functionality of expressed COUP-TFI and the functional similarity between COUP-TFI and COUP-TFII. Expression of COUP-TFI in COUP-TFII-ablated uterus suppressed aberrant estrogen receptor-alpha activities and rescued implantation and decidualization defects of COUP-TFII mutants, suggesting that COUP-TFI and COUP-TFII are able to functionally compensate for each other in the uterus. A toolbox currently under construction will contain ES cell lines for overexpressing all 48 nuclear receptors and selected 10 coregulators. Upon completion, it will be a very valuable resource for the scientific community. Several ES cells are currently available for distribution.


Subject(s)
Embryonic Stem Cells/metabolism , Nuclear Receptor Coactivators/metabolism , Receptors, Cytoplasmic and Nuclear/metabolism , Alleles , Animals , COUP Transcription Factor I/metabolism , COUP Transcription Factor II/deficiency , COUP Transcription Factor II/metabolism , Cell Line , Female , Humans , Mice , Uterus/metabolism , Uterus/pathology
20.
Mol Endocrinol ; 24(5): 930-40, 2010 May.
Article in English | MEDLINE | ID: mdl-20219888

ABSTRACT

Synchrony between embryo competency and uterine receptivity is essential for successful implantation. Mice with ablation of chicken ovalbumin upstream promoter-transcription factor II (COUP-TFII) in the uterus (PR(Cre/+);COUP-TFII(flox/flox)) exhibit implantation defects and increased estrogen receptor (ER)alpha activity in the luminal epithelium, suggesting high ERalpha activity may disrupt the window of uterine receptivity. To determine whether increased ERalpha activity in the PR(Cre/+);COUP-TFII(flox/flox) uterus is the cause of defective implantation, we assessed whether inhibition of ERalpha activity could rescue the PR(Cre/+);COUP-TFII(flox/flox) uterine implantation defect. ICI 182,780 (ICI), a pure ERalpha antagonist, was administered to PR(Cre/+);COUP-TFII(flox/flox) mutant and COUP-TFII(flox/flox) control mice during the receptive period, and the number of implantation sites was examined. COUP-TFII(flox/flox) control mice treated with oil or ICI showed the normal number of implantation sites. As expected, no implantation sites were observed in PR(Cre/+);COUP-TFII(flox/flox) mutant mice treated with oil, consistent with previous observations. In contrast, implantation sites were greatly increased in ICI-treated PR(Cre/+);COUP-TFII(flox/flox) mutant mice, albeit at a reduced number in comparison with the control mice. ICI treatment was also able to restore the expression of Wnt4 and bone morphogenetic protein 2, important for endometrial decidualization in the PR(Cre/+);COUP-TFII(flox/flox) mutant mice. To confirm that the rescue of embryo attachment and decidualization is a consequence of a reduced ERalpha activity upon ICI treatment, we showed a reduction of the expression of ERalpha target genes in PR(Cre/+);COUP-TFII(flox/flox) mutant mice. Because COUP-TFII was also shown in our laboratory to be important for placentation during pregnancy, we asked whether ICI treatment could also rescue the placentation defect to allow full-term pregnancy in these mice. We found that whereas mice were born in COUP-TFII(flox/flox) control mice given ICI, no pups were born in the PR(Cre/+);COUP-TFII(flox/flox) mutant mice, suggesting that the increased ERalpha activity is not the reason for placentation defects. These results demonstrate that during the periimplantation period, COUP-TFII regulates embryo attachment and decidualization through controlling ERalpha activity. However, COUP-TFII expression is still required in the postimplantation period to facilitate placentation.


Subject(s)
COUP Transcription Factor II/physiology , Embryo Implantation/physiology , Estrogen Receptor alpha/metabolism , Animals , COUP Transcription Factor II/genetics , Embryo Implantation/drug effects , Embryo Implantation/genetics , Estradiol/analogs & derivatives , Estradiol/pharmacology , Estrogen Receptor alpha/antagonists & inhibitors , Female , Fulvestrant , Immunohistochemistry , Male , Mice , Mice, Mutant Strains , Oligonucleotide Array Sequence Analysis , Pregnancy , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL