Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters








Publication year range
1.
Front Bioeng Biotechnol ; 10: 845716, 2022.
Article in English | MEDLINE | ID: mdl-35372320

ABSTRACT

To enhance bone regeneration, the use of bone morphogenetic protein (BMP)-2 is an attractive option. Unfortunately, the dose-dependent side effects prevent its widespread use. Therefore, a novel osteogenic agent using a different mechanism of action than BMP-2 is highly desirable. Previous reports demonstrated that prostaglandin E2 receptor 4 (EP4) agonists have potent osteogenic effects on non-human cells and are one of the potential alternatives for BMP-2. Here, we investigated the effects of an EP4 agonist (AKDS001) on human cells with a rat heterotopic xenograft model of human bone. Bone formation in the xenograft model was significantly enhanced by AKDS001 treatment. Histomorphometric analysis showed that the mode of bone formation by AKDS001 was minimodeling rather than remodeling. In cultured human mesenchymal stem cells, AKDS001 enhanced osteogenic differentiation and mineralization via the cAMP/PKA pathway. In cultured human preosteoclasts, AKDS001 suppressed bone resorption by inhibiting differentiation into mature osteoclasts. Thus, we conclude that AKDS001 can enhance bone formation in grafted autogenous bone by minimodeling while maintaining the volume of grafted bone. The combined use of an EP4 agonist and autogenous bone grafting may be a novel treatment option to enhance bone regeneration. However, we should be careful in interpreting the results because male xenografts were implanted in male rats in the present study. It remains to be seen whether females can benefit from the positive effects of AKDS001 MS by using female xenografts implanted in female rats in clinically relevant animal models.

2.
Sci Rep ; 10(1): 207, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31937792

ABSTRACT

Using an efficient gene targeting approach, we developed a novel mouse line that expresses the tetracycline-controlled transactivator (tTA) from the constitutively active Eef1a1 locus in a Cre recombinase-inducible manner. The temporally and spatially controlled expression of the EF1-LSL-tTA knockin and activation of tTA-driven responder transgenes was tested using four transgenic lines that express Cre under tissue-specific promoters of the pancreas, mammary gland and other secretory tissues, as well as an interferon-inducible promoter. In all models, the endogenous Eef1a1 promoter facilitated a cell-type-specific activation of target genes at high levels without exogenous enhancer elements. The applicability of the EF1-LSL-tTA strain for biological experiments was tested in two studies related to mammary gland development and tumorigenesis. First, we validated the crucial role of active STAT5 as a survival factor for functionally differentiated epithelial cells by expressing a hyperactive STAT5 mutant in the mammary gland during postlactational remodeling. In a second experiment, we assessed the ability of the EF1-tTA to initiate tumor formation through upregulation of mutant KRAS. The collective results show that the EF1-LSL-tTA knockin line is a versatile genetic tool that can be applied to constitutively express transgenes in specific cell types to examine their biological functions at defined developmental stages.


Subject(s)
Gene Expression Regulation/drug effects , Peptide Elongation Factor 1/metabolism , Tetracycline/pharmacology , Transgenes/physiology , Animals , Anti-Bacterial Agents/pharmacology , Female , Genes, Reporter , Integrases/metabolism , Male , Mice , Mice, Transgenic , Peptide Elongation Factor 1/genetics , Promoter Regions, Genetic , Tissue Distribution , Trans-Activators
3.
Cancer Med ; 7(2): 445-453, 2018 02.
Article in English | MEDLINE | ID: mdl-29282901

ABSTRACT

Salivary gland cancer is an aggressive and painful cancer, but a rare tumor type accounting for only ~0.5% of cancer cases. Tumors of the salivary gland exhibit heterogeneous histologic and genetic features and they are subdivided into different subtypes, with adenoid cystic carcinomas (ACC) being one of the most abundant. Treatment of ACC patients is afflicted by high recurrence rates, the high potential of the tumors to metastasize, as well as the poor response of ACC to chemotherapy. A prerequisite for the development of targeted therapies is insightful genetic information for driver core cancer pathways. Here, we developed a transgenic mouse model toward establishment of a preclinical model. There is currently no available mouse model for adenoid cystic carcinomas as a rare disease entity to serve as a test system to block salivary gland tumors with targeted therapy. Based on tumor genomic data of ACC patients, a key role for the activation of the PI3K-AKT-mTOR pathway was suggested in tumors of secretory glands. Therefore, we investigated the role of Akt3 expression in tumorigenesis and report that Akt3 overexpression results in ACC of salivary glands with 100% penetrance, while abrogation of transgenic Akt3 expression could revert the phenotype. In summary, our findings validate a novel mouse model to study ACC and highlight the druggable potential of AKT3 in the treatment of salivary gland patients.


Subject(s)
Carcinoma, Adenoid Cystic/pathology , Doxycycline/administration & dosage , Proto-Oncogene Proteins c-akt/metabolism , Salivary Gland Neoplasms/pathology , Animals , Anti-Bacterial Agents/administration & dosage , Carcinoma, Adenoid Cystic/drug therapy , Carcinoma, Adenoid Cystic/enzymology , Humans , Mice , Mice, Transgenic , Prognosis , Salivary Gland Neoplasms/drug therapy , Salivary Gland Neoplasms/enzymology
4.
Genesis ; 54(11): 582-588, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27671227

ABSTRACT

The biological functions of the Janus kinase 1 (JAK1) are suggested to be pleiotropic since this signal transducer is ubiquitously expressed and coupled to a variety of cytokine receptors. Consequently, mice that are deficient in this tyrosine kinase were reported to die shortly after birth. To facilitate studies that address the biological and molecular functions of JAK1 during postnatal development, we performed gene targeting in embryonic stem cells and generated a Cre/lox-based conditional knockout mouse model. Expression of Cre recombinase in the germline converted the Jak1 conditional knockout allele (Jak1fl ) into a null allele (Jak1- ) that when subsequently crossed into homozygosity led to a complete absence of the JAK1 protein in developing embryos. JAK1 deficient embryos were visibly smaller starting at E15.5. Newborn pups exhibited signs of apnea and died within hours after birth. The examination of fibroblasts from conditional knockout embryos and their littermate wildtype controls expressing JAK1 showed that lack of this Janus kinase resulted in an impaired tyrosine phosphorylation and activation of the downstream Signal Transducers and Activators of Transcription (STATs) 1, 3, and 6. JAK1 conditional knockout mice will be an invaluable tool to study cytokine signaling during normal development and disease progression in adult animals.


Subject(s)
Embryonic Development/genetics , Janus Kinase 1/genetics , Mice, Knockout , Alleles , Animals , Gene Expression Regulation, Developmental , Gene Targeting , Integrases/genetics , Mice , Mouse Embryonic Stem Cells/metabolism , STAT Transcription Factors/genetics
5.
Mol Cell Biol ; 36(11): 1673-90, 2016 06 01.
Article in English | MEDLINE | ID: mdl-27044867

ABSTRACT

Despite a wealth of knowledge about the significance of individual signal transducers and activators of transcription (STATs), essential functions of their upstream Janus kinases (JAKs) during postnatal development are less well defined. Using a novel mammary gland-specific JAK1 knockout model, we demonstrate here that this tyrosine kinase is essential for the activation of STAT1, STAT3, and STAT6 in the mammary epithelium. The loss of JAK1 uncouples interleukin-6-class ligands from their downstream effector, STAT3, which leads to the decreased expression of STAT3 target genes that are associated with the acute-phase response, inflammation, and wound healing. Consequently, JAK1-deficient mice exhibit impaired apoptosis and a significant delay in mammary gland remodeling. Using RNA sequencing, we identified several new JAK1 target genes that are upregulated during involution. These include Bmf and Bim, which are known regulators of programmed cell death. Using a BMF/BIM-double-knockout epithelial transplant model, we further validated that the synergistic action of these proapoptotic JAK1 targets is obligatory for the remodeling of the mammary epithelium. The collective results of this study suggest that JAK1 has nonredundant roles in the activation of particular STAT proteins and this tyrosine kinase is essential for coupling inflammatory cytokine signals to the cell death machinery in the differentiated mammary epithelium.


Subject(s)
Cytokines/metabolism , Janus Kinase 1/genetics , Mammary Glands, Animal/pathology , STAT Transcription Factors/genetics , Sequence Analysis, RNA/methods , Animals , Apoptosis , Cells, Cultured , Female , Janus Kinase 1/metabolism , Mammary Glands, Animal/immunology , Mice , STAT1 Transcription Factor/genetics , STAT3 Transcription Factor/genetics , STAT6 Transcription Factor/genetics , Signal Transduction , Transcriptional Activation
6.
Am J Pathol ; 185(12): 3202-10, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26475415

ABSTRACT

The Janus kinase (JAK) system is involved in numerous cell signaling processes and is highly expressed in cardiac tissue. The JAK isoform JAK2 is activated by numerous factors known to influence cardiac function and pathologic conditions. However, although abundant, the role of JAK2 in the regulation or maintenance of cardiac homeostasis remains poorly understood. Using the Cre-loxP system, we generated a cardiac-specific deletion of Jak2 in the mouse to assess the effect on cardiac function with animals followed up for a 4-month period after birth. These animals had marked mortality during this period, although at 4 months mortality in male mice (47%) was substantially higher compared with female mice (30%). Both male and female cardiac Jak2-deleted mice had hypertrophy, dilated cardiomyopathy, and severe left ventricular dysfunction, including a marked reduction in ejection fractions as assessed by serial echocardiography, although the responses in females were somewhat less severe. Defective cardiac function was associated with altered protein levels of sarcoplasmic reticulum calcium-regulatory proteins particularly in hearts from male mice that had depressed levels of SERCA2 and phosphorylated phospholamban. In contrast, SERCA2 was unchanged in hearts of female mice, whereas phosphorylated phospholamban was increased. Our findings suggest that cardiac JAK2 is critical for maintaining normal heart function, and its ablation produces a severe pathologic phenotype composed of myocardial remodeling, heart failure, and pronounced mortality.


Subject(s)
Cardiomegaly/enzymology , Janus Kinase 2/physiology , Ventricular Dysfunction, Left/enzymology , Ventricular Remodeling/physiology , Animals , Cardiomegaly/genetics , Cardiomegaly/pathology , Cardiomegaly/physiopathology , Female , Gene Deletion , Genotype , Janus Kinase 2/deficiency , Janus Kinase 2/genetics , Male , Mice, Knockout , Ventricular Dysfunction, Left/genetics , Ventricular Dysfunction, Left/pathology , Ventricular Dysfunction, Left/physiopathology , Ventricular Remodeling/genetics
7.
Methods Mol Biol ; 1267: 47-71, 2015.
Article in English | MEDLINE | ID: mdl-25636464

ABSTRACT

Breast cancer is the most common cause of cancer death in women worldwide. This malignancy is a complex disease, which is defined by an intrinsic heterogeneity on the histopathological and molecular level as well as response to therapy and outcome. In addition to classical histopathological features, breast cancer can be categorized into at least five major subtypes based on comprehensive gene expression profiling: luminal A, luminal B, basal-like, ERBB2-positive, and normal-like breast cancer. Genetically engineered mouse models can serve as tools to study the molecular underpinnings for this disease. Given the genetic complexity that drives the initiation and progression of individual breast cancer subtypes, it is evident that certain models can reflect only particular aspects of this malignancy. In this book chapter, we will primarily focus on advances in modeling breast cancer at defined stages of carcinogenesis using genetically engineered mice. We will discuss the ability as well as shortcomings of these models to faithfully recapitulate the spectrum of human breast cancer subtypes.


Subject(s)
Disease Models, Animal , Mammary Neoplasms, Experimental , Animals , Female , Gene Knockout Techniques , Genetic Engineering , Humans , Ligands , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Neoplasm Metastasis , Oncogenes/genetics , Organ Specificity
8.
Methods Mol Biol ; 1194: 21-35, 2014.
Article in English | MEDLINE | ID: mdl-25064096

ABSTRACT

Conditional knockout mouse models are powerful tools to examine the biological and molecular function(s) of genes in specific tissues. The general procedure to generate such genetically engineered mouse models consists of three main steps. The first step is to find the appropriate genomic clone of the gene of interest and to design the cloning and Southern blot strategies. The second step is the cloning of the gene-targeting vector with all its essential components including positive and negative selection cassettes and the insertion of LoxP sites. Although conventional methods are still being widely used for DNA cloning, we describe in this book chapter the use of λ Red phage-based homologous recombination in Escherichia coli to capture the genomic DNA of the gene of interest and to assemble the gene-targeting vector. This new method provides several advantages as it does not require the presence of restriction sites within the gene of interest to insert LoxP-flanked DNA fragments. In the final step, the gene-targeting vector is transferred into embryonic stem (ES) cells, and successfully targeted ES cell clones are injected into mouse blastocysts to generate conditional knockout mice.


Subject(s)
Gene Knockout Techniques/methods , Animals , Blastocyst/cytology , Blotting, Southern , Chromosomes, Artificial, Bacterial/genetics , Clone Cells/metabolism , DNA, Bacterial/genetics , Embryonic Stem Cells/cytology , Embryonic Stem Cells/metabolism , Gene Targeting , Genetic Loci/genetics , Genetic Vectors/genetics , Genomics , Mice , Mice, Knockout
9.
Stem Cells ; 32(7): 1878-89, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24677703

ABSTRACT

Jak2, a member of the Janus kinase family of nonreceptor protein tyrosine kinases, is activated in response to a variety of cytokines, and functions in survival and proliferation of cells. An activating JAK2V617F mutation has been found in most patients with myeloproliferative neoplasms, and patients treated with Jak2 inhibitors show significant hematopoietic toxicities. However, the role of Jak2 in adult hematopoietic stem cells (HSCs) has not been clearly elucidated. Using a conditional Jak2 knockout allele, we have found that Jak2 deletion results in rapid loss of HSCs/progenitors leading to bone marrow failure and early lethality in adult mice. Jak2 deficiency causes marked impairment in HSC function, and the mutant HSCs are severely defective in reconstituting hematopoiesis in recipient animals. Jak2 deficiency also causes significant apoptosis and loss of quiescence in HSC-enriched LSK (Lin(-)Sca-1(+)c-Kit(+)) cells. Jak2-deficient LSK cells exhibit elevated reactive oxygen species levels and enhanced p38 MAPK activation. Mutant LSK cells also show defective Stat5, Erk, and Akt activation in response to thrombopoietin and stem cell factor. Gene expression analysis reveals significant downregulation of genes related to HSC quiescence and self-renewal in Jak2-deficient LSK cells. These data suggest that Jak2 plays a critical role in the maintenance and function of adult HSCs.


Subject(s)
Adult Stem Cells/enzymology , Hematopoietic Stem Cells/enzymology , Janus Kinase 2/physiology , Adult Stem Cells/physiology , Anemia, Aplastic , Animals , Bone Marrow Diseases , Bone Marrow Failure Disorders , Cell Proliferation , Cell Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Hemoglobinuria, Paroxysmal/enzymology , Mice, Inbred C57BL , Mice, Transgenic , Reactive Oxygen Species/metabolism , Stem Cell Factor/physiology , Thrombopoietin/physiology
10.
BMC Cancer ; 14: 195, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24628780

ABSTRACT

BACKGROUND: The serine-threonine kinase AKT1 plays essential roles during normal mammary gland development as well as the initiation and progression of breast cancer. AKT1 is generally considered a ubiquitously expressed gene, and its persistent activation is transcriptionally controlled by regulatory elements characteristic of housekeeping gene promoters. We recently identified a novel Akt1 transcript in mice (Akt1m), which is induced by growth factors and their signal transducers of transcription from a previously unknown promoter. The purpose of this study was to examine whether normal and neoplastic human breast epithelial cells express an orthologous AKT1m transcript and whether its expression is deregulated in cancer cells. METHODS: Initial sequence analyses were performed using the UCSC Genome Browser and GenBank to assess the potential occurrence of an AKT1m transcript variant in human cells and to identify conserved promoter sequences that are orthologous to the murine Akt1m. Quantitative RT-PCR was used to determine the transcriptional activation of AKT1m in mouse mammary tumors as well as 41 normal and neoplastic human breast epithelial cell lines and selected primary breast cancers. RESULTS: We identified four new AKT1 transcript variants in human breast cancer cells that are orthologous to the murine Akt1m and that encode the full-length kinase. These transcripts originate from an alternative promoter that is conserved between humans and mice. Akt1m is upregulated in the majority of luminal-type and basal-type mammary cancers in four different genetically engineered mouse models. Similarly, a subset of human breast cancer cell lines and primary breast cancers exhibited a higher expression of orthologous AKT1m transcripts. CONCLUSIONS: The existence of an alternative promoter that drives the expression of the unique AKT1m transcript may provide a mechanism by which the levels of AKT1 can be temporally and spatially regulated at particular physiological states, such as cancer, where a heightened activity of this kinase is required.


Subject(s)
BRCA1 Protein/genetics , Breast Neoplasms/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/genetics , RNA Isoforms/analysis , Alternative Splicing , Animals , Base Sequence , Breast Neoplasms/pathology , Cell Line, Tumor , Conserved Sequence , Female , Gene Expression Regulation, Neoplastic , Humans , Mammary Neoplasms, Experimental , Mice , Molecular Sequence Data , Proto-Oncogene Proteins c-akt/metabolism , RNA Splice Sites
11.
Mol Cell Biol ; 34(7): 1363-77, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24469394

ABSTRACT

Stat5 (signal transducer and activator of transcription 5) is an essential mediator of cytokine receptor signaling and plays important roles in the proliferation of alveolar progenitors and the survival of functionally differentiated epithelial cells in the mammary gland. A deregulated expression and activation of Stat5 leads to precocious alveolar development in the absence of pregnancy hormones, impaired mammary gland remodeling following the cessation of lactation, and mammary tumor formation. We reported previously that Stat5 induces the transcription of the Akt1 gene from a novel promoter. In this report, we provide experimental evidence that Akt1 is an essential mediator for the biological function of Stat5 as a survival factor. Additionally, Stat5 controls the expression of the regulatory and catalytic subunits of the phosphatidylinositol 3-kinase (PI3K) (p85α and p110α), thereby greatly augmenting signaling through the prosurvival PI3K/Akt pathway. In agreement with this model, we observed that the constitutive activation of Stat5 cooperates with the loss of function of the tumor suppressor PTEN by accelerating the formation of preneoplastic lesions and mammary tumors. The mammary gland-specific ablation of Stat5 is sufficient to prevent mammary carcinogenesis in a genuine mouse model for Cowden syndrome. Therefore, targeting the Jak2/Stat5 pathway might be a suitable strategy to prevent breast cancer in patients that carry a mutant PTEN allele.


Subject(s)
Mammary Glands, Animal/growth & development , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism , Animals , Cell Line , Class I Phosphatidylinositol 3-Kinases/genetics , Class I Phosphatidylinositol 3-Kinases/metabolism , Class Ia Phosphatidylinositol 3-Kinase/genetics , Class Ia Phosphatidylinositol 3-Kinase/metabolism , Female , Gene Knockout Techniques , Hamartoma Syndrome, Multiple/etiology , Hamartoma Syndrome, Multiple/genetics , Hamartoma Syndrome, Multiple/metabolism , Humans , Janus Kinase 2/metabolism , Mammary Neoplasms, Experimental/etiology , Mammary Neoplasms, Experimental/genetics , Mice , Mice, Knockout , Mice, Mutant Strains , Mice, Transgenic , PTEN Phosphohydrolase/deficiency , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Phosphatidylinositol 3-Kinases/genetics , Precancerous Conditions/etiology , Precancerous Conditions/genetics , Precancerous Conditions/metabolism , Pregnancy , Proto-Oncogene Proteins c-akt/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , STAT5 Transcription Factor/deficiency , STAT5 Transcription Factor/genetics , Signal Transduction
12.
Mol Cell Endocrinol ; 382(1): 583-592, 2014 Jan 25.
Article in English | MEDLINE | ID: mdl-23562856

ABSTRACT

In response to the ligand-mediated activation of cytokine receptors, cells decide whether to proliferate or to undergo differentiation. D-type Cyclins (Cyclin D1, D2, or D3) and their associated Cyclin-dependent kinases (CDK4, CDK6) connect signals from cytokines to the cell cycle machinery, and they propel cells through the G1 restriction point and into the S phase, after which growth factor stimulation is no longer essential to complete cell division. D-type Cyclins are upregulated in many human malignancies including breast cancer to promote an uncontrolled proliferation of cancer cells. After summarizing important aspects of the cytokine-mediated transcriptional regulation and the posttranslational modification of D-type Cyclins, this review will highlight the physiological significance of these cell cycle regulators during normal mammary gland development as well as the initiation and promotion of breast cancer. Although the vast majority of published reports focus almost exclusively on the role of Cyclin D1 in breast cancer, we summarize here previous and recent findings that demonstrate an important contribution of the remaining two members of this Cyclin family, in particular Cyclin D3, for the growth of ErbB2-associated breast cancer cells in humans and in mouse models. New data from genetically engineered models as well as the pharmacological inhibition of CDK4/6 suggest that targeting the combined functions of D-type Cyclins could be a suitable strategy for the treatment of ErbB2-positive and potentially other types of breast cancer.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cyclins/metabolism , Cytokines/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , Signal Transduction , Animals , Cell Proliferation , Female , Humans
13.
Anat Rec (Hoboken) ; 297(2): 200-7, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24347432

ABSTRACT

The purpose of this study was to determine the timing of the onset of adult-type sulcal length asymmetry during postnatal development of the male cynomolgus monkey cerebrum. The monkey brain has already reached adult size by 3 months of age, although the body weight only represents 1/8 of the adult body weight by that time. The fronto-occipital length and the cerebral width also reached adult levels by that postnatal age with no left/right bias. Consistently, lengths of the major primary sulci reached adult levels by 3 months of age, and then decreased slightly in sexually mature monkeys (4-6.5 years of age). Asymmetry quotient analysis showed that sulcal length asymmetry patterns gradually changed during postnatal development. The male adult pattern of sulcal length asymmetry was acquired after 24 months of age. In particular, age-dependent rightward lateralization of the arcuate sulcal length was revealed during cerebral maturation by three-way ANOVA. The results suggest that the regional difference in cerebral maturation from adolescence to young adulthood modifies the sulcal morphology with characteristic asymmetric patterns in male cynomolgus monkeys.


Subject(s)
Cerebrum/anatomy & histology , Cerebrum/growth & development , Macaca fascicularis/anatomy & histology , Aging/pathology , Animals , Body Weight , Female , Male , Organ Size , Sex Factors
14.
Cancer Res ; 73(6): 1821-30, 2013 Mar 15.
Article in English | MEDLINE | ID: mdl-23467612

ABSTRACT

The initiation and progression of pancreatic ductal adenocarcinoma (PDAC) is governed by a series of genetic and epigenetic changes, but it is still unknown whether these alterations are required for the maintenance of primary and metastatic PDAC. We show here that the c-Myc oncogene is upregulated throughout the entire process of neoplastic progression in human PDAC and in genetically engineered mice that express mutant Kras. To experimentally address whether c-Myc is essential for the growth and survival of cancer cells, we developed a novel mouse model that allows a temporally and spatially controlled expression of this oncogene in pancreatic progenitors and derived lineages of the exocrine pancreas. Unlike previous reports, upregulation of c-Myc was sufficient to induce the formation of adenocarcinomas after a short latency without additional genetic manipulation of cell survival pathways. Deficiency in Cdkn2a increased the rate of metastasis but had no effect on tumor latency or c-Myc-mediated cancer maintenance. Despite a macroscopically complete regression of primary, metastatic, and transplantable tumors following the ablation of c-Myc, some cancer cells remained dormant. A significant number of these residual neoplastic cells expressed cancer stem cell markers, and re-expression of exogenous c-Myc in these cells led to rapid cancer recurrence. Collectively, the results of this study suggest that c-Myc plays a significant role in the progression and maintenance of PDAC, but besides targeting this oncogene or its downstream effectors, additional therapeutic strategies are necessary to eradicate residual cancer cells to prevent disease recurrence.


Subject(s)
Disease Models, Animal , Pancreatic Neoplasms/pathology , Animals , Cell Transformation, Neoplastic/genetics , Flow Cytometry , Genes, myc , Genes, p16 , Mice , Neoplasm Recurrence, Local , Pancreatic Neoplasms/genetics
15.
Nat Immunol ; 14(4): 364-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23435120

ABSTRACT

Dendritic cells (DCs) are critical in immune responses, linking innate and adaptive immunity. We found here that DC-specific deletion of the transcription factor STAT5 was not critical for development but was required for T helper type 2 (TH2), but not TH1, allergic responses in both the skin and lungs. Loss of STAT5 in DCs led to the inability to respond to thymic stromal lymphopoietin (TSLP). STAT5 was required for TSLP-dependent DC activation, including upregulation of the expression of costimulatory molecules and chemokine production. Furthermore, TH2 responses in mice with DC-specific loss of STAT5 resembled those seen in mice deficient in the receptor for TSLP. Our results show that the TSLP-STAT5 axis in DCs is a critical component for the promotion of type 2 immunity at barrier surfaces.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , STAT5 Transcription Factor/metabolism , Th2 Cells/immunology , Animals , Cell Differentiation , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/cytology , Dermatitis, Contact/immunology , Dermatitis, Contact/metabolism , Dermis/immunology , Dermis/metabolism , Female , Homeostasis/immunology , Janus Kinases/metabolism , Lung/immunology , Lung/metabolism , Mice , Mice, Knockout , STAT5 Transcription Factor/genetics , Signal Transduction , Th1 Cells/immunology , Thymic Stromal Lymphopoietin
16.
PLoS One ; 7(8): e43778, 2012.
Article in English | MEDLINE | ID: mdl-22952764

ABSTRACT

We have generated a new and improved transgenic mouse strain that permits a temporally controlled expression of transgenes throughout mammary gland development. High expression of the tetracycline-regulatible transactivator (tTA) under control of the mouse mammary tumor virus long terminal repeat (MMTV-LTR) is restricted to mammary epithelial cells and the salivary gland. The novel MMTV-tTA mouse strain induces a sustained transactivation of responder transgenes, which can be swiftly suppressed through administration of doxycycline (Dox). An important characteristic of this strain is its expression in early progenitor cells of mammary gland anlagen beginning at day 13.5 of embryonic development. We show here that the MMTV-tTA can be used in combination with GFP reporter strains to visualize CK8/CK14-dual positive progenitors in newborn females and their derived basal and luminal epithelial cell lineages in adult females. Our observations suggest that the novel MMTV-tTA can be utilized to express exogenous proteins in multipotent mammary progenitors during the earliest stages of mammary gland development to assess their biological significance throughout mammogenesis. Moreover, we demonstrate that the expression of the MMTV-tTA is sustained during mammary gland tumorigenesis in female mice expressing wildtype ErbB2. This makes this strain particular valuable to target the expression of exogenous proteins into developing mammary tumors to assess their significance in biological processes, such as tumor cell growth and survival, metabolism, and metastasis.


Subject(s)
Embryo, Mammalian/metabolism , Mammary Glands, Animal/metabolism , Mammary Tumor Virus, Mouse/genetics , Mice, Transgenic/genetics , Terminal Repeat Sequences/genetics , Tetracycline/pharmacology , Trans-Activators/genetics , Animals , Cell Line , Epithelial Cells/metabolism , Female , Gene Deletion , Gene Expression , Genes, erbB-2/genetics , Integrases/genetics , Ligands , Mammary Neoplasms, Experimental/genetics , Mice , Transcriptional Activation/drug effects
17.
Cancer Res ; 71(24): 7513-24, 2011 Dec 15.
Article in English | MEDLINE | ID: mdl-22037875

ABSTRACT

Cyclin D1 regulates cell proliferation and is a candidate molecular target for breast cancer therapy. This study addresses whether Cyclin D1 is indispensable for ErbB2-associated mammary tumor initiation and progression using a breast cancer model in which this cell-cycle regulator can be genetically ablated prior to or after neoplastic transformation. Deficiency in Cyclin D1 delayed tumor onset but did not prevent the occurrence of mammary cancer in mice overexpressing wild-type ErbB2. The lack of Cyclin D1 was associated with a compensatory upregulation of Cyclin D3, which explains why the targeted downregulation of Cyclin D1 in established mammary tumors had no effect on cancer cell proliferation. Cyclin D1 and D3 are overexpressed in human breast cancer cell lines and primary invasive breast cancers, and Cyclin D3 frequently exceeded the expression of Cyclin D1 in ErbB2-positive cases. The simultaneous inhibition of both cyclins in mammary tumor cells reduced cancer cell proliferation in vitro and decreased the tumor burden in vivo. Collectively, the results of this study suggest that only the combined inhibition of Cyclin D1 and D3 might be a suitable strategy for breast cancer prevention and therapy.


Subject(s)
Cyclin D1/metabolism , Cyclin D3/metabolism , Mammary Neoplasms, Animal/metabolism , Receptor, ErbB-2/metabolism , Animals , Blotting, Western , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cells, Cultured , Cyclin D1/genetics , Cyclin D3/genetics , Disease Progression , Female , Humans , Immunohistochemistry , Mammary Neoplasms, Animal/genetics , Mammary Neoplasms, Animal/pathology , Mice , Mice, Knockout , Mice, Transgenic , NIH 3T3 Cells , RNA Interference , Receptor, ErbB-2/genetics
18.
Genesis ; 49(1): 24-35, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21254334

ABSTRACT

The mammary gland is a developmentally dynamic, hormone-responsive organ that undergoes proliferation and differentiation within the secretory epithelial compartment during pregnancy. The epithelia are maintained by pro-survival signals (e.g., Stat5, Akt1) during lactation, but undergo apoptosis during involution through inactivation of cell survival pathways and upregulation of pro-apoptotic proteins. To assess if the survival signals in the functionally differentiated mammary epithelial cells can override a pro-apoptotic signal, we generated transgenic mice that express Bax under the whey acidic protein (WAP) promoter. WAP-Bax females exhibited a lactation defect and were unable to nourish their offspring. Mammary glands demonstrated: (1) a reduction in epithelial content, (2) hallmark signs of mitochondria-mediated cell death, (3) an increase in apoptotic cells by TUNEL assay, and (4) precocious Stat3 activation. This suggests that upregulation of a single pro-apoptotic factor of the Bcl-2 family is sufficient to initiate apoptosis of functionally differentiated mammary epithelial cells in vivo.


Subject(s)
Apoptosis , Mammary Glands, Animal/embryology , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis/genetics , Cell Differentiation/genetics , Cell Differentiation/physiology , Epithelial Cells/physiology , Female , Genetic Vectors , Immunohistochemistry , Lactation/genetics , Mammary Glands, Animal/physiology , Mice , Mice, Transgenic , Milk Proteins/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-akt/metabolism , STAT5 Transcription Factor/metabolism , Up-Regulation/genetics , bcl-2-Associated X Protein/genetics
19.
Proc Natl Acad Sci U S A ; 107(45): 19455-60, 2010 Nov 09.
Article in English | MEDLINE | ID: mdl-20974963

ABSTRACT

Thymic stromal lymphopoietin (TSLP) is a type I cytokine that plays essential roles in allergic/inflammatory skin and airway disorders, in helminth infections, and in regulating intestinal immunity. TSLP signals via IL-7Rα and a specific TSLPR subunit that is highly related to the common cytokine receptor γ chain, γ(c). Although TSLP has effects on a broad range of hematopoetic cells and can induce STAT5 phosphorylation, TSLP was reported to not signal via JAK kinases, and the mechanism by which TSLP regulates STAT5 phosphorylation has been unclear. We now demonstrate the role of JAK1 and JAK2 in TSLP-mediated STAT5 phosphorylation in mouse and human primary CD4(+) T cells, in contrast to the known activation of JAK1 and JAK3 by the related cytokine, IL-7. We also show that just as JAK1 interacts with IL-7Rα, JAK2 is associated with TSLPR protein. Moreover, we demonstrate the importance of STAT5 activation for TSLP-mediated survival and proliferation of CD4(+) T cells. These findings clarify the basis for TSLP-mediated signaling and provide an example wherein a cytokine uses JAK1 and JAK2 to mediate the activation of STAT5.


Subject(s)
Cytokines/metabolism , Interleukin-7/metabolism , Janus Kinase 1/metabolism , Janus Kinase 2/metabolism , STAT5 Transcription Factor/metabolism , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , Cell Proliferation , Cell Survival/immunology , Cells, Cultured , Humans , Mice , Phosphorylation , Signal Transduction , Thymic Stromal Lymphopoietin
20.
Mol Cell Biol ; 30(12): 2957-70, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20385773

ABSTRACT

The signal transducer and activator of transcription 5 (Stat5) plays a pivotal role in the proliferation, secretory differentiation, and survival of mammary epithelial cells. However, there is little information about Stat5 target genes that facilitate these biological processes. We provide here experimental evidence that the prolactin-mediated phosphorylation of Stat5 regulates the transcriptional activation of the Akt1 gene. Stat5 binds to consensus sequences within the Akt1 locus in a growth factor-dependent manner to initiate transcription of a unique Akt1 mRNA from a distinct promoter, which is only active in the mammary gland. Elevating the levels of active Akt1 restores the expression of cyclin D1 and proliferation of Jak2-deficient mammary epithelial cells, which provides evidence that Akt1 acts downstream of Jak/Stat signaling. The ligand-inducible expression of Stat5 in transgenic females mediates a sustained upregulation of Akt1 in mammary epithelial cells during the onset of postlactational involution. Stat5-expressing mammary glands exhibit a delay in involution despite induction of proapoptotic signaling events. Collectively, the results of the present study elucidate an underlying mechanism by which active Stat5 mediates evasion from apoptosis and self-sufficiency in growth signals.


Subject(s)
Epithelial Cells/cytology , Epithelial Cells/enzymology , Mammary Glands, Animal/cytology , Promoter Regions, Genetic/genetics , Proto-Oncogene Proteins c-akt/genetics , STAT5 Transcription Factor/metabolism , Transcriptional Activation/genetics , Animals , Apoptosis/drug effects , Cell Line , Cell Proliferation/drug effects , Cell Survival/drug effects , Cell Survival/genetics , Consensus Sequence , Cyclin D1/metabolism , Doxycycline/pharmacology , Epithelial Cells/drug effects , Female , Gene Expression Regulation, Enzymologic/drug effects , Janus Kinase 2/deficiency , Janus Kinase 2/metabolism , Lactation/drug effects , Lactation/genetics , Mice , Models, Biological , Organ Specificity/drug effects , Organ Specificity/genetics , Protein Binding/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Receptors, Prolactin/metabolism , Signal Transduction/drug effects , Transcriptional Activation/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL