Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
Add more filters










Publication year range
1.
Mol Cancer Res ; 18(6): 822-834, 2020 06.
Article in English | MEDLINE | ID: mdl-32152233

ABSTRACT

The PI3K signaling pathway is frequently mutated in head and neck squamous cell carcinoma (HNSCC), often via gain-of-function (GOF) mutations in the PIK3CA gene. Here, we present novel genetically engineered mouse models (GEMM) carrying a GOF allele Loxp-STOP-Loxp(LSL)-PIK3CAH1047R (E20) alone or in combination with heterozygous LSL-p53+/R172H (p53) mutation with tissue-specific expression to interrogate the role of oncogenic PIK3CA in transformation of upper aerodigestive track epithelium. We demonstrated that the GOF PIK3CA mutation promoted progression of 4-nitroquinoline 1-oxide-induced oral squamous cell carcinoma (OSCC) in both E20 single mutant and E20/p53 double mutant mice, with frequent distal metastasis detected only in E20/p53 GEMM. Similar to in human OSCC, loss of p16 was associated with progression of OSCC in these mice. RNA-seq analyses revealed that among the common genes differentially expressed in primary OSCC cell lines derived from E20, p53, and E20/p53 GEMMs compared with those from the wild-type mice, genes associated with proliferation and cell cycle were predominantly represented, which is consistent with the progressive loss of p16 detected in these GEMMs. Importantly, all of these OSCC primary cell lines exhibited enhanced sensitivity to BYL719 and cisplatin combination treatment in comparison with cisplatin alone in vitro and in vivo, regardless of p53 and/or p16 status. Given the prevalence of mutations in p53 and the PI3K pathways in HNSCC in conjunction with loss of p16 genetically or epigenetically, this universal increased sensitivity to cisplatin and BYL719 combination therapy in cancer cells with PIK3CA mutation represents an opportunity to a subset of patients with HNSCC. IMPLICATIONS: Our results suggest that combination therapy of cisplatin and PI3K inhibitor may be worthy of consideration in patients with HNSCC with PIK3CA mutation.


Subject(s)
4-Nitroquinoline-1-oxide/toxicity , Class I Phosphatidylinositol 3-Kinases/genetics , Head and Neck Neoplasms/pathology , Mutation , Squamous Cell Carcinoma of Head and Neck/secondary , Tumor Suppressor Protein p53/genetics , Animals , Carcinogens/toxicity , Disease Progression , Head and Neck Neoplasms/chemically induced , Head and Neck Neoplasms/genetics , Mice , Mice, Nude , Squamous Cell Carcinoma of Head and Neck/chemically induced , Squamous Cell Carcinoma of Head and Neck/genetics
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.
Proc Natl Acad Sci U S A ; 116(41): 20679-20688, 2019 10 08.
Article in English | MEDLINE | ID: mdl-31548432

ABSTRACT

The presence of progenitor or stem cells in the adult pancreas and their potential involvement in homeostasis and cancer development remain unresolved issues. Here, we show that mouse centroacinar cells can be identified and isolated by virtue of the mitochondrial enzyme Aldh1b1 that they uniquely express. These cells are necessary and sufficient for the formation of self-renewing adult pancreatic organoids in an Aldh1b1-dependent manner. Aldh1b1-expressing centroacinar cells are largely quiescent, self-renew, and, as shown by genetic lineage tracing, contribute to all 3 pancreatic lineages in the adult organ under homeostatic conditions. Single-cell RNA sequencing analysis of these cells identified a progenitor cell population, established its molecular signature, and determined distinct differentiation pathways to early progenitors. A distinct feature of these progenitor cells is the preferential expression of small GTPases, including Kras, suggesting that they might be susceptible to Kras-driven oncogenic transformation. This finding and the overexpression of Aldh1b1 in human and mouse pancreatic cancers, driven by activated Kras, prompted us to examine the involvement of Aldh1b1 in oncogenesis. We demonstrated genetically that ablation of Aldh1b1 completely abrogates tumor development in a mouse model of KrasG12D-induced pancreatic cancer.


Subject(s)
Aldehyde Dehydrogenase 1 Family/metabolism , Aldehyde Dehydrogenase, Mitochondrial/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Transformation, Neoplastic/pathology , Mutation , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Stem Cells/pathology , Aldehyde Dehydrogenase 1 Family/genetics , Aldehyde Dehydrogenase, Mitochondrial/genetics , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Differentiation , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Signal Transduction , Single-Cell Analysis , Stem Cells/metabolism
4.
J Natl Cancer Inst ; 106(12)2014 Dec.
Article in English | MEDLINE | ID: mdl-25306215

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDA) is frequently driven by oncogenic KRAS(KRAS*) mutations. We developed a mouse model of KRAS*-induced PDA and, based on genetic results demonstrating that KRAS* tumorigenicity depends on Myc activity, we evaluated the therapeutic potential of an orally administered anti-Myc drug. METHODS: We tested the efficacy of Mycro3, a small-molecule inhibitor of Myc-Max dimerization, in the treatment of mouse PDA (n = 9) and also of xenografts of human pancreatic cancer cell lines (NOD/SCID mice, n = 3-12). Tumor responses to the drug were evaluated by PET/CT imaging, and histological, immunohistochemical, molecular and microarray analyses. The Student's t test was used for differences between groups. All statistical tests were two-sided. RESULTS: Transgenic overexpression of KRAS* in the pancreas resulted in pancreatic intraepithelial neoplasia in two-week old mice, which developed invasive PDA a week later and became moribund at one month. However, this aggressive form of pancreatic tumorigenesis was effectively prevented by genetic ablation of Myc specifically in the pancreas. We then treated moribund, PDA-bearing mice daily with the Mycro3 Myc-inhibitor. The mice survived until killed at two months. PET/CT image analysis (n = 5) demonstrated marked shrinkage of PDA, while immunohistochemical analyses showed an increase in cancer cell apoptosis and reduction in cell proliferation (treated/untreated proliferation index ratio: 0.29, P < .001, n = 3, each group). Tumor growth was also drastically attenuated in Mycro3-treated NOD/SCID mice (n = 12) carrying orthotopic or heterotopic xenografts of human pancreatic cancer cells (eg, mean tumor weight ± SD of treated heterotopic xenografts vs vehicle-treated controls: 15.2±5.8 mg vs 230.2±43.9 mg, P < .001). CONCLUSION: These results provide strong justification for eventual clinical evaluation of anti-Myc drugs as potential chemotherapeutic agents for the treatment of PDA.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins c-myc/antagonists & inhibitors , Proto-Oncogene Proteins p21(ras)/genetics , Thiazoles/pharmacology , Animals , Cell Proliferation/drug effects , Gene Expression Regulation, Neoplastic , Gene Knock-In Techniques , Immunohistochemistry , Male , Mice , Mice, Inbred NOD , Mice, SCID , Mice, Transgenic , Positron-Emission Tomography , Proto-Oncogene Proteins c-myc/genetics , Tomography, X-Ray Computed , Up-Regulation , Xenograft Model Antitumor Assays
5.
Am J Physiol Renal Physiol ; 302(11): F1362-73, 2012 Jun 01.
Article in English | MEDLINE | ID: mdl-22461304

ABSTRACT

The adult kidney contains a population of low-cycling cells that resides in the papilla. These cells retain for long periods S-phase markers given as a short pulse early in life; i.e., they are label-retaining cells (LRC). In previous studies in adult rat and mice, we found that shortly after acute kidney injury many of the quiescent papillary LRC started proliferating (Oliver JA, Klinakis A, Cheema FH, Friedlander J, Sampogna RV, Martens TP, Liu C, Efstratiadis A, Al-Awqati Q. J Am Soc Nephrol 20: 2315-2327, 2009; Oliver JA, Maarouf O, Cheema FH, Martens TP, Al-Awqati Q. J Clin Invest 114: 795-804, 2004) and, with cell-tracking experiments, we found upward migration of some papillary cells including LRC (Oliver JA, Klinakis A, Cheema FH, Friedlander J, Sampogna RV, Martens TP, Liu C, Efstratiadis A, Al-Awqati Q. J Am Soc Nephrol 20: 2315-2327, 2009). To identify molecular cues involved in the activation (i.e., proliferation and/or migration) of the papillary LRC that follows injury, we isolated these cells from the H2B-GFP mice and found that they migrated and proliferated in response to the cytokine stromal cell-derived factor-1 (SDF-1). Moreover, in a papillary organ culture assay, the cell growth out of the upper papilla was dependent on the interaction of SDF-1 with its receptor Cxcr4. Interestingly, location of these two proteins in the kidney revealed a complementary location, with SDF-1 being preferentially expressed in the medulla and Cxcr4 more abundant in the papilla. Blockade of Cxcr4 in vivo prevented mobilization of papillary LRC after transient kidney ischemic injury and worsened its functional consequences. The data indicate that the SDF-1/Cxcr4 axis is a critical regulator of papillary LRC activation following transient kidney injury and during organ repair.


Subject(s)
Acute Kidney Injury/pathology , Chemokine CXCL12/pharmacology , Kidney Diseases/pathology , Kidney Medulla/growth & development , Acute Kidney Injury/physiopathology , Animals , Blotting, Western , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Separation , Cells, Cultured , Chemotaxis/drug effects , Female , Immunohistochemistry , Indicators and Reagents , Kidney Diseases/physiopathology , Kidney Medulla/pathology , Kidney Medulla/physiopathology , Male , Mice , Mice, Inbred C57BL , Organ Culture Techniques , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, CXCR4/antagonists & inhibitors , Receptors, CXCR4/metabolism
6.
Nature ; 473(7346): 230-3, 2011 May 12.
Article in English | MEDLINE | ID: mdl-21562564

ABSTRACT

Notch signalling is a central regulator of differentiation in a variety of organisms and tissue types. Its activity is controlled by the multi-subunit γ-secretase (γSE) complex. Although Notch signalling can play both oncogenic and tumour-suppressor roles in solid tumours, in the haematopoietic system it is exclusively oncogenic, notably in T-cell acute lymphoblastic leukaemia, a disease characterized by Notch1-activating mutations. Here we identify novel somatic-inactivating Notch pathway mutations in a fraction of patients with chronic myelomonocytic leukaemia (CMML). Inactivation of Notch signalling in mouse haematopoietic stem cells (HSCs) results in an aberrant accumulation of granulocyte/monocyte progenitors (GMPs), extramedullary haematopoieisis and the induction of CMML-like disease. Transcriptome analysis revealed that Notch signalling regulates an extensive myelomonocytic-specific gene signature, through the direct suppression of gene transcription by the Notch target Hes1. Our studies identify a novel role for Notch signalling during early haematopoietic stem cell differentiation and suggest that the Notch pathway can play both tumour-promoting and -suppressive roles within the same tissue.


Subject(s)
Gene Expression Regulation, Neoplastic , Genes, Tumor Suppressor/physiology , Leukemia, Myelomonocytic, Chronic/genetics , Leukemia, Myelomonocytic, Chronic/pathology , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction , Animals , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation , Cells, Cultured , Gene Expression Profiling , Gene Silencing , Granulocyte-Macrophage Progenitor Cells/cytology , Granulocyte-Macrophage Progenitor Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/metabolism , Humans , Mice , Mice, Inbred C57BL , Mutation , Receptors, Notch/deficiency , Transcription Factor HES-1 , Tumor Cells, Cultured
7.
Mol Cancer Ther ; 10(4): 697-707, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21447712

ABSTRACT

Inhibition of the insulin-like growth factor 1 receptor (Igf1r) is an approach being taken in clinical trials to overcome the dismal outcome for metastatic alveolar rhabdomyosarcoma (ARMS), an aggressive muscle cancer of children and young adults. In our study, we address the potential mechanism(s) of Igf1r inhibitor resistance that might be anticipated for patients. Using a genetically engineered mouse model of ARMS, validated for active Igf1r signaling, we show that the prototypic Igf1r inhibitor NVP-AEW541 can inhibit cell growth and induce apoptosis in vitro in association with decreased Akt and Mapk phosphorylation. However, drug resistance in vivo is more common and is accompanied by Igf1r overexpression, Mapk reactivation, and Her2 overexpression. Her2 is found to form heterodimers with Igf1r in resistant primary tumor cell cultures, and stimulation with Igf2 leads to Her2 phosphorylation. The Her2 inhibitor lapatinib cooperates with NVP-AEW541 to reduce Igf1r phosphorylation and to inhibit cell growth even though lapatinib alone has little effect on growth. These results point to the potential therapeutic importance of simultaneous targeting of Igf1r and Her2 to abrogate resistance.


Subject(s)
Pyrimidines/pharmacology , Pyrroles/pharmacology , Receptor, IGF Type 1/antagonists & inhibitors , Rhabdomyosarcoma/metabolism , Animals , Apoptosis/drug effects , Blotting, Western , Cell Cycle/drug effects , Cell Proliferation/drug effects , Child , Chorioallantoic Membrane/drug effects , Chorioallantoic Membrane/pathology , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Embryo, Mammalian/drug effects , Embryo, Mammalian/pathology , Gene Expression Regulation, Neoplastic , Humans , Lapatinib , Mice , Phosphorylation/drug effects , Quail , Quinazolines/pharmacology , RNA Interference , Receptor, ErbB-2/antagonists & inhibitors , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptor, IGF Type 1/genetics , Receptor, IGF Type 1/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Rhabdomyosarcoma/genetics , Rhabdomyosarcoma/pathology , Tumor Burden/drug effects , Tumor Cells, Cultured , Young Adult
8.
J Biol Chem ; 285(52): 41044-50, 2010 Dec 24.
Article in English | MEDLINE | ID: mdl-20947509

ABSTRACT

Signaling by receptor tyrosine kinases regulates pancreatic ß cell function. Inactivation of insulin receptor (InsR), IGF1 receptor (Igf1r), or Irs1 in ß cells impairs insulin secretion. Conversely, Irs2 ablation impairs ß cell replication. In this study, we examined aspects of the Igf1r regulatory signaling cascade in ß cells. To examine genetically the involvement of Irs1 and Irs2 in Igf1r signaling, we generated double mutant mice lacking Igf1r specifically in pancreatic ß cells in an Irs1- or Irs2-null background. We show that Igf1r/Irs1 double mutants do not differ phenotypically from Irs1 single mutants and exhibit hyperinsulinemia, while maintaining normal ß cell mass and glucose tolerance. In contrast, lack of Igf1r function in ß cells aggravates the consequences of Irs2 ablation in double mutants and results in lethal diabetes by 6 weeks of age. This additivity of phenotypic manifestations indicates that Irs2 serves a pathway that is largely independent of Igf1r signaling. Consistent with the view that the latter is the InsR pathway, we show that combined ß cell-specific knock-out of both Insr and Igf1r results in a phenocopy of double mutants lacking Igf1r and Irs2. We conclude that Igf1r signals primarily through Irs1 and affects insulin secretion, whereas ß cell proliferation is mainly regulated by InsR using Irs2 as a downstream signaling effector. The insulin and IGF pathways appear to control ß cell functions independently and selectively.


Subject(s)
Insulin Receptor Substrate Proteins/metabolism , Insulin-Secreting Cells/metabolism , Receptor, IGF Type 1/metabolism , Signal Transduction/physiology , Animals , Cell Proliferation , Diabetes Mellitus/genetics , Diabetes Mellitus/mortality , Glucose/genetics , Glucose/metabolism , Humans , Hyperinsulinism/genetics , Hyperinsulinism/metabolism , Insulin/genetics , Insulin/metabolism , Insulin Receptor Substrate Proteins/genetics , Mice , Mice, Knockout , Receptor, IGF Type 1/genetics , Receptor, Insulin/genetics , Receptor, Insulin/metabolism
9.
J Am Soc Nephrol ; 20(11): 2315-27, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19762493

ABSTRACT

The kidney papilla contains a population of cells with several characteristics of adult stem cells, including the retention of proliferation markers during long chase periods (i.e., they are label-retaining cells [LRCs]). To determine whether the papillary LRCs generate new cells in the normal adult kidney, we examined cell proliferation throughout the kidney and found that the upper papilla is a site of enhanced cell cycling. Using genetically modified mice that conditionally expressed green fluorescence protein fused to histone 2B, we observed that the LRCs of the papilla proliferated only in its upper part, where they associate with "chains" of cycling cells. The papillary LRCs decreased in number with age, suggesting that the cells migrated to the upper papilla before entering the cell cycle. To test this directly, we marked papillary cells with vital dyes in vivo and found that some cells in the kidney papilla, including LRCs, migrated toward other parts of the kidney. Acute kidney injury enhanced both cell migration and proliferation. These results suggest that during normal homeostasis, LRCs of the kidney papilla (or their immediate progeny) migrate to the upper papilla and form a compartment of rapidly proliferating cells, which may play a role in repair after ischemic injury.


Subject(s)
Cell Movement , Cell Proliferation , Kidney/cytology , Age Factors , Animals , Kidney/growth & development , Rats , Staining and Labeling
10.
Nature ; 459(7249): 1000-4, 2009 Jun 18.
Article in English | MEDLINE | ID: mdl-19536265

ABSTRACT

T-cell acute lymphoblastic leukaemia (T-ALL) is a blood malignancy afflicting mainly children and adolescents. T-ALL patients present at diagnosis with increased white cell counts and hepatosplenomegaly, and are at an increased risk of central nervous system (CNS) relapse. For that reason, T-ALL patients usually receive cranial irradiation in addition to intensified intrathecal chemotherapy. The marked increase in survival is thought to be worth the considerable side-effects associated with this therapy. Such complications include secondary tumours, neurocognitive deficits, endocrine disorders and growth impairment. Little is known about the mechanism of leukaemic cell infiltration of the CNS, despite its clinical importance. Here we show, using T-ALL animal modelling and gene-expression profiling, that the chemokine receptor CCR7 (ref. 5) is the essential adhesion signal required for the targeting of leukaemic T-cells into the CNS. Ccr7 gene expression is controlled by the activity of the T-ALL oncogene Notch1 and is expressed in human tumours carrying Notch1-activating mutations. Silencing of either CCR7 or its chemokine ligand CCL19 (ref. 6) in an animal model of T-ALL specifically inhibits CNS infiltration. Furthermore, murine CNS-targeting by human T-ALL cells depends on their ability to express CCR7. These studies identify a single chemokine-receptor interaction as a CNS 'entry' signal, and open the way for future pharmacological targeting. Targeted inhibition of CNS involvement in T-ALL could potentially decrease the intensity of CNS-targeted therapy, thus reducing its associated short- and long-term complications.


Subject(s)
Central Nervous System/metabolism , Central Nervous System/pathology , Leukemia, T-Cell/metabolism , Leukemia, T-Cell/pathology , Receptors, CCR7/metabolism , Signal Transduction , Animals , Cell Adhesion , Cell Line, Tumor , Chemokine CCL19/deficiency , Chemokine CCL19/metabolism , Chemokine CCL21/metabolism , Humans , Mice , Mice, Inbred C57BL , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptors, CCR7/deficiency
11.
Proc Natl Acad Sci U S A ; 106(7): 2359-64, 2009 Feb 17.
Article in English | MEDLINE | ID: mdl-19174523

ABSTRACT

Considering the strong association between dysregulated insulin-like growth factor (IGF) signaling and various human cancers, we have used an expedient combination of genetic analysis and pharmacological treatment to evaluate the potential of the type 1 IGF receptor (Igf1r) for targeted anticancer therapy in a mouse model of mammary tumorigenesis. In this particular strain of genetically modified animals, histopathologically heterogeneous invasive carcinomas exhibiting up-regulation of the Igf1r gene developed extremely rapidly by mammary gland-specific overexpression of constitutively active oncogenic Kras* (mutant Kras(G12D)). Immunophenotyping data and expression profiling analyses showed that, except for a minor luminal component, these mouse tumors resembled basal-like human breast cancers. This is a group of aggressive tumors of poor prognosis for which there is no targeted therapy currently available, and it includes a subtype correlating with KRAS locus amplification. Conditional ablation of Igf1r in the mouse mammary epithelium increased the latency of Kras*-induced tumors very significantly (approximately 11-fold in comparison with the intact model), whereas treatment of tumor-bearing animals by administration of picropodophyllin (PPP), a specific Igf1r inhibitor, resulted in a dramatic decrease in tumor mass of the main forms of basal-like carcinomas. PPP also was effective against xenografts of the human basal-like cancer cell line MDA-MB-231, which carries a KRAS(G13D) mutation.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/therapy , Gene Expression Regulation, Neoplastic , Receptor, IGF Type 1/metabolism , Animals , Disease Models, Animal , Female , Gene Expression Profiling , Humans , Immunophenotyping , Mice , Mice, Transgenic , Models, Biological , Podophyllotoxin/analogs & derivatives , Podophyllotoxin/pharmacology , Signal Transduction , ras Proteins/metabolism
12.
Am J Pathol ; 174(1): 276-86, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19095950

ABSTRACT

Mutations in the phosphatase and tensin homologue (PTEN)/phosphatidylinositol-3 kinase-alpha (PI3K) signaling pathway are frequently found in human cancer. In addition, Pten(+/-) mice develop tumors in multiple organs because of the activation of the PI3K signaling cascade. Because activation of PI3K signaling leads to feedback inhibition of insulin receptor substrate-2 (IRS2) expression, an upstream activator of PI3K, we therefore anticipated that IRS2 expression would be low in tumors that lack PTEN. Surprisingly, however, an elevation of IRS2 was often detected in tumor samples in which PTEN levels were compromised. To determine the potential contribution of Irs2 to tumor progression, Pten(+/-) mice were crossed with Irs2(+/-) mice. Deletion of Irs2 did not affect the initiation of neoplasia found in Pten(+/-) mice but suppressed cancer cell growth, proliferation, and invasion through the basement membrane. Deletion of Irs2 also attenuated the expression of Myc in prostatic intraepithelial neoplasia in Pten(+/-) mice. In addition, the expression levels of IRS2 and MYC were highly correlated in human prostate cancer, and IRS2 could stimulate MYC expression in cultured cells. Our findings provide evidence that the PI3K-activating adaptor Irs2 contributes to tumor progression in Pten(+/-) mice by stimulating both Myc and DNA synthesis.


Subject(s)
Insulin Receptor Substrate Proteins/genetics , Insulin Receptor Substrate Proteins/metabolism , Neoplasms/genetics , Neoplasms/metabolism , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Disease Progression , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrial Neoplasms/pathology , Female , Humans , Immunohistochemistry , In Situ Hybridization , Male , Mice , Mice, Mutant Strains , Neoplasms/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Proto-Oncogene Proteins c-myc/metabolism , Transfection
13.
Proc Natl Acad Sci U S A ; 105(49): 19378-83, 2008 Dec 09.
Article in English | MEDLINE | ID: mdl-19033454

ABSTRACT

The mammalian insulin-like growth factor 1 (IGF1), which is a member of a major growth-promoting signaling system, is produced by many tissues and functions throughout embryonic and postnatal development in an autocrine/paracrine fashion. In addition to this local action, IGF1 secreted by the liver and circulating in the plasma presumably acts systemically as a classical hormone. However, an endocrine role of IGF1 in growth control was disputed on the basis of the results of a conditional, liver-specific Igf1 gene knockout in mice, which reduced significantly the level of serum IGF1, but did not affect average body weight. Because alternate interpretations of these negative data were tenable, we addressed genetically the question of hormonal IGF1 action by using a positive experimental strategy based on the features of the cre/loxP recombination system. Thus, we generated bitransgenic mice carrying in an Igf1 null background a dormant Igf1 cDNA placed downstream of a transcriptional "stop" DNA sequence flanked by loxP sites (floxed) and also a cre transgene driven by a liver-specific promoter. The Igf1 cDNA, which was inserted by knock-in into the mutated and inactive Igf1 locus itself to ensure proper transcriptional regulation, was conditionally expressed from cognate promoters exclusively in the liver after Cre-mediated excision of the floxed block. Our genetic study demonstrated that the endocrine IGF1 plays a very significant role in mouse growth, as its action contributes approximately30% of the adult body size and sustains postnatal development, including the reproductive functions of both mouse sexes.


Subject(s)
Endocrine System/growth & development , Endocrine System/physiology , Gene Expression Regulation, Developmental/physiology , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/metabolism , Animals , Base Sequence , Body Size/physiology , Female , Fertility/physiology , Gene Knock-In Techniques , Male , Mice , Mice, Transgenic , Molecular Sequence Data , Phenotype
14.
Neuron ; 57(6): 847-57, 2008 Mar 27.
Article in English | MEDLINE | ID: mdl-18367086

ABSTRACT

Olfactory neurons project their axons to spatially invariant glomeruli in the olfactory bulb, forming an ordered pattern of innervation comprising the olfactory sensory map. A mirror symmetry exists within this map, such that neurons expressing a given receptor typically project to one glomerulus on the medial face and one glomerulus on the lateral face of the bulb. The mechanisms underlying an olfactory neuron's choice to project medially versus laterally remain largely unknown, however. Here we demonstrate that insulin-like growth factor (IGF) signaling is required for sensory innervation of the lateral olfactory bulb. Mutations that eliminate IGF signaling cause axons destined for targets in the lateral bulb to shift to ectopic sites on the ventral-medial surface. Using primary cultures of olfactory and cerebellar neurons, we further show that IGF is a chemoattractant for axon growth cones. Together these observations reveal a role of IGF signaling in sensory map formation and axon guidance.


Subject(s)
Axons/physiology , Insulin-Like Growth Factor II/metabolism , Insulin-Like Growth Factor I/metabolism , Olfactory Pathways/physiology , Olfactory Receptor Neurons/cytology , Signal Transduction/physiology , Animals , Axons/drug effects , Cerebellum/cytology , Chemotactic Factors/pharmacology , Chromones/pharmacology , Embryo, Mammalian , Enzyme Inhibitors/pharmacology , Gene Expression Regulation, Developmental/physiology , In Vitro Techniques , Insulin-Like Growth Factor I/genetics , Insulin-Like Growth Factor I/pharmacology , Insulin-Like Growth Factor II/genetics , Insulin-Like Growth Factor II/pharmacology , Mice , Mice, Transgenic , Morpholines/pharmacology , Mutation/physiology , Olfactory Bulb/cytology , Olfactory Bulb/embryology , Olfactory Bulb/growth & development , Olfactory Marker Protein/genetics , Olfactory Marker Protein/metabolism , Olfactory Pathways/embryology , Olfactory Receptor Neurons/drug effects , Olfactory Receptor Neurons/physiology
15.
Cell Cycle ; 6(4): 418-29, 2007 Feb 15.
Article in English | MEDLINE | ID: mdl-17329972

ABSTRACT

The involvement of Myc in the development of human breast cancer is known for quite some time, but an analogous role of Notch signaling is only now emerging from fragmented pieces of information. Recently, a Notch/Myc relationship in oncogenesis was revealed from a mouse model, in which mammary tumors are induced by the intracellular domain of Notch1 (N1(IC)). In fact, a combination of genetic and molecular data demonstrated that Myc is a direct transcriptional target of Notch1 participating as an indispensable downstream effector in N1(IC)-induced tumorigenic action. A medically-relevant correlative observation based on immunophenotyping was the coexpression of Notch1 and Myc in a significant fraction of human breast cancer specimens. A Notch1/Myc oncogenic association was also observed in T-cell acute lymphoblastic leukemia.


Subject(s)
Breast Neoplasms/genetics , Carcinoma/genetics , Mammary Neoplasms, Experimental/genetics , Proto-Oncogene Proteins c-myc/physiology , Receptor, Notch1/physiology , Animals , Animals, Genetically Modified/metabolism , Breast/metabolism , Breast Neoplasms/metabolism , Carcinoma/metabolism , Evolution, Molecular , Humans , Leukemia-Lymphoma, Adult T-Cell/genetics , Mammary Glands, Animal/metabolism , Mammary Neoplasms, Experimental/metabolism , Mice , Phylogeny , Proto-Oncogene Proteins c-myc/genetics , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Notch1/genetics , Receptor, Notch1/metabolism
16.
Dev Biol ; 298(1): 327-33, 2006 Oct 01.
Article in English | MEDLINE | ID: mdl-16905129

ABSTRACT

The insulin-like growth factors (IGFs) play a major role in regulating the systemic growth of mammals. However, it is unclear to what extent their systemic and/or local functions act in concert with other local growth factors controlling the sizes of individual organs. We have specifically addressed whether growth control of the skeleton by IGFs interacts genetically with that by Indian hedgehog (Ihh), a locally produced growth signal for the endochondral skeleton. Here, we report that disruption of both IGF and Ihh signaling resulted in additive reduction in the size of the embryonic skeleton. Thus, IGF and Ihh signaling appear to control the growth of the skeleton in parallel pathways.


Subject(s)
Intracellular Signaling Peptides and Proteins/physiology , Osteogenesis , Signal Transduction , Skeleton , Somatomedins/physiology , Animals , Cartilage/metabolism , Cell Proliferation , Chondrocytes/physiology , Growth Plate/ultrastructure , Hedgehog Proteins , Intracellular Signaling Peptides and Proteins/genetics , Mice , Mice, Knockout , Somatomedins/genetics
17.
Proc Natl Acad Sci U S A ; 103(24): 9262-7, 2006 Jun 13.
Article in English | MEDLINE | ID: mdl-16751266

ABSTRACT

To explore the potential involvement of aberrant Notch1 signaling in breast cancer pathogenesis, we have used a transgenic mouse model. In these animals, mouse mammary tumor virus LTR-driven expression of the constitutively active intracellular domain of the Notch1 receptor (N1(IC)) causes development of lactation-dependent mammary tumors that regress upon gland involution but progress to nonregressing, invasive adenocarcinomas in subsequent pregnancies. Up-regulation of Myc in these tumors prompted a genetic investigation of a potential Notch1/Myc functional relationship in breast carcinogenesis. Conditional ablation of Myc in the mammary epithelium prevented the induction of regressing N1(IC) neoplasms and also reduced the incidence of nonregressing carcinomas, which developed with significantly increased latency. Molecular analyses revealed that both the mouse and human Myc genes are direct transcriptional targets of N1(IC) acting through its downstream Cbf1 transcriptional effector. Consistent with this mechanistic link, Notch1 and Myc expression is positively correlated by immunostaining in 38% of examined human breast carcinomas.


Subject(s)
Mammary Neoplasms, Experimental/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology , Animals , Base Sequence , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Immunophenotyping , Mammary Neoplasms, Experimental/genetics , Mammary Neoplasms, Experimental/pathology , Mice , Mice, Transgenic , Molecular Sequence Data , Pregnancy , Promoter Regions, Genetic , Proto-Oncogene Proteins c-myc/genetics , Receptor, Notch1/genetics , Survival Rate , Transcription, Genetic
18.
Carcinogenesis ; 27(5): 1054-67, 2006 May.
Article in English | MEDLINE | ID: mdl-16401639

ABSTRACT

NEU (ERBB2) and other members of the epidermal growth factor receptor (EGFR) family have been implicated in human prostate cancer (CAP) development and progression to an androgen-independent state, but the extent of involvement and precise role of this signaling pathway remain unclear. To begin addressing such open questions in an animal model, we have developed a transgenic line in which an oncogenic Neu cDNA (Neu*) driven by the probasin gene promoter is overexpressed in the mouse prostate and causes development of prostatic intraepithelial neoplasia (PIN) that progresses to invasive carcinoma. Expression profiling using microarrays, which was selectively validated and extended by immunophenotyping of Neu*-induced PIN and CAP, led to the identification of some novel biomarkers and also revealed increased expression of Egfr, Erbb3 and phosphorylated androgen receptor. In view of this information from our mouse model, which can be used to analyze further the role of Erbb signaling in prostatic tumorigenesis, we examined human prostate cancer tissue arrays by immunohistochemistry. Based on statistical analyses of the results, we propose the testable hypothesis that ERBB3, shown to be expressed in 86% of the human CAP cases that we examined, is the pivotal element of the Erbb pathway promoting tumorigenesis by heterodimerization with NEU or EGFR, while a NEU/EGFR dimer does not appear to play a significant role in CAP.


Subject(s)
Adenocarcinoma/genetics , Gene Expression Regulation, Neoplastic , Prostatic Intraepithelial Neoplasia/genetics , Receptor, ErbB-2/genetics , Transgenes , Adenocarcinoma/metabolism , Animals , ErbB Receptors/genetics , Gene Expression Profiling/methods , Humans , Immunophenotyping , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phosphorylation , Prostatic Intraepithelial Neoplasia/metabolism , Receptor, ErbB-3/genetics , Time Factors
19.
Mol Cell Biol ; 25(5): 1869-78, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15713641

ABSTRACT

Studies of Drosophila and mammals have revealed the importance of insulin signaling through phosphatidylinositol 3-kinase and the serine/threonine kinase Akt/protein kinase B for the regulation of cell, organ, and organismal growth. In mammals, three highly conserved proteins, Akt1, Akt2, and Akt3, comprise the Akt family, of which the first two are required for normal growth and metabolism, respectively. Here we address the function of Akt3. Like Akt1, Akt3 is not required for the maintenance of normal carbohydrate metabolism but is essential for the attainment of normal organ size. However, in contrast to Akt1-/- mice, which display a proportional decrease in the sizes of all organs, Akt3-/- mice present a selective 20% decrease in brain size. Moreover, although Akt1- and Akt3-deficient brains are reduced in size to approximately the same degree, the absence of Akt1 leads to a reduction in cell number, whereas the lack of Akt3 results in smaller and fewer cells. Finally, mammalian target of rapamycin signaling is attenuated in the brains of Akt3-/- but not Akt1-/- mice, suggesting that differential regulation of this pathway contributes to an isoform-specific regulation of cell growth.


Subject(s)
Brain/enzymology , Brain/growth & development , Oncogene Proteins/physiology , Protein Serine-Threonine Kinases/physiology , Animals , Blood Glucose/analysis , Body Weight/genetics , Body Weight/physiology , Brain/cytology , Female , Glucose/metabolism , Glucose Tolerance Test , Insulin/blood , Insulin/metabolism , Male , Mice , Mice, Knockout , Myocardium/cytology , Oncogene Proteins/genetics , Organ Size/genetics , Organ Size/physiology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/physiology , Proto-Oncogene Proteins c-akt , Ribosomal Protein S6 Kinases/metabolism
20.
Am J Pathol ; 165(2): 695-705, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15277242

ABSTRACT

Deregulation of Notch signaling, which normally affects a broad spectrum of cell fates, has been implicated in various neoplastic conditions. Here we describe a transgenic mouse model, which demonstrates that expression of a constitutively active form of the Notch1 receptor in the mammary epithelium induces the rapid development of pregnancy/lactation-dependent neoplasms that consistently exhibit a characteristic histopathological pattern. These signature tumors retain the ability to respond to apoptotic stimuli and regress on initiation of mammary gland involution, but eventually appear to progress in subsequent pregnancies to nonregressing malignant adenocarcinomas. Additionally, we present evidence indicating that cyclin D1 is an in vivo target of Notch signals in the mammary glands and demonstrate that we can effectively inhibit Hras1-driven, cyclin D1-dependent mammary oncogenesis by transgenic expression of the Notch antagonist Deltex.


Subject(s)
Cell Transformation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic , Mammary Glands, Animal/growth & development , Mammary Neoplasms, Animal/genetics , Mammary Tumor Virus, Mouse/physiology , Membrane Proteins/metabolism , Signal Transduction/physiology , Adenocarcinoma/pathology , Animals , Apoptosis , Carrier Proteins/metabolism , Cyclin D1 , Female , Genes, ras , Humans , Lactation , Male , Mammary Glands, Animal/embryology , Mammary Neoplasms, Animal/pathology , Mammary Tumor Virus, Mouse/genetics , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Pregnancy , Receptors, Notch
SELECTION OF CITATIONS
SEARCH DETAIL
...