Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Int J Mol Sci ; 25(12)2024 Jun 16.
Article in English | MEDLINE | ID: mdl-38928326

ABSTRACT

Diagnostic markers are desperately needed for the early detection of pancreatic ductal adenocarcinoma (PDA). We describe sets of markers expressed in temporal order in mouse models during pancreatitis, PDA initiation and progression. Cell type specificity and the differential expression of PDA markers were identified by screening single cell (sc) RNAseq from tumor samples of a mouse model for PDA (KIC) at early and late stages of PDA progression compared to that of a normal pancreas. Candidate genes were identified from three sources: (1) an unsupervised screening of the genes preferentially expressed in mouse PDA tumors; (2) signaling pathways that drive PDA, including the Ras pathway, calcium signaling, and known cancer genes, or genes encoding proteins that were identified by differential mass spectrometry (MS) of mouse tumors and conditioned media from human cancer cell lines; and (3) genes whose expression is associated with poor or better prognoses (PAAD, oncolnc.org). The developmental progression of PDA was detected in the temporal order of gene expression in the cancer cells of the KIC mice. The earliest diagnostic markers were expressed in epithelial cancer cells in early-stage, but not late-stage, PDA tumors. Other early markers were expressed in the epithelium of both early- and late-state PDA tumors. Markers that were expressed somewhat later were first elevated in the epithelial cancer cells of the late-stage tumors, then in both epithelial and mesenchymal cells, or only in mesenchymal cells. Stromal markers were differentially expressed in early- and/or late-stage PDA neoplasia in fibroblast and hematopoietic cells (lymphocytes and/or macrophages) or broadly expressed in cancer and many stromal cell types. Pancreatitis is a risk factor for PDA in humans. Mouse models of pancreatitis, including caerulein treatment and the acinar-specific homozygous deletion of differentiation transcription factors (dTFs), were screened for the early expression of all PDA markers identified in the KIC neoplasia. Prognostic markers associated with a more rapid decline were identified and showed differential and cell-type-specific expression in PDA, predominately in late-stage epithelial and/or mesenchymal cancer cells. Select markers were validated by immunohistochemistry in mouse and human samples of a normal pancreas and those with early- and late-stage PDA. In total, we present 2165 individual diagnostic and prognostic markers for disease progression to be tested in humans from pancreatitis to late-stage PDA.


Subject(s)
Biomarkers, Tumor , Carcinoma, Pancreatic Ductal , Pancreatic Neoplasms , Pancreatitis , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/diagnosis , Carcinoma, Pancreatic Ductal/pathology , Pancreatitis/metabolism , Pancreatitis/genetics , Pancreatitis/pathology , Pancreatitis/diagnosis , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Humans , Prognosis , Gene Expression Regulation, Neoplastic , Disease Models, Animal , Cell Line, Tumor , Disease Progression
2.
PLoS One ; 18(10): e0291512, 2023.
Article in English | MEDLINE | ID: mdl-37796967

ABSTRACT

Proper maintenance of mature cellular phenotypes is essential for stable physiology, suppression of disease states, and resistance to oncogenic transformation. We describe the transcriptional regulatory roles of four key DNA-binding transcription factors (Ptf1a, Nr5a2, Foxa2 and Gata4) that sit at the top of a regulatory hierarchy controlling all aspects of a highly differentiated cell-type-the mature pancreatic acinar cell (PAC). Selective inactivation of Ptf1a, Nr5a2, Foxa2 and Gata4 individually in mouse adult PACs rapidly altered the transcriptome and differentiation status of PACs. The changes most emphatically included transcription of the genes for the secretory digestive enzymes (which conscript more than 90% of acinar cell protein synthesis), a potent anabolic metabolism that provides the energy and materials for protein synthesis, suppressed and properly balanced cellular replication, and susceptibility to transformation by oncogenic KrasG12D. The simultaneous inactivation of Foxa2 and Gata4 caused a greater-than-additive disruption of gene expression and uncovered their collaboration to maintain Ptf1a expression and control PAC replication. A measure of PAC dedifferentiation ranked the effects of the conditional knockouts as Foxa2+Gata4 > Ptf1a > Nr5a2 > Foxa2 > Gata4. Whereas the loss of Ptf1a or Nr5a2 greatly accelerated Kras-mediated transformation of mature acinar cells in vivo, the absence of Foxa2, Gata4, or Foxa2+Gata4 together blocked transformation completely, despite extensive dedifferentiation. A lack of correlation between PAC dedifferentiation and sensitivity to oncogenic KrasG12D negates the simple proposition that the level of differentiation determines acinar cell resistance to transformation.


Subject(s)
Pancreas, Exocrine , Pancreatic Neoplasms , Mice , Animals , Acinar Cells/metabolism , Epithelium/metabolism , Transcription Factors/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Phenotype , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism
3.
Cell Calcium ; 94: 102340, 2021 03.
Article in English | MEDLINE | ID: mdl-33601151

ABSTRACT

The recent paper by Pfeil et al., "Heterotrimeric G Protein Subunit Gαq Is a Master Switch for Gßγ-Mediated Calcium Mobilization by Gi-Coupled GPCRs", opens another path from biochemical in vitro reconstitution to understanding the complex regulation of calcium signaling inside the cell.


Subject(s)
Calcium , GTP-Binding Protein alpha Subunits, Gq-G11 , Calcium/metabolism , Calcium Signaling , GTP-Binding Protein alpha Subunits, Gq-G11/metabolism , Phospholipase C beta/metabolism
4.
Am J Pathol ; 188(3): 616-626, 2018 03.
Article in English | MEDLINE | ID: mdl-29248457

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is a deadly cancer that resists efforts to identify better chemotherapeutics. PDA is associated with chronic pancreatitis and acinar cell dedifferentiation. This reduces enzyme production by the exocrine pancreas, resulting in digestive insufficiencies. Malabsorption of partially digested food causes bloating, overfilled intestines, abdominal pain, excessive feces, steatorrhea, and malnutrition. These maladies affect quality of life and restrict treatment options for pancreatitis and PDA. Here, we characterize health benefits and risks of dietary pancreatic enzymes in three mouse models of PDA-KC, KCR8-16, and KIC. KC expresses oncogenic KrasG12D in pancreatic tissue whereas KCR8-16 also has deletions of the Rgs8 and Rgs16 genes. Rgs proteins inhibit the release of digestive enzymes evoked by G-protein-coupled-receptor agonists. KC and KCR8-16 mice developed dedifferentiated exocrine pancreata within 2 months of age and became malnourished, underweight, hypoglycemic, and hypothermic. KC mice adapted but KCR8-16 mice rapidly transitioned to starvation after mild metabolic challenges. Dietary pancreatic enzyme supplements reversed these symptoms in KC and KCR8-16 animals, and extended survival. Therefore, we tested the benefits of pancreatic enzymes in an aggressive mouse model of PDA (KIC). Median survival improved with dietary pancreatic enzyme supplements and was extended further when combined with warfarin and gemcitabine chemotherapy. However, dietary pancreatic enzymes stimulated tumor growth in the terminal stages of disease progression in KIC mice.


Subject(s)
Carcinoma, Pancreatic Ductal/complications , Malnutrition/drug therapy , Pancreatic Neoplasms/complications , Animals , Blood Glucose , Carcinoma, Pancreatic Ductal/pathology , Disease Models, Animal , Disease Progression , Eating , Female , Insulin/blood , Male , Malnutrition/etiology , Malnutrition/pathology , Mice , Pancreatic Neoplasms/pathology
5.
Mol Cell Biol ; 36(24): 3033-3047, 2016 12 15.
Article in English | MEDLINE | ID: mdl-27697859

ABSTRACT

Maintenance of cell type identity is crucial for health, yet little is known of the regulation that sustains the long-term stability of differentiated phenotypes. To investigate the roles that key transcriptional regulators play in adult differentiated cells, we examined the effects of depletion of the developmental master regulator PTF1A on the specialized phenotype of the adult pancreatic acinar cell in vivo Transcriptome sequencing and chromatin immunoprecipitation sequencing results showed that PTF1A maintains the expression of genes for all cellular processes dedicated to the production of the secretory digestive enzymes, a highly attuned surveillance of unfolded proteins, and a heightened unfolded protein response (UPR). Control by PTF1A is direct on target genes and indirect through a ten-member transcription factor network. Depletion of PTF1A causes an imbalance that overwhelms the UPR, induces cellular injury, and provokes acinar metaplasia. Compromised cellular identity occurs by derepression of characteristic stomach genes, some of which are also associated with pancreatic ductal cells. The loss of acinar cell homeostasis, differentiation, and identity is directly relevant to the pathologies of pancreatitis and pancreatic adenocarcinoma.


Subject(s)
Acinar Cells/cytology , Gene Expression Profiling/methods , Pancreas, Exocrine/cytology , Transcription Factors/genetics , Transcription, Genetic , Acinar Cells/metabolism , Animals , Cell Differentiation , Gene Expression Regulation , Gene Knockout Techniques , Homeostasis , Mice , Pancreas, Exocrine/metabolism , Protein Unfolding , Sequence Analysis, RNA/methods , Transcription Factors/metabolism , Unfolded Protein Response
6.
Dis Model Mech ; 8(10): 1201-11, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26438693

ABSTRACT

Pancreatic ductal adenocarcinoma (PDA) is the fourth leading cause of cancer-related deaths in the United States, and is projected to be second by 2025. It has the worst survival rate among all major cancers. Two pressing needs for extending life expectancy of affected individuals are the development of new approaches to identify improved therapeutics, addressed herein, and the identification of early markers. PDA advances through a complex series of intercellular and physiological interactions that drive cancer progression in response to organ stress, organ failure, malnutrition, and infiltrating immune and stromal cells. Candidate drugs identified in organ culture or cell-based screens must be validated in preclinical models such as KIC (p48(Cre);LSL-Kras(G12D);Cdkn2a(f/f)) mice, a genetically engineered model of PDA in which large aggressive tumors develop by 4 weeks of age. We report a rapid, systematic and robust in vivo screen for effective drug combinations to treat Kras-dependent PDA. Kras mutations occur early in tumor progression in over 90% of human PDA cases. Protein kinase and G-protein coupled receptor (GPCR) signaling activates Kras. Regulators of G-protein signaling (RGS) proteins are coincidence detectors that can be induced by multiple inputs to feedback-regulate GPCR signaling. We crossed Rgs16::GFP bacterial artificial chromosome (BAC) transgenic mice with KIC mice and show that the Rgs16::GFP transgene is a Kras(G12D)-dependent marker of all stages of PDA, and increases proportionally to tumor burden in KIC mice. RNA sequencing (RNA-Seq) analysis of cultured primary PDA cells reveals characteristics of embryonic progenitors of pancreatic ducts and endocrine cells, and extraordinarily high expression of the receptor tyrosine kinase Axl, an emerging cancer drug target. In proof-of-principle drug screens, we find that weanling KIC mice with PDA treated for 2 weeks with gemcitabine (with or without Abraxane) plus inhibitors of Axl signaling (warfarin and BGB324) have fewer tumor initiation sites and reduced tumor size compared with the standard-of-care treatment. Rgs16::GFP is therefore an in vivo reporter of PDA progression and sensitivity to new chemotherapeutic drug regimens such as Axl-targeted agents. This screening strategy can potentially be applied to identify improved therapeutics for other cancers.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Drug Evaluation, Preclinical , Pancreatic Neoplasms/drug therapy , Albumin-Bound Paclitaxel/pharmacology , Albumin-Bound Paclitaxel/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Biological Assay , Carcinogenesis/pathology , Carcinoma, Pancreatic Ductal/pathology , Cell Proliferation , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Deoxycytidine/therapeutic use , Genes, Reporter , Green Fluorescent Proteins/metabolism , Humans , Mice , Pancreatic Neoplasms/pathology , Proto-Oncogene Proteins/antagonists & inhibitors , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , RGS Proteins/metabolism , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Receptor Protein-Tyrosine Kinases/metabolism , Gemcitabine , Axl Receptor Tyrosine Kinase , Pancreatic Neoplasms
7.
Development ; 141(22): 4385-94, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25371369

ABSTRACT

The timing and gene regulatory logic of organ-fate commitment from within the posterior foregut of the mammalian endoderm is largely unexplored. Transient misexpression of a presumed pancreatic-commitment transcription factor, Ptf1a, in embryonic mouse endoderm (Ptf1a(EDD)) dramatically expanded the pancreatic gene regulatory network within the foregut. Ptf1a(EDD) temporarily suppressed Sox2 broadly over the anterior endoderm. Pancreas-proximal organ territories underwent full tissue conversion. Early-stage Ptf1a(EDD) rapidly expanded the endogenous endodermal Pdx1-positive domain and recruited other pancreas-fate-instructive genes, thereby spatially enlarging the potential for pancreatic multipotency. Early Ptf1a(EDD) converted essentially the entire glandular stomach, rostral duodenum and extrahepatic biliary system to pancreas, with formation of many endocrine cell clusters of the type found in normal islets of Langerhans. Sliding the Ptf1a(EDD) expression window through embryogenesis revealed differential temporal competencies for stomach-pancreas respecification. The response to later-stage Ptf1a(EDD) changed radically towards unipotent, acinar-restricted conversion. We provide strong evidence, beyond previous Ptf1a inactivation or misexpression experiments in frog embryos, for spatiotemporally context-dependent activity of Ptf1a as a potent gain-of-function trigger of pro-pancreatic commitment.


Subject(s)
Endoderm/embryology , Gastrointestinal Tract/embryology , Gene Expression Regulation, Developmental/physiology , Organogenesis/physiology , Pancreas/embryology , Transcription Factors/metabolism , Animals , Endoderm/metabolism , Fluorescent Antibody Technique , Gastrointestinal Tract/metabolism , Gene Expression Regulation, Developmental/genetics , Gene Regulatory Networks/genetics , Histological Techniques , Mice , Microscopy, Confocal , Organogenesis/genetics , SOXB1 Transcription Factors/metabolism
8.
Development ; 141(16): 3123-33, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25063451

ABSTRACT

The orphan nuclear receptor NR5A2 is necessary for the stem-like properties of the epiblast of the pre-gastrulation embryo and for cellular and physiological homeostasis of endoderm-derived organs postnatally. Using conditional gene inactivation, we show that Nr5a2 also plays crucial regulatory roles during organogenesis. During the formation of the pancreas, Nr5a2 is necessary for the expansion of the nascent pancreatic epithelium, for the subsequent formation of the multipotent progenitor cell (MPC) population that gives rise to pre-acinar cells and bipotent cells with ductal and islet endocrine potential, and for the formation and differentiation of acinar cells. At birth, the NR5A2-deficient pancreas has defects in all three epithelial tissues: a partial loss of endocrine cells, a disrupted ductal tree and a >90% deficit of acini. The acinar defects are due to a combination of fewer MPCs, deficient allocation of those MPCs to pre-acinar fate, disruption of acinar morphogenesis and incomplete acinar cell differentiation. NR5A2 controls these developmental processes directly as well as through regulatory interactions with other pancreatic transcriptional regulators, including PTF1A, MYC, GATA4, FOXA2, RBPJL and MIST1 (BHLHA15). In particular, Nr5a2 and Ptf1a establish mutually reinforcing regulatory interactions and collaborate to control developmentally regulated pancreatic genes by binding to shared transcriptional regulatory regions. At the final stage of acinar cell development, the absence of NR5A2 affects the expression of Ptf1a and its acinar specific partner Rbpjl, so that the few acinar cells that form do not complete differentiation. Nr5a2 controls several temporally distinct stages of pancreatic development that involve regulatory mechanisms relevant to pancreatic oncogenesis and the maintenance of the exocrine phenotype.


Subject(s)
Acinar Cells/cytology , Gene Expression Regulation, Developmental , Pancreas/embryology , Pancreas/growth & development , Receptors, Cytoplasmic and Nuclear/physiology , Stem Cells/cytology , Animals , Base Sequence , Cell Differentiation , Cell Lineage , Cell Proliferation , Male , Mice , Mice, Transgenic , Mutation , Phenotype , Receptors, Cytoplasmic and Nuclear/genetics , Transgenes
9.
J Clin Invest ; 122(10): 3516-28, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23006325

ABSTRACT

Pancreatic agenesis is a human disorder caused by defects in pancreas development. To date, only a few genes have been linked to pancreatic agenesis in humans, with mutations in pancreatic and duodenal homeobox 1 (PDX1) and pancreas-specific transcription factor 1a (PTF1A) reported in only 5 families with described cases. Recently, mutations in GATA6 have been identified in a large percentage of human cases, and a GATA4 mutant allele has been implicated in a single case. In the mouse, Gata4 and Gata6 are expressed in several endoderm-derived tissues, including the pancreas. To analyze the functions of GATA4 and/or GATA6 during mouse pancreatic development, we generated pancreas-specific deletions of Gata4 and Gata6. Surprisingly, loss of either Gata4 or Gata6 in the pancreas resulted in only mild pancreatic defects, which resolved postnatally. However, simultaneous deletion of both Gata4 and Gata6 in the pancreas caused severe pancreatic agenesis due to disruption of pancreatic progenitor cell proliferation, defects in branching morphogenesis, and a subsequent failure to induce the differentiation of progenitor cells expressing carboxypeptidase A1 (CPA1) and neurogenin 3 (NEUROG3). These studies address the conserved and nonconserved mechanisms underlying GATA4 and GATA6 function during pancreas development and provide a new mouse model to characterize the underlying developmental defects associated with pancreatic agenesis.


Subject(s)
GATA4 Transcription Factor/physiology , GATA6 Transcription Factor/physiology , Gene Expression Regulation, Developmental/genetics , Organogenesis/genetics , Pancreas/embryology , Animals , Basic Helix-Loop-Helix Transcription Factors/analysis , Binding Sites , Carboxypeptidases A/analysis , Cell Differentiation , Cell Division , Cell Lineage , Disease Models, Animal , Endoderm/metabolism , Epithelial Cells/pathology , GATA4 Transcription Factor/deficiency , GATA4 Transcription Factor/genetics , GATA6 Transcription Factor/deficiency , GATA6 Transcription Factor/genetics , Gene Knockdown Techniques , Genotype , Gestational Age , Hyperglycemia/congenital , Hyperglycemia/genetics , Insulin/metabolism , Insulin Secretion , Mice , Nerve Tissue Proteins/analysis , Organ Specificity , Pancreas/abnormalities , Pancreas/pathology , Transcription, Genetic
10.
Mol Cell Biol ; 28(17): 5458-68, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18606784

ABSTRACT

The basic helix-loop-helix (bHLH) transcription factor PTF1a is critical to the development of the embryonic pancreas. It is required early for the formation of the undifferentiated tubular epithelium of the nascent pancreatic rudiment and then becomes restricted to the differentiating acinar cells, where it directs the transcriptional activation of the secretory digestive enzyme genes. Here we report that the complex temporal and spatial expression of Ptf1a is controlled by at least three separable gene-flanking regions. A 14.8-kb control domain immediately downstream of the last Ptf1a exon is highly conserved among mammals and directs expression in the dorsal part of the spinal cord but has very little activity in the embryonic or neonatal pancreas. A 13.4-kb proximal promoter domain initiates limited expression in cells that begin the acinar differentiation program. The activity of the proximal promoter domain is complemented by an adjacent 2.3-kb autoregulatory enhancer that is able to activate a heterologous minimal promoter with high-level penetrance in the pancreases of transgenic mice. During embryonic development, the enhancer initiates expression in the early precursor epithelium and then superinduces expression in acinar cells at the onset of their development. The enhancer contains two evolutionarily conserved binding sites for the active form of PTF1a, a trimeric complex composed of PTF1a, one of the common bHLH E proteins, and either RBPJ or RBPJL. The two sites are essential for acinar cell-specific transcription in transfected cell lines and mice. In mature acinar cells, the enhancer and PTF1a establish an autoregulatory loop that reinforces and maintains Ptf1a expression. Indeed, the trimeric PTF1 complex forms dual autoregulatory loops with the Ptf1a and Rbpjl genes that may maintain the stable phenotype of pancreatic acinar cells.


Subject(s)
Gene Expression Regulation, Developmental , Pancreas/embryology , Pancreas/growth & development , Transcription Factors/genetics , Transcription, Genetic , 5' Flanking Region/genetics , Animals , Base Sequence , Binding Sites , Cell Line , Conserved Sequence , Electrophoretic Mobility Shift Assay , Enhancer Elements, Genetic/genetics , Epithelium/metabolism , Humans , Mice , Models, Genetic , Molecular Sequence Data , Pancreas/cytology , Promoter Regions, Genetic/genetics , Protein Binding , Rats , Vertebrates/genetics
11.
Dev Biol ; 286(1): 225-37, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16126192

ABSTRACT

The homeoprotein PDX1 is expressed throughout pancreatic development and is thought to play important roles at multiple stages. We describe the properties of a tet-off regulatory scheme to manage the expression of Pdx1 in utero. Cessation of Pdx1 expression at increasingly later gestational times blocked pancreatic development at progressive and morphologically distinct stages and provided the opportunity to assess the requirement for Pdx1 at each stage. Embryonic PDX1 is depleted below effective levels within 1 day of the initiation of doxycycline treatment of pregnant mice. We show that PDX1, which is necessary for early pancreatic development, is also required later for the genesis of acinar tissue, the compartment of the pancreas that produces digestive enzymes. Without PDX1, acini do not form; the precursor epithelium continues to grow and branch, creating a truncated ductal tree comprising immature duct-like cells. The bHLH factor PTF1a, a critical regulator of acinar development, is not expressed and cells producing digestive enzymes are rare. This approach should be generally applicable to study the in vivo functions of other developmental regulators with multiple, temporally distinct roles.


Subject(s)
Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Pancreas/embryology , Pancreas/metabolism , Trans-Activators/genetics , Trans-Activators/metabolism , Animals , Doxycycline/pharmacology , Female , Gene Expression Regulation, Developmental , Genes, Reporter , Gestational Age , Mice , Mice, Knockout , Mice, Transgenic , Pancreas/drug effects , Pregnancy , Trans-Activators/deficiency
12.
Proc Natl Acad Sci U S A ; 99(19): 12236-41, 2002 Sep 17.
Article in English | MEDLINE | ID: mdl-12221286

ABSTRACT

To investigate the role of the HOX-like homeoprotein PDX1 in the formation and maintenance of the pancreas, we have genetically engineered mice so that the only source of PDX1 is a transgene that can be controlled by the application of tetracycline or its analogue doxycycline. In these mice the coding region for the tetracycline-regulated transactivator (tTA(off)) has replaced the coding region of the endogenous Pdx1 gene to ensure correct temporal and spatial expression of the regulatable transactivator. In the absence of doxycycline, tTA(off) activates the transcription of a bicistronic transgene encoding PDX1 and an enhanced green fluorescent protein reporter, which acts as a visual marker of transgene expression in living cells. Expression of the transgene-encoded PDX1 rescues the Pdx1-null phenotype; the pancreata of these mice develop and function normally. The rescue is conditional; doxycycline-mediated repression of the transgenic Pdx1 throughout gestation recapitulates the Pdx1 null phenotype. Moreover, application of doxycycline at mid-pancreogenesis blocks further development. Adult animals of the rescue genotype that were treated with doxycycline for 3 weeks shut off Pdx1 expression, decreased insulin production, and lost the ability to maintain glucose homeostasis. These results demonstrate the feasibility of controlling the formation of an organ during embryogenesis in utero and the maintenance of the mature organ through the experimental manipulation of a key developmental regulator.


Subject(s)
Carrier Proteins , Homeodomain Proteins/physiology , Pancreas/growth & development , Pancreas/physiology , Trans-Activators/physiology , Animals , Bacterial Proteins/genetics , Doxycycline/pharmacology , Female , Gene Expression/drug effects , Genes, Homeobox , Genes, Reporter , Glucose/metabolism , Green Fluorescent Proteins , Homeodomain Proteins/genetics , Insulin/biosynthesis , Luminescent Proteins/genetics , Mice , Mice, Knockout , Mice, Transgenic , Pancreas/drug effects , Pancreas/embryology , Phenotype , Pregnancy , Tetracycline/pharmacology , Trans-Activators/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...