Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Proc Natl Acad Sci U S A ; 113(35): 9928-33, 2016 08 30.
Article in English | MEDLINE | ID: mdl-27543332

ABSTRACT

The atomic-level mechanisms by which G protein-coupled receptors (GPCRs) transmit extracellular ligand binding events through their transmembrane helices to activate intracellular G proteins remain unclear. Using a comprehensive library of mutations covering all 352 residues of the GPCR CXC chemokine receptor 4 (CXCR4), we identified 41 amino acids that are required for signaling induced by the chemokine ligand CXCL12 (stromal cell-derived factor 1). CXCR4 variants with each of these mutations do not signal properly but remain folded, based on receptor surface trafficking, reactivity to conformationally sensitive monoclonal antibodies, and ligand binding. When visualized on the structure of CXCR4, the majority of these residues form a continuous intramolecular signaling chain through the transmembrane helices; this chain connects chemokine binding residues on the extracellular side of CXCR4 to G protein-coupling residues on its intracellular side. Integrated into a cohesive model of signal transmission, these CXCR4 residues cluster into five functional groups that mediate (i) chemokine engagement, (ii) signal initiation, (iii) signal propagation, (iv) microswitch activation, and (v) G protein coupling. Propagation of the signal passes through a "hydrophobic bridge" on helix VI that coordinates with nearly every known GPCR signaling motif. Our results agree with known conserved mechanisms of GPCR activation and significantly expand on understanding the structural principles of CXCR4 signaling.


Subject(s)
Protein Conformation , Receptors, CXCR4/chemistry , Receptors, CXCR4/metabolism , Signal Transduction , Amino Acid Sequence , Binding Sites/genetics , Chemokine CXCL12/chemistry , Chemokine CXCL12/metabolism , HEK293 Cells , Humans , Ligands , Models, Molecular , Mutation , Protein Binding , Protein Multimerization , Receptors, CXCR4/genetics , Sequence Homology, Amino Acid
2.
PLoS One ; 5(8): e12311, 2010 Aug 19.
Article in English | MEDLINE | ID: mdl-20808819

ABSTRACT

BACKGROUND: Pancreas organogenesis is the result of well-orchestrated and balanced activities of transcription factors. The homeobox transcription factor PDX-1 plays a crucial role in the development and function of the pancreas, both in the maintenance of progenitor cells and in determination and maintenance of differentiated endocrine cells. However, the activity of homeobox transcription factors requires coordination with co-factors, such as PBX and MEIS proteins. PBX and MEIS proteins belong to the family of three amino acid loop extension (TALE) homeodomain proteins. In a previous study we found that PDX-1 negatively regulates the transcriptional activity of the ductal specific keratin 19 (Krt19). In this study, we investigate the role of different domains of PDX-1 and elucidate the functional interplay of PDX-1 and MEIS1 necessary for Krt19 regulation. METHODOLOGY/PRINCIPAL FINDINGS: Here, we demonstrate that PDX-1 exerts a dual manner of regulation of Krt19 transcriptional activity. Deletion studies highlight that the NH(2)-terminus of PDX-1 is functionally relevant for the down-regulation of Krt19, as it is required for DNA binding of PDX-1 to the Krt19 promoter. Moreover, this effect occurs independently of PBX. Second, we provide insight on how PDX-1 regulates the Hox co-factor MEIS1 post-transcriptionally. We find specific binding of MEIS1 and MEIS2 to the Krt19 promoter using IP-EMSA, and siRNA mediated silencing of Meis1, but not Meis2, reduces transcriptional activation of Krt19 in primary pancreatic ductal cells. Over-expression of PDX-1 leads to a decreased level of MEIS1 protein, and this decrease is prevented by inhibition of the proteasome. CONCLUSIONS/SIGNIFICANCE: Taken together, our data provide evidence for a dual mechanism of how PDX-1 negatively regulates Krt19 ductal specific gene expression. These findings imply that transcription factors may efficiently regulate target gene expression through diverse, non-redundant mechanisms.


Subject(s)
DNA/metabolism , Homeodomain Proteins/metabolism , Keratin-19/metabolism , Neoplasm Proteins/metabolism , Pancreatic Ducts/metabolism , Trans-Activators/metabolism , Amino Acid Sequence , Animals , Cell Line , Down-Regulation , Genes, Reporter/genetics , Homeodomain Proteins/chemistry , Keratin-19/genetics , Mice , Mice, Inbred C57BL , Myeloid Ecotropic Viral Integration Site 1 Protein , Organ Specificity , Promoter Regions, Genetic/genetics , Protein Isoforms/metabolism , Protein Structure, Tertiary , Substrate Specificity , Trans-Activators/chemistry , Transcription, Genetic , Transcriptional Activation
3.
Mol Biol Cell ; 20(22): 4838-44, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19793922

ABSTRACT

Embryonic development of the pancreas is marked by an early phase of dramatic morphogenesis, in which pluripotent progenitor cells of the developing pancreatic epithelium give rise to the full array of mature exocrine and endocrine cell types. The genetic determinants of acinar and islet cell lineages are somewhat well defined; however, the molecular mechanisms directing ductal formation and differentiation remain to be elucidated. The complex ductal architecture of the pancreas is established by a reiterative program of progenitor cell expansion and migration known as branching morphogenesis, or tubulogenesis, which proceeds in mouse development concomitantly with peak Pdx1 transcription factor expression. We therefore evaluated Pdx1 expression with respect to lineage-specific markers in embryonic sections of the pancreas spanning this critical period of duct formation and discovered an unexpected population of nonislet Pdx1-positive cells displaying physical traits of branching. We then established a 3D cell culture model of branching morphogenesis using primary pancreatic duct cells and identified a transient surge of Pdx1 expression exclusive to branching cells. From these observations we propose that Pdx1 might be involved temporally in a program of gene expression sufficient to facilitate the biochemical and morphological changes necessary for branching morphogenesis.


Subject(s)
Homeodomain Proteins/metabolism , Morphogenesis/physiology , Pancreatic Ducts/embryology , Trans-Activators/metabolism , Animals , Biomarkers/metabolism , Cell Lineage , Cells, Cultured , Gene Expression Regulation, Developmental , Homeodomain Proteins/genetics , Mice , Mice, Knockout , Pancreatic Ducts/cytology , Pancreatic Ducts/physiology , Stem Cells/physiology , Trans-Activators/genetics
4.
Cell Cycle ; 8(4): 665-70, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19223768

ABSTRACT

In mammalian oocytes, meiosis arrests at prophase I. Meiotic resumption requires activation of Maturation-Promoting Factor (MPF), comprised of a catalytic Cyclin-dependent kinase-1 (Cdk1) and a regulatory subunit cyclin B and results in germinal vesicle breakdown (GVBD). Cyclic AMP (cAMP)-mediated Protein Kinase A (PKA) activity sustains prophase arrest by inhibiting Cdk1. However, the link between PKA activity and MPF inhibition remains unclear. Cdc25 phosphatases can activate Cdks by removing inhibitory phosphates from Cdks. Thus one method for sustaining prophase arrest could be inhibition of the activity of the Cdc25 protein required for MPF activation. Indeed, studies in Xenopus identify Cdc25C as a target of PKA activity in meiosis. However, in mice, studies suggest that Cdc25B is the phosphatase essential for GVBD and, therefore, the likely target of PKA activity. To assess these questions, we targeted a potential PKA substrate, a highly conserved serine 321 residue of Cdc25B and evaluated the effect on oocyte maturation. A Cdc25B-Ser321Ala point mutant mRNA induces GVBD when injected into prophase-arrested oocytes more rapidly than wild type mRNA. Using fluorescently-tagged proteins we also determined that the mutant protein enters the nucleus more rapidly than its wildtype counterpart. These data suggest that phosphorylation of the Ser321 residue plays a key role in the negative regulation and localization of Cdc25B during prophase arrest. PKA also phosphorylates a wildtype Cdc25B protein but not a Ser321Ala mutant protein in vitro. Mutation of Ser321 in Cdc25B also affects its association with a sequestering protein, 14-3-3. Our studies suggest that Cdc25B is a direct target of PKA in prophase-arrested oocytes and that Cdc25B phosphorylation results in its inhibition and sequestration by the 14-3-3 protein.


Subject(s)
Cyclic AMP-Dependent Protein Kinases/metabolism , Meiosis/physiology , Oocytes/physiology , cdc25 Phosphatases/metabolism , 14-3-3 Proteins/metabolism , Animals , Cell Line , Cyclic AMP-Dependent Protein Kinases/genetics , Female , Mesothelin , Mice , Mutagenesis, Site-Directed , Phosphorylation , Serine/metabolism , cdc25 Phosphatases/genetics
5.
Cancer Prev Res (Phila) ; 1(7): 503-6, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19138999

ABSTRACT

Pancreatic ductal adenocarcinoma is the overwhelmingly predominant form of pancreatic cancer and the second most common type of gastrointestinal cancer (behind colorectal cancer) in the United States. Recent exciting advances in two areas of pancreatic ductal adenocarcinoma (i.e., the development and characterization of genetically engineered mouse models and the dissection of the genetic basis of hereditary forms in families) have been illuminating. These preclinical models and clinical syndromes provide the first tangible basis for progress in screening and prevention in high-risk populations and in the development of molecular diagnostics and experimental therapeutics.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Genetic Predisposition to Disease , Pancreatic Neoplasms/genetics , Animals , Biomarkers, Tumor/genetics , Carcinoma, Pancreatic Ductal/prevention & control , Disease Models, Animal , Genetic Testing , Humans , Mice , Pancreatic Neoplasms/prevention & control , Pancreatitis/etiology
6.
J Biol Chem ; 281(50): 38385-95, 2006 Dec 15.
Article in English | MEDLINE | ID: mdl-17056599

ABSTRACT

Keratin 19 is a member of the cytokeratin family that is critical for maintenance of cellular architecture and organization, especially of epithelia. The pancreas has three distinct cell types, ductal, acinar, and islet, each with different functions. Embryologically, the pancreatic and duodenal homeobox 1 (PDX1) homeodomain protein is critical for the initiation of all pancreatic lineages; however, the later differentiation of the endocrine pancreas is uniquely dependent upon high PDX1 expression, whereas PDX1 is down-regulated in the ductal and acinar cell lineages. We find that this down-regulation may be required for normal ductal expression of cytokeratin K19. The K19 promoter-reporter gene assay demonstrates that ectopic PDX1 inhibits K19 reporter gene activity in primary pancreatic ductal cells. This is reinforced by our findings that retrovirally mediated stable transduction of PDX1 in primary pancreatic ductal cells suppresses K19 expression, and short interfering RNA to PDX1 in Min6 insulinoma cells results in the induction of normally undetectable K19. Complementary functional and biochemical approaches led to the unexpected finding that a multimeric complex of PDX1 and two members of the TALE homeodomain factor family, MEIS1a and PBX1b, regulates K19 gene transcription through a specific cis-regulatory element (-341 to -325) upstream of the K19 transcription start site. These data suggest a unifying mechanism whereby PDX1, myeloid ecotropic viral insertion site (MEIS), and pre-B-cell leukemia transcription factor 1 (PBX) may regulate ductal and acinar lineage specification during pancreatic development. Specifically, concomitant PDX1 suppression and MEIS isoform expression result in proper ductal and acinar lineage specification. Furthermore, PDX1 may inhibit the ductal differentiation program in the pancreatic endocrine compartment, particularly beta cells.


Subject(s)
Homeodomain Proteins/physiology , Keratin-19/genetics , Pancreas/metabolism , Promoter Regions, Genetic , Trans-Activators/physiology , Animals , Base Sequence , Cell Line , DNA Primers , Genes, Reporter , Insulinoma/genetics , Insulinoma/pathology , Mice , Myeloid Ecotropic Viral Integration Site 1 Protein , Neoplasm Proteins/physiology , Pancreas/cytology
SELECTION OF CITATIONS
SEARCH DETAIL
...