Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
Add more filters










Publication year range
1.
Nat Commun ; 10(1): 632, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30733432

ABSTRACT

To reveal how cells exit human pluripotency, we designed a CRISPR-Cas9 screen exploiting the metabolic and epigenetic differences between naïve and primed pluripotent cells. We identify the tumor suppressor, Folliculin(FLCN) as a critical gene required for the exit from human pluripotency. Here we show that FLCN Knock-out (KO) hESCs maintain the naïve pluripotent state but cannot exit the state since the critical transcription factor TFE3 remains active in the nucleus. TFE3 targets up-regulated in FLCN KO exit assay are members of Wnt pathway and ESRRB. Treatment of FLCN KO hESC with a Wnt inhibitor, but not ESRRB/FLCN double mutant, rescues the cells, allowing the exit from the naïve state. Using co-immunoprecipitation and mass spectrometry analysis we identify unique FLCN binding partners. The interactions of FLCN with components of the mTOR pathway (mTORC1 and mTORC2) reveal a mechanism of FLCN function during exit from naïve pluripotency.


Subject(s)
Mechanistic Target of Rapamycin Complex 1/metabolism , Mechanistic Target of Rapamycin Complex 2/metabolism , Wnt Signaling Pathway/physiology , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/genetics , Basic Helix-Loop-Helix Leucine Zipper Transcription Factors/metabolism , CRISPR-Cas Systems/genetics , CRISPR-Cas Systems/physiology , Cell Line , Estrone/genetics , Estrone/metabolism , Humans , Immunoprecipitation , Mechanistic Target of Rapamycin Complex 1/genetics , Mechanistic Target of Rapamycin Complex 2/genetics , Proteomics , Receptors, Estrogen/genetics , Receptors, Estrogen/metabolism , Wnt Signaling Pathway/genetics
2.
Oncogene ; 36(22): 3119-3136, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28092677

ABSTRACT

Aberrant regulation of WNT/ß-catenin signaling has a crucial role in the onset and progression of cancers, where the effects are not always predictable depending on tumor context. In melanoma, for example, models of the disease predict differing effects of the WNT/ß-catenin pathway on metastatic progression. Understanding the processes that underpin the highly context-dependent nature of WNT/ß-catenin signaling in tumors is essential to achieve maximal therapeutic benefit from WNT inhibitory compounds. In this study, we have found that expression of the tumor suppressor, phosphatase and tensin homolog deleted on chromosome 10 (PTEN), alters the invasive potential of melanoma cells in response to WNT/ß-catenin signaling, correlating with differing metabolic profiles. This alters the bioenergetic potential and mitochondrial activity of melanoma cells, triggered through regulation of pro-survival autophagy. Thus, WNT/ß-catenin signaling is a regulator of catabolic processes in cancer cells, which varies depending on the metabolic requirements of tumors.


Subject(s)
Melanoma/metabolism , Mitochondria/metabolism , PTEN Phosphohydrolase/genetics , Wnt Signaling Pathway , beta Catenin/metabolism , Animals , Humans , Male , Melanoma/enzymology , Melanoma/genetics , Mice , Mice, Inbred NOD , Mice, SCID , PTEN Phosphohydrolase/metabolism
3.
Oncogene ; 33(7): 899-908, 2014 Feb 13.
Article in English | MEDLINE | ID: mdl-23416978

ABSTRACT

Developmental and cancer models show Wnt/ß-catenin-dependent signaling mediates diverse phenotypic outcomes in the pancreas that are dictated by context, duration and strength of activation. While generally assumed to be pro-tumorigenic, it is unclear to what extent dysregulation of Wnt/ß-catenin signaling impacts tumor progression in pancreatic adenocarcinoma (PDAC). In the present study, Wnt/ß-catenin activity was characterized across a spectrum of PDAC cell lines and primary tumors. Reporter and gene expression-based assays revealed wide heterogeneity in Wnt/ß-catenin transcriptional activity across PDAC cell lines and patient tumors, as well as variable responsiveness to exogenous Wnt ligand stimulation. An experimentally generated, pancreas-specific gene expression signature of Wnt/ß-catenin transcriptional activation was used to stratify pathway activation across a cohort of resected, early-stage PDAC tumors (N=41). In this cohort, higher Wnt/ß-catenin activation was found to significantly correlate with lymphvascular invasion and worse disease-specific survival (median survival time 20.3 versus 43.9 months, log-rank P=0.03). Supporting the importance of Wnt ligand in mediating autocrine Wnt signaling, Wnt/ß-catenin activity was significantly inhibited in PDAC cell lines by WLS gene silencing and the small-molecule inhibitor IWP-2, both of which functionally block Wnt ligand processing and secretion. Transcriptional profiling revealed elevated expression of WNT7B occurred in PDAC cell lines with high levels of cell autonomous Wnt/ß-catenin activity. Gene-knockdown studies in AsPC-1 and HPAF-2 cell lines confirmed WNT7B-mediated cell autonomous Wnt/ß-catenin activation, as well as an anchorage-independent growth phenotype. Our findings indicate WNT7B can serve as a primary determinant of differential Wnt/ß-catenin activation in PDAC. Disrupting the interaction between Wnt ligands and their receptors may be a particularly suitable approach for therapeutic modulation of Wnt/ß-catenin signaling in PDAC and other cancer contexts where Wnt activation is mediated by ligand expression rather than mutations in canonical pathway members.


Subject(s)
Carcinoma, Pancreatic Ductal/metabolism , Cell Proliferation , Pancreatic Neoplasms/metabolism , Wnt Proteins/physiology , Wnt Signaling Pathway , Aged , Autocrine Communication , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Adhesion , Cell Line, Tumor , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Prognosis , Proportional Hazards Models , Transcription, Genetic , Transcriptome
4.
Transl Psychiatry ; 3: e301, 2013 Sep 03.
Article in English | MEDLINE | ID: mdl-24002087

ABSTRACT

Wnt signaling, which encompasses multiple biochemical pathways that regulate neural development downstream of extracellular Wnt glycoprotein ligands, has been suggested to contribute to major psychiatric disorders including autism spectrum disorders (ASD). We used next-generation sequencing and Sequenom genotyping technologies to resequence 10 Wnt signaling pathway genes in 198 ASD patients and 240 matched controls. Results for single-nucleotide polymorphisms (SNPs) of interest were confirmed in a second set of 91 ASD and 144 control samples. We found a significantly increased burden of extremely rare missense variants predicted to be deleterious by PolyPhen-2, distributed across seven genes in the ASD sample (3.5% in ASD vs 0.8% in controls; Fisher's exact test, odds ratio (OR)=4.37, P=0.04). We also found a missense variant in WNT1 (S88R) that was overrepresented in the ASD sample (8 A/T in 267 ASD (minor allele frequency (MAF)=1.69%) vs 1 A/T in 377 controls (MAF=0.13%), OR=13.0, Fisher's exact test, P=0.0048; OR=8.2 and P=0.053 after correction for population stratification). Functional analysis revealed that WNT1-S88R is more active than wild-type WNT1 in assays for the Wnt/ß-catenin signaling pathway. Our findings of a higher burden in ASD of rare missense variants distributed across 7 of 10 Wnt signaling pathway genes tested, and of a functional variant at the WNT1 locus associated with ASD, support that dysfunction of this pathway contributes to ASD susceptibility. Given recent findings of common molecular mechanisms in ASD, schizophrenia and affective disorders, these loci merit scrutiny in other psychiatric conditions as well.


Subject(s)
Child Development Disorders, Pervasive/genetics , Wnt Signaling Pathway/genetics , Wnt1 Protein/genetics , Case-Control Studies , Gene Frequency , Genotype , Humans , Mutation, Missense/genetics , Polymorphism, Single Nucleotide , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/genetics
6.
Oncogene ; 31(32): 3696-708, 2012 Aug 09.
Article in English | MEDLINE | ID: mdl-22179838

ABSTRACT

By analyzing public data sets of gene expression in human breast cancers we observed that increased levels of transcripts encoding the planar cell polarity (PCP) proteins SCRIB and VANGL1 correlate with increased risk of patient relapse. Experimentally, we found that reducing expression of SCRIB by short-hairpin RNAs (shRNAs) reduces the growth of human breast cancer cells in xenograft assays. To investigate SCRIB-associated proteins that might participate in the responses of breast cancer cells to altered levels of SCRIB, we used mass spectrometry and confocal microscopy. These studies reveal that SCRIB is present in at least two unique protein complexes: (1) a complex of SCRIB, ARHGEF, GIT and PAK (p21-activated kinase), and (2) a complex of SCRIB, NOS1AP and VANGL. Focusing on NOS1AP, we observed that NOS1AP colocalizes with both SCRIB and VANGL1 along cellular protrusions in metastatic breast cancer cells, but does not colocalize with either SCRIB or VANGL1 at cell junctions in normal breast cells. We investigated the effects of shRNA-mediated knockdown of NOS1AP and SCRIB in vitro, and found that reducing NOS1AP and SCRIB slows breast cancer cell migration and prevents the establishment of leading-trailing polarity. We also find that reduction of NOS1AP enhances anchorage-independent growth. Collectively these data point to the relevance of NOS1AP and SCRIB protein complexes in breast cancer.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , Breast Neoplasms/metabolism , Carrier Proteins/metabolism , Cell Movement , Cell Polarity , Membrane Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation , Cell Surface Extensions/metabolism , Disease Progression , Female , Gene Knockdown Techniques , Humans , Kaplan-Meier Estimate , Lymphatic Metastasis , Membrane Proteins/genetics , Mice , Mice, Nude , Multiprotein Complexes/metabolism , Neoplasm Transplantation , Protein Interaction Mapping , Protein Interaction Maps , Protein Transport , RNA Interference , Tumor Burden , Tumor Suppressor Proteins/genetics
7.
J Periodontal Res ; 47(3): 309-19, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22150562

ABSTRACT

BACKGROUND AND OBJECTIVE: Bone morphogenetic protein 2 (BMP2)-induced osteogenic differentiation has been shown to occur through the canonical Wnt/ßcatenin pathway, whereas factors promoting canonical Wnt signaling in cementoblasts inhibit cell differentiation and promote cell proliferation in vitro. The aim of this study was to investigate whether putative precursor cells of cementoblasts, dental follicle cells (murine SVF4 cells), when stimulated with BMP2, would exhibit changes in genes/proteins associated with the Wnt/ß-catenin pathway. MATERIAL AND METHODS: SVF4 cells were stimulated with BMP2, and the following assays were carried out: (i) Wnt/ß-catenin pathway activation assessed by western blotting, ß-catenin/transcription factor (TCF) reporter assays and expression of the lymphoid enhancer-binding factor-1 (Lef1), transcription factor 7 (Tcf7), Wnt inhibitor factor 1 (Wif1) and Axin2 (Axin2) genes; and (ii) cementoblast/osteoblast differentiation assessed by mineralization in vitro, and by the mRNA levels of runt-related transcription factor 2 (Runx2), osterix (Osx), alkaline phosphatase (Alp), osteocalcin (Ocn) and bone sialoprotein (Bsp), determined by quantitative PCR after treatment with wingless-type MMTV integration site family, member 3A (WNT3A) and knockdown of ß-catenin. RESULTS: WNT3A induced ß-catenin nuclear translocation and up-regulated the transcriptional activity of a canonical Wnt-responsive reporter, suggesting that the Wnt/ß-catenin pathway functions in SVF4 cells. Activation of Wnt signaling with WNT3A suppressed BMP2-mediated induction of cementoblast/osteoblast maturation of SVF4 cells. However, ß-catenin knockdown showed that the BMP2-induced expression of cementoblast/osteoblast differentiation markers requires endogenous ß-catenin. WNT3A down-regulated transcripts for Runx2, Alp and Ocn in SVF4 cells compared with untreated cells. In contrast, BMP2 induction of Bsp transcripts occurred independently of Wnt/ß-catenin signaling. CONCLUSION: These data suggest that stabilization of ß-catenin by WNT3A inhibits BMP2-mediated induction of cementoblast/osteoblast differentiation in SVF4 cells, although BMP2 requires endogenous Wnt/ß-catenin signaling to promote cell maturation.


Subject(s)
Bone Morphogenetic Protein 2/physiology , Dental Sac/cytology , Wnt Signaling Pathway/physiology , Adaptor Proteins, Signal Transducing , Alkaline Phosphatase/analysis , Animals , Axin Protein/analysis , Bone Morphogenetic Protein 2/drug effects , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Differentiation/physiology , Cell Line , Cell Proliferation , Core Binding Factor Alpha 1 Subunit/analysis , Dental Cementum/drug effects , Dental Cementum/physiology , Dental Sac/drug effects , Extracellular Matrix Proteins/analysis , Gene Knockdown Techniques , Hepatocyte Nuclear Factor 1-alpha , Intercellular Signaling Peptides and Proteins/analysis , Lymphoid Enhancer-Binding Factor 1/analysis , Mice , Osteoblasts/drug effects , Osteoblasts/physiology , Osteocalcin/analysis , Osteogenesis/physiology , Osteopontin/analysis , Sp7 Transcription Factor , T Cell Transcription Factor 1/analysis , Transcription Factors/analysis , Transcription, Genetic/drug effects , Wnt Signaling Pathway/drug effects , Wnt3A Protein/pharmacology , Zinc Fingers , beta Catenin/genetics
8.
Oncogene ; 25(57): 7545-53, 2006 Dec 04.
Article in English | MEDLINE | ID: mdl-17143299

ABSTRACT

In order to function properly, the brain must be wired correctly during critical periods in early development. Mistakes in this process are hypothesized to occur in disorders like autism and schizophrenia. Later in life, signaling pathways are essential in maintaining proper communication between neuronal and non-neuronal cells, and disrupting this balance may result in disorders like Alzheimer's disease. The Wnt/beta-catenin pathway has a well-established role in cancer. Here, we review recent evidence showing the involvement of Wnt/beta-catenin signaling in neurodevelopment as well as in neurodegenerative diseases. We suggest that the onset/development of such pathological conditions may involve the additive effect of genetic variation within Wnt signaling components and of molecules that modulate the activity of this signaling cascade.


Subject(s)
Gene Expression Regulation , Nervous System Diseases/metabolism , Wnt Proteins/physiology , Alzheimer Disease/metabolism , Animals , Apolipoproteins E/metabolism , Autistic Disorder , Genetic Variation , Humans , Neurodegenerative Diseases/metabolism , Polymorphism, Genetic , Schizophrenia/genetics , Schizophrenia/metabolism , Signal Transduction , Wnt Proteins/metabolism
9.
Dev Cell ; 1(1): 103-14, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11703928

ABSTRACT

In vertebrates, wnt8 has been implicated in the early patterning of the mesoderm. To determine directly the embryonic requirements for wnt8, we generated a chromosomal deficiency in zebrafish that removes the bicistronic wnt8 locus. We report that homozygous mutants exhibit pronounced defects in dorso-ventral mesoderm patterning and in the antero-posterior neural pattern. Despite differences in their signaling activities, either coding region of the bicistronic RNA can rescue the deficiency phenotype. Specific interference of wnt8 translation by morpholino antisense oligomers phenocopies the deficiency, and interference with wnt8 translation in ntl and spt mutants produces embryos lacking trunk and tail. These data demonstrate that the zebrafish wnt8 locus is required during gastrulation to pattern both the mesoderm and the neural ectoderm properly.


Subject(s)
Body Patterning/physiology , Proteins/genetics , Transcription, Genetic/physiology , Zebrafish Proteins/genetics , Zebrafish/embryology , Amino Acid Sequence , Animals , Cytoskeletal Proteins , Ectoderm/physiology , Fetal Proteins , Gene Expression Regulation, Developmental , Head/embryology , Mesoderm/physiology , Molecular Sequence Data , Mutagenesis/physiology , Oligonucleotides, Antisense , Open Reading Frames , Phenotype , RNA, Messenger/genetics , T-Box Domain Proteins/genetics , Tail/embryology , Wnt Proteins
10.
Biochem Biophys Res Commun ; 287(3): 589-93, 2001 Sep 28.
Article in English | MEDLINE | ID: mdl-11563835

ABSTRACT

Wnts are secreted ligands with diverse roles in animal development. Wnts bind to cell surface membrane proteins termed Frizzleds. Molecular cloning of members of the Frizzled family revealed hydropathy plots with seven putative, transmembrane-spanning regions, conserved in Frizzleds characterized in mice, humans, flies, and worms. Understanding how Frizzled translates binding of their cognate Wnts into intracellular signals controlling aspects of development has been an elusive goal. Earlier observations gathered from a variety of model systems provided compelling, but indirect, support that the Frizzled receptors may be members of the superfamily of G-protein-coupled receptors that possess seven transmembrane-spanning domains. Search for a linkage between Frizzled and possible downstream heterotrimeric G-proteins has been advanced by the use of bacterial toxins, antisense DNA, and novel chimeric receptor constructs. New data establish that Frizzleds are indeed bona fide G-protein-coupled receptors. Frizzled-1 couples via G-proteins Go and Gq to the canonical beta-catenin-Lef-Tcf pathway. Frizzled-2 couples via Gq and Gt to downstream effectors including calcium mobilization. Frizzleds and G-proteins might once have been considered strange bedfellows, not likely partners in signaling. The new data, consistent with the properties known for virtually all members of the G-protein-coupled receptors, reveal a more classic romance of signaling elements controlling aspects of early development.


Subject(s)
GTP-Binding Proteins/metabolism , GTP-Binding Proteins/physiology , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators , Zebrafish Proteins , Animals , Coculture Techniques , Cytoskeletal Proteins/metabolism , Dimerization , Ligands , Models, Biological , Protein Binding , Proto-Oncogene Proteins/physiology , Receptors, Neurotransmitter/metabolism , Wnt Proteins , beta Catenin
11.
Mech Dev ; 106(1-2): 61-76, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11472835

ABSTRACT

Convergent extension movements are the main driving force of Xenopus gastrulation. A fine-tuned regulation of cadherin-mediated cell-cell adhesion is thought to be required for this process. Members of the Wnt family of extracellular glycoproteins have been shown to modulate cadherin-mediated cell-cell adhesion, convergent extension movements, and cell differentiation. Here we show that endogenous Wnt/beta-catenin signaling activity is essential for convergent extension movements due to its effect on gene expression rather than on cadherins. Our data also suggest that XLEF-1 rather than XTCF-3 is required for convergent extension movements and that XLEF-1 functions in this context in the Wnt/beta-catenin pathway to regulate Xnr-3. In contrast, activation of the Wnt/Ca2+ pathway blocks convergent extension movements, with potential regulation of the Wnt/beta-catenin pathway at two different levels. PKC, activated by the Wnt/Ca2+ pathway, blocks the Wnt/beta-catenin pathway upstream of beta-catenin and phosphorylates Dishevelled. CamKII, also activated by the Wnt/Ca2+ pathway, inhibits the Wnt/beta-catenin signaling cascade downstream of beta-catenin. Thus, an opposing cross-talk of two distinct Wnt signaling cascades regulates convergent extension movements in Xenopus.


Subject(s)
Calcium Signaling , Cytoskeletal Proteins/metabolism , Embryo, Nonmammalian/physiology , Gastrula/physiology , Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Trans-Activators , Xenopus Proteins , Zebrafish Proteins , Animals , Calcium/metabolism , Cell Movement , Embryo, Nonmammalian/cytology , Gastrula/cytology , Gene Expression Regulation, Developmental , High Mobility Group Proteins/genetics , High Mobility Group Proteins/metabolism , In Vitro Techniques , Lymphoid Enhancer-Binding Factor 1 , Protein Kinase C/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Wnt Proteins , Wnt-5a Protein , Xenopus , beta Catenin
12.
Science ; 292(5522): 1718-22, 2001 Jun 01.
Article in English | MEDLINE | ID: mdl-11387477

ABSTRACT

The frizzled receptors, which mediate development and display seven hydrophobic, membrane-spanning segments, are cell membrane-localized. We constructed a chimeric receptor with the ligand-binding and transmembrane segments from the beta2-adrenergic receptor (beta2AR) and the cytoplasmic domains from rat Frizzled-1 (Rfz1). Stimulation of mouse F9 clones expressing the chimera (beta2AR-Rfz1) with the beta-adrenergic agonist isoproterenol stimulated stabilization of beta-catenin, activation of a beta-catenin-sensitive promoter, and formation of primitive endoderm. The response was blocked by inactivation of pertussis toxin-sensitive, heterotrimeric guanine nucleotide-binding proteins (G proteins) and by depletion of Galphaq and Galphao. Thus, G proteins are elements of Wnt/Frizzled-1 signaling to the beta-catenin-lymphoid-enhancer factor (LEF)-T cell factor (Tcf) pathway.


Subject(s)
Cytoskeletal Proteins/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Receptors, Neurotransmitter/metabolism , Signal Transduction , Trans-Activators , Transcription Factors/metabolism , Xenopus Proteins , Zebrafish Proteins , Amino Acid Sequence , Animals , Cloning, Molecular , Embryo, Nonmammalian/metabolism , Endoderm/physiology , Frizzled Receptors , Gene Expression Regulation/drug effects , Genes, Reporter , Guanosine Triphosphate/metabolism , Isoproterenol/metabolism , Isoproterenol/pharmacology , Mice , Molecular Sequence Data , Pertussis Toxin , Propranolol/metabolism , Propranolol/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Receptors, Adrenergic, beta-2/chemistry , Receptors, Adrenergic, beta-2/genetics , Receptors, Adrenergic, beta-2/metabolism , Receptors, G-Protein-Coupled , Receptors, Neurotransmitter/chemistry , Receptors, Neurotransmitter/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/metabolism , Transfection , Tumor Cells, Cultured , Virulence Factors, Bordetella/pharmacology , Wnt Proteins , Xenopus , beta Catenin
13.
Mol Cell Biol ; 21(5): 1866-73, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11238923

ABSTRACT

We have determined that I-mfa, an inhibitor of several basic helix-loop-helix (bHLH) proteins, and XIC, a Xenopus ortholog of human I-mf domain-containing protein that shares a highly conserved cysteine-rich C-terminal domain with I-mfa, inhibit the activity and DNA binding of the HMG box transcription factor XTcf3. Ectopic expression of I-mfa or XIC in early Xenopus embryos inhibited dorsal axis specification, the expression of the Tcf3/beta-catenin-regulated genes siamois and Xnr3, and the ability of beta-catenin to activate reporter constructs driven by Lef/Tcf binding sites. I-mfa domain proteins can regulate both the Wnt signaling pathway and a subset of bHLH proteins, possibly coordinating the activities of these two critical developmental pathways.


Subject(s)
Cell Cycle Proteins , HMGB Proteins , Myogenic Regulatory Factors/metabolism , Trans-Activators , Transcription Factors/metabolism , Tumor Suppressor Proteins , Xenopus Proteins , Xenopus/embryology , Amino Acid Sequence , Animals , Binding Sites , Blotting, Western , Cyclin-Dependent Kinase Inhibitor p27 , Cytoskeletal Proteins/metabolism , Gene Expression Regulation, Developmental , Gene Library , Genes, Reporter , Homeodomain Proteins/metabolism , Mice , Microtubule-Associated Proteins/metabolism , Molecular Sequence Data , Myogenic Regulatory Factors/chemistry , Precipitin Tests , Protein Binding , Protein Structure, Tertiary , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Signal Transduction , TCF Transcription Factors , Transcription Factor 7-Like 1 Protein , Transforming Growth Factor beta/metabolism , beta Catenin
14.
Dev Biol ; 229(1): 102-18, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11133157

ABSTRACT

Patterning the neural plate in vertebrates depends on complex interactions between a variety of secreted growth factors. Here we describe a novel secreted factor in zebrafish, named mdk2, related to the midkine family of heparin-binding growth factors that is involved in posterior neural development. mdk2 is expressed shortly after the onset of gastrulation in the presumptive neural plate cells of the epiblast, and this expression is enhanced by exogenous retinoic acid. Ectopic expression of mdk2 enhances neural crest cell fates at the lateral edges of the caudal neural plate, concomitant with a repression of anterior structures and mesendodermal and ectodermal markers. Reciprocally, ectopic expression of a dominant negative mdk2 results in severe deficiencies of structures posterior to the midbrain-hindbrain boundary, with negligible effects on anterior structures. In these embryos, the expression of hindbrain and neural crest markers is strongly reduced, and the formation of posterior primary moto- and sensory neurons is blocked. Analyses in mutant zebrafish embryos shows that expression of mdk2 is independent of FGF8 and nodal-related-1 signaling, but is under negative control of BMP signaling. These data support the hypothesis that mdk2 participates in posterior neural development in zebrafish.


Subject(s)
Carrier Proteins/metabolism , Cytokines , Nerve Growth Factors/metabolism , Nervous System/embryology , Neurons/cytology , Amino Acid Sequence , Animals , Body Patterning , Cell Differentiation , Embryonic Induction , Gastrula , Head/embryology , Midkine , Molecular Sequence Data , Mutation , Neural Crest/embryology , Sequence Homology, Amino Acid , Zebrafish
15.
Physiol Genomics ; 2(2): 37-48, 2000 Mar 13.
Article in English | MEDLINE | ID: mdl-11015580

ABSTRACT

The zebrafish has become a popular model system for the study of vertebrate developmental biology because of its numerous strengths as a molecular genetic and embryological system. To determine the requirement for specific genes during embryogenesis, it is necessary to generate organisms carrying loss-of-function mutations. This can be accomplished in zebrafish through a reverse genetic approach. This review discusses the current techniques for generating mutations in known genes in zebrafish. These techniques include the generation of chromosomal deletions and the subsequent identification of complementation groups within deletions through noncomplementation assays. In addition, this review will discuss methods currently being evaluated that may improve the methods for finding mutations in a known sequence, including screening for randomly induced small deletions within genes and screening for randomly induced point mutations within specific genes.


Subject(s)
Genetic Techniques , Zebrafish/embryology , Zebrafish/genetics , Animals , Blastocyst/radiation effects , Chromosome Deletion , DNA Mutational Analysis/methods , Gene Targeting , Genetic Complementation Test , Genetic Testing/methods , Heteroduplex Analysis , Models, Biological , Mutagenesis, Site-Directed , Polymerase Chain Reaction , Sequence Deletion
16.
Dev Growth Differ ; 42(4): 347-57, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10969734

ABSTRACT

In spite of abundant evidence that Wnts play essential roles in embryonic induction and patterning, little is known about the expression or activities of Wnt receptors during embryogenesis. The isolation and expression of two maternal Xenopus frizzled genes, Xfrizzled-1 and Xfrizzled-7, is described. It is also demonstrated that both can activate the Wnt/beta-catenin signaling pathway as monitored by the induction of specific target genes. Activation of the beta-Catenin pathway has previously been shown to be necessary and sufficient for specifying the dorsal axis of Xenopus. beta-Catenin is thought to work through the cell-autonomous induction of the homeobox genes siamois and twin, that in turn bind to and activate the promoter of another homeobox gene, goosecoid. However, it was found that the beta-catenin pathway regulated the expression of both endogenous goosecoid, and a goosecoid promoter construct, in a cell non-autonomous manner. These data demonstrate that maternal Frizzleds can activate the Wnt/beta-catenin pathway in Xenopus embryos, and that induction of a known downstream gene can occur in a cell non-autonomous manner.


Subject(s)
Cytoskeletal Proteins/physiology , Embryo, Nonmammalian/metabolism , Homeodomain Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, G-Protein-Coupled , Receptors, Neurotransmitter/metabolism , Repressor Proteins , Signal Transduction/physiology , Trans-Activators , Transcription Factors , Xenopus Proteins , Xenopus laevis/embryology , Zebrafish Proteins , Amino Acid Sequence , Animals , Cell-Free System , Cells, Cultured , DNA Primers/chemistry , Evolution, Molecular , Female , Frizzled Receptors , Gene Expression Regulation, Developmental , Goosecoid Protein , Homeodomain Proteins/genetics , Luciferases/metabolism , Microinjections , Microscopy, Confocal , Molecular Sequence Data , Proto-Oncogene Proteins/metabolism , RNA/metabolism , Receptors, Cell Surface/genetics , Receptors, Neurotransmitter/genetics , Reverse Transcriptase Polymerase Chain Reaction , Sequence Homology, Amino Acid , Supine Position/physiology , Wnt Proteins , Xenopus laevis/physiology , beta Catenin
17.
J Biol Chem ; 275(42): 32649-57, 2000 Oct 20.
Article in English | MEDLINE | ID: mdl-10915780

ABSTRACT

The cyclin D1 gene encodes the regulatory subunit of a holoenzyme that phosphorylates and inactivates the pRB tumor suppressor protein. Cyclin D1 is overexpressed in 20-30% of human breast tumors and is induced both by oncogenes including those for Ras, Neu, and Src, and by the beta-catenin/lymphoid enhancer factor (LEF)/T cell factor (TCF) pathway. The ankyrin repeat containing serine-threonine protein kinase, integrin-linked kinase (ILK), binds to the cytoplasmic domain of beta(1) and beta(3) integrin subunits and promotes anchorage-independent growth. We show here that ILK overexpression elevates cyclin D1 protein levels and directly induces the cyclin D1 gene in mammary epithelial cells. ILK activation of the cyclin D1 promoter was abolished by point mutation of a cAMP-responsive element-binding protein (CREB)/ATF-2 binding site at nucleotide -54 in the cyclin D1 promoter, and by overexpression of either glycogen synthase kinase-3beta (GSK-3beta) or dominant negative mutants of CREB or ATF-2. Inhibition of the PI 3-kinase and AKT/protein kinase B, but not of the p38, ERK, or JNK signaling pathways, reduced ILK induction of cyclin D1 expression. ILK induced CREB transactivation and CREB binding to the cyclin D1 promoter CRE. Wnt-1 overexpression in mammary epithelial cells induced cyclin D1 mRNA and targeted overexpression of Wnt-1 in the mammary gland of transgenic mice increased both ILK activity and cyclin D1 levels. We conclude that the cyclin D1 gene is regulated by the Wnt-1 and ILK signaling pathways and that ILK induction of cyclin D1 involves the CREB signaling pathway in mammary epithelial cells.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinases/metabolism , Cyclic AMP Response Element-Binding Protein/metabolism , Cyclin D1/genetics , Gene Expression Regulation , Protein Serine-Threonine Kinases/metabolism , Zebrafish Proteins , Activating Transcription Factor 2 , Animals , Breast Neoplasms , CD18 Antigens/physiology , Cell Line , Epithelial Cells/metabolism , Female , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3 , Glycogen Synthase Kinases , Humans , Integrin beta1/physiology , Mammary Glands, Animal/cytology , Mammary Glands, Animal/metabolism , Mice , Mice, Transgenic , Promoter Regions, Genetic , Protein Serine-Threonine Kinases/genetics , Protein Subunits , Protein-Tyrosine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Signal Transduction , Transcription Factors/metabolism , Transfection , Tumor Cells, Cultured , Wnt Proteins , Wnt1 Protein
18.
Bioessays ; 22(8): 708-16, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10918301

ABSTRACT

Neural crest cells are multipotent progenitors, capable of producing diverse cell types upon differentiation. Recent studies have identified significant heterogeneity in both the fates produced and genes expressed by different premigratory crest cells. While these cells may be specified toward particular fates prior to migration, transplant studies show that some may still be capable of respecification at this time. Here we summarize evidence that extracellular signals in the local environment may act to specify premigratory crest and thus generate diversity in the population. Three main classes of signals-Wnts, BMP2/BMP4 and TGFbeta1,2,3-have been shown to directly influence the production of particular neural crest cell fates, and all are expressed near the premigratory crest. This system may therefore provide a good model for integration of multiple signaling pathways during embryonic cell fate specification.


Subject(s)
Neural Crest/cytology , Zebrafish Proteins , Animals , Bone Morphogenetic Proteins/metabolism , Neural Crest/metabolism , Neuronal Plasticity , Proto-Oncogene Proteins/metabolism , Signal Transduction , Stem Cells/cytology , Stem Cells/metabolism , Transforming Growth Factor beta/metabolism , Wnt Proteins
19.
Dev Biol ; 222(2): 376-91, 2000 Jun 15.
Article in English | MEDLINE | ID: mdl-10837126

ABSTRACT

A new bHLH gene from mouse that we call pMesogenin1 (referring to paraxial mesoderm-specific expression and regulatory capacities) and its candidate ortholog from Xenopus were isolated and studied comparatively. In both organisms the gene is specifically expressed in unsegmented paraxial mesoderm and its immediate progenitors. A striking feature of pMesogenin1 expression is that it terminates abruptly in presumptive somites (somitomeres). Somitomeres rostral to the pMesogenin1 domain strongly upregulate expression of pMesogenin's closest known paralogs, MesP1 and MesP2 (Thylacine1/2 in Xenopus). Subsequently, the most rostral somitomere becomes a new somite and expression of MesP1/2 is sharply downregulated before this transition. Thus, expression patterns of these bHLH genes, together with that of an additional bHLH gene in the mouse, Paraxis, collectively define discrete but highly dynamic prepatterned subdomains of the paraxial mesoderm. In functional assays, we show that pMesogenin1 from either mouse or frog can efficiently drive nonmesodermal cells to assume a phenotype with molecular and cellular characteristics of early paraxial mesoderm. Among genes induced by added pMesogenin1 is Xwnt-8, a signaling factor that induces a similar repertoire of marker genes and a similar cellular phenotype. Additional target genes induced by pMesogenin1 are ESR4/5, regulators known to play a significant role in segmentation of paraxial mesoderm (W. C. Jen et al., 1999, Genes Dev. 13, 1486-1499). pMesogenin1 differs from other known mesoderm-inducing transcription factors because it does not also activate a dorsal (future axial) mesoderm phenotype, suggesting that pMesogenin1 is involved in specifying paraxial mesoderm. In the context of the intact frog embryo, ectopic pMesogenin1 also actively suppressed axial mesoderm markers and disrupted normal formation of notochord. In addition, we found evidence for cross-regulatory interactions between pMesogenin1 and T-box transcription factors, a family of genes normally expressed in a broader pattern and known to induce multiple types of mesoderm. Based on our results and results from prior studies of related bHLH genes, we propose that pMesogenin1 and its closest known relatives, MesP1/2 (in mouse) and Thylacine1/2 (in Xenopus), comprise a bHLH subfamily devoted to formation and segmentation of paraxial mesoderm.


Subject(s)
DNA-Binding Proteins/genetics , Embryonic and Fetal Development , Gene Expression Regulation, Developmental , Mesoderm/physiology , Transcription Factors/genetics , Amino Acid Sequence , Animals , Basic Helix-Loop-Helix Transcription Factors , Body Patterning , Gene Library , Helix-Loop-Helix Motifs , In Situ Hybridization , Mice , Mice, Inbred Strains , Molecular Sequence Data , Phenotype , Phylogeny , Reverse Transcriptase Polymerase Chain Reaction , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Transcription Factors/chemistry , Xenopus laevis
20.
Trends Genet ; 16(7): 279-83, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10858654

ABSTRACT

Members of the vertebrate Wnt family have been subdivided into two functional classes according to their biological activities. Some Wnts signal through the canonical Wnt-1/wingless pathway by stabilizing cytoplasmic beta-catenin. By contrast other Wnts stimulate intracellular Ca2+ release and activate two kinases, CamKII and PKC, in a G-protein-dependent manner. Moreover, putative Wnt receptors belonging to the Frizzled gene family have been identified that preferentially couple to the two prospective pathways in the absence of ectopic Wnt ligand and that might account for the signaling specificity of the Wnt pathways. As Ca2+ release was the first described feature of the noncanonical pathway, and as Ca2+ probably plays a key role in the activation of CamKII and PKC, we have named this Wnt pathway the Wnt/Ca2+ pathway.


Subject(s)
Calcium/metabolism , Proto-Oncogene Proteins/metabolism , Vertebrates/metabolism , Zebrafish Proteins , Animals , Wnt Proteins , Wnt1 Protein
SELECTION OF CITATIONS
SEARCH DETAIL
...