Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Mol Ecol ; 26(7): 2006-2018, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27662109

ABSTRACT

Previous studies have shown that the response of bacterial communities to disturbances depends on their environmental history. Historically fluctuating habitats host communities that respond better to disturbance than communities of historically stable habitats. However, the exact ecological mechanism that drives this dependency remains unknown. Here, we experimentally demonstrate that modifications of niche optima and niche breadths of the community members are driving this dependency of bacterial responses to past environmental conditions. First, we develop a novel, simple method to calculate the niche optima and breadths of bacterial taxa regarding single environmental gradients. Then, we test this method on sediment bacterial communities of three habitats, one historically stable and less loaded and two historically more variable and more loaded habitats in terms of historical chlorophyll-α water concentration, that we subject to hypoxia via organic matter addition ex situ. We find that communities containing bacterial taxa differently adapted to hypoxia show different structural and functional responses, depending on the sediment's environmental history. Specifically, in the historically less fluctuating and loaded sediments where we find more taxa poorly adapted to hypoxic conditions, communities change a lot over time and organic matter is not degraded efficiently. The opposite is true for the historically more fluctuating and loaded sediments where we find more taxa well adapted to hypoxia. Based on the community responses observed here, we also propose an alternative calculation of community resistance that takes into account how rapidly the communities respond to disturbances and not just the initial and final states of the community.


Subject(s)
Bacteria/classification , Ecosystem , Eutrophication , Geologic Sediments/microbiology , Acclimatization , Estuaries , Greece , Population Dynamics , Seawater
2.
Eye (Lond) ; 28(12): 1452-60, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25233824

ABSTRACT

UNLABELLED: PURPORSE: To determine the associated balance of forces of the vitreofoveal interface in focal vitreomacular traction evolving to full-thickness macular hole (FTMH) and to link/explain the observed changes in the context of mathematical and physics models. PATIENTS AND METHODS: This is a multicenter, prospective, and observational case series conducted at the Vitreoretinal Department of three different referral centers. Fellow eyes of patients with unilateral idiopathic FTMH were included. Eighty-nine patients were included in the analysis. The fellow normal eye of the study patients was imaged with spectral-domain optical coherence tomography. The main outcome measure was the optical-coherence-tomography-defined characteristics of the vitreofoveal interface and their analysis with mathematical and physics models at the end of follow-up period. RESULTS: Of the included 89 patients (66 women and 23 men; mean age±SD, 68.5 years±9.8), 10 (11.2%) developed FTMH at the fellow eye at the end of the follow-up period. We observed two types of vitreofoveal attachment. A V-shaped (cord-like) configuration and a U-shaped configuration. The eyes with the V-shaped attachment demonstrated initial structural changes in the outer foveal layers and the eyes with the U-shaped attachment showed inner morphological changes. CONCLUSION: We hypothesize that the type (V- or U-shaped) of the vitreofoveal attachment may affect the type and location of the initial structural change leading to the formation of FTMH from the stage of the focal vitreomacular traction.


Subject(s)
Models, Theoretical , Retinal Diseases/diagnosis , Retinal Perforations/diagnosis , Vitreous Detachment/diagnosis , Aged , Endotamponade , Female , Fluorocarbons/administration & dosage , Humans , Male , Middle Aged , Prone Position , Prospective Studies , Retinal Perforations/classification , Tissue Adhesions , Tomography, Optical Coherence , Vitrectomy
3.
Oncogene ; 32(47): 5409-20, 2013 Nov 21.
Article in English | MEDLINE | ID: mdl-23524585

ABSTRACT

Glioblastoma multiforme (GBM) is an aggressive brain malignancy characterized by high heterogeneity and invasiveness. It is increasingly accepted that the refractory feature of GBM to current therapies stems from the existence of few tumorigenic cells that sustain tumor growth and spreading, the so-called glioma-initiating cells (GICs). Previous studies showed that cytokines of the bone morphogenetic protein (BMP) family induce differentiation of the GICs, and thus act as tumor suppressors. Molecular pathways that explain this behavior of BMP cytokines remain largely elusive. Here, we show that BMP signaling induces Smad-dependent expression of the transcriptional regulator Snail in a rapid and sustained manner. Consistent with its already established promigratory function in other cell types, we report that Snail silencing decreases GBM cell migration. Consequently, overexpression of Snail increases GBM invasiveness in a mouse xenograft model. Surprisingly, we found that Snail depletes the GBM capacity to form gliomaspheres in vitro and to grow tumors in vivo, both of which are important features shared by GICs. Thus Snail, acting downstream of BMP signaling, dissociates the invasive capacity of GBM cells from their tumorigenic potential.


Subject(s)
Brain Neoplasms/pathology , Glioblastoma/pathology , Transcription Factors/metabolism , Animals , Bone Morphogenetic Proteins/metabolism , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Cell Differentiation/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Gene Expression Regulation, Neoplastic , Glioblastoma/genetics , Glioblastoma/metabolism , Humans , Mice , Neoplastic Stem Cells/metabolism , RNA Interference , RNA, Small Interfering , Signal Transduction/genetics , Smad1 Protein/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , Smad4 Protein/genetics , Smad5 Protein/genetics , Snail Family Transcription Factors , Transcription Factors/genetics
4.
Oncogene ; 31(23): 2862-75, 2012 Jun 07.
Article in English | MEDLINE | ID: mdl-21986943

ABSTRACT

In the present study we analyzed the regulation of the two isoforms of the RhoA-specific guanine nucleotide exchange factor Net1 by transforming growth factor-ß (TGF-ß) in keratinocytes. We report that short-term TGF-ß treatment selectively induced Net1 isoform2 (Net1A) but not Net1 isoform1. This led to upregulation of cytoplasmic Net1A protein levels that were necessary for TGF-ß-mediated RhoA activation. Smad signaling and the MAPK/ERK kinase (MEK)/extracellular signal-regulated kinase (ERK) pathway were involved in Net1A upregulation by TGF-ß. Interestingly, long-term TGF-ß treatment resulted in Net1 mRNA downregulation and Net1A protein degradation by the proteasome. Furthermore, we identified the microRNA miR-24 as a novel post-transcriptional regulator of Net1A expression. Silencing of Net1A resulted in disruption of E-cadherin- and zonula occludens-1 (ZO-1)-mediated junctions, as well as expression of the transcriptional repressor of E-cadherin, Slug and the mesenchymal markers N-cadherin, plasminogen activator inhibitor-1 (PAI-1) and fibronectin, indicating that late TGF-ß-induced downregulation of Net1A is involved in epithelial-to-mesenchymal transition (EMT). Finally, miR-24 was found to be implicated in the regulation of the EMT program in response to TGF-ß and was shown to be directly involved in the TGF-ß-induced breast cancer cell invasiveness through Net1A regulation. Our results emphasize the importance of Net1 isoform2 in the short- and long-term TGF-ß-mediated regulation of EMT.


Subject(s)
Epithelial-Mesenchymal Transition , Gene Expression Regulation , MicroRNAs/genetics , Oncogene Proteins/genetics , RNA Interference , Transforming Growth Factor beta/pharmacology , rhoA GTP-Binding Protein/metabolism , Cadherins/metabolism , Cells, Cultured , Extracellular Signal-Regulated MAP Kinases/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Humans , Keratinocytes/cytology , Keratinocytes/metabolism , Kidney/cytology , Kidney/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Oncogene Proteins/metabolism , Phosphorylation , Protein Isoforms , Proteolysis , Signal Transduction
5.
Genes Immun ; 12(6): 415-27, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21562577

ABSTRACT

Human leukocyte antigen (HLA) class II haplotypes are established risk factors in type 1 diabetes (T1D). The heterozygous DQ2/8 genotype confers the highest risk, whereas the DQ6/8 genotype is protective. We hypothesized that DQ2/8 trans-molecules composed of α and ß chains from DQ2 and DQ8 express unique ß-cell epitopes, whereas DQ6 may interfere with peptide binding to DQ8. Here we show that a single insulin epitope (InsB13-21) within the T1D prototype antigenic InsB6-22 peptide can bind to both cis- and trans-dimers, although these molecules display different peptide binding patterns. DQ6 binds a distinct insulin epitope (InsB6-14). The phenotype of DQ8-restricted T cells from a T1D patient changed from proinflammatory to anti-inflammatory in the presence of DQ6. Our data provide new insights into both susceptible and protective mechanism of DQ, where protecting HLA molecules bind autoantigens in a different (competing) binding register leading to 'epitope stealing', thereby inducing a regulatory, rather than a pathogenic immune response.


Subject(s)
Diabetes Mellitus, Type 1/metabolism , HLA-DQ Antigens/genetics , Islets of Langerhans/immunology , Adolescent , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Diabetes Mellitus, Type 1/genetics , Diabetes Mellitus, Type 1/immunology , Epitopes, B-Lymphocyte/immunology , Genetic Predisposition to Disease , Heterozygote , Homozygote , Humans , Insulin/genetics , Male , Protein Binding , Syndecans/metabolism , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Thymosin/metabolism
6.
Br J Dermatol ; 164(2): 263-72, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20977447

ABSTRACT

BACKGROUND: Epidermolytic ichthyosis (EI) is a skin fragility disorder caused by mutations in genes encoding suprabasal keratins 1 and 10. While the aetiology of EI is known, model systems are needed for pathophysiological studies and development of novel therapies. OBJECTIVES: To generate immortalized keratinocyte lines from patients with EI for studies of EI cell pathology and the effects of chemical chaperones as putative therapies. METHODS: We derived keratinocytes from three patients with EI and one healthy control and established immortalized keratinocytes using human papillomavirus 16-E6/E7. Growth and differentiation characteristics, ability to regenerate organotypic epidermis, keratin expression, formation of cytoskeletal aggregates, and responses to heat shock and chemical chaperones were assessed. RESULTS: The cell lines EH11 (K1_p.Val176_Lys197del), EH21 (K10_p.156Arg>Gly), EH31 (K10_p.Leu161_Asp162del) and NKc21 (wild-type) currently exceed 160 population doublings and differentiate when exposed to calcium. At resting state, keratin aggregates were detected in 9% of calcium-differentiated EH31 cells, but not in any other cell line. Heat stress further increased this proportion to 30% and also induced aggregates in 3% of EH11 cultures. Treatment with trimethylamine N-oxide and 4-phenylbutyrate (4-PBA) reduced the fraction of aggregate-containing cells and affected the mRNA expression of keratins 1 and 10 while 4-PBA also modified heat shock protein 70 (HSP70) expression. Furthermore, in situ proximity ligation assay suggested a colocalization between HSP70 and keratins 1 and 10. Reconstituted epidermis from EI cells cornified but EH21 and EH31 cells produced suprabasal cytolysis, closely resembling the in vivo phenotype. CONCLUSIONS: These immortalized cell lines represent a useful model for studying EI biology and novel therapies.


Subject(s)
Cell Line/pathology , Hyperkeratosis, Epidermolytic/pathology , Keratinocytes/pathology , Adolescent , Adult , Cell Line/drug effects , Cell Transformation, Viral , Epidermis/growth & development , Epidermis/metabolism , HSP70 Heat-Shock Proteins/metabolism , Hot Temperature , Humans , Hyperkeratosis, Epidermolytic/physiopathology , Keratinocytes/drug effects , Keratinocytes/physiology , Keratins/metabolism , Male , Methylamines/pharmacology , Models, Biological , Phenotype , Phenylbutyrates/pharmacology , Stress, Physiological , Young Adult
7.
Oncogene ; 27(9): 1218-30, 2008 Feb 21.
Article in English | MEDLINE | ID: mdl-17724470

ABSTRACT

To better understand the dual, tumour-suppressive and tumour-promoting function of transforming growth factor-beta (TGFbeta), we analysed mammary epithelial NMuMG cells in response to short and long-term TGFbeta exposure. NMuMG cells became proliferation-arrested and apoptotic after exposure to TGFbeta for 2-5 days, whereas surviving cells underwent epithelial-mesenchymal transition (EMT). After chronic TGFbeta exposure (2-3 weeks), however, NMuMG cells became resistant to proliferation arrest and apoptosis, showing sustained EMT instead (TD cells). EMT was fully reversed by a pharmacologic TGFbeta-receptor-I kinase inhibitor or withdrawal of TGFbeta for 6-12 days. Interestingly, both cell cycle arresting/proapoptotic (Smads, p38 kinase) and antiapoptotic, proliferation and EMT-promoting signalling pathways (PI3K-PKB/Akt, ERK) were co-suppressed to low, but significant levels. Except for PI3K-Akt, TGFbeta-dependent downregulation of these signalling pathways in transdifferentiated (TD) cells was fully reversed upon TGFbeta withdrawal, together with partial re-induction of proliferation arrest and apoptosis. Co-injection of non-tumorigenic NMuMG cells with tumour-forming CHO cells oversecreting exogenous TGFbeta1 (CHO-TGFbeta1) allowed outgrowth of epithelioid cells in CHO-TGFbeta1 cell-induced tumours. These epithelial islands enhanced CHO-TGFbeta1 tumour cell proliferation, possibly due to chemokines (for example, JE/MCP-1) secreted by NMuMG/TD cells. We conclude that suppression of antiproliferative, proapoptotic TGFbeta signalling in TD cells may permit TGFbeta-dependent proliferation, survival and EMT-enhancing signalling pathways to act at low levels. Thus, TGFbeta may modulate its own signalling to facilitate switching from tumour suppression to tumour progression.


Subject(s)
Apoptosis , Cell Transformation, Neoplastic/pathology , Epithelial Cells/pathology , Mammary Glands, Animal/pathology , Mesoderm/pathology , Signal Transduction/physiology , Smad Proteins/antagonists & inhibitors , Transforming Growth Factor beta/physiology , Animals , Apoptosis/genetics , CHO Cells , Cell Culture Techniques , Cell Line , Cell Transformation, Neoplastic/metabolism , Cricetinae , Cricetulus , Epithelial Cells/cytology , Epithelial Cells/metabolism , Female , Genes, Tumor Suppressor/physiology , Mammary Glands, Animal/metabolism , Mesoderm/cytology , Mesoderm/metabolism , Mice , Smad Proteins/physiology , Transforming Growth Factor beta/genetics
8.
Oncol Rep ; 15(4): 983-996, 2006.
Article in English | MEDLINE | ID: mdl-16525688

ABSTRACT

Microarray technology allows gene expression profiling at a global level. Many algorithms for the normalization of raw microarray data have been proposed, but no attempt has yet been made to propose operationally verifiable criteria for their comparative evaluation, which is necessary for the selection of the most appropriate method for a given dataset. This study develops a set of operational criteria for assessing the impact of various normalization algorithms in terms of accuracy (bias), precision (variance) and over-fitting (information reduction). The use of these criteria is illustrated by applying the three most widely used algorithms (global median normalization, spiked-in based normalization and lowess) on a specifically designed, multiply-controlled dataset.


Subject(s)
Algorithms , Gene Expression Profiling/statistics & numerical data , Oligonucleotide Array Sequence Analysis , Humans , Reproducibility of Results , Sensitivity and Specificity
9.
Tissue Antigens ; 59(2): 101-12, 2002 Feb.
Article in English | MEDLINE | ID: mdl-12028536

ABSTRACT

Human CD4 T cell responses to an epitope of hGAD65 (GAD = glutamic acid decarboxylase), residues 555-567, are modulated by interaction with an altered peptide ligand containing modifications at TCR contact residues. Using different HLA-DR4 molecules with polymorphisms at sites corresponding to peptide binding pockets p1 and p9, we tested the effect of additional modifications in the altered peptide ligand (APL) designed to increase the avidity of the MHC-peptide interaction and therefore the efficiency of TCR signaling. Modification of the peptide or the MHC molecule which enhanced the p1 interaction also enhanced the antagonist activity of the modified APL. In contrast, modifications at p9 led to a reversal in APL function, resulting in agonist activity. Molecular homology modeling of these MHC-peptide interactions suggests a structural basis for this functional dichotomy in which topographically remote variations lead to unique interaction effects.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Glutamate Decarboxylase/immunology , Isoenzymes/immunology , T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/chemistry , Glutamate Decarboxylase/chemistry , Humans , Isoenzymes/chemistry , Models, Molecular , Peptides/immunology , Peptides/metabolism , Polymorphism, Genetic
10.
J Biol Chem ; 276(31): 29116-25, 2001 Aug 03.
Article in English | MEDLINE | ID: mdl-11384995

ABSTRACT

In the present study we present evidence for the critical role of Sp1 in the mechanism of transactivation of the human cell cycle inhibitor p21(WAF1/Cip1) (p21) gene promoter by the tumor suppressor p53 protein. We found that the distal p53-binding site of the p21 promoter acts as an enhancer on the homologous or heterologous promoters in hepatoma HepG2 cells. In transfection experiments, p53 transactivated the p21 promoter in HaCaT cells that express Sp1 but have a mutated p53 form. In contrast, p53 could not transactivate the p21 promoter in the Drosophila embryo-derived Schneider's SL2 cells that lack endogenous Sp1 or related factors. Cotransfection of SL2 cells with p53 and Sp1 resulted in a synergistic transactivation of the p21 promoter. Synergistic transactivation was greatly decreased in SL2 cells and HaCaT cells by mutations in either the p53-binding site or in the -82/-77 Sp1-binding site indicating functional cooperation between Sp1 and p53 in the transactivation of the p21 promoter. Synergistic transactivation was also decreased by mutations in the transactivation domain of p53. Physical interactions between Sp1 and p53 proteins were established by glutathione S-transferase pull-down and coimmunoprecipitation assays. By using deletion mutants we found that the DNA binding domain of Sp1 is required for its physical interaction with p53. In conclusion, Sp1 must play a critical role in regulating important biological processes controlled by p53 via p21 gene activation such as DNA repair, cell growth, differentiation, and apoptosis.


Subject(s)
Cyclins/genetics , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Transcriptional Activation , Tumor Suppressor Protein p53/metabolism , Animals , Base Sequence , Binding Sites , Carcinoma, Hepatocellular , Cell Line , Cloning, Molecular , Cyclin-Dependent Kinase Inhibitor p21 , Drosophila melanogaster , Enzyme Inhibitors , Escherichia coli , Genes, Reporter , Genes, p53 , Humans , Keratinocytes , Liver Neoplasms , Molecular Sequence Data , Mutagenesis, Site-Directed , Oligonucleotide Probes , Osteosarcoma , Recombinant Fusion Proteins/metabolism , Recombinant Proteins/metabolism , Sp1 Transcription Factor/genetics , Transfection , Tumor Cells, Cultured , Tumor Suppressor Protein p53/genetics
11.
Mol Biol Cell ; 12(4): 1079-91, 2001 Apr.
Article in English | MEDLINE | ID: mdl-11294908

ABSTRACT

Smad proteins are cytoplasmic signaling effectors of transforming growth factor-beta (TGF-beta) family cytokines and regulate gene transcription in the nucleus. Receptor-activated Smads (R-Smads) become phosphorylated by the TGF-beta type I receptor. Rapid and precise transport of R-Smads to the nucleus is of crucial importance for signal transduction. By focusing on the R-Smad Smad3 we demonstrate that 1) only activated Smad3 efficiently enters the nucleus of permeabilized cells in an energy- and cytosol-dependent manner. 2) Smad3, via its N-terminal domain, interacts specifically with importin-beta1 and only after activation by receptor. In contrast, the unique insert of exon3 in the N-terminal domain of Smad2 prevents its association with importin-beta1. 3) Nuclear import of Smad3 in vivo requires the action of the Ran GTPase, which mediates release of Smad3 from the complex with importin-beta1. 4) Importin-beta1, Ran, and p10/NTF2 are sufficient to mediate import of activated Smad3. The data describe a pathway whereby Smad3 phosphorylation by the TGF-beta receptor leads to enhanced interaction with importin-beta1 and Ran-dependent import and release into the nucleus. The import mechanism of Smad3 shows distinct features from that of the related Smad2 and the structural basis for this difference maps to the divergent sequences of their N-terminal domains.


Subject(s)
Cell Nucleus/metabolism , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Nucleocytoplasmic Transport Proteins , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , ran GTP-Binding Protein/metabolism , Active Transport, Cell Nucleus , Animals , Binding Sites , Carrier Proteins/metabolism , Cell Line , Cell Line, Transformed , Humans , Karyopherins , Mice , Signal Transduction , Smad2 Protein , Smad3 Protein , Tumor Cells, Cultured
12.
J Biol Chem ; 276(27): 24627-37, 2001 Jul 06.
Article in English | MEDLINE | ID: mdl-11323414

ABSTRACT

Transforming growth factor beta (TGF-beta) signals through three high affinity cell surface receptors, TGF-beta type I, type II, and type III receptors. The type III receptor, also known as betaglycan, binds to the type II receptor and is thought to act solely by "presenting" the TGF-beta ligand to the type II receptor. The short cytoplasmic domain of the type III receptor is thought to have no role in TGF-beta signaling because deletion of this domain has no effect on association with the type II receptor, or with the presentation role of the type III receptor. Here we demonstrate that the cytoplasmic domains of the type III and type II receptors interact specifically in a manner dependent on the kinase activity of the type II receptor and the ability of the type II receptor to autophosphorylate. This interaction results in the phosphorylation of the cytoplasmic domain of the type III receptor by the type II receptor. The type III receptor with the cytoplasmic domain deleted is able to bind TGF-beta, to bind the type II receptor, and to enhance TGF-beta binding to the type II receptor but is unable to enhance TGF-beta2 signaling, determining that the cytoplasmic domain is essential for some functions of the type III receptor. The type III receptor functions by selectively binding the autophosphorylated type II receptor via its cytoplasmic domain, thus promoting the preferential formation of a complex between the autophosphorylated type II receptor and the type I receptor and then dissociating from this active signaling complex. These studies, for the first time, elucidate important functional roles of the cytoplasmic domain of the type III receptor and demonstrate that these roles are essential for regulating TGF-beta signaling.


Subject(s)
Activin Receptors, Type I , Cytoplasm/metabolism , Proteoglycans/physiology , Receptors, Transforming Growth Factor beta/physiology , Signal Transduction , Transforming Growth Factor beta/physiology , Animals , COS Cells , Models, Molecular , Phosphorylation , Protein Serine-Threonine Kinases/physiology , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Structure-Activity Relationship
13.
Cytogenet Cell Genet ; 95(3-4): 157-62, 2001.
Article in English | MEDLINE | ID: mdl-12063393

ABSTRACT

Transforming growth factor-beta (TGF-beta) superfamily related growth factors signal by binding to transmembrane type I and type II receptor serine/threonine kinases (RSTK), which phosphorylate intracellular Smad transcription factors in response to ligand binding. Here we describe the cloning of the human type I RSTK activin receptor-like kinase 7 (ALK7), an orthologue of the previously identified rat ALK7. Nodal, a TGF-beta member expressed during embryonic development and implicated in developmental events like mesoderm formation and left-right axis specification, was recently shown to signal through ALK7. We found ALK7 mRNA to be most abundantly expressed in human brain, pancreas and colon. A cDNA encoding the open reading frame of ALK7 was obtained from a human brain cDNA library. Furthermore, a P1 artificial chromosome (PAC) clone containing the human ALK7 gene was isolated and fluorescent in situ hybridization (FISH) on metaphase chromosomes identified the gene locus as chromosome 2q24.1-->q3. To test the functionality of the ALK7 signaling, we generated recombinant adenoviruses containing a constitutively active form of ALK7 (Ad-caALK7), which is capable of activating downstream targets in a ligand independent manner. Infection with Ad-caALK7 of MIN6 insulinoma cells, in which ALK7 has previously been shown to be endogenously expressed, led to a marked increase in the phosphorylation of Smad2, a signaling molecule also used by TGF-betas and activins.


Subject(s)
Activin Receptors, Type I/genetics , Brain Chemistry/genetics , Protein Serine-Threonine Kinases/genetics , Activin Receptors, Type I/metabolism , Amino Acid Sequence , Chromosome Mapping , Cloning, Molecular , DNA-Binding Proteins/metabolism , Gene Expression , Gene Library , Humans , Insulinoma/genetics , Molecular Sequence Data , Phosphorylation , Protein Serine-Threonine Kinases/metabolism , Signal Transduction/genetics , Smad2 Protein , Trans-Activators/metabolism , Tumor Cells, Cultured
14.
J Cell Sci ; 114(Pt 24): 4359-69, 2001 Dec.
Article in English | MEDLINE | ID: mdl-11792802

ABSTRACT

Smad proteins transduce signals from transforming growth factor-beta (TGF-beta) superfamily ligands that regulate cell proliferation, differentiation and death through activation of receptor serine/threonine kinases. Phosphorylation of receptor-activated Smads (R-Smads) leads to formation of complexes with the common mediator Smad (Co-Smad), which are imported to the nucleus. Nuclear Smad oligomers bind to DNA and associate with transcription factors to regulate expression of target genes. Alternatively, nuclear R-Smads associate with ubiquitin ligases and promote degradation of transcriptional repressors, thus facilitating target gene regulation by TGF-beta. Smads themselves can also become ubiquitinated and are degraded by proteasomes. Finally, the inhibitory Smads (I-Smads) block phosphorylation of R-Smads by the receptors and promote ubiquitination and degradation of receptor complexes, thus inhibiting signalling.


Subject(s)
Activin Receptors, Type I/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Transforming Growth Factor beta/metabolism , Signal Transduction/physiology , Transforming Growth Factor beta/physiology , Activin Receptors, Type I/physiology , Animals , DNA-Binding Proteins/physiology , Humans , Phosphoproteins/physiology , Protein Serine-Threonine Kinases , Receptor Protein-Tyrosine Kinases/physiology , Receptor, Transforming Growth Factor-beta Type I , Receptor, Transforming Growth Factor-beta Type II , Receptors, Transforming Growth Factor beta/physiology , Smad Proteins , Smad2 Protein , Smad3 Protein , Smad4 Protein , Smad5 Protein , Smad8 Protein , Trans-Activators/physiology
15.
Oncogene ; 19(38): 4396-404, 2000 Sep 07.
Article in English | MEDLINE | ID: mdl-10980615

ABSTRACT

Smads, the intracellular effectors of transforming growth factor-beta (TGF-beta) family members, are somatically mutated at high frequency in particular types of human cancers. Certain of these mutations affect the Smad amino-terminal domain, which, in the case of Smad3 and Smad4, binds DNA. We investigated the functional consequences of four missense mutations in the Smad4 amino-terminal domain found in human tumors. The mutant proteins were found to have impaired abilities to bind DNA although they were fully capable of forming complexes with Smad3. All four Smad4 mutants showed decreased protein stability compared to wild-type Smad4. Two of the Smad4 mutants (G65V and P130S) were translocated to the nucleus and were capable of transactivating a Smad-dependent promoter in a ligand-dependent manner. In contrast, the L43S and R100T mutants were not translocated efficiently to the nucleus and consequently resulted in severely defective transcriptional responses to TGF-beta. Moreover, we demonstrate here the critical importance of two basic residues in the beta-hairpin loop of Smad3 or Smad4 for DNA binding, consistent with predictions from the Smad3 crystal structure. In addition, our results reveal that in the TGF-beta-induced heteromeric signaling complex, loss of DNA binding of Smad4 can be compensated by Smad3, however, both Smad3 and Smad4 are needed for efficient DNA binding and signaling. In conclusion, mutations in the amino-terminal domain of Smad4, that are found in cancer, show loss of multiple functional properties which may contribute to tumorigenesis.


Subject(s)
DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Mutation, Missense , Trans-Activators/genetics , Trans-Activators/metabolism , Transforming Growth Factor beta/metabolism , Amino Acid Sequence , Biological Transport , Cell Nucleus/metabolism , DNA-Binding Proteins/drug effects , Genes, Tumor Suppressor , Genetic Complementation Test , Humans , Molecular Sequence Data , Smad3 Protein , Smad4 Protein , Trans-Activators/drug effects , Transcription, Genetic , Transforming Growth Factor beta/pharmacology , Tumor Cells, Cultured/pathology
16.
Int Immunol ; 12(8): 1157-66, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10917890

ABSTRACT

The association of celiac disease (CD) with HLA-DQ2 and HLA-DQ8 is indicative of preferential mucosal T cell recognition of gluten fragments bound to either DQ allele. We have recently identified two gluten-derived, HLA-DQ8-restricted T cell stimulatory peptides, one each from gliadin and glutenin, recognized by specific T cell clones derived from the small intestine of CD patients. We have now performed molecular modeling and examined the fine specificity of these peptides in complex with HLA-DQ8. There is only one binding register for both peptides, with glutamine residues at the p1 and p9 anchor positions. Both T cell clones recognize substituted peptides at p1 and p9, but poorly so at p2-p8, especially the gliadin-specific clone. Contrasting patterns of recognition of p9Gln --> Glu peptide variants (both predicted as better DQ8 binders by modeling) were observed: enhancement of recognition for the gliadin peptide, yet complete absence thereof for the glutenin peptide. The double-substituted gliadin peptide variant p1/9Gln --> Glu, which can also arise by pepsin/acid/transglutaminase treatment, shows a considerable increase in sensitivity of recognition, consistent with better binding of this peptide to DQ8, as predicted by energy minimization. Surprisingly, the two native peptides are also recognized by their respective T cell clones in the context of the related molecule HLA-DQ9 (beta57Asp(+)). The p1/9Gln --> Glu gliadin peptide variant is likewise recognized, albeit with a 10-fold lower sensitivity, the first reported p9Glu binding in a beta57Asp(+) MHC II allele. Our results have important implications for the pathogenesis of autoimmune disease and the possible manipulation of aberrant responses thereof.


Subject(s)
Autoimmune Diseases/immunology , Celiac Disease/immunology , Epitopes/immunology , Genes, MHC Class II , Gliadin/immunology , Glutens/analogs & derivatives , Glutens/immunology , HLA-DQ Antigens/genetics , Alleles , Amino Acid Sequence , Amino Acid Substitution , Antigen Presentation , Autoantigens/immunology , Autoimmune Diseases/genetics , Binding Sites , Celiac Disease/genetics , Epitopes/chemistry , Genetic Predisposition to Disease , Gliadin/chemistry , Glutens/chemistry , HLA-DQ Antigens/chemistry , HLA-DQ Antigens/immunology , Intestinal Mucosa/immunology , Lymphocyte Activation , Models, Molecular , Molecular Mimicry , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/immunology , Polymorphism, Genetic , Protein Conformation , Sequence Alignment , Sequence Homology, Amino Acid , Structure-Activity Relationship
17.
J Biol Chem ; 275(38): 29244-56, 2000 Sep 22.
Article in English | MEDLINE | ID: mdl-10878024

ABSTRACT

Transforming growth factor-beta (TGF-beta) inhibits cell cycle progression, in part through up-regulation of gene expression of the p21(WAF1/Cip1) (p21) cell cycle inhibitor. Previously we have reported that the intracellular effectors of TGF-beta, Smad3 and Smad4, functionally cooperate with Sp1 to activate the human p21 promoter in hepatoma HepG2 cells. In this study we show that Smad3 and Smad4 when overexpressed in HaCaT keratinocytes lead to activation of the p21 promoter. Activation requires the binding sites for the ubiquitous transcription factor Sp1 on the proximal promoter. Induction of the endogenous HaCaT p21 gene by TGF-beta1 is further enhanced after overexpression of Smad3 and Smad4, whereas dominant negative mutants of Smad3 and Smad4 and the inhibitory Smad7 all inhibit p21 induction by TGF-beta1 in a dose-dependent manner. We show that Sp1 expressed in the Sp1-deficient Drosophila SL-2 cells binds to the proximal p21 promoter sequences, whereas Smad proteins do not. In support of this finding, we show that DNA-binding domain mutants of Smad3 and Smad4 are capable of transactivating the p21 promoter as efficiently as wild type Smads. Co-expression of Smad3 with Smad4 and Sp1 in SL-2 cells or co-incubation of phosphorylated Smad3, Smad4, and Sp1 in vitro results in enhanced binding of Sp1 to the p21 proximal promoter sequences. We demonstrate that Sp1 physically and directly interacts with Smad2, Smad3, and weakly with Smad4 via their amino-terminal (Mad-Homology 1) domain. Finally, by using GAL4 fusion proteins we show that the glutamine-rich sequences in the transactivation domain of Sp1 contribute to the cooperativity with Smad proteins. In conclusion, Smad proteins play important roles in regulation of the p21 gene by TGF-beta, and the functional cooperation of Smad proteins with Sp1 involves the physical interaction of these two types of transcription factors.


Subject(s)
Cyclins/metabolism , DNA-Binding Proteins/metabolism , Keratinocytes/metabolism , Signal Transduction/drug effects , Sp1 Transcription Factor/metabolism , Trans-Activators/metabolism , Transforming Growth Factor beta/pharmacology , Cell Line , Cyclin-Dependent Kinase Inhibitor p21 , Cyclins/genetics , DNA-Binding Proteins/genetics , Dose-Response Relationship, Drug , Gene Expression Regulation/drug effects , Humans , Smad3 Protein , Smad4 Protein , Sp1 Transcription Factor/genetics , Trans-Activators/genetics
18.
Diabetologia ; 43(5): 609-24, 2000 May.
Article in English | MEDLINE | ID: mdl-10855536

ABSTRACT

AIMS/HYPOTHESIS: We modelled the three-dimensional structure of I-A(g7), the chief genetic component of diabetes in non-obese diabetic mice, to understand the unusual properties of this molecule. METHODS: Modelling was done, in complex with established antigenic peptides, based on the structure of I-A(k). RESULTS: The selectivity of the I-A(g7) molecule changes greatly at pockets 9 and 6 but hardly at all at pockets 1, 4 and 7, between endosomal pH (5.0) and extracellular pH (7.0), in agreement with previous results. This selectivity is attributed to the unique combination of beta9His, beta56His and beta57Ser. The positive charges in and around pocket 9 at pH 5, favour binding by negatively charged residues. At pH 7 however, the uncharged alpha68, beta9 and beta56 histidines favour the accommodation of the bulky residues lysine, arginine, phenylalanine and tyrosine at pocket 9. The combination of beta9His and alpha66Glu is responsible for the pH-dependent selectivity at pocket 6. Furthermore, the lack of repulsion between beta56His and alpha76Arg at pH 7 leads to a more stable ternary complex. CONCLUSION/INTERPRETATION: These results reconcile previous conflicts over the peptide binding ability of I-A(g7) and its motif. They furthermore provide possible explanations for the short lifetime of cell-surface I-A(g7) complexes in vivo, the higher threshold of thymic negative selection and inherent self-reactivity shown by immunocytes in these mice and the protection from diabetes afforded to them by several transgenically expressed mouse class II alleles. This contributes to an understanding of the pathogenesis of Type I (insulin-dependent) diabetes mellitus in this animal.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Histocompatibility Antigens Class II/chemistry , Models, Molecular , Amino Acid Sequence , Animals , Binding Sites , Chemical Phenomena , Chemistry, Physical , Crystallization , Electrochemistry , Histocompatibility Antigens Class II/metabolism , Hydrogen-Ion Concentration , Mice , Mice, Inbred NOD , Molecular Sequence Data , Sequence Alignment , Structure-Activity Relationship
19.
J Cell Sci ; 112 ( Pt 24): 4557-68, 1999 Dec.
Article in English | MEDLINE | ID: mdl-10574705

ABSTRACT

The capacities of different transforming growth factor-(beta) (TGF-(beta)) superfamily members to drive epithelial to mesenchymal transdifferentiation of the murine mammary epithelial cell line NMuMG were investigated. TGF-(beta)1, but not activin A or osteogenic protein-1 (OP-1)/bone morphogenetic protein-7 (BMP-7), was able to induce morphological transformation of NMuMG cells as shown by reorganisation of the actin cytoskeleton and relocalisation/downregulation of E-cadherin and (beta)-catenin, an effect that was abrogated by the more general serine/threonine kinase and protein kinase C inhibitor, staurosporine. TGF-(beta)1 bound to TGF-(beta) type I receptor (T(beta)R-I)/ALK-5 and T(beta)R-II, but not to activin type I receptor (ActR-I)/ALK-2. Activin A bound to ActR-IB/ALK-4 and ActR-II, and BMP-7 bound to ActR-I/ALK-2, BMP type I receptor (BMPR-I)/ALK-3, ActR-II and BMPR-II. TGF-(beta)1 and BMP-7 activated the Smad-binding element (SBE)(4) promoter with equal potency, whereas activin A had no effect. Transfection of constitutively active (CA)-ALK-4 activated the 3TP promoter to the same extent as TGF-(beta)1 and CA-ALK-5 indicating that activin signalling downstream of type I receptors was functional in NMuMG cells. In agreement with this, activin A induced low levels of plasminogen activator inhibitor I expression compared to the high induction by TGF-(beta)1. In contrast to activin A and BMP-7, TGF-(beta)1 strongly induced Smad2 phosphorylation. Consistent with these findings, TGF-(beta)1 induced the nuclear accumulation of Smad2 and/or Smad3. In addition, NMuMG cells transiently infected with adenoviral vectors expressing high level CA-ALK-5 exhibited full transdifferentiation. On the other hand, infections with low level CA-ALK-5, which alone did not result in transdifferentiation, together with Smad2 and Smad4, or with Smad3 and Smad4 led to transdifferentiation. In conclusion, TGF-(beta)1 signals potently and passes the activation threshold to evoke NMuMG cell transdifferentiation. The TGF-(beta) type I receptor (ALK-5) and its effector Smad proteins mediate the epithelial to mesenchymal transition. Activin A does not induce mesenchymal transformation, presumably because the number of activin receptors is limited, while BMP-7-initiated signalling cannot mediate transdifferentiation.


Subject(s)
Activin Receptors, Type I , Cell Differentiation/physiology , Mammary Glands, Animal/cytology , Protein Serine-Threonine Kinases/physiology , Receptors, Transforming Growth Factor beta/physiology , Trans-Activators/physiology , Activins , Animals , Bone Morphogenetic Protein 7 , Bone Morphogenetic Proteins/physiology , Cell Differentiation/drug effects , Down-Regulation , Enzyme Inhibitors/pharmacology , Epithelial Cells/cytology , Inhibins/physiology , Luciferases/genetics , Mesoderm/cytology , Mice , Protein Kinase Inhibitors , Receptor, Transforming Growth Factor-beta Type I , Transforming Growth Factor beta/physiology , Tumor Cells, Cultured
20.
J Biol Chem ; 274(41): 29572-81, 1999 Oct 08.
Article in English | MEDLINE | ID: mdl-10506225

ABSTRACT

The cell cycle inhibitor protein p21(WAF1/Cip1) (p21) is a critical downstream effector in p53-dependent mechanisms of growth control and p53-independent pathways of terminal differentiation. We have recently reported that the transforming growth factor-beta pathway-specific Smad3 and Smad4 proteins transactivate the human p21 promoter via a short proximal region, which contains multiple binding sites for the ubiquitous transcription factor Sp1. In the present study we show that the Sp1-occupied promoter region mediates transactivation of the p21 promoter by c-Jun and the related proteins JunB, JunD, and ATF-2. By using gel electrophoretic mobility shift assays we show that this region does not contain a binding site for c-Jun. In accordance with the DNA binding data, c-Jun was unable to transactivate the p21 promoter when overexpressed in the Sp1-deficient Drosophila-derived SL2 cells. Coexpression of c-Jun and Sp1 in these cells resulted in a strong synergistic transactivation of this promoter. In addition, a chimeric promoter consisting of six tandem high affinity Sp1-binding sites fused with the CAT gene was transactivated by overexpressed c-Jun in HepG2 cells. The above data propose functional cooperation between c-Jun and Sp1. Physical interactions between the two factors were demonstrated in vitro by using GST-Sp1 hybrid proteins expressed in bacteria and in vitro transcribed-translated c-Jun. The region of c-Jun mediating interaction with Sp1 was mapped within the basic region leucine zipper domain. In vivo, functional interactions between c-Jun and Sp1 were demonstrated using a GAL4-based transactivation assay. Overexpressed c-Jun transactivated a chimeric promoter consisting of five tandem GAL4-binding sites only when coexpressed with GAL4-Sp1-(83-778) fusion proteins in HepG2 cells. By utilizing the same assay, we found that the glutamine-rich segment of the B domain of Sp1 (Bc, amino acids 424-542) was sufficient for c-Jun-induced transactivation of the p21 promoter. In conclusion, our data support a mechanism of superactivation of Sp1 by c-Jun, which is based on physical and functional interactions between these two transcription factors on the human p21 and possibly other Sp1-dependent promoters.


Subject(s)
Cyclins/genetics , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/metabolism , Saccharomyces cerevisiae Proteins , Sp1 Transcription Factor/metabolism , Trans-Activators/pharmacology , Animals , Base Sequence , Cyclin-Dependent Kinase Inhibitor p21 , DNA-Binding Proteins/metabolism , Drosophila/genetics , Fungal Proteins/genetics , Gene Expression Regulation , Genes, Reporter , Humans , Protein Binding , Recombinant Fusion Proteins , Transcription Factors/genetics , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...