Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Int J Dev Neurosci ; 83(7): 581-599, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37574439

ABSTRACT

Cortical development depends on neuronal migration of both excitatory and inhibitory interneurons. Neuronal migration disorders (NMDs) are conditions characterised by anatomical cortical defects leading to varying degrees of neurocognitive impairment, developmental delay and seizures. Refractory epilepsy affects 15 million people worldwide, and it is thought that cortical developmental disorders are responsible for 25% of childhood cases. However, little is known about the epidemiology of these disorders, nor are their aetiologies fully understood, though many are associated with sporadic genetic mutations. In this review, we aim to highlight X-linked NMDs including lissencephaly, periventricular nodular heterotopia and polymicrogyria because of their mostly familial inheritance pattern. We focus on the most prominent genes responsible: including DCX, ARX, FLNA, FMR1, L1CAM, SRPX2, DDX3X, NSHDL, CUL4B and OFD1, outlining what is known about their prevalence among NMDs, and the underlying pathophysiology. X-linked disorders are important to recognise clinically, as females often have milder phenotypes. Consequently, there is a greater chance they survive to reproductive age and risk passing the mutations down. Effective genetic screening is important to prevent and treat these conditions, and for this, we need to know gene mutations and have a clear understanding of the function of the genes involved. This review summarises the knowledge base and provides clear direction for future work by both scientists and clinicians alike.


Subject(s)
Epilepsy , Malformations of Cortical Development, Group II , Female , Humans , Epilepsy/genetics , Sex Factors , Genetic Testing , Mutation , Malformations of Cortical Development, Group II/complications , Malformations of Cortical Development, Group II/genetics , Fragile X Mental Retardation Protein/genetics , Cullin Proteins/genetics
2.
Cancers (Basel) ; 15(4)2023 Feb 07.
Article in English | MEDLINE | ID: mdl-36831384

ABSTRACT

Arkadia (RNF111) is a positive regulator of the TGF-ß signaling that mediates the proteasome-dependent degradation of negative factors of the pathway. It is classified as an E3 ubiquitin ligase and a SUMO-targeted ubiquitin ligase (STUBL), implicated in various pathological conditions including cancer and fibrosis. The enzymatic (ligase) activity of Arkadia is located at its C-terminus and involves the RING domain. Notably, E3 ligases require E2 enzymes to perform ubiquitylation. However, little is known about the cooperation of Arkadia with various E2 enzymes and the type of ubiquitylation that they mediate. In the present work, we study the interaction of Arkadia with the E2 partners UbcH5B and UbcH13, as well as UbcH7. Through NMR spectroscopy, we found that the E2-Arkadia interaction surface is similar in all pairs examined. Nonetheless, the requirements and factors that determine an enzymatically active E2-Arkadia complex differ in each case. Furthermore, we revealed that the cooperation of Arkadia with different E2s results in either monoubiquitylation or polyubiquitin chain formation via K63, K48, or K11 linkages, which can determine the fate of the substrate and lead to distinct biological outcomes.

3.
Int J Mol Sci ; 23(18)2022 Sep 13.
Article in English | MEDLINE | ID: mdl-36142504

ABSTRACT

Arkadia is a positive regulator of the TGFß-SMAD2/3 pathway, acting through its C-terminal RING-H2 domain and targeting for degradation of its negative regulators. Here we explore the role of regions outside the RING domain (non-RING elements) of Arkadia on the E2-E3 interaction. The contribution of the non-RING elements was addressed using Arkadia RING 68 aa and Arkadia 119 aa polypeptides. The highly conserved NRGA (asparagine-arginine-glycine-alanine) and TIER (threonine-isoleucine-glutamine-arginine) motifs within the 119 aa Arkadia polypeptide, have been shown to be required for pSMAD2/3 substrate recognition and ubiquitination in vivo. However, the role of the NRGA and TIER motifs in the enzymatic activity of Arkadia has not been addressed. Here, nuclear magnetic resonance interaction studies with the E2 enzyme, UBCH5B, C85S UBCH5B-Ub oxyester hydrolysis, and auto-ubiquitination assays were used to address the role of the non-RING elements in E2-E3 interaction and in the enzymatic activity of the RING. The results support that the non-RING elements including the NRGA and TIER motifs are required for E2-E3 recognition and interaction and for efficient auto-ubiquitination. Furthermore, while Arkadia isoform-2 and its close homologue Arkadia 2C are known to interact with free ubiquitin, the results here showed that Arkadia isoform-1 does not interact with free ubiquitin.


Subject(s)
Isoleucine , Ubiquitin-Protein Ligases , Alanine/metabolism , Arginine/metabolism , Asparagine/metabolism , Glutamine/metabolism , Glycine/metabolism , Isoleucine/metabolism , Threonine/metabolism , Transforming Growth Factor beta/metabolism , Ubiquitin/metabolism , Ubiquitin-Conjugating Enzymes/metabolism , Ubiquitin-Protein Ligases/metabolism , Ubiquitination
4.
Front Mol Biosci ; 9: 844129, 2022.
Article in English | MEDLINE | ID: mdl-35281275

ABSTRACT

Single nucleotide polymorphisms (SNPs) are genetic variations which can play a vital role in the study of human health. SNP studies are often used to identify point mutations that are associated with diseases. Arkadia (RNF111) is an E3 ubiquitin ligase that enhances transforming growth factor-beta (TGF-ß) signaling by targeting negative regulators for degradation. Dysregulation of the TGF-ß pathway is implicated in cancer because it exhibits tumor suppressive activity in normal cells while in tumor cells it promotes invasiveness and metastasis. Τhe SNP CGT > TGT generated an amino-acid (aa) substitution of Arginine 957 to Cysteine on the enzymatic RING domain of Arkadia. This was more prevalent in a tumor than in a normal tissue sample of a patient with colorectal cancer. This prompted us to investigate the effect of this mutation in the structure and activity of Arkadia RING. We used nuclear magnetic resonance (NMR) to analyze at an atomic-level the structural and dynamic properties of the R957C Arkadia RING domain, while ubiquitination and luciferase assays provided information about its enzymatic functionality. Our study showed that the R957C mutation changed the electrostatic properties of the RING domain however, without significant effects on the structure of its core region. However, the functional studies revealed that the R957C Arkadia exhibits significantly increased enzymatic activity supporting literature data that Arkadia within tumor cells promotes aggressive and metastatic behavior.

5.
J Exp Med ; 218(11)2021 11 01.
Article in English | MEDLINE | ID: mdl-34473197

ABSTRACT

TGF-ß signaling is fundamental for both Th17 and regulatory T (Treg) cell differentiation. However, these cells differ in requirements for downstream signaling components, such as SMAD effectors. To further characterize mechanisms that distinguish TGF-ß signaling requirements for Th17 and Treg cell differentiation, we investigated the role of Arkadia (RNF111), an E3 ubiquitin ligase that mediates TGF-ß signaling during development. Inactivation of Arkadia in CD4+ T cells resulted in impaired Treg cell differentiation in vitro and loss of RORγt+FOXP3+ iTreg cells in the intestinal lamina propria, which increased susceptibility to microbiota-induced mucosal inflammation. In contrast, Arkadia was dispensable for Th17 cell responses. Furthermore, genetic ablation of two Arkadia substrates, the transcriptional corepressors SKI and SnoN, rescued Arkadia-deficient iTreg cell differentiation both in vitro and in vivo. These results reveal distinct TGF-ß signaling modules governing Th17 and iTreg cell differentiation programs that could be targeted to selectively modulate T cell functions.


Subject(s)
Cell Differentiation/immunology , Proto-Oncogene Proteins/immunology , Signal Transduction/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Transforming Growth Factor beta/immunology , Ubiquitin-Protein Ligases/immunology , Animals , Inflammation/immunology , Mice , Mice, Inbred C57BL , Ubiquitin/immunology
6.
J Mol Biol ; 429(15): 2373-2386, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28647409

ABSTRACT

Arkadia (Rnf111) is an E3 ubiquitin ligase that plays a central role in the amplification of transforming growth factor beta (TGF-ß) signaling responses by targeting for degradation the negative regulators of the pathway, Smad6 and Smad7, and the nuclear co-repressors Ski and Skil (SnoN). Arkadia's function in vivo depends on the really interesting new gene (RING)-H2 interaction with the E2 enzyme UbcH5b in order to ligate ubiquitin chains on its substrates. A conserved tryptophan (W972) in the C-terminal α-helix is widely accepted as essential for E2 recruitment and interaction and thus also for E3 enzymatic activity. The present NMR-driven study provides an atomic-level investigation of the structural and dynamical properties of two W972 Arkadia RING mutants, attempting to illuminate for the first time the differences between a functional and a nonfunctional mutant W972A and W972R, respectively. A TGF-ß-responsive promoter driving luciferase was used to assay for Arkadia function in vivo. These experiments showed that the Arkadia W972A mutant has the same activity as wild-type (WT) Arkadia in enhancing TGF-ß signaling responses, while W972R does not. Only minor structural differences exist between the W972A RING domain and WT-RING. In contrast, the W972R mutant hardly interacts with E2. The loss of function correlates with structural changes in the C-terminal α-helix and an increase in the distance between the Zn(II) ions. Our data show that the position occupied by W972 within WT Arkadia is critical for the function of RING and that it depends on the nature of the residue at this position.


Subject(s)
Mutant Proteins/chemistry , Mutant Proteins/metabolism , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Alanine/genetics , Alanine/metabolism , Amino Acid Substitution , Arginine/genetics , Arginine/metabolism , Genes, Reporter , Luciferases/analysis , Magnetic Resonance Spectroscopy , Mutant Proteins/genetics , Nuclear Proteins/genetics , Protein Conformation , Signal Transduction , Transforming Growth Factor beta/metabolism , Tryptophan/genetics , Tryptophan/metabolism , Ubiquitin-Protein Ligases/genetics
7.
Science ; 345(6204): 1254927, 2014 Sep 26.
Article in English | MEDLINE | ID: mdl-25258086

ABSTRACT

Development requires tissue growth as well as cell diversification. To address how these processes are coordinated, we analyzed the development of molecularly distinct domains of neural progenitors in the mouse and chick neural tube. We show that during development, these domains undergo changes in size that do not scale with changes in overall tissue size. Our data show that domain proportions are first established by opposing morphogen gradients and subsequently controlled by domain-specific regulation of differentiation rate but not differences in proliferation rate. Regulation of differentiation rate is key to maintaining domain proportions while accommodating both intra- and interspecies variations in size. Thus, the sequential control of progenitor specification and differentiation elaborates pattern without requiring that signaling gradients grow as tissues expand.


Subject(s)
Body Patterning , Cell Differentiation , Neural Tube/embryology , Spinal Cord/embryology , Stem Cells/cytology , Animals , Chick Embryo , Mice , Models, Biological , Neural Tube/cytology
8.
PLoS Biol ; 11(4): e1001538, 2013.
Article in English | MEDLINE | ID: mdl-23610558

ABSTRACT

Little is known about extrinsic signals required for the advancement of motor neuron (MN) axons, which extend over long distances in the periphery to form precise connections with target muscles. Here we present that Rnf165 (Arkadia-like; Arkadia2; Ark2C) is expressed specifically in the nervous system and that its loss in mice causes motor innervation defects that originate during development and lead to wasting and death before weaning. The defects range from severe reduction of motor axon extension as observed in the dorsal forelimb to shortening of presynaptic branches of the phrenic nerve, as observed in the diaphragm. Molecular functional analysis showed that in the context of the spinal cord Ark2C enhances transcriptional responses of the Smad1/5/8 effectors, which are activated (phosphorylated) downstream of Bone Morphogenetic Protein (BMP) signals. Consistent with Ark2C-modulated BMP signaling influencing motor axons, motor pools in the spinal cord were found to harbor phosphorylated Smad1/5/8 (pSmad) and treatment of primary MN with BMP inhibitor diminished axon length. In addition, genetic reduction of BMP-Smad signaling in Ark2C (+/-) mice caused the emergence of Ark2C (-/-) -like dorsal forelimb innervation deficits confirming that enhancement of BMP-Smad responses by Ark2C mediates efficient innervation. Together the above data reveal an involvement of BMP-Smad signaling in motor axon advancement.


Subject(s)
Bone Morphogenetic Proteins/physiology , Motor Neurons/physiology , Nerve Tissue Proteins/metabolism , Signal Transduction , Smad Proteins/physiology , Ubiquitin-Protein Ligases/metabolism , Animals , Axons/physiology , Body Patterning , Cell Enlargement , Forelimb/innervation , Gene Expression , HEK293 Cells , Humans , Mice , Mice, 129 Strain , Mice, Knockout , Muscle, Skeletal/innervation , Muscular Atrophy/genetics , Mutagenesis, Insertional , Nerve Tissue Proteins/genetics , Phenotype , Phrenic Nerve/pathology , Proteolysis , Spinal Cord/metabolism , Transcriptional Activation , Ubiquitin-Protein Ligases/genetics , Ubiquitination
9.
EMBO J ; 32(7): 982-95, 2013 Apr 03.
Article in English | MEDLINE | ID: mdl-23455154

ABSTRACT

The INK4/ARF locus regulates senescence and is frequently altered in cancer. In normal cells, the INK4/ARF locus is found silenced by Polycomb repressive complexes (PRCs). Which are the mechanisms responsible for the recruitment of PRCs to INK4/ARF and their other target genes remains unclear. In a genetic screen for transcription factors regulating senescence, we identified the homeodomain-containing protein HLX1 (H2.0-like homeobox 1). Expression of HLX1 extends cellular lifespan and blunts oncogene-induced senescence. Using quantitative proteomics, we identified p16(INK4a) as the key target mediating the effects of HLX1 in senescence. HLX1 represses p16(INK4a) transcription by recruiting PRCs and HDAC1. This mechanism has broader implications, as HLX1 also regulates a subset of PRC targets besides p16(INK4a). Finally, sampling members of the Homeobox family, we identified multiple genes with ability to repress p16(INK4a). Among them, we found HOXA9 (Homeobox A9), a putative oncogene in leukaemia, which also recruits PRCs and HDAC1 to regulate p16(INK4a). Our results reveal an unexpected and conserved interplay between homeodomain-containing proteins and PRCs with implications in senescence, development and cancer.


Subject(s)
Cellular Senescence/physiology , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression Regulation/physiology , Homeodomain Proteins/metabolism , Polycomb-Group Proteins/metabolism , Transcription Factors/metabolism , Cyclin-Dependent Kinase Inhibitor p16/genetics , HeLa Cells , Histone Deacetylase 1/genetics , Histone Deacetylase 1/metabolism , Homeodomain Proteins/genetics , Humans , Polycomb-Group Proteins/genetics , Transcription Factors/genetics
10.
PLoS One ; 8(1): e55186, 2013.
Article in English | MEDLINE | ID: mdl-23390484

ABSTRACT

The Transforming Growth Factor-ß (TGF-ß) signaling pathway is one of the major pathways essential for normal embryonic development and tissue homeostasis, with anti-tumor but also pro-metastatic properties in cancer. This pathway directly regulates several target genes that mediate its downstream functions, however very few microRNAs (miRNAs) have been identified as targets. miRNAs are modulators of gene expression with essential roles in development and a clear association with diseases including cancer. Little is known about the transcriptional regulation of the primary transcripts (pri-miRNA, pri-miR) from which several mature miRNAs are often derived. Here we present the identification of miRNAs regulated by TGF-ß signaling in mouse embryonic stem (ES) cells and early embryos. We used an inducible ES cell system to maintain high levels of the TGF-ß activated/phosphorylated Smad2/3 effectors, which are the transcription factors of the pathway, and a specific inhibitor that blocks their activation. By performing short RNA deep-sequencing after 12 hours Smad2/3 activation and after 16 hours inhibition, we generated a database of responsive miRNAs. Promoter/enhancer analysis of a subset of these miRNAs revealed that the transcription of pri-miR-181c/d and the pri-miR-341∼3072 cluster were found to depend on activated Smad2/3. Several of these miRNAs are expressed in early mouse embryos, when the pathway is known to play an essential role. Treatment of embryos with TGF-ß inhibitor caused a reduction of their levels confirming that they are targets of this pathway in vivo. Furthermore, we showed that pri-miR-341∼3072 transcription also depends on FoxH1, a known Smad2/3 transcription partner during early development. Together, our data show that miRNAs are regulated directly by the TGF-ß/Smad2/3 pathway in ES cells and early embryos. As somatic abnormalities in functions known to be regulated by the TGF-ß/Smad2/3 pathway underlie tumor suppression and metastasis, this research also provides a resource for miRNAs involved in cancer.


Subject(s)
Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , MicroRNAs/biosynthesis , Signal Transduction/physiology , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Transforming Growth Factor beta/metabolism , Animals , Cells, Cultured , Embryo, Mammalian/cytology , Embryonic Stem Cells/cytology , Gene Expression Regulation, Developmental/drug effects , Gene Expression Regulation, Developmental/physiology , Mice , Multigene Family , Neoplasms/embryology , Neoplasms/metabolism , Signal Transduction/drug effects , Transcription, Genetic/drug effects , Transcription, Genetic/physiology , Transforming Growth Factor beta/pharmacology
11.
Proteins ; 80(5): 1484-9, 2012 May.
Article in English | MEDLINE | ID: mdl-22411132

ABSTRACT

Arkadia (Rnf111), an E3 Ubiquitin (Ub) ligase, amplifies TGF-ß signaling responses by targeting for degradation of the negative regulators Smad6/7 and the SnoN/Ski transcriptional repressors when they block the TGF-ß effectors Smad2/3. The E3 ligase activity of Arkadia depends on its C-terminal RING-H2 domain that constitutes the docking site for the E2 Ub-conjugating enzyme carrying the activated Ub. We determined the nuclear magnetic resonance solution structure of Arkadia's RING-H2 domain and revealed a (ß)ßßα fold, fully consistent with the expected "cross-brace" mode of Zn(II)-ligation. In addition, the interaction of the Arkadia RING-H2 domain with its E2 partner enzyme (UbcH5b) was examined through chemical shift perturbation. Proteins 2012. © 2012 Wiley Periodicals, Inc.


Subject(s)
Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism , Humans , Models, Molecular , Nuclear Magnetic Resonance, Biomolecular , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Conformation , Protein Interaction Domains and Motifs
12.
Cancer Res ; 71(20): 6438-49, 2011 Oct 15.
Article in English | MEDLINE | ID: mdl-21998011

ABSTRACT

TGF-ß signaling provides tumor protection against colorectal cancer (CRC). Mechanisms that support its tumor-suppressive properties remain unclear. The ubiquitin ligase Arkadia/RNF111 enhances TGF-ß signaling responses by targeting repressors of the pathway for degradation. The corepressors SnoN/Ski, critical substrates of Arkadia, complex with the activated TGF-ß signaling effectors Smad2/3 (pSmad2/3) on the promoters of target genes and block their transcription. Arkadia degrades this complex including pSmad2/3 and unblocks the promoter. Here, we report that Arkadia is expressed highly in the mouse colonic epithelium. Heterozygous Akd(+/-) mice are normal but express less Arkadia. This leads to reduced expression of several TGF-ß target genes, suggesting that normal levels of Arkadia are required for efficient signaling responses. Critically, Akd(+/-) mice exhibit increased susceptibility to azoxymethane/dextran sodium sulfate carcinogen-induced CRC, as they develop four-fold more tumors than wild-type mice. Akd(+/-) tumors also exhibit a more aggressive pathology, higher proliferation index, and reduced cytostasis. Therefore, Arkadia functions as a tumor suppressor whose peak expression is required to suppress CRC development and progression. The accumulation of nuclear SnoN and pSmad2, along with the downregulation of TGF-ß target genes observed in Akd(+/-) colon and tumors, suggest that tumor-suppressing properties of Arkadia are mediated by its ability to derepress TGF-ß signaling. Consistent with this likelihood, we identified mutations in primary colorectal tumors from human patients that reduce Arkadia function and are associated with the accumulation of nuclear SNON. Collectively, our findings reveal that Arkadia enhances TGF-ß signaling responses and supports its tumor-suppressing properties in CRC.


Subject(s)
Adenocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Nuclear Proteins/metabolism , Transforming Growth Factor beta/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , Amino Acid Sequence , Animals , Azoxymethane/toxicity , Base Sequence , Colon/metabolism , Colorectal Neoplasms/chemically induced , Gene Expression Regulation, Neoplastic , HEK293 Cells , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Molecular Sequence Data , Mutation , Nuclear Proteins/genetics , Proto-Oncogene Proteins/metabolism , Severity of Illness Index , Signal Transduction , Smad2 Protein/metabolism , Ubiquitin-Protein Ligases/genetics
13.
PLoS One ; 6(5): e20197, 2011.
Article in English | MEDLINE | ID: mdl-21637844

ABSTRACT

The evolutionarily conserved Hox family of homeodomain transcription factors plays fundamental roles in regulating cell specification along the anterior posterior axis during development of all bilaterian animals by controlling cell fate choices in a highly localized, extracellular signal and cell context dependent manner. Some studies have established downstream target genes in specific systems but their identification is insufficient to explain either the ability of Hox genes to direct homeotic transformations or the breadth of their patterning potential. To begin delineating Hox gene function in neural development we used a mouse ES cell based system that combines efficient neural differentiation with inducible Hoxb1 expression. Gene expression profiling suggested that Hoxb1 acted as both activator and repressor in the short term but predominantly as a repressor in the long run. Activated and repressed genes segregated in distinct processes suggesting that, in the context examined, Hoxb1 blocked differentiation while activating genes related to early developmental processes, wnt and cell surface receptor linked signal transduction and cell-to-cell communication. To further elucidate aspects of Hoxb1 function we used loss and gain of function approaches in the mouse and chick embryos. We show that Hoxb1 acts as an activator to establish the full expression domain of CRABPI and II in rhombomere 4 and as a repressor to restrict expression of Lhx5 and Lhx9. Thus the Hoxb1 patterning activity includes the regulation of the cellular response to retinoic acid and the delay of the expression of genes that commit cells to neural differentiation. The results of this study show that ES neural differentiation and inducible Hox gene expression can be used as a sensitive model system to systematically identify Hox novel target genes, delineate their interactions with signaling pathways in dictating cell fate and define the extent of functional overlap among different Hox genes.


Subject(s)
Cell Differentiation/genetics , Embryonic Stem Cells/cytology , Homeodomain Proteins/metabolism , Neurons/cytology , Animals , Cell Differentiation/drug effects , Chick Embryo , Embryo, Mammalian/drug effects , Embryo, Mammalian/metabolism , Embryonic Stem Cells/metabolism , Gene Expression Profiling , Gene Expression Regulation, Developmental/drug effects , Homeodomain Proteins/genetics , LIM-Homeodomain Proteins , Mice , Nerve Tissue Proteins/metabolism , Neurons/drug effects , Neurons/metabolism , Receptors, Retinoic Acid/genetics , Receptors, Retinoic Acid/metabolism , Repressor Proteins/genetics , Repressor Proteins/metabolism , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Rhombencephalon/drug effects , Rhombencephalon/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics , Transcription Factors/metabolism , Tretinoin/pharmacology
14.
J Vis Exp ; (49)2011 Mar 03.
Article in English | MEDLINE | ID: mdl-21403637

ABSTRACT

BMPs are responsible for a wide range of developmental and biological effects. BMP receptors activate (phosphorylate) the Smad1/5/8 effectors, which then, form a complex with Smad4 and translocate to the nucleus where they function as transcription factors to initiate BMP specific downstream effects (1). Traditional immuno-fluorescence techniques with antibodies against phospho-Smad peptides exhibit low sensitivity, high background and offer gross quantification as they rely on intensity of the antibody signal particularly if this is photosensitive fluorescent. In addition, phospho-Smads may not all be in complex with Smad4 and engaged in active transcription. In situ PLA is a technology capable of detecting protein interactions with high specificity and sensitivity (2-4). This new technology couples antibody recognition with the amplification of DNA surrogate of the protein. It generates a localized, discrete signal in a form of spots revealing the exact position of the recognition event. The number of signals can be counted and compared providing a measurement. We applied in situ PLA, using the Duolink kit, with a combination of antibodies that allows the detection of the BMP signaling effectors phospho-Smad1/5/8 and Smad4 only when these are in proximity i.e. in a complex, which occurs only with signaling activation. This allowed for the first time, the visualization and measurement of endogenous BMP signaling with high specificity and sensitivity in a time course experiment under BMP4 stimulation.


Subject(s)
Biological Assay/methods , Bone Morphogenetic Protein 4/metabolism , Protein Interaction Mapping/methods , Animals , Bone Morphogenetic Protein 4/antagonists & inhibitors , Bone Morphogenetic Protein 4/pharmacology , COS Cells , Chlorocebus aethiops , HEK293 Cells , Humans , Phosphorylation , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Smad Proteins/metabolism
15.
Immunobiology ; 216(3): 343-50, 2011 Mar.
Article in English | MEDLINE | ID: mdl-20961651

ABSTRACT

The pleiotropic cytokine transforming growth factor-ß (TGF-ß) signals through different pathways among which the Smad- and the MAP-Kinase pathways are already well characterized. Both pathways utilize adaptor/chaperone molecules that facilitate or modulate the intracellular signaling events. Two of the proteins shown in vitro to play a role in Smad-dependent signaling are the TGF-ß Receptor Associated Protein-1 (TRAP1, also TGFBRAP1) and its homologue VPS39, also known as Vam6 and TRAP1-Like-Protein (TLP). We generated mice deficient for TRAP1 and VPS39/TLP, respectively. Absence of TRAP1 protein results in death at either of two defined timepoints during embryogenesis, before the blastula stage or during gastrulation, whereas most of the VPS39 deficient mice die before E6.5. Heterozygous mice show no overt phenotype. In summary, our data indicate that TRAP1 and VPS39 are nonredundant and essentially required for early embryonic development.


Subject(s)
Blastula/embryology , Embryonic Development , Gastrula/embryology , Guanine Nucleotide Exchange Factors/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Transforming Growth Factor beta/metabolism , Animals , Animals, Genetically Modified , Autophagy-Related Proteins , Blotting, Northern , Blotting, Western , Cells, Cultured , Gene Expression , Genotype , Guanine Nucleotide Exchange Factors/genetics , HSP90 Heat-Shock Proteins , Intracellular Signaling Peptides and Proteins/genetics , Mice , Polymerase Chain Reaction , Signal Transduction , Vesicular Transport Proteins
16.
PLoS One ; 4(1): e4268, 2009.
Article in English | MEDLINE | ID: mdl-19172185

ABSTRACT

The Transforming Growth Factor (TGF) beta signalling family includes morphogens, such as Nodal and Activin, with important functions in vertebrate development. The concentration of the morphogen is critical for fate decisions in the responding cells. Smad2 and Smad3 are effectors of the Nodal/Activin branch of TGFbeta signalling: they are activated by receptors, enter the nucleus and directly transcribe target genes. However, there have been no studies correlating levels of Smad2/3 activation with expression patterns of endogenous target genes in a developmental context over time. We used mouse Embryonic Stem (ES) cells to create a system whereby levels of activated Smad2/3 can be manipulated by an inducible constitutively active receptor (Alk4*) and an inhibitor (SB-431542) that blocks specifically Smad2/3 activation. The transcriptional responses were analysed by microarrays at different time points during activation and repression. We identified several genes that follow faithfully and reproducibly the Smad2/3 activation profile. Twenty-seven of these were novel and expressed in the early embryo downstream of Smad2/3 signalling. As they responded to Smad2/3 activation in the absence of protein synthesis, they were considered direct. These immediate responsive genes included negative intracellular feedback factors, like SnoN and I-Smad7, which inhibit the transcriptional activity of Smad2/3. However, their activation did not lead to subsequent repression of target genes over time, suggesting that this type of feedback is inefficient in ES cells or it is counteracted by mechanisms such as ubiquitin-mediated degradation by Arkadia. Here we present an ES cell system along with a database containing the expression profile of thousands of genes downstream of Smad2/3 activation patterns, in the presence or absence of protein synthesis. Furthermore, we identify primary target genes that follow proportionately and with high sensitivity changes in Smad2/3 levels over 15-30 hours. The above system and resource provide tools to study morphogen function in development.


Subject(s)
Embryonic Stem Cells/metabolism , Gene Expression Regulation, Developmental , Smad2 Protein/metabolism , Smad3 Protein/metabolism , Activin Receptors, Type I/metabolism , Animals , Benzamides/pharmacology , Dioxoles/pharmacology , Flow Cytometry , Mice , Models, Biological , Nodal Protein/metabolism , Signal Transduction , Transcription, Genetic , Transforming Growth Factor beta/metabolism , Ubiquitin/metabolism , Ubiquitin-Protein Ligases
17.
Cell Oncol ; 31(1): 41-51, 2009.
Article in English | MEDLINE | ID: mdl-19096149

ABSTRACT

BACKGROUND: The TGF-beta signaling repressors SnoN and Ski have been critically implicated in human cancer. METHODS: To explore the role of SnoN and Ski in the development and progression of colorectal cancer we examined their protein expression profile by immunohistochemistry in a series of human colorectal adenomas, carcinomas and lymph node metastases. The mRNA expression of SnoN was also quantified by Real-Time RT-PCR. RESULTS: SnoN and Ski were overexpressed both in adenomas with severe dysplasia and colorectal carcinomas. Protein expression was cytoplasmic and nuclear with predominant cytoplasmic localization. The subcellular localization was related differently to pathologic variables of colorectal carcinomas. Although there was no significant association of protein levels with tumor invasion and metastasis, a significant correlation of nuclear SnoN and Ski with beta-catenin pathway was observed. Moreover, SnoN mRNA did not differ in carcinomas as compared to normal control and there was no correlation between SnoN protein and mRNA levels. CONCLUSION: Our findings suggest that SnoN and Ski exert oncogenic effects in human colorectal carcinogenesis and their overexpression is implicated in early stage disease.


Subject(s)
Colorectal Neoplasms/metabolism , DNA-Binding Proteins/metabolism , Down-Regulation , Intracellular Signaling Peptides and Proteins/metabolism , Proto-Oncogene Proteins/metabolism , Transforming Growth Factor beta/metabolism , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , DNA-Binding Proteins/genetics , Disease Progression , Gene Expression , Humans , Intracellular Signaling Peptides and Proteins/genetics , Proto-Oncogene Proteins/genetics , Signal Transduction , Transforming Growth Factor beta/genetics
18.
Biochem Biophys Res Commun ; 378(3): 498-502, 2009 Jan 16.
Article in English | MEDLINE | ID: mdl-19032943

ABSTRACT

E3 ubiquitin ligases play a key role in the recognition of target proteins and the degradation by 26S proteasomes. Arkadia is the first example of an E3 ubiquitin ligase that positively regulates TGF-beta family signaling. It has been shown to induce ubiquitin-dependent degradation of negative regulators of TGF-beta signaling through its C-terminal RING domain. Structural analysis of Arkadia RING domain is needed to elucidate its enzymatic properties. For such studies efficient production of pure and correctly folded Arkadia protein is required. Here we report the recombinant expression in Escherichia coli and purification of the C-terminal RING domain of Arkadia. NMR analysis of the soluble construct reveals a stable folded protein suitable for high resolution structural studies.


Subject(s)
RING Finger Domains , Recombinant Proteins/biosynthesis , Ubiquitin-Protein Ligases/biosynthesis , Ubiquitin/biosynthesis , Amino Acid Sequence , Animals , Escherichia coli/genetics , Mice , Molecular Sequence Data , Nuclear Magnetic Resonance, Biomolecular , Protein Folding , Recombinant Proteins/chemistry , Recombinant Proteins/isolation & purification , Transforming Growth Factor beta1/metabolism , Ubiquitin/chemistry , Ubiquitin/isolation & purification , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/isolation & purification , Zinc/chemistry
19.
Stem Cells ; 26(1): 3-16, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17932425

ABSTRACT

The delineation of regulatory networks involved in early endocrine pancreas specification will play a crucial role in directing the differentiation of embryonic stem cells toward the mature phenotype of beta cells for cell therapy of type 1 diabetes. The transcription factor Ngn3 is required for the specification of the endocrine lineage, but its direct targets and the scope of biological processes it regulates remain elusive. We show that stepwise differentiation of embryonic stem cells using successive in vivo patterning signals can lead to simultaneous induction of Ptf1a and Pdx1 expression. In this cellular context, Ngn3 induction results in upregulation of its known direct target genes within 12 hours. Microarray gene expression profiling at distinct time points following Ngn3 induction suggested novel and diverse roles of Ngn3 in pancreas endocrine cell specification. Induction of Ngn3 expression results in regulation of the Wnt, integrin, Notch, and transforming growth factor beta signaling pathways and changes in biological processes affecting cell motility, adhesion, the cytoskeleton, the extracellular matrix, and gene expression. Furthermore, the combination of in vivo patterning signals and inducible Ngn3 expression enhances ESC differentiation toward the pancreas endocrine lineage. This is shown by strong upregulation of endocrine lineage terminal differentiation markers and strong expression of the hormones glucagon, somatostatin, and insulin. Importantly, all insulin(+) cells are also C-peptide(+), and glucose-dependent insulin release was 10-fold higher than basal levels. These data suggest that bona fide pancreas endocrine cells have been generated and that timely induction of Ngn3 expression can play a decisive role in directing ESC differentiation toward the endocrine lineage.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Differentiation/physiology , Embryonic Stem Cells/cytology , Islets of Langerhans/embryology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Animals , Cell Lineage , Cells, Cultured , Fluorescent Antibody Technique , Gene Expression Profiling , Gene Expression Regulation , Gene Expression Regulation, Developmental , Genetic Vectors , Homeodomain Proteins/biosynthesis , Islets of Langerhans/cytology , Mice , Oligonucleotide Array Sequence Analysis , Trans-Activators/biosynthesis , Transcription Factors/biosynthesis , Transgenes
20.
Dev Biol ; 310(1): 85-98, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17719572

ABSTRACT

The transcription factor, Sox1 has been implicated in the maintenance of neural progenitor cell status, but accumulating evidence suggests that this is only part of its function. This study examined the role of Sox1 expression in proliferation, lineage commitment, and differentiation by telencephalic neural progenitor cells in vitro and in vivo, and further clarified the pattern of Sox1 expression in postnatal and adult mouse brain. Telencephalic neural progenitor cells isolated from Sox1 null embryos formed neurospheres normally, but were specifically deficient in neuronal differentiation. Conversely, overexpression of Sox1 in the embryonic telencephalon in vivo both expanded the progenitor pool and biased neural progenitor cells towards neuronal lineage commitment. Sox1 mRNA and protein were found to be persistently expressed in the postnatal and adult brain in both differentiated and neurogenic regions. Importantly, in differentiated regions Sox1 co-labeled only with neuronal markers. These observations, coupled with previous studies, suggest that Sox1 expression by early embryonic progenitor cells initially helps to maintain the cells in cell cycle, but that continued expression subsequently promotes neuronal lineage commitment.


Subject(s)
DNA-Binding Proteins/physiology , Gene Expression Regulation, Developmental , High Mobility Group Proteins/physiology , Stem Cells/physiology , Telencephalon/cytology , Animals , Cell Cycle/genetics , Cell Differentiation , Cell Lineage/genetics , Mice , Nerve Tissue Proteins/genetics , SOXB1 Transcription Factors , Stem Cells/cytology , Telencephalon/growth & development
SELECTION OF CITATIONS
SEARCH DETAIL
...