Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Nat Commun ; 12(1): 7047, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857764

ABSTRACT

Eph receptor tyrosine kinases play a key role in cell-cell communication. Lack of structural information on the entire multi-domain intracellular region of any Eph receptor has hindered understanding of their signaling mechanisms. Here, we use integrative structural biology to investigate the structure and dynamics of the EphA2 intracellular region. EphA2 promotes cancer malignancy through a poorly understood non-canonical form of signaling involving serine/threonine phosphorylation of the linker connecting its kinase and SAM domains. We show that accumulation of multiple linker negative charges, mimicking phosphorylation, induces cooperative changes in the EphA2 intracellular region from more closed to more extended conformations and perturbs the EphA2 juxtamembrane segment and kinase domain. In cells, linker negative charges promote EphA2 oligomerization. We also identify multiple kinases catalyzing linker phosphorylation. Our findings suggest multiple effects of linker phosphorylation on EphA2 signaling and imply that coordination of different kinases is necessary to promote EphA2 non-canonical signaling.


Subject(s)
Receptor, EphA2/chemistry , Serine/chemistry , Sterile Alpha Motif/genetics , Threonine/chemistry , A549 Cells , Amino Acid Sequence , Binding Sites , Cell Line, Tumor , Crystallography, X-Ray , Gene Expression , HEK293 Cells , Humans , Models, Molecular , Molecular Mimicry , PC-3 Cells , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Receptor, EphA2/genetics , Receptor, EphA2/metabolism , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Sequence Alignment , Sequence Homology, Amino Acid , Serine/genetics , Serine/metabolism , Static Electricity , Substrate Specificity , Threonine/genetics , Threonine/metabolism
2.
Biochem J ; 478(17): 3351-3371, 2021 09 17.
Article in English | MEDLINE | ID: mdl-34431498

ABSTRACT

EphB6 and EphA10 are two poorly characterised pseudokinase members of the Eph receptor family, which collectively serves as mediators of contact-dependent cell-cell communication to transmit extracellular cues into intracellular signals. As per their active counterparts, EphB6 and EphA10 deregulation is strongly linked to proliferative diseases. However, unlike active Eph receptors, whose catalytic activities are thought to initiate an intracellular signalling cascade, EphB6 and EphA10 are classified as catalytically dead, raising the question of how non-catalytic functions contribute to Eph receptor signalling homeostasis. In this study, we have characterised the biochemical properties and topology of the EphB6 and EphA10 intracellular regions comprising the juxtamembrane (JM) region, pseudokinase and SAM domains. Using small-angle X-ray scattering and cross-linking-mass spectrometry, we observed high flexibility within their intracellular regions in solution and a propensity for interaction between the component domains. We identified tyrosine residues in the JM region of EphB6 as EphB4 substrates, which can bind the SH2 domains of signalling effectors, including Abl, Src and Vav3, consistent with cellular roles in recruiting these proteins for downstream signalling. Furthermore, our finding that EphB6 and EphA10 can bind ATP and ATP-competitive small molecules raises the prospect that these pseudokinase domains could be pharmacologically targeted to counter oncogenic signalling.


Subject(s)
Receptors, Eph Family/chemistry , Receptors, Eph Family/metabolism , Signal Transduction/genetics , Sterile Alpha Motif/genetics , src Homology Domains/genetics , Adenosine Triphosphate/metabolism , Animals , Humans , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Kinase Inhibitors/metabolism , Receptors, Eph Family/genetics , Recombinant Proteins/metabolism , Sf9 Cells , Spodoptera/cytology , Tyrosine/metabolism
3.
Nat Commun ; 11(1): 5609, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33154383

ABSTRACT

Polycomb Group (PcG) proteins organize chromatin at multiple scales to regulate gene expression. A conserved Sterile Alpha Motif (SAM) in the Polycomb Repressive Complex 1 (PRC1) subunit Polyhomeotic (Ph) has been shown to play an important role in chromatin compaction and large-scale chromatin organization. Ph SAM forms helical head to tail polymers, and SAM-SAM interactions between chromatin-bound Ph/PRC1 are believed to compact chromatin and mediate long-range interactions. To understand the underlying mechanism, here we analyze the effects of Ph SAM on chromatin in vitro. We find that incubation of chromatin or DNA with a truncated Ph protein containing the SAM results in formation of concentrated, phase-separated condensates. Ph SAM-dependent condensates can recruit PRC1 from extracts and enhance PRC1 ubiquitin ligase activity towards histone H2A. We show that overexpression of Ph with an intact SAM increases ubiquitylated H2A in cells. Thus, SAM-induced phase separation, in the context of Ph, can mediate large-scale compaction of chromatin into biochemical compartments that facilitate histone modification.


Subject(s)
DNA-Binding Proteins/chemistry , Drosophila Proteins/chemistry , Polycomb Repressive Complex 1/chemistry , Polycomb-Group Proteins/metabolism , Sterile Alpha Motif/physiology , Animals , Cell Compartmentation , Cell Line , Cell Nucleus/metabolism , Chromatin/metabolism , DNA/metabolism , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drosophila , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Histones/metabolism , Polycomb Repressive Complex 1/genetics , Polycomb Repressive Complex 1/metabolism , Polymerization , Sterile Alpha Motif/genetics , Ubiquitination
4.
Curr Genet ; 66(6): 1101-1115, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32656574

ABSTRACT

Boi1 and Boi2 are paralogous proteins essential for bud formation in budding yeast. So far, the domains that target Boi1/Boi2 to the polarity sites and function in bud formation are not well understood. Here, we report that a coiled-coil domain of Boi2 cooperates with the adjacent PH domain to confer Boi2's bud-cortex localization and major function in cell growth. The PH domain portion of the PH-CC bi-domain interacts with the Rho GTPases Cdc42 and Rho3 and both interactions are independent of the GTP/GDP-bound state of each GTPase. Interestingly, high-copy RHO3 and BOI2 but not CDC42 suppressed the growth defect of RGA1-C538 overexpression and the sec15-1 mutant and this BOI2 function depends on RHO3, suggesting that Boi2 may function in the Rho3 pathway. The SAM domain of Boi2 plays an essential role in high-copy suppression of the two mutants as well as in the early bud-neck localization of Boi2. The SAM domain and the CC domain also interact homotypically. They are likely involved in the formation of Boi2-containing protein complex. Our results provide new insights in the localization and function of Boi2 and highlight the importance of the PH-CC bi-domain and the SAM domain in Boi2's localization and function.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Cell Polarity/genetics , Saccharomyces cerevisiae Proteins/genetics , rho GTP-Binding Proteins/genetics , Amino Acid Sequence/genetics , Phenotype , Protein Binding/genetics , Saccharomyces cerevisiae/genetics , Sterile Alpha Motif/genetics
5.
J Biol Chem ; 294(40): 14512-14525, 2019 10 04.
Article in English | MEDLINE | ID: mdl-31337708

ABSTRACT

The human microbiota plays a central role in human physiology. This complex ecosystem is a promising but untapped source of bioactive compounds and antibiotics that are critical for its homeostasis. However, we still have a very limited knowledge of its metabolic and biosynthetic capabilities. Here we investigated an enigmatic biosynthetic gene cluster identified previously in the human gut symbiont Ruminococcus gnavus This gene cluster which encodes notably for peptide precursors and putative radical SAM enzymes, has been proposed to be responsible for the biosynthesis of ruminococcin C (RumC), a ribosomally synthesized and posttranslationally modified peptide (RiPP) with potent activity against the human pathogen Clostridium perfringens By combining in vivo and in vitro approaches, including recombinant expression and purification of the respective peptides and proteins, enzymatic assays, and LC-MS analyses, we determined that RumC is a sulfur-to-α-carbon thioether-containing peptide (sactipeptide) with an unusual architecture. Moreover, our results support that formation of the thioether bridges follows a processive order, providing mechanistic insights into how radical SAM (AdoMet) enzymes install posttranslational modifications in RiPPs. We also found that the presence of thioether bridges and removal of the leader peptide are required for RumC's antimicrobial activity. In summary, our findings provide evidence that production of the anti-Clostridium peptide RumC depends on an R. gnavus operon encoding five potential RumC precursor peptides and two radical SAM enzymes, uncover key RumC structural features, and delineate the sequence of posttranslational modifications leading to its formation and antimicrobial activity.


Subject(s)
Bacteriocins/chemistry , Clostridiales/genetics , Clostridium perfringens/genetics , Gastrointestinal Microbiome/genetics , Peptides/genetics , Amino Acid Sequence/genetics , Bacteriocins/biosynthesis , Bacteriocins/genetics , Clostridiales/enzymology , Clostridium perfringens/chemistry , Clostridium perfringens/pathogenicity , Humans , Multigene Family/genetics , Peptide Biosynthesis/genetics , Peptides/chemistry , Protein Processing, Post-Translational/genetics , Ribosomes/genetics , Sterile Alpha Motif/genetics , Sulfides/chemistry , Symbiosis/genetics
6.
Mol Genet Genomic Med ; 7(3): e00560, 2019 03.
Article in English | MEDLINE | ID: mdl-30632316

ABSTRACT

BACKGROUND: We describe a patient presenting with pachygyria, epilepsy, developmental delay, short stature, failure to thrive, facial dysmorphisms, and multiple osteochondromas. METHODS: The patient underwent extensive genetic testing and analysis in an attempt to diagnose the cause of his condition. Clinical testing included metaphase karyotyping, array comparative genomic hybridization, direct sequencing and multiplex ligation-dependent probe amplification and trio-based exome sequencing. Subsequently, research-based whole transcriptome sequencing was conducted to determine whether it might shed light on the undiagnosed phenotype. RESULTS: Clinical exome sequencing of patient and parent samples revealed a maternally inherited splice-site variant in the doublecortin (DCX) gene that was classified as likely pathogenic and diagnostic of the patient's neurological phenotype. Clinical array comparative genome hybridization analysis revealed a 16p13.3 deletion that could not be linked to the patient phenotype based on affected genes. Further clinical testing to determine the cause of the patient's multiple osteochondromas was unrevealing despite extensive profiling of the most likely causative genes, EXT1 and EXT2, including mutation screening by direct sequence analysis and multiplex ligation-dependent probe amplification. Whole transcriptome sequencing identified a SAMD12-EXT1 fusion transcript that could have resulted from a chromosomal deletion, leading to the loss of EXT1 function. Re-review of the clinical array comparative genomic hybridization results indicated a possible unreported mosaic deletion affecting the SAMD12 and EXT1 genes that corresponded precisely to the introns predicted to be affected by a fusion-causing deletion. The existence of the mosaic deletion was subsequently confirmed clinically by an increased density copy number array and orthogonal methodologies CONCLUSIONS: While mosaic mutations and deletions of EXT1 and EXT2 have been reported in the context of multiple osteochondromas, to our knowledge, this is the first time that transcriptomics technologies have been used to diagnose a patient via fusion transcript analysis in the congenital disease setting.


Subject(s)
Exostoses, Multiple Hereditary/genetics , Gene Fusion , N-Acetylglucosaminyltransferases/genetics , Nerve Tissue Proteins/genetics , Child , Exostoses, Multiple Hereditary/pathology , Gene Deletion , Humans , Male , RNA, Messenger/genetics , Sterile Alpha Motif/genetics
7.
Nat Genet ; 50(4): 581-590, 2018 04.
Article in English | MEDLINE | ID: mdl-29507423

ABSTRACT

Epilepsy is a common neurological disorder, and mutations in genes encoding ion channels or neurotransmitter receptors are frequent causes of monogenic forms of epilepsy. Here we show that abnormal expansions of TTTCA and TTTTA repeats in intron 4 of SAMD12 cause benign adult familial myoclonic epilepsy (BAFME). Single-molecule, real-time sequencing of BAC clones and nanopore sequencing of genomic DNA identified two repeat configurations in SAMD12. Intriguingly, in two families with a clinical diagnosis of BAFME in which no repeat expansions in SAMD12 were observed, we identified similar expansions of TTTCA and TTTTA repeats in introns of TNRC6A and RAPGEF2, indicating that expansions of the same repeat motifs are involved in the pathogenesis of BAFME regardless of the genes in which the expanded repeats are located. This discovery that expansions of noncoding repeats lead to neuronal dysfunction responsible for myoclonic tremor and epilepsy extends the understanding of diseases with such repeat expansion.


Subject(s)
DNA Repeat Expansion , Epilepsies, Myoclonic/genetics , Microsatellite Repeats , Nerve Tissue Proteins/genetics , Sterile Alpha Motif/genetics , Adult , Age of Onset , Autoantigens/genetics , Base Sequence , Epilepsies, Myoclonic/etiology , Epilepsies, Myoclonic/pathology , Female , Genomic Instability , Guanine Nucleotide Exchange Factors/genetics , Humans , Introns , Male , Pedigree , Purkinje Cells/pathology , RNA-Binding Proteins/genetics , Sequence Analysis, DNA
8.
Arch Biochem Biophys ; 619: 1-9, 2017 04 01.
Article in English | MEDLINE | ID: mdl-28235466

ABSTRACT

The alpha splice variant of p73 (p73α), a homologue of the tumour suppressor p53, has at its C terminus a sterile alpha motif (SAM); this domain, SAMp73, is involved in lipid binding and it is thought to mediate in protein-protein interactions. As SAMp73 is a 68-residue-long helical bundle, it could be a good model to study the (2,2,2-trifluoroethanol) TFE-induced conformational transitions of α-helical proteins. Furthermore, as SAMp73 binds to lipids through a well-known polypeptide patch, we can test whether TFE is a good mimic of lipids and membranes. To address those questions, we used several biophysical probes, namely, fluorescence, circular dichroism, 1D, 2D and 3D-NMR spectroscopies, and dynamic light scattering. The TFE-induced conformational transition of SAMp73 was complex, involving several species as detected by the biophysical probes. The last TFE-induced transition occurred at a concentration of TFE of ∼20% (v/v), where the protein lost its compactness. None of those TFE-induced species accumulated during the two-state folding of SAMp73 in aqueous solution. The final state at 40% TFE was highly helical, but its structure was not rigid. For SAMp73, TFE did not properly mimic a membrane-like environment, since at very low TFE concentrations, other residues, together with those known to interact with lipids, were also affected by the co-solvent. Comparison with studies on isolated peptides, comprising the helical regions of SAMp73, suggests that peptides were good models of the intact protein in TFE.


Subject(s)
Sterile Alpha Motif/genetics , Trifluoroethanol/chemistry , Tumor Protein p73/chemistry , Cell Membrane/metabolism , Circular Dichroism , Humans , Light , Magnetic Resonance Spectroscopy , Peptides/chemistry , Protein Binding , Protein Domains , Protein Folding , Protein Interaction Mapping , Protein Structure, Secondary , Scattering, Radiation , Solvents/chemistry , Spectrometry, Fluorescence
9.
Oncotarget ; 7(37): 60535-60554, 2016 Sep 13.
Article in English | MEDLINE | ID: mdl-27528034

ABSTRACT

The aberrant epigenetic silencing of tumor suppressor genes (TSGs) plays a major role during carcinogenesis and regaining these dormant functions by engineering of sequence-specific epigenome editing tools offers a unique opportunity for targeted therapies. However, effectively normalizing the expression and regaining tumor suppressive functions of silenced TSGs by artificial transcription factors (ATFs) still remains a major challenge. Herein we describe novel combinatorial strategies for the potent reactivation of two class II TSGs, MASPIN and REPRIMO, in cell lines with varying epigenetic states, using the CRISPR/dCas9 associated system linked to a panel of effector domains (VP64, p300, VPR and SAM complex), as well as with protein-based ATFs, Zinc Fingers and TALEs. We found that co-delivery of multiple effector domains using a combination of CRISPR/dCas9 and TALEs or SAM complex maximized activation in highly methylated promoters. In particular, CRISPR/dCas9 VPR with SAM upregulated MASPIN mRNA (22,145-fold change) in H157 lung cancer cells, with accompanying re-expression of MASPIN protein, which led to a concomitant inhibition of cell proliferation and induction of apoptotic cell death. Consistently, CRISPR/dCas9 VP64 with SAM upregulated REPRIMO (680-fold change), which led to phenotypic reprogramming in AGS gastric cancer cells. Altogether, our results outlined novel sequence-specific, combinatorial epigenome editing approaches to reactivate highly methylated TSGs as a promising therapy for cancer and other diseases.


Subject(s)
CRISPR-Cas Systems/genetics , Homeodomain Proteins/genetics , Lung Neoplasms/metabolism , RNA, Long Noncoding/genetics , Repressor Proteins/genetics , Sterile Alpha Motif/genetics , Stomach Neoplasms/metabolism , Zinc Fingers/genetics , Apoptosis , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Proliferation , DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Glycoproteins/genetics , Glycoproteins/metabolism , Humans , Lung Neoplasms/genetics , Serpins/genetics , Serpins/metabolism , Stomach Neoplasms/genetics , Transcriptional Activation , Tumor Suppressor Proteins/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL