Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Int J Biol Macromol ; 262(Pt 1): 129926, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38331062

ABSTRACT

The emergence of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) posed a threat to public health and the global economy, necessitating the development of various vaccination strategies. Mutations in the SPIKE protein gene, a crucial component of mRNA and adenovirus-based vaccines, raised concerns about vaccine efficacy, prompting the need for rapid vaccine updates. To address this, we leveraged PeptiCRAd, an oncolytic vaccine based on tumor antigen decorated oncolytic adenoviruses, creating a vaccine platform called PeptiVAX. First, we identified multiple CD8 T-cell epitopes from highly conserved regions across coronaviruses, expanding the range of T-cell responses to non-SPIKE proteins. We designed short segments containing the predicted epitopes presented by common HLA-Is in the global population. Testing the immunogenicity, we characterized T-cell responses to candidate peptides in peripheral blood mononuclear cells (PBMCs) from pre-pandemic healthy donors and ICU patients. As a proof of concept in mice, we selected a peptide with epitopes predicted to bind to murine MHC-I haplotypes. Our technology successfully elicited peptide-specific T-cell responses, unaffected by the use of unarmed adenoviral vectors or adeno-based vaccines encoding SPIKE. In conclusion, PeptiVAX represents a fast and adaptable SARS-CoV-2 vaccine delivery system that broadens T-cell responses beyond the SPIKE protein, offering potential benefits for vaccine effectiveness.


Subject(s)
COVID-19 , Viral Vaccines , Humans , Mice , Animals , COVID-19 Vaccines , COVID-19/prevention & control , Spike Glycoprotein, Coronavirus/genetics , Leukocytes, Mononuclear , SARS-CoV-2 , Peptides/chemistry , Epitopes, T-Lymphocyte
2.
Comput Struct Biotechnol J ; 20: 4870-4884, 2022.
Article in English | MEDLINE | ID: mdl-36147664

ABSTRACT

Transcriptome level expression data connected to the spatial organization of the cells and molecules would allow a comprehensive understanding of how gene expression is connected to the structure and function in the biological systems. The spatial transcriptomics platforms may soon provide such information. However, the current platforms still lack spatial resolution, capture only a fraction of the transcriptome heterogeneity, or lack the throughput for large scale studies. The strengths and weaknesses in current ST platforms and computational solutions need to be taken into account when planning spatial transcriptomics studies. The basis of the computational ST analysis is the solutions developed for single-cell RNA-sequencing data, with advancements taking into account the spatial connectedness of the transcriptomes. The scRNA-seq tools are modified for spatial transcriptomics or new solutions like deep learning-based joint analysis of expression, spatial, and image data are developed to extract biological information in the spatially resolved transcriptomes. The computational ST analysis can reveal remarkable biological insights into spatial patterns of gene expression, cell signaling, and cell type variations in connection with cell type-specific signaling and organization in complex tissues. This review covers the topics that help choosing the platform and computational solutions for spatial transcriptomics research. We focus on the currently available ST methods and platforms and their strengths and limitations. Of the computational solutions, we provide an overview of the analysis steps and tools used in the ST data analysis. The compatibility with the data types and the tools provided by the current ST analysis frameworks are summarized.

3.
Biotechnol J ; 17(6): e2100213, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35174641

ABSTRACT

BACKGROUND AND AIM: Single-cell RNA sequencing (scRNA-seq) is a powerful method utilising transcriptomic data for detailed characterisation of heterogeneous cell populations. The use of oligonucleotide-labelled antibodies for targeted proteomics addresses the shortcomings of the scRNA-seq-only based approach by improving detection of low expressing targets. However, optimisation of large antibody panels is challenging and depends on the availability of co-functioning oligonucleotide-labelled antibodies. MAIN METHODS AND RESULTS: We present here a simple adjustable oligonucleotide-antibody conjugation method which enables a desired level of oligo-conjugation per antibody. The mean labelling in the produced antibody batches varied from 1 to 6 oligos per antibody. In the scRNA-seq multimodal experiment, the highest sensitivity was seen with moderate antibody labelling as the high activation and/or labelling was detrimental to antibody performance. The conjugates were also tested for compatibility with the fixation and freeze storage protocols. The oligo-antibody signal was stable in fixed cells indicating the feasibility of a stain, fix, store, and analyse later type of workflow for multimodal scRNA-seq. CONCLUSIONS AND IMPLICATIONS: Optimised oligo-labelling will improve detection of weak protein targets in scRNA-seq multimodal experiments and reduce sequencing costs due to a more balanced amplification of different antibody signals in CITE-seq libraries. Furthermore, the use of a pre-stain, fix, run later protocol will allow for flexibility, facilitate sample pooling, and ease logistics in scRNA-seq multimodal experiments.


Subject(s)
Single-Cell Analysis , Transcriptome , Antibodies/genetics , Gene Expression Profiling , Oligonucleotides/genetics , Proteomics , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods
4.
Clin Immunol ; 231: 108851, 2021 10.
Article in English | MEDLINE | ID: mdl-34508889

ABSTRACT

Almost all patients with autoimmune polyendocrine syndrome type 1 (APS-1) have neutralizing antibodies against type 1 interferons (IFN), important mediators of antiviral defense. Recently, neutralizing anti-IFN antibodies were shown to be a risk factor of severe COVID-19. Here we show in a cohort of 44 patients with APS-1 that higher titers of neutralizing anti-IFNα4 antibodies are associated with a higher and earlier incidence of VZV reactivation (herpes zoster). The patients also present with uncommonly severe clinical sequelae of herpetic infections. APS-1 patients had decreased humoral immune responses to varicella zoster virus, but cellular responses were comparable to healthy controls. These results suggest that blocking the type I interferon pathway in patients with APS-1 patients leads to a clinically significant immune deficiency, and susceptibility to herpesviruses should be taken into account when treating patients with APS-1.


Subject(s)
Herpesvirus 3, Human , Polyendocrinopathies, Autoimmune/complications , Varicella Zoster Virus Infection/complications , Adolescent , Adult , Aged , Child , Cohort Studies , Female , Humans , Immunity, Cellular , Interferon-alpha/immunology , Male , Middle Aged , Polyendocrinopathies, Autoimmune/immunology , Risk Factors , Varicella Zoster Virus Infection/pathology , Young Adult
5.
Data Brief ; 35: 106751, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33553521

ABSTRACT

T cell receptor (TCR) is a heterodimer consisting of TCRα and TCRß chains that are generated by somatic recombination of multiple gene segments. Nascent TCR repertoire undergoes thymic selections where non-functional and potentially autoreactive receptors are removed. During the last years, the development of high-throughput sequencing technology has allowed a large scale assessment of TCR repertoire and multiple analysis tools are now also available. In our recent manuscript, Human thymic T cell repertoire is imprinted with strong convergence to shared sequences[1], we show highly overlapping thymic TCR repertoires in unrelated individuals. In the current Data in Brief article, we provide a more detailed characterization of the basic features of these thymic and related peripheral blood TCR repertoires. The thymus samples were collected from eight infants undergoing corrective cardiac surgery, two of whom were monozygous twins [2]. In parallel with the surgery, a small aliquot of peripheral blood was drawn from four of the donors. Genomic DNA was extracted from mechanically released thymocytes and circulating leukocytes. The sequencing of TCRα and TCRß repertoires was performed at ImmunoSEQ platform (Adaptive Biotechnologies). The obtained repertoire data were analysed applying relevant features from immunoSEQ® 3.0 Analyzer (Adaptive Biotechnologies) and a freely available VDJTools software package for programming language R [3]. The current data analysis displays the basic features of the sequenced repertoires including observed TCR diversity, various descriptive TCR diversity measures, and V and J gene usage. In addition, multiple methods to calculate repertoire overlap between two individuals are applied. The raw sequence data provide a large database of reference TCRs in healthy individuals at an early developmental stage. The data can be exploited to improve existing computational models on TCR repertoire behaviour as well as in the generation of new models.

6.
Mol Immunol ; 127: 112-123, 2020 11.
Article in English | MEDLINE | ID: mdl-32961421

ABSTRACT

A highly diverse repertoire of T cell antigen receptors (TCR) is created in the thymus by recombination of gene segments and the insertion or deletion of nucleotides at the junctions. Using next-generation TCR sequencing we define here the features of recombination and selection in the human TCRα and TCRß locus, and show that a strikingly high proportion of the repertoire is shared by unrelated individuals. The thymic TCRα nucleotide repertoire was more diverse than TCRß, with 4.1 × 106 vs. 0.81 × 106 unique clonotypes, and contained nonproductive clonotypes at a higher frequency (69.2% vs. 21.2%). The convergence of distinct nucleotide clonotypes to the same amino acid sequences was higher in TCRα than in TCRß repertoire (1.45 vs. 1.06 nucleotide sequences per amino acid sequence in thymus). The gene segment usage was biased, and generally all individuals favored the same genes in both TCRα and TCRß loci. Despite the high diversity, a large fraction of the repertoire was found in more than one donor. The shared fraction was bigger in TCRα than TCRß repertoire, and more common in in-frame sequences than in nonproductive sequences. Thus, both biases in rearrangement and thymic selection are likely to contribute to the generation of shared repertoire in humans.


Subject(s)
Genomic Imprinting , T-Lymphocytes/immunology , Thymus Gland/cytology , Base Sequence , Clone Cells , Complementarity Determining Regions/genetics , Female , Genetic Variation , Humans , Infant , Infant, Newborn , Male , Mutagenesis, Insertional , Probability , Receptors, Antigen, T-Cell, alpha-beta/genetics , Recombination, Genetic/genetics
7.
Methods Mol Biol ; 2184: 31-45, 2020.
Article in English | MEDLINE | ID: mdl-32808216

ABSTRACT

Transcriptome analysis at a single-cell level with single-cell RNA sequencing (scRNA-seq) is a powerful method for detailed characterization of heterogeneous cell populations. Recent developments have enabled parallel analysis of both transcript and protein levels by using antibodies conjugated to barcoded oligonucleotides. These antibodies enable protein levels to be converted into nucleotide format, allowing the sequencing-based detection of both modalities at single-cell level. Here we present a simple and reliable method for conjugation of oligonucleotides with antibodies and a protocol for their use in single-cell transcriptome sequencing.


Subject(s)
Antibodies/genetics , Gene Expression Profiling/methods , Membrane Proteins/genetics , Single-Cell Analysis/methods , Transcriptome/genetics , Cells, Cultured , Humans , Leukocytes, Mononuclear/metabolism , Oligonucleotides/genetics , Sequence Analysis, RNA/methods
8.
Front Immunol ; 11: 2020, 2020.
Article in English | MEDLINE | ID: mdl-32849667

ABSTRACT

Background: Live viral vaccines are generally contraindicated in patients with combined immunodeficiency including cartilage-hair hypoplasia (CHH); however, they may be tolerated in milder syndromes. We evaluated the safety and efficacy of live viral vaccines in patients with CHH. Methods: We analyzed hospital and immunization records of 104 patients with CHH and measured serum antibodies to measles, mumps, rubella, and varicella zoster virus (VZV) in all patients who agreed to blood sampling (n = 50). We conducted a clinical trial (ClinicalTrials.gov identifier: NCT02383797) of live VZV vaccine on five subjects with CHH who lacked varicella history, had no clinical symptoms of immunodeficiency, and were seronegative for VZV; humoral and cellular immunologic responses were assessed post-immunization. Results: A large proportion of patients have been immunized with live viral vaccines, including measles-mumps-rubella (MMR) (n = 40, 38%) and VZV (n = 10, 10%) vaccines, with no serious adverse events. Of the 50 patients tested for antibodies, previous immunization has been documented with MMR (n = 22), rubella (n = 2) and measles (n = 1) vaccines. Patients with CHH demonstrated seropositivity rates of 96%/75%/91% to measles, mumps and rubella, respectively, measured at a medium of 24 years post-immunization. Clinical trial participants developed humoral and cellular responses to VZV vaccine. One trial participant developed post-immunization rash and knee swelling, both resolved without treatment. Conclusion: No serious adverse events have been recorded after immunization with live viral vaccines in Finnish patients with CHH. Patients generate humoral and cellular immune response to live viral vaccines. Immunization with live vaccines may be considered in selected CHH patients with no or clinically mild immunodeficiency.


Subject(s)
Hair/abnormalities , Herpesvirus 3, Human/immunology , Hirschsprung Disease/immunology , Immunologic Deficiency Syndromes/immunology , Measles-Mumps-Rubella Vaccine/immunology , Osteochondrodysplasias/congenital , Primary Immunodeficiency Diseases/immunology , Viral Vaccines/immunology , Antibodies, Viral/blood , Cells, Cultured , Cohort Studies , Enzyme-Linked Immunospot Assay , Hair/immunology , Hirschsprung Disease/genetics , Humans , Immunity, Cellular , Immunity, Humoral , Immunologic Deficiency Syndromes/genetics , Interferon-gamma/metabolism , Osteochondrodysplasias/genetics , Osteochondrodysplasias/immunology , Primary Immunodeficiency Diseases/genetics , RNA, Long Noncoding/genetics , Vaccination
9.
Mol Cell Proteomics ; 15(10): 3270-3281, 2016 10.
Article in English | MEDLINE | ID: mdl-27440912

ABSTRACT

The Src Homology-3 (SH3) domains are ubiquitous protein modules that mediate important intracellular protein interactions via binding to short proline-rich consensus motifs in their target proteins. The affinity and specificity of such core SH3 - ligand contacts are typically modest, but additional binding interfaces can give rise to stronger and more specific SH3-mediated interactions. To understand how commonly such robust SH3 interactions occur in the human protein interactome, and to identify these in an unbiased manner we have expressed 324 predicted human SH3 ligands as full-length proteins in mammalian cells, and screened for their preferred SH3 partners using a phage display-based approach. This discovery platform contains an essentially complete repertoire of the ∼300 human SH3 domains, and involves an inherent binding threshold that ensures selective identification of only SH3 interactions with relatively high affinity. Such strong and selective SH3 partners could be identified for only 19 of these 324 predicted ligand proteins, suggesting that the majority of human SH3 interactions are relatively weak, and thereby have capacity for only modest inherent selectivity. The panel of exceptionally robust SH3 interactions identified here provides a rich source of leads and hypotheses for further studies. However, a truly comprehensive characterization of the human SH3 interactome will require novel high-throughput methods based on function instead of absolute binding affinity.


Subject(s)
Proteome/analysis , src-Family Kinases/chemistry , src-Family Kinases/metabolism , Amino Acid Sequence , Binding Sites , Humans , Ligands , Peptide Library , Protein Binding , Protein Interaction Maps , Proteome/chemistry , src Homology Domains
10.
PLoS One ; 10(3): e0121301, 2015.
Article in English | MEDLINE | ID: mdl-25825872

ABSTRACT

A disintegrin and metalloproteinases (ADAMs) constitute a protein family essential for extracellular signaling and regulation of cell adhesion. Catalytic activity of ADAMs and their predicted potential for Src-homology 3 (SH3) domain binding show a strong correlation. Here we present a comprehensive characterization of SH3 binding capacity and preferences of the catalytically active ADAMs 8, 9, 10, 12, 15, 17, and 19. Our results revealed several novel interactions, and also confirmed many previously reported ones. Many of the identified SH3 interaction partners were shared by several ADAMs, whereas some were ADAM-specific. Most of the ADAM-interacting SH3 proteins were adapter proteins or kinases, typically associated with sorting and endocytosis. Novel SH3 interactions revealed in this study include TOCA1 and CIP4 as preferred partners of ADAM8, and RIMBP1 as a partner of ADAM19. Our results suggest that common as well as distinct mechanisms are involved in regulation and execution of ADAM signaling, and provide a useful framework for addressing the pathways that connect ADAMs to normal and aberrant cell behavior.


Subject(s)
ADAM Proteins/metabolism , src Homology Domains , Carrier Proteins/metabolism , HEK293 Cells , Humans , Protein Binding , Sorting Nexins/metabolism
11.
J Cell Sci ; 128(6): 1139-49, 2015 Mar 15.
Article in English | MEDLINE | ID: mdl-25616895

ABSTRACT

Cranial neural crest (CNC) cells are a transient population of stem cells that originate at the border of the neural plate and the epidermis, and migrate ventrally to contribute to most of the facial structures including bones, cartilage, muscles and ganglia. ADAM13 is a cell surface metalloprotease that is essential for CNC cell migration. Here, we show in Xenopus laevis embryos that the Wnt receptor Fz4 binds to the cysteine-rich domain of ADAM13 and negatively regulates its proteolytic activity in vivo. Gain of Fz4 function inhibits CNC cell migration and can be rescued by gain of ADAM13 function. Loss of Fz4 function also inhibits CNC cell migration and induces a reduction of mature ADAM13, together with an increase in the ADAM13 cytoplasmic fragment that is known to translocate into the nucleus to regulate gene expression. We propose that Fz4 associates with ADAM13 during its transport to the plasma membrane to regulate its proteolytic activity.


Subject(s)
ADAM Proteins/metabolism , Embryo, Nonmammalian/metabolism , Frizzled Receptors/metabolism , Gene Expression Regulation, Developmental , Membrane Proteins/metabolism , Neural Crest/metabolism , Wnt Proteins/metabolism , Xenopus Proteins/metabolism , ADAM Proteins/genetics , Animals , COS Cells , Cell Membrane/metabolism , Cell Movement , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Chlorocebus aethiops , Embryo, Nonmammalian/cytology , Fluorescent Antibody Technique , Frizzled Receptors/genetics , HEK293 Cells , Humans , Immunoprecipitation , In Situ Hybridization , Membrane Proteins/genetics , Neural Crest/cytology , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Wnt Proteins/genetics , Xenopus Proteins/genetics , Xenopus laevis/genetics , Xenopus laevis/growth & development , Xenopus laevis/metabolism
12.
Sci Signal ; 6(290): ra76, 2013 Aug 27.
Article in English | MEDLINE | ID: mdl-23982207

ABSTRACT

Itk (interleukin-2-inducible T cell kinase) and Btk (Bruton's tyrosine kinase) are nonreceptor tyrosine kinases of the Tec family that signal downstream of the T cell receptor (TCR) and B cell receptor (BCR), respectively. Despite their high sequence similarity and related signaling roles, Btk is a substantially more active kinase than Itk. We showed that substitution of 6 of the 619 amino acid residues of Itk with the corresponding residues of Btk (and vice versa) was sufficient to completely switch the activities of Itk and Btk. The substitutions responsible for the swap in activity are all localized to the activation segment of the kinase domain. Nuclear magnetic resonance and hydrogen-deuterium exchange mass spectrometry analyses revealed that Itk and Btk had distinct protein dynamics in this region, which could explain the differences in catalytic efficiency between these kinases. Introducing Itk with enhanced activity into T cells led to enhanced and prolonged TCR signaling compared to that in cells with wild-type Itk. These findings imply that evolutionary pressures have led to Tec kinases having distinct enzymatic properties, depending on the cellular context. We suggest that the weaker catalytic activities of T cell-specific kinases serve to regulate cellular activation and prevent aberrant immune responses.


Subject(s)
Protein-Tyrosine Kinases/metabolism , Receptors, Antigen, B-Cell/metabolism , Receptors, Antigen, T-Cell/metabolism , Signal Transduction , Agammaglobulinaemia Tyrosine Kinase , Amino Acid Sequence , Animals , Binding Sites/genetics , Biocatalysis , Blotting, Western , Deuterium Exchange Measurement , Kinetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Molecular , Molecular Sequence Data , Mutation , Phosphorylation , Protein Structure, Tertiary , Protein-Tyrosine Kinases/chemistry , Protein-Tyrosine Kinases/genetics , Sequence Homology, Amino Acid , Sf9 Cells , T-Lymphocytes/metabolism , Tyrosine/genetics , Tyrosine/metabolism
13.
J Cell Sci ; 123(Pt 1): 118-27, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-20016072

ABSTRACT

Dystroglycan is a ubiquitously expressed cell adhesion protein. Its principal role has been determined as a component of the dystrophin-glycoprotein complex of muscle, where it constitutes a key component of the costameric cell adhesion system. To investigate more fundamental aspects of dystroglycan function in cell adhesion, we examined the role of dystroglycan in the dynamics and assembly of cellular adhesions in myoblasts. We show that beta-dystroglycan is recruited to adhesion structures and, based on staining for vinculin, that overexpression or depletion of dystroglycan affects both size and number of fibrillar adhesions. Knockdown of dystroglycan increases the size and number of adhesions, whereas overexpression decreases the number of adhesions. Dystroglycan knockdown or overexpression affects the ability of cells to adhere to different substrates, and has effects on cell migration that are consistent with effects on the formation of fibrillar adhesions. Using an SH3 domain proteomic screen, we identified vinexin as a binding partner for dystroglycan. Furthermore, we show that dystroglycan can interact indirectly with vinculin by binding to the vinculin-binding protein vinexin, and that this interaction has a role in dystroglycan-mediated cell adhesion and spreading. For the first time, we also demonstrate unequivocally that beta-dystroglycan is a resident of focal adhesions.


Subject(s)
Dystroglycans/metabolism , Focal Adhesions/metabolism , Myoblasts/metabolism , Animals , Cell Adhesion , Cell Line, Transformed , Cell Surface Extensions/genetics , Cell Surface Extensions/metabolism , Cloning, Molecular , Dystroglycans/genetics , Mice , Microscopy, Fluorescence , Myoblasts/pathology , Protein Binding/genetics , Protein Transport/genetics , RNA, Small Interfering/genetics , Transfection , Vinculin/metabolism
14.
J Cell Biochem ; 108(4): 877-85, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-19718658

ABSTRACT

A Disintegrin And Metalloprotease (ADAM15) is a member of the adamalysin protein family and has been associated with cancer, possibly via its role in ectodomain shedding of cadherins. Alternative mRNA splicing generates several ADAM15 isoforms containing different combinations of putative Src homology-3 (SH3) domain binding sites in their cytosolic tails. Here we present a comprehensive characterization of SH3 binding potential of different ADAM15 isoforms. Alternative use of ADAM15 exons was found to profoundly influence selection of SH3-containing cellular partner proteins, including the avid interactions with nephrocystin and sorting nexin-33 (SNX33 a.k.a. SNX30). Specifically, strong co-precipitation of nephrocystin from cell lysates was specific to ADAM15 isoforms i4, i5, and i6. These isoforms contain one or both of the two almost identical proline-rich regions encoded by exons 20 and 21, wherein the residues RxLPxxP were found to be indispensable for nephrocystin SH3 binding. Similarly, robust cellular association with SNX33 was observed only for ADAM15 isoforms containing the most carboxyterminal proline cluster lacking in isoforms i1 and i3. Thus, alternative mRNA splicing provides a versatile mechanism for regulation of intracellular protein interactions and thereby likely the cellular functions of ADAM15, which could explain the association with cancer of some but not all ADAM15 isoforms.


Subject(s)
ADAM Proteins/biosynthesis , ADAM Proteins/genetics , Adaptor Proteins, Signal Transducing/chemistry , Alternative Splicing , Carrier Proteins/chemistry , Membrane Proteins/biosynthesis , Membrane Proteins/chemistry , Membrane Proteins/genetics , Vesicular Transport Proteins/chemistry , Amino Acid Motifs , Cell Line , Cytoskeletal Proteins , Humans , Proline/chemistry , Promoter Regions, Genetic , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , RNA, Messenger/metabolism , Sorting Nexins , src Homology Domains
15.
PLoS One ; 3(11): e3638, 2008.
Article in English | MEDLINE | ID: mdl-18982058

ABSTRACT

BACKGROUND: Dystroglycan is a ubiquitously expressed cell adhesion receptor best understood in its role as part of the dystrophin glycoprotein complex of mature skeletal muscle. Less is known of the role of dystroglycan in more fundamental aspects of cell adhesion in other cell types, nor of its role in myoblast cell adhesion. PRINCIPAL FINDINGS: We have examined the role of dystroglycan in the early stages of myoblast adhesion and spreading and found that dystroglycan initially associates with other adhesion proteins in large puncta morphologically similar to podosomes. Using a human SH3 domain phage display library we identified Tks5, a key regulator of podosomes, as interacting with beta-dystroglycan. We verified the interaction by immunoprecipitation, GST-pulldown and immunfluorescence localisation. Both proteins localise to puncta during early phases of spreading, but importantly following stimulation with phorbol ester, also localise to structures indistinguishable from podosomes. Dystroglycan overexpression inhibited podosome formation by sequestering Tks5 and Src. Mutation of dystroglycan tyrosine 890, previously identified as a Src substrate, restored podosome formation. CONCLUSIONS: We propose therefore, that Src-dependent phosphorylation of beta-dystroglycan results in the formation of a Src/dystroglycan complex that drives the SH3-mediated association between dystroglycan and Tks5 which together regulate podosome formation in myoblasts.


Subject(s)
Cell Adhesion/physiology , Cell Surface Extensions/metabolism , Dystroglycans/metabolism , Myoblasts/cytology , Phosphoproteins/metabolism , src-Family Kinases/metabolism , Actins/metabolism , Animals , Biomarkers/metabolism , Cell Surface Extensions/ultrastructure , Cells, Cultured , Cortactin/metabolism , Dystroglycans/genetics , Humans , Mice , Myoblasts/physiology , Phosphate-Binding Proteins , Phosphoproteins/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , src Homology Domains , src-Family Kinases/antagonists & inhibitors , src-Family Kinases/genetics
16.
J Mol Biol ; 382(1): 167-78, 2008 Sep 26.
Article in English | MEDLINE | ID: mdl-18644376

ABSTRACT

We have determined the solution structure of epidermal growth factor receptor pathway substrate 8 (Eps8) L1 Src homology 3 (SH3) domain in complex with the PPVPNPDYEPIR peptide from the CD3epsilon cytoplasmic tail. Our structure reveals the distinct structural features that account for the unusual specificity of the Eps8 family SH3 domains for ligands containing a PxxDY motif instead of canonical PxxP ligands. The CD3epsilon peptide binds Eps8L1 SH3 in a class II orientation, but neither adopts a polyproline II helical conformation nor engages the first proline-binding pocket of the SH3 ligand binding interface. Ile531 of Eps8L1 SH3, instead of Tyr or Phe residues typically found in this position in SH3 domains, renders this hydrophobic pocket smaller and nonoptimal for binding to conventional PxxP peptides. A positively charged arginine at position 512 in the n-Src loop of Eps8L1 SH3 plays a key role in PxxDY motif recognition by forming a salt bridge to D7 of the CD3epsilon peptide. In addition, our structural model suggests a hydrogen bond between the hydroxyl group of the aromatic ring of Y8 and the carboxyl group of E496, thus explaining the critical role of the PxxDY motif tyrosine residue in binding to Eps8 family SH3. These finding have direct implications also for understanding the atypical binding specificity of the amino-terminal SH3 of the Nck family proteins.


Subject(s)
Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/metabolism , src Homology Domains , Amino Acid Motifs , Amino Acid Sequence , CD3 Complex/metabolism , Calorimetry , Ligands , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Interaction Mapping , Protein Structure, Secondary , Sequence Alignment , Structure-Activity Relationship
17.
BMC Mol Biol ; 8: 90, 2007 Oct 15.
Article in English | MEDLINE | ID: mdl-17937806

ABSTRACT

BACKGROUND: ADAM15 is a metalloprotease-disintegrin implicated in ectodomain shedding and cell adhesion. Aberrant ADAM15 expression has been associated with human cancer and other disorders. We have previously shown that the alternative splicing of ADAM15 transcripts is mis-regulated in cancer cells. To gain a better understanding of ADAM15 regulation, its genomic organization and regulatory elements as well as the alternative exon use in human tissues were characterized. RESULTS: Human ADAM15, flanked by the FLJ32785/DCST1 and ephrin-A4 genes, spans 11.4 kb from the translation initiation codon to the polyadenylation signal, being the shortest multiple-exon ADAM gene. The gene contains 23 exons varying from 63 to 316 bp and 22 introns from 79 to 1283 bp. The gene appeared to have several transcription start sites and their location suggested the promoter location within a CpG island proximal to the translation start. Reporter expression experiments confirmed the location of functional GC-rich, TATAless and CAATless promoter, with the most critical transcription-supporting elements located -266 to -23 bp relative to the translation start. Normal human tissues showed different complex patterns of at least 13 different ADAM15 splice variants arising from the alternative use of the cytosolic-encoding exons 19, 20a/b, and 21a/b. The deduced ADAM15 protein isoforms have different combinations of cytosolic regulatory protein interaction motifs. CONCLUSION: Characterization of human ADAM15 gene and identification of elements involved in the regulation of transcription and alternative splicing provide important clues for elucidation of physiological and pathological roles of ADAM15. The present results also show that the alternative exon use is a physiological post-transcriptional mechanism regulating ADAM15 expression in human tissues.


Subject(s)
ADAM Proteins/genetics , Alternative Splicing/physiology , Exons/physiology , Gene Expression Regulation, Enzymologic/physiology , Membrane Proteins/genetics , ADAM Proteins/biosynthesis , Amino Acid Motifs/genetics , Cell Adhesion/physiology , Cell Line , Codon, Initiator/physiology , CpG Islands/physiology , Ephrin-A4/genetics , Ephrin-A4/metabolism , Humans , Membrane Proteins/biosynthesis , Neoplasms/enzymology , Neoplasms/genetics , Organ Specificity/physiology , Promoter Regions, Genetic/physiology , Protein Isoforms/biosynthesis , Protein Isoforms/genetics
18.
EMBO Rep ; 7(2): 186-91, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16374509

ABSTRACT

We have determined the human genome to contain 296 different Src homology-3 (SH3) domains and cloned them into a phage-display vector. This provided a powerful and unbiased system for simultaneous assaying of the complete human SH3 proteome for the strongest binding to target proteins of interest, without the limitations posed by short linear peptide ligands or confounding variables of more indirect methods for protein interaction screening. Studies involving three ligand proteins, human immunodeficiency virus-1 Nef, p21-activated kinase (PAK)2 and ADAM15, showed previously reported as well as novel SH3 partners with nanomolar affinities specific for them. This argues that SH3 domains may have a more dominant role in directing cellular protein interactions than has been assumed. Besides showing potentially important new SH3-directed interactions, these studies also led to the discovery of novel signalling proteins, such as the PAK2-binding adaptor protein POSH2 and the ADAM15-binding sorting nexin family member SNX30.


Subject(s)
Peptide Library , Proteins/metabolism , Proteome , src Homology Domains/physiology , ADAM Proteins/genetics , ADAM Proteins/metabolism , Carrier Proteins/genetics , Carrier Proteins/metabolism , Gene Products, nef/genetics , Gene Products, nef/metabolism , Genetic Vectors , Glutathione Transferase/metabolism , Humans , Ligands , Membrane Proteins/genetics , Membrane Proteins/metabolism , Protein Binding , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proteins/genetics , Recombinant Fusion Proteins/metabolism , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism , Vesicular Transport Proteins/genetics , Vesicular Transport Proteins/metabolism , p21-Activated Kinases , src Homology Domains/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...