Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 13.902
Filter
1.
Methods Mol Biol ; 2836: 253-281, 2024.
Article in English | MEDLINE | ID: mdl-38995545

ABSTRACT

Interactomics is bringing a deluge of data regarding protein-protein interactions (PPIs) which are involved in various molecular processes in all types of cells. However, this information does not easily translate into direct and precise molecular interfaces. This limits our understanding of each interaction network and prevents their efficient modulation. A lot of the detected interactions involve recognition of short linear motifs (SLiMs) by a folded domain while others rely on domain-domain interactions. Functional SLiMs hide among a lot of spurious ones, making deeper analysis of interactomes tedious. Hence, actual contacts and direct interactions are difficult to identify.Consequently, there is a need for user-friendly bioinformatic tools, enabling rapid molecular and structural analysis of SLiM-based PPIs in a protein network. In this chapter, we describe the use of the new webserver SLiMAn to help digging into SLiM-based PPIs in an interactive fashion.


Subject(s)
Computational Biology , Internet , Protein Interaction Mapping , Software , Protein Interaction Mapping/methods , Computational Biology/methods , Protein Interaction Domains and Motifs , Proteins/chemistry , Proteins/metabolism , Protein Interaction Maps , Amino Acid Motifs , Humans , Databases, Protein , Protein Binding
2.
Mol Cell ; 84(14): 2648-2664.e10, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38955181

ABSTRACT

The essential Mediator (MED) coactivator complex plays a well-understood role in regulation of basal transcription in all eukaryotes, but the mechanism underlying its role in activator-dependent transcription remains unknown. We investigated modulation of metazoan MED interaction with RNA polymerase II (RNA Pol II) by antagonistic effects of the MED26 subunit and the CDK8 kinase module (CKM). Biochemical analysis of CKM-MED showed that the CKM blocks binding of the RNA Pol II carboxy-terminal domain (CTD), preventing RNA Pol II interaction. This restriction is eliminated by nuclear receptor (NR) binding to CKM-MED, which enables CTD binding in a MED26-dependent manner. Cryoelectron microscopy (cryo-EM) and crosslinking-mass spectrometry (XL-MS) revealed that the structural basis for modulation of CTD interaction with MED relates to a large intrinsically disordered region (IDR) in CKM subunit MED13 that blocks MED26 and CTD interaction with MED but is repositioned upon NR binding. Hence, NRs can control transcription initiation by priming CKM-MED for MED26-dependent RNA Pol II interaction.


Subject(s)
Cryoelectron Microscopy , Cyclin-Dependent Kinase 8 , Mediator Complex , Protein Binding , RNA Polymerase II , RNA Polymerase II/metabolism , RNA Polymerase II/genetics , Mediator Complex/metabolism , Mediator Complex/genetics , Mediator Complex/chemistry , Humans , Cyclin-Dependent Kinase 8/metabolism , Cyclin-Dependent Kinase 8/genetics , Animals , Intrinsically Disordered Proteins/metabolism , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/chemistry , Binding Sites , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , HEK293 Cells , Protein Interaction Domains and Motifs
3.
Methods Mol Biol ; 2796: 105-118, 2024.
Article in English | MEDLINE | ID: mdl-38856898

ABSTRACT

Ion channels are transmembrane proteins essential for cellular functions and are important drug targets. Surface plasmon resonance (SPR) is a powerful technique for investigating protein-protein and protein-small molecule ligand interactions. SPR has been underutilized for studies of ion channels, even though it could provide a wealth of information on the mechanisms of ion channel regulation and aid in ion channel drug discovery. Here we provide a detailed description of the use of SPR technology for investigating inter-domain interactions in KCNH potassium-selective and voltage-gated ion channels.


Subject(s)
Surface Plasmon Resonance , Surface Plasmon Resonance/methods , Humans , Protein Binding , Ion Channels/metabolism , Ion Channels/chemistry , Ether-A-Go-Go Potassium Channels/metabolism , Ether-A-Go-Go Potassium Channels/chemistry , Protein Interaction Domains and Motifs , Ligands , Animals
4.
Int J Mol Sci ; 25(12)2024 Jun 18.
Article in English | MEDLINE | ID: mdl-38928400

ABSTRACT

The emergence of coronavirus disease 2019 (COVID-19) posed a major challenge to healthcare systems worldwide, especially as mutations in the culprit Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) complicated the development of vaccines and antiviral drugs. Therefore, the search for natural products with broad anti-SARS-CoV-2 capabilities is an important option for the prevention and treatment of similar infectious diseases. Lectins, which are widely recognized as antiviral agents, could contribute to the development of anti-SARS-CoV-2 drugs. This study evaluated the binding affinity of six lectins (including the cyanobacterial lectin from Microcystis viridis NIES-102 (MVL), and Jacalin, a lectin from the breadfruit, Artocarpus altilis) to the receptor binding domain (RBD) of the spike protein on the original (wild) SARS-CoV-2 and three of its mutants: Alpha, Delta, and Omicron. MVL and Jacalin showed distinct binding affinity to the RBDs of the four SARS-CoV-2 strains. The remaining four lectins (DB1, ConA, PHA-M and CSL3) showed no such binding affinity. Although the glycan specificities of MVL and Jacalin were different, they showed the same affinity for the spike protein RBDs of the four SARS-CoV-2 strains, in the order of effectiveness Alpha > Delta > original > Omicron. The verification of glycan-specific inhibition revealed that both lectins bind to RBDs by glycan-specific recognition, but, in addition, MVL binds to RBDs through protein-protein interactions.


Subject(s)
Lectins , Microcystis , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Lectins/metabolism , Lectins/chemistry , SARS-CoV-2/metabolism , SARS-CoV-2/genetics , Microcystis/metabolism , Humans , COVID-19/virology , COVID-19/metabolism , Antiviral Agents/pharmacology , Antiviral Agents/metabolism , Protein Interaction Domains and Motifs , Cyanobacteria/metabolism , Plant Lectins/metabolism , Plant Lectins/chemistry , Binding Sites , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Mutation
5.
Mol Cell ; 84(14): 2601-2617.e12, 2024 Jul 25.
Article in English | MEDLINE | ID: mdl-38925115

ABSTRACT

The evolutionarily conserved HIRA/Hir histone chaperone complex and ASF1a/Asf1 co-chaperone cooperate to deposit histone (H3/H4)2 tetramers on DNA for replication-independent chromatin assembly. The molecular architecture of the HIRA/Hir complex and its mode of histone deposition have remained unknown. Here, we report the cryo-EM structure of the S. cerevisiae Hir complex with Asf1/H3/H4 at 2.9-6.8 Å resolution. We find that the Hir complex forms an arc-shaped dimer with a Hir1/Hir2/Hir3/Hpc2 stoichiometry of 2/4/2/4. The core of the complex containing two Hir1/Hir2/Hir2 trimers and N-terminal segments of Hir3 forms a central cavity containing two copies of Hpc2, with one engaged by Asf1/H3/H4, in a suitable position to accommodate a histone (H3/H4)2 tetramer, while the C-terminal segments of Hir3 harbor nucleic acid binding activity to wrap DNA around the Hpc2-assisted histone tetramer. The structure suggests a model for how the Hir/Asf1 complex promotes the formation of histone tetramers for their subsequent deposition onto DNA.


Subject(s)
Cell Cycle Proteins , Cryoelectron Microscopy , Histone Chaperones , Histones , Protein Binding , Saccharomyces cerevisiae Proteins , Saccharomyces cerevisiae , Histones/metabolism , Histones/chemistry , Histones/genetics , Saccharomyces cerevisiae Proteins/metabolism , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/genetics , Saccharomyces cerevisiae Proteins/ultrastructure , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Cell Cycle Proteins/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/genetics , Histone Chaperones/metabolism , Histone Chaperones/chemistry , Histone Chaperones/genetics , Models, Molecular , Molecular Chaperones/metabolism , Molecular Chaperones/chemistry , Molecular Chaperones/genetics , Protein Multimerization , Binding Sites , Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/genetics , Protein Interaction Domains and Motifs
6.
Curr Genet ; 70(1): 8, 2024 Jun 24.
Article in English | MEDLINE | ID: mdl-38913087

ABSTRACT

The Byr2 kinase of fission yeast Schizosaccharomyces pombe is recruited to the membrane with the assistance of Ras1. Byr2 is also negatively regulated by 14-3-3 proteins encoded by rad24 and rad25. We conducted domain and mutational analysis of Byr2 to determine which region is critical for its binding to 14-3-3 proteins. Rad24 and Rad25 bound to both the Ras interaction domain in the N-terminus and to the C-terminal catalytic domain of Byr2. When amino acid residues S87 and T94 of the Ras-interacting domain of Byr2 were mutated to alanine, Rad24 could no longer bind to Byr2. S402, S566, S650, and S654 mutations in the C-terminal domain of Byr2 also abolished its interaction with Rad24 and Rad25. More than three mutations in the C-terminal domain were required to abolish completely its interaction with 14-3-3 protein, suggesting that multiple residues are involved in this interaction. Expression of the N-terminal domain of Byr2 in wild-type cells lowered the mating ratio, because it likely blocked the interaction of Byr2 with Ste4 and Ras1, whereas expression of the catalytic domain of Byr2 increased the mating ratio as a result of freeing from intramolecular regulation by the N-terminal domain of Byr2. The S87A and T94A mutations of Byr2 increased the mating ratio and attenuated inhibition of Byr2 by Rad24; therefore, these two amino acids are critical for its regulation by Rad24. S566 of Byr2 is critical for activity of Byr2 but not for its interaction with 14-3-3 proteins. In this study, we show that 14-3-3 proteins interact with two separate domains in Byr2 as negative regulators.


Subject(s)
14-3-3 Proteins , Protein Binding , Schizosaccharomyces pombe Proteins , Schizosaccharomyces , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Schizosaccharomyces pombe Proteins/genetics , Schizosaccharomyces pombe Proteins/metabolism , Schizosaccharomyces pombe Proteins/chemistry , Schizosaccharomyces/genetics , Schizosaccharomyces/metabolism , Mutation , DNA Mutational Analysis , Protein Domains/genetics , Protein Interaction Domains and Motifs , Cell Cycle Proteins , Intracellular Signaling Peptides and Proteins
7.
Int J Mol Sci ; 25(12)2024 Jun 13.
Article in English | MEDLINE | ID: mdl-38928241

ABSTRACT

Human infection with the coronavirus disease 2019 (COVID-19) is mediated by the binding of the spike protein of the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) to the human angiotensin-converting enzyme 2 (ACE2). The frequent mutations in the receptor-binding domain (RBD) of the spike protein induced the emergence of variants with increased contagion and can hinder vaccine efficiency. Hence, it is crucial to better understand the binding mechanisms of variant RBDs to human ACE2 and develop efficient methods to characterize this interaction. In this work, we present an approach that uses machine learning to analyze the molecular dynamics simulations of RBD variant trajectories bound to ACE2. Along with the binding free energy calculation, this method was used to characterize the major differences in ACE2-binding capacity of three SARS-CoV-2 RBD variants-namely the original Wuhan strain, Omicron BA.1, and the more recent Omicron BA.5 sublineages. Our analyses assessed the differences in binding free energy and shed light on how it affects the infectious rates of different variants. Furthermore, this approach successfully characterized key binding interactions and could be deployed as an efficient tool to predict different binding inhibitors to pave the way for new preventive and therapeutic strategies.


Subject(s)
Angiotensin-Converting Enzyme 2 , COVID-19 , Machine Learning , Molecular Dynamics Simulation , Protein Binding , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , SARS-CoV-2/metabolism , SARS-CoV-2/genetics , Angiotensin-Converting Enzyme 2/metabolism , Angiotensin-Converting Enzyme 2/chemistry , Humans , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , COVID-19/virology , COVID-19/metabolism , Binding Sites , Mutation , Protein Interaction Domains and Motifs
8.
Nucleic Acids Res ; 52(W1): W313-W317, 2024 Jul 05.
Article in English | MEDLINE | ID: mdl-38783158

ABSTRACT

Among the myriad of protein-protein interactions occurring in living organisms, a substantial amount involves small linear motifs (SLiMs) recognized by structured domains. However, predictions of SLiM-based networks are tedious, due to the abundance of such motifs and a high portion of false positive hits. For this reason, a webserver SLiMAn (Short Linear Motif Analysis) was developed to focus the search on the most relevant SLiMs. Using SLiMAn, one can navigate into a given (meta-)interactome and tune a variety of parameters associated to each type of SLiMs in attempt to identify functional ELM motifs and their recognition domains. The IntAct and BioGRID databases bring experimental information, while IUPred and AlphaFold provide boundaries of folded and disordered regions. Post-translational modifications listed in PhosphoSite+ are highlighted. Links to PubMed accelerate scrutiny into the literature, to support (or not) putative pairings. Dedicated visualization features are also incorporated, such as Cytoscape for macromolecular networks and BINANA for intermolecular contacts within structural models generated by SCWRL 3.0. The use of SLiMAn 2.0 is illustrated on a simple example. It is freely available at https://sliman2.cbs.cnrs.fr.


Subject(s)
Peptides , Software , Peptides/chemistry , Peptides/metabolism , Protein Interaction Mapping/methods , Protein Interaction Maps , Databases, Protein , Humans , Amino Acid Motifs , Proteins/chemistry , Proteins/metabolism , Internet , Protein Interaction Domains and Motifs , Protein Processing, Post-Translational
9.
Proteins ; 92(8): 984-997, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38641972

ABSTRACT

Glycoside hydrolase (GH) family 13 is among the main families of enzymes acting on starch; recently, subfamily 47 of GH13 (GH13_47) has been established. The crystal structure and function of a GH13_47 enzyme from Bacteroides ovatus has only been reported to date. This enzyme has α-amylase activity, while the GH13_47 enzymes comprise approximately 800-900 amino acid residues which are almost double those of typical α-amylases. It is important to know how different the GH13_47 enzymes are from other α-amylases. Rhodothermus marinus JCM9785, a thermophilic bacterium, possesses a gene for the GH13_47 enzyme, which is designated here as RmGH13_47A. Its structure has been predicted to be composed of seven domains: N1, N2, N3, A, B, C, and D. We constructed a plasmid encoding Gly266-Glu886, which contains the N3, A, B, and C domains and expressed the protein in Escherichia coli. The enzyme hydrolyzed starch and pullulan by a neopullulanase-type action. Additionally, the enzyme acted on maltotetraose, and saccharides with α-1,6-glucosidic linkages were observed in the products. Following the replacement of the catalytic residue Asp563 with Ala, the crystal structure of the variant D563A in complex with the enzymatic products from maltotetraose was determined; as a result, electron density for an α-1,6-branched pentasaccharide was observed in the catalytic pocket, and Ile762 and Asp763 interacted with the branched chain of the pentasaccharide. These findings suggest that RmGH13_47A is an α-amylase that prefers α-1,6-branched parts of starch to produce oligosaccharides.


Subject(s)
Bacterial Proteins , Models, Molecular , Rhodothermus , alpha-Amylases , Rhodothermus/enzymology , Rhodothermus/genetics , alpha-Amylases/chemistry , alpha-Amylases/metabolism , alpha-Amylases/genetics , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , Glucans/metabolism , Glucans/chemistry , Substrate Specificity , Starch/metabolism , Starch/chemistry , Amino Acid Sequence , Oligosaccharides/metabolism , Oligosaccharides/chemistry , Catalytic Domain , Protein Binding , Escherichia coli/genetics , Escherichia coli/metabolism , Hydrolysis , Protein Interaction Domains and Motifs , Crystallography, X-Ray , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Cloning, Molecular , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/metabolism , Glycoside Hydrolases/genetics , Binding Sites , Protein Conformation, alpha-Helical , Maltose/analogs & derivatives
10.
Sci Data ; 11(1): 402, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38643260

ABSTRACT

This dataset represents a collection of pocket-centric structural data related to protein-protein interactions (PPIs) and PPI-related ligand binding sites. The dataset includes high-quality structural information on more than 23,000 pockets, 3,700 proteins on more than 500 organisms, and nearly 3500 ligands that can aid researchers in the fields of bioinformatics, structural biology, and drug discovery. It encompasses a diverse set of PPI complexes with more than 1,700 unique protein families including some with associated ligands, enabling detailed investigations into molecular interactions at the atomic level. This article introduces an indispensable resource designed to unlock the full potential of PPIs while pioneering a novel metric for pocket similarity for hypothesizing protein partners repurposing.


Subject(s)
Drug Discovery , Protein Interaction Domains and Motifs , Proteins , Binding Sites , Ligands , Proteins/chemistry
11.
FEBS Lett ; 598(7): 725-742, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38439692

ABSTRACT

Protein-protein interactions (PPIs) are often mediated by short linear motifs (SLiMs) in one protein and domain in another, known as domain-motif interactions (DMIs). During the past decade, SLiMs have been studied to find their role in cellular functions such as post-translational modifications, regulatory processes, protein scaffolding, cell cycle progression, cell adhesion, cell signalling and substrate selection for proteasomal degradation. This review provides a comprehensive overview of the current PPI detection techniques and resources, focusing on their relevance to capturing interactions mediated by SLiMs. We also address the challenges associated with capturing DMIs. Moreover, a case study analysing the BioGrid database as a source of DMI prediction revealed significant known DMI enrichment in different PPI detection methods. Overall, it can be said that current high-throughput PPI detection methods can be a reliable source for predicting DMIs.


Subject(s)
Protein Interaction Mapping , Proteins , Protein Interaction Domains and Motifs , Proteins/metabolism , Databases, Protein
12.
Proteins ; 92(7): 808-818, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38333996

ABSTRACT

Isopentenyl phosphate kinases (IPKs) have recently garnered attention for their central role in biocatalytic "isoprenol pathways," which seek to reduce the synthesis of the isoprenoid precursors to two enzymatic steps. Furthermore, the natural promiscuity of IPKs toward non-natural alkyl-monophosphates (alkyl-Ps) as substrates has hinted at the isoprenol pathways' potential to access novel isoprenoids with potentially useful activities. However, only a handful of IPK crystal structures have been solved to date, and even fewer of these contain non-natural substrates bound in the active site. The current study sought to elucidate additional ternary complexes bound to non-natural substrates using the IPK homolog from Thermococcus paralvinellae (TcpIPK). Four such structures were solved, each bound to a different non-natural alkyl-P and the phosphoryl donor substrate/product adenosine triphosphate (ATP)/adenosine diphosphate (ADP). As expected, the quaternary, tertiary, and secondary structures of TcpIPK closely resembled those of IPKs published previously, and kinetic analysis of a novel alkyl-P substrate highlighted the potentially dramatic effects of altering the core scaffold of the natural substrate. Even more interesting, though, was the discovery of a trend correlating the position of two α helices in the active site with the magnitude of an IPK homolog's reaction rate for the natural reaction. Overall, the current structures of TcpIPK highlight the importance of continued structural analysis of the IPKs to better understand and optimize their activity with both natural and non-natural substrates.


Subject(s)
Adenosine Triphosphate , Catalytic Domain , Thermococcus , Substrate Specificity , Thermococcus/enzymology , Adenosine Triphosphate/metabolism , Adenosine Triphosphate/chemistry , Crystallography, X-Ray , Models, Molecular , Protein Binding , Kinetics , Archaeal Proteins/chemistry , Archaeal Proteins/metabolism , Archaeal Proteins/genetics , Hemiterpenes/metabolism , Hemiterpenes/chemistry , Protein Interaction Domains and Motifs , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Recombinant Proteins/genetics , Protein Conformation, alpha-Helical , Adenosine Diphosphate/metabolism , Adenosine Diphosphate/chemistry , Cloning, Molecular , Gene Expression , Protein Conformation, beta-Strand , Amino Acid Sequence , Escherichia coli/genetics , Escherichia coli/metabolism , Escherichia coli/enzymology , Protein Kinases
13.
Proteins ; 92(7): 797-807, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38314653

ABSTRACT

Antibody light chain amyloidosis is a disorder in which protein aggregates, mainly composed of immunoglobulin light chains, deposit in diverse tissues impairing the correct functioning of organs. Interestingly, due to the high susceptibility of antibodies to mutations, AL amyloidosis appears to be strongly patient-specific. Indeed, every patient will display their own mutations that will make the proteins involved prone to aggregation thus hindering the study of this disease on a wide scale. In this framework, determining the molecular mechanisms that drive the aggregation could pave the way to the development of patient-specific therapeutics. Here, we focus on a particular patient-derived light chain, which has been experimentally characterized. We investigated the early phases of the aggregation pathway through extensive full-atom molecular dynamics simulations, highlighting a structural rearrangement and the exposure of two hydrophobic regions in the aggregation-prone species. Next, we moved to consider the pathological dimerization process through docking and molecular dynamics simulations, proposing a dimeric structure as a candidate pathological first assembly. Overall, our results shed light on the first phases of the aggregation pathway for a light chain at an atomic level detail, offering new structural insights into the corresponding aggregation process.


Subject(s)
Molecular Dynamics Simulation , Protein Folding , Protein Multimerization , Humans , Immunoglobulin Light Chains/chemistry , Immunoglobulin Light Chains/metabolism , Immunoglobulin Light Chains/genetics , Hydrophobic and Hydrophilic Interactions , Protein Aggregation, Pathological/metabolism , Protein Aggregates , Mutation , Molecular Docking Simulation , Immunoglobulin Light-chain Amyloidosis/metabolism , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs
14.
mSphere ; 9(3): e0069623, 2024 Mar 26.
Article in English | MEDLINE | ID: mdl-38376217

ABSTRACT

Fungal infections cause a large health burden but are treated by only a handful of antifungal drug classes. Chromatin factors have emerged as possible targets for new antifungals. These targets include the reader proteins, which interact with posttranslationally modified histones to influence DNA transcription and repair. The YEATS domain is one such reader recognizing both crotonylated and acetylated histones. Here, we performed a detailed structure/function analysis of the Candida albicans YEATS domain reader Yaf9, a subunit of the NuA4 histone acetyltransferase and the SWR1 chromatin remodeling complex. We have previously demonstrated that the homozygous deletion mutant yaf9Δ/Δ displays growth defects and is avirulent in mice. Here we show that a YEATS domain mutant expected to inactivate Yaf9's chromatin binding does not display strong phenotypes in vitro, nor during infection of immune cells or in a mouse systemic infection model, with only a minor virulence reduction in vivo. In contrast to the YEATS domain mutation, deletion of the C-terminal domain of Yaf9, a protein-protein interaction module necessary for its interactions with SWR1 and NuA4, phenocopies the null mutant. This shows that the C-terminal domain is essential for Yaf9 roles in vitro and in vivo, including C. albicans virulence. Our study informs on the strategies for therapeutic targeting of Yaf9, showing that approaches taken for the mammalian YEATS domains by disrupting their chromatin binding might not be effective in C. albicans, and provides a foundation for studying YEATS proteins in human fungal pathogens.IMPORTANCEThe scarcity of available antifungal drugs and rising resistance demand the development of therapies with new modes of action. In this context, chromatin regulation may be a target for novel antifungal therapeutics. To realize this potential, we must better understand the roles of chromatin regulators in fungal pathogens. Toward this goal, here, we studied the YEATS domain chromatin reader Yaf9 in Candida albicans. Yaf9 uses the YEATS domain for chromatin binding and a C-terminal domain to interact with chromatin remodeling complexes. By constructing mutants in these domains and characterizing their phenotypes, our data indicate that the Yaf9 YEATS domain might not be a suitable therapeutic drug target. Instead, the Yaf9 C-terminal domain is critical for C. albicans virulence. Collectively, our study informs how a class of chromatin regulators performs their cellular and pathogenesis roles in C. albicans and reveals strategies to inhibit them.


Subject(s)
Chromatin , Saccharomyces cerevisiae Proteins , Humans , Animals , Mice , Chromatin/genetics , Histones/genetics , Candida albicans/genetics , Candida albicans/metabolism , Saccharomyces cerevisiae Proteins/genetics , Antifungal Agents , Homozygote , Sequence Deletion , Transcription Factors/genetics , Transcription Factors/metabolism , Protein Interaction Domains and Motifs , Mammals
15.
J Biol Chem ; 300(3): 105699, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38301891

ABSTRACT

DEC205 (CD205) is one of the major endocytic receptors on dendritic cells and has been widely used as a receptor target in immune therapies. It has been shown that DEC205 can recognize dead cells through keratins in a pH-dependent manner. However, the mechanism underlying the interaction between DEC205 and keratins remains unclear. Here we determine the crystal structures of an N-terminal fragment of human DEC205 (CysR∼CTLD3). The structural data show that DEC205 shares similar overall features with the other mannose receptor family members such as the mannose receptor and Endo180, but the individual domains of DEC205 in the crystal structure exhibit distinct structural features that may lead to specific ligand binding properties of the molecule. Among them, CTLD3 of DEC205 adopts a unique fold of CTLD, which may correlate with the binding of keratins. Furthermore, we examine the interaction of DEC205 with keratins by mutagenesis and biochemical assays based on the structural information and identify an XGGGX motif on keratins that can be recognized by DEC205, thereby providing insights into the interaction between DEC205 and keratins. Overall, these findings not only improve the understanding of the diverse ligand specificities of the mannose receptor family members at the molecular level but may also give clues for the interactions of keratins with their binding partners in the corresponding pathways.


Subject(s)
Keratins , Lectins, C-Type , Models, Molecular , Humans , Dendritic Cells/metabolism , Lectins, C-Type/chemistry , Lectins, C-Type/genetics , Lectins, C-Type/metabolism , Ligands , Mannose Receptor/chemistry , Mutagenesis , Protein Binding , Protein Folding , Protein Structure, Tertiary , Protein Interaction Domains and Motifs , Crystallography, X-Ray
16.
Nucleic Acids Res ; 52(6): 3106-3120, 2024 Apr 12.
Article in English | MEDLINE | ID: mdl-38364856

ABSTRACT

Chromatin accessibility plays a critical role in the regulation of cell fate decisions. Although gene expression changes have been extensively profiled at the single-cell level during early embryogenesis, the dynamics of chromatin accessibility at cis-regulatory elements remain poorly studied. Here, we used a plate-based single-cell ATAC-seq method to profile the chromatin accessibility dynamics of over 10 000 nuclei from zebrafish embryos. We investigated several important time points immediately after zygotic genome activation (ZGA), covering key developmental stages up to dome. The results revealed key chromatin signatures in the first cell fate specifications when cells start to differentiate into enveloping layer (EVL) and yolk syncytial layer (YSL) cells. Finally, we uncovered many potential cell-type specific enhancers and transcription factor motifs that are important for the cell fate specifications.


Subject(s)
Chromatin , Embryonic Development , Zebrafish , Animals , Chromatin/genetics , Chromatin/metabolism , Egg Yolk/metabolism , Embryo, Nonmammalian/embryology , Embryo, Nonmammalian/metabolism , Embryonic Development/genetics , Zebrafish/embryology , Zebrafish/genetics , Single-Cell Analysis , Protein Interaction Domains and Motifs/genetics , Transcription Factors/genetics , Transcription Factors/metabolism
17.
Science ; 383(6685): 890-897, 2024 Feb 23.
Article in English | MEDLINE | ID: mdl-38386755

ABSTRACT

Recordings of the physiological history of cells provide insights into biological processes, yet obtaining such recordings is a challenge. To address this, we introduce a method to record transient cellular events for later analysis. We designed proteins that become labeled in the presence of both a specific cellular activity and a fluorescent substrate. The recording period is set by the presence of the substrate, whereas the cellular activity controls the degree of the labeling. The use of distinguishable substrates enabled the recording of successive periods of activity. We recorded protein-protein interactions, G protein-coupled receptor activation, and increases in intracellular calcium. Recordings of elevated calcium levels allowed selections of cells from heterogeneous populations for transcriptomic analysis and tracking of neuronal activities in flies and zebrafish.


Subject(s)
Calcium , Cell Physiological Phenomena , Cells , Staining and Labeling , Animals , Coloring Agents , Gene Expression Profiling , Zebrafish , Cells/chemistry , Protein Interaction Domains and Motifs
18.
Science ; 383(6678): 101-108, 2024 01 05.
Article in English | MEDLINE | ID: mdl-38175886

ABSTRACT

ß-arrestins (ßarrs) are multifunctional proteins involved in signaling and regulation of seven transmembrane receptors (7TMRs), and their interaction is driven primarily by agonist-induced receptor activation and phosphorylation. Here, we present seven cryo-electron microscopy structures of ßarrs either in the basal state, activated by the muscarinic receptor subtype 2 (M2R) through its third intracellular loop, or activated by the ßarr-biased decoy D6 receptor (D6R). Combined with biochemical, cellular, and biophysical experiments, these structural snapshots allow the visualization of atypical engagement of ßarrs with 7TMRs and also reveal a structural transition in the carboxyl terminus of ßarr2 from a ß strand to an α helix upon activation by D6R. Our study provides previously unanticipated molecular insights into the structural and functional diversity encoded in 7TMR-ßarr complexes with direct implications for exploring novel therapeutic avenues.


Subject(s)
Protein Interaction Domains and Motifs , Receptors, G-Protein-Coupled , beta-Arrestins , beta-Arrestins/chemistry , Cryoelectron Microscopy , Receptors, G-Protein-Coupled/chemistry , Signal Transduction , Protein Conformation, beta-Strand , Protein Conformation, alpha-Helical , Humans
19.
Nucleic Acids Res ; 52(D1): D154-D163, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-37971293

ABSTRACT

We present a major update of the HOCOMOCO collection that provides DNA binding specificity patterns of 949 human transcription factors and 720 mouse orthologs. To make this release, we performed motif discovery in peak sets that originated from 14 183 ChIP-Seq experiments and reads from 2554 HT-SELEX experiments yielding more than 400 thousand candidate motifs. The candidate motifs were annotated according to their similarity to known motifs and the hierarchy of DNA-binding domains of the respective transcription factors. Next, the motifs underwent human expert curation to stratify distinct motif subtypes and remove non-informative patterns and common artifacts. Finally, the curated subset of 100 thousand motifs was supplied to the automated benchmarking to select the best-performing motifs for each transcription factor. The resulting HOCOMOCO v12 core collection contains 1443 verified position weight matrices, including distinct subtypes of DNA binding motifs for particular transcription factors. In addition to the core collection, HOCOMOCO v12 provides motif sets optimized for the recognition of binding sites in vivo and in vitro, and for annotation of regulatory sequence variants. HOCOMOCO is available at https://hocomoco12.autosome.org and https://hocomoco.autosome.org.


Subject(s)
Databases, Genetic , Gene Expression Regulation , Protein Interaction Domains and Motifs , Transcription Factors , Animals , Humans , Mice , Binding Sites/genetics , Nucleotide Motifs , Transcription Factors/genetics , Transcription Factors/metabolism , Internet , Protein Interaction Domains and Motifs/genetics
20.
J Mol Biol ; 436(7): 168371, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-37977297

ABSTRACT

Spindlin1 is a histone reader with three Tudor-like domains and its transcriptional co-activator activity could be attenuated by SPINDOC. The first two Tudors are involved in histone methylation readout, while the function of Tudor 3 is largely unknown. Here our structural and binding studies revealed an engagement mode of SPINDOC-Spindlin1, in which a hydrophobic motif of SPINDOC, DOCpep3, stably interacts with Spindlin1 Tudor 3, and two neighboring K/R-rich motifs, DOCpep1 and DOCpep2, bind to the acidic surface of Spindlin1 Tudor 2. Although DOCpep3-Spindlin1 engagement is compatible with histone readout, an extended SPINDOC fragment containing the K/R-rich region attenuates histone or TCF4 binding by Spindlin1 due to introduced competition. This inhibitory effect is more pronounced for weaker binding targets but not for strong ones such as H3 "K4me3-K9me3" bivalent mark. Further ChIP-seq and RT-qPCR indicated that SPINDOC could promote genomic relocation of Spindlin1, thus modulate downstream gene transcription. Collectively, we revealed multivalent engagement between SPINDOC and Spindlin1, in which a hydrophobic motif acts as the primary binding site for stable SPINDOC-Spindlin1 association, while K/R-rich region modulates the target selectivity of Spindlin1 via competitive inhibition, therefore attenuating the transcriptional co-activator activity of Spindlin1.


Subject(s)
Cell Cycle Proteins , Co-Repressor Proteins , Gene Expression Regulation , Histones , Microtubule-Associated Proteins , Phosphoproteins , Protein Interaction Domains and Motifs , Transcription, Genetic , Tudor Domain , Binding Sites , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Co-Repressor Proteins/chemistry , Co-Repressor Proteins/metabolism , Histones/metabolism , Methylation , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/metabolism , Phosphoproteins/chemistry , Phosphoproteins/metabolism , Protein Binding , Humans , Protein Interaction Mapping
SELECTION OF CITATIONS
SEARCH DETAIL