Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 129
Filter
Add more filters

Publication year range
1.
Cell ; 179(4): 923-936.e11, 2019 10 31.
Article in English | MEDLINE | ID: mdl-31675499

ABSTRACT

Tight junctions are cell-adhesion complexes that seal tissues and are involved in cell polarity and signaling. Supra-molecular assembly and positioning of tight junctions as continuous networks of adhesion strands are dependent on the membrane-associated scaffolding proteins ZO1 and ZO2. To understand how zona occludens (ZO) proteins organize junction assembly, we performed quantitative cell biology and in vitro reconstitution experiments. We discovered that ZO proteins self-organize membrane-attached compartments via phase separation. We identified the multivalent interactions of the conserved PDZ-SH3-GuK supra-domain as the driver of phase separation. These interactions are regulated by phosphorylation and intra-molecular binding. Formation of condensed ZO protein compartments is sufficient to specifically enrich and localize tight-junction proteins, including adhesion receptors, cytoskeletal adapters, and transcription factors. Our results suggest that an active-phase transition of ZO proteins into a condensed membrane-bound compartment drives claudin polymerization and coalescence of a continuous tight-junction belt.


Subject(s)
Tight Junctions/genetics , Zonula Occludens Proteins/genetics , Zonula Occludens-1 Protein/genetics , Zonula Occludens-2 Protein/genetics , Animals , Binding Sites/genetics , Cell Adhesion/genetics , Cell Polarity/genetics , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Membrane Proteins/genetics , PDZ Domains/genetics , Phosphoproteins/genetics , Phosphorylation/genetics , Protein Binding/genetics , Signal Transduction/genetics , Tight Junctions/metabolism , Zonula Occludens Proteins/chemistry , Zonula Occludens Proteins/ultrastructure , Zonula Occludens-1 Protein/chemistry , Zonula Occludens-1 Protein/ultrastructure , Zonula Occludens-2 Protein/chemistry , Zonula Occludens-2 Protein/ultrastructure , src Homology Domains/genetics
2.
Nature ; 604(7904): 175-183, 2022 04.
Article in English | MEDLINE | ID: mdl-35388192

ABSTRACT

Allosteric communication between distant sites in proteins is central to biological regulation but still poorly characterized, limiting understanding, engineering and drug development1-6. An important reason for this is the lack of methods to comprehensively quantify allostery in diverse proteins. Here we address this shortcoming and present a method that uses deep mutational scanning to globally map allostery. The approach uses an efficient experimental design to infer en masse the causal biophysical effects of mutations by quantifying multiple molecular phenotypes-here we examine binding and protein abundance-in multiple genetic backgrounds and fitting thermodynamic models using neural networks. We apply the approach to two of the most common protein interaction domains found in humans, an SH3 domain and a PDZ domain, to produce comprehensive atlases of allosteric communication. Allosteric mutations are abundant, with a large mutational target space of network-altering 'edgetic' variants. Mutations are more likely to be allosteric closer to binding interfaces, at glycine residues and at specific residues connecting to an opposite surface within the PDZ domain. This general approach of quantifying mutational effects for multiple molecular phenotypes and in multiple genetic backgrounds should enable the energetic and allosteric landscapes of many proteins to be rapidly and comprehensively mapped.


Subject(s)
Allosteric Site , PDZ Domains , Proteins , Allosteric Regulation/genetics , PDZ Domains/genetics , Protein Binding/genetics , Proteins/chemistry , Thermodynamics
3.
J Biol Chem ; 296: 100473, 2021.
Article in English | MEDLINE | ID: mdl-33639163

ABSTRACT

The type II sodium-dependent phosphate cotransporter (NPT2A) mediates renal phosphate uptake. The NPT2A is regulated by parathyroid hormone (PTH) and fibroblast growth factor 23, which requires Na+/H+ exchange regulatory factor-1 (NHERF1), a multidomain PDZ-containing phosphoprotein. Phosphocycling controls the association between NHERF1 and the NPT2A. Here, we characterize the critical involvement of G protein-coupled receptor kinase 6A (GRK6A) in mediating PTH-sensitive phosphate transport by targeted phosphorylation coupled with NHERF1 conformational rearrangement, which in turn allows phosphorylation at a secondary site. GRK6A, through its carboxy-terminal PDZ recognition motif, binds NHERF1 PDZ1 with greater affinity than PDZ2. However, the association between NHERF1 PDZ2 and GRK6A is necessary for PTH action. Ser162, a PKCα phosphorylation site in PDZ2, regulates the binding affinity between PDZ2 and GRK6A. Substitution of Ser162 with alanine (S162A) blocks the PTH action but does not disrupt the interaction between NHERF1 and the NPT2A. Replacement of Ser162 with aspartic acid (S162D) abrogates the interaction between NHERF1 and the NPT2A and concurrently PTH action. We used amber codon suppression to generate a phosphorylated Ser162(pSer162)-PDZ2 variant. KD values determined by fluorescence anisotropy indicate that incorporation of pSer162 increased the binding affinity to the carboxy terminus of GRK6A 2-fold compared with WT PDZ2. Molecular dynamics simulations predict formation of an electrostatic network between pSer162 and Asp183 of PDZ2 and Arg at position -1 of the GRK6A PDZ-binding motif. Our results suggest that PDZ2 plays a regulatory role in PTH-sensitive NPT2A-mediated phosphate transport and phosphorylation of Ser162 in PDZ2 modulates the interaction with GRK6A.


Subject(s)
G-Protein-Coupled Receptor Kinases/metabolism , Phosphoproteins/metabolism , Sodium-Hydrogen Exchangers/metabolism , Biological Transport , Fibroblast Growth Factor-23 , Fibroblast Growth Factors/metabolism , G-Protein-Coupled Receptor Kinases/genetics , Humans , Ion Transport , Molecular Dynamics Simulation , PDZ Domains/genetics , Parathyroid Hormone/metabolism , Phosphates/metabolism , Phosphoproteins/genetics , Phosphorylation , Protein Binding , Protein Conformation , Sodium-Hydrogen Exchangers/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/genetics , Sodium-Phosphate Cotransporter Proteins, Type IIa/metabolism
4.
FASEB J ; 35(8): e21681, 2021 08.
Article in English | MEDLINE | ID: mdl-34196428

ABSTRACT

The sodium/iodide symporter (NIS) expresses at the basolateral plasma membrane of the thyroid follicular cell and mediates iodide accumulation required for normal thyroid hormonogenesis. Loss-of-function NIS variants cause congenital hypothyroidism due to impaired iodide accumulation in thyroid follicular cells underscoring the significance of NIS for thyroid physiology. Here we report novel findings derived from the thorough characterization of the nonsense NIS mutant p.R636* NIS-leading to a truncated protein missing the last eight amino acids-identified in twins with congenital hypothyroidism. R636* NIS is severely mislocalized into intracellular vesicular compartments due to the lack of a conserved carboxy-terminal type 1 PDZ-binding motif. As a result, R636* NIS is barely targeted to the plasma membrane and therefore iodide transport is reduced. Deletion of the PDZ-binding motif causes NIS accumulation into late endosomes and lysosomes. Using PDZ domain arrays, we revealed that the PDZ-domain containing protein SCRIB binds to the carboxy-terminus of NIS by a PDZ-PDZ interaction. Furthermore, in CRISPR/Cas9-based SCRIB deficient cells, NIS expression at the basolateral plasma membrane is compromised, leading to NIS localization into intracellular vesicular compartments. We conclude that the PDZ-binding motif is a plasma membrane retention signal that participates in the polarized expression of NIS by selectively interacting with the PDZ-domain containing protein SCRIB, thus retaining the transporter at the basolateral plasma membrane. Our data provide insights into the molecular mechanisms that regulate NIS expression at the plasma membrane, a topic of great interest in the thyroid cancer field considering the relevance of NIS-mediated radioactive iodide therapy for differentiated thyroid carcinoma.


Subject(s)
Membrane Proteins/metabolism , Symporters/metabolism , Tumor Suppressor Proteins/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Codon, Nonsense , Congenital Hypothyroidism/genetics , Congenital Hypothyroidism/metabolism , Conserved Sequence , Dogs , Endosomes/metabolism , HEK293 Cells , Humans , Lysosomes/metabolism , Madin Darby Canine Kidney Cells , Membrane Proteins/chemistry , Membrane Proteins/genetics , Models, Molecular , Mutagenesis, Site-Directed , PDZ Domains/genetics , Protein Structure, Secondary , Rats , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Symporters/chemistry , Symporters/genetics , Thyroid Gland/metabolism , Tumor Suppressor Proteins/chemistry , Tumor Suppressor Proteins/genetics
5.
Cell Microbiol ; 23(4): e13299, 2021 04.
Article in English | MEDLINE | ID: mdl-33277762

ABSTRACT

The Helicobacter pylori HtrA protein (HtrAHp ) is an important virulence factor involved in the infection process by proteolysis of components of the tight (claudin-8 and occludin) and adherens junctions (E-cadherin) between epithelial cells. As a protease and chaperone, HtrAHp is involved in protein quality control, which is particularly important under stress conditions. HtrAHp contains a protease domain and two C-terminal PDZ domains (PDZ1 and PDZ2). In the HtrA protein family, the PDZ domains are proposed to play important roles, including regulation of proteolytic activity. We therefore mutated the PDZ1 and PDZ2 domains in HtrAHp and studied the maintenance of proteolytic activity, assembly and rearrangement of the corresponding oligomeric forms. Our in vitro experiments demonstrated that at least PDZ1 is important for efficient substrate cleavage, while both PDZ domains are dispensable for the chaperone-like activity. However, in living H. pylori cells, only the mutant containing at least PDZ1, but not PDZ2, ensured bacterial growth under stressful conditions. Moreover, we can demonstrate that PDZ1 is crucial for HtrAHp oligomerization. Interestingly, all truncated proteolytically active HtrAHp variants were functional in the in vitro infection assay and caused damage to the E-cadherin-based adherens junctions. These findings provide valuable new insights into the function of HtrAHp in an important pathogen of humans.


Subject(s)
Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Helicobacter pylori/enzymology , Helicobacter pylori/genetics , Molecular Chaperones/metabolism , PDZ Domains/genetics , Serine Proteases/genetics , Serine Proteases/metabolism , Bacterial Proteins/chemistry , Helicobacter pylori/pathogenicity , Humans , Mutation , Protein Folding , Proteolysis , Serine Proteases/chemistry , Virulence Factors
6.
Exp Cell Res ; 408(2): 112865, 2021 11 15.
Article in English | MEDLINE | ID: mdl-34637763

ABSTRACT

Protein homeostasis (proteostasis) in multicellular organisms depends on the maintenance of force-bearing and force-generating cellular structures. Within myofibrillar Z-discs of striated muscle, isoforms of synaptopodin-2 (SYNPO2/myopodin) act as adapter proteins that are engaged in proteostasis of the actin-crosslinking protein filamin C (FLNc) under mechanical stress. SYNPO2 directly binds F-actin, FLNc and α-actinin and thus contributes to the architectural features of the actin cytoskeleton. By its association with autophagy mediating proteins, i.e. BAG3 and VPS18, SYNPO2 is also engaged in protein quality control and helps to target mechanical unfolded and damaged FLNc for degradation. Here we show that deficiency of all SYNPO2-isoforms in myotubes leads to decreased myofibrillar stability and deregulated autophagy under mechanical stress. In addition, isoform-specific proteostasis functions were revealed. The PDZ-domain containing variant SYNPO2b and the shorter, PDZ-less isoform SYNPO2e both localize to Z-discs. Yet, SYNPO2e is less stably associated with the Z-disc than SYNPO2b, and is dynamically transferred into FLNc-containing myofibrillar lesions under mechanical stress. SYNPO2e also recruits BAG3 into these lesions via interaction with the WW domain of BAG3. Our data provide evidence for a role of myofibrillar lesions as a transient quality control compartment essential to prevent and repair contraction-induced myofibril damage in muscle and indicate an important coordinating activity for SYNPO2 therein.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Apoptosis Regulatory Proteins/genetics , Microfilament Proteins/genetics , Muscle, Skeletal/metabolism , Stress, Mechanical , Vesicular Transport Proteins/genetics , Actin Cytoskeleton/genetics , Actinin/genetics , Actins/genetics , Animals , Autophagy/genetics , Cell Line , Cytoskeleton/genetics , Humans , Mice , Muscle Fibers, Skeletal/metabolism , Muscle, Striated/metabolism , Myofibrils/genetics , Myofibrils/metabolism , PDZ Domains/genetics , Protein Isoforms/genetics , Synaptophysin/genetics
7.
Mol Pharmacol ; 100(3): 271-282, 2021 09.
Article in English | MEDLINE | ID: mdl-34330822

ABSTRACT

G protein-coupled receptor 30 (GPR30) is a membrane receptor reported to bind 17ß-estradiol (E2) and mediate rapid nongenomic estrogen responses, hence also named G protein-coupled estrogen receptor. G-1 is a proposed GPR30-specific agonist that has been used to implicate the receptor in several pathophysiological events. However, controversy surrounds the role of GPR30 in G-1 and E2 responses. We investigated GPR30 activity in the absence and presence of G-1 and E2 in several eukaryotic systems ex vivo and in vitro in the absence and presence of the receptor. Ex vivo activity was addressed using the caudal artery from wild-type (WT) and GPR30 knockout (KO) mice, and in vitro activity was addressed using a HeLa cell line stably expressing a synthetic multifunctional promoter (nuclear factor κB, signal transducer and activator of transcription, activator protein 1)-luciferase construct (HFF11 cells) and a human GPR30-inducible T-REx system (T-REx HFF11 cells), HFF11 and human embryonic kidney 293 cells transiently expressing WT GPR30 and GPR30 lacking the C-terminal PDZ (postsynaptic density-95/discs-large /zonula occludens-1 homology) motif SSAV, and yeast Saccharomyces cerevisiae transformed to express GPR30. WT and KO arteries exhibited similar contractile responses to 60 mM KCl and 0.3 µM cirazoline, and G-1 relaxed both arteries with the same potency and efficacy. Furthermore, expression of GPR30 did not introduce any responses to 1 µM G-1 and 0.1 µM E2 in vitro. On the other hand, receptor expression caused considerable ligand-independent activity in vitro, which was receptor PDZ motif-dependent in mammalian cells. We conclude from these results that GPR30 exhibits ligand-independent activity in vitro but no G-1- or E2-stimulated activity in any of the systems used. SIGNIFICANCE STATEMENT: Much controversy surrounds 17ß-estradiol (E2) and G-1 as G protein-coupled receptor 30 (GPR30) agonists. We used several recombinant eukaryotic systems ex vivo and in vitro with and without GPR30 expression to address the role of this receptor in responses to these proposed agonists. Our results show that GPR30 exhibits considerable ligand-independent activity in vitro but no G-1- or E2-stimulated activity in any of the systems used. Thus, classifying GPR30 as an estrogen receptor and G-1 as a specific GPR30 agonist is unfounded.


Subject(s)
Cyclopentanes/pharmacology , Estradiol/pharmacology , Quinolines/pharmacology , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Arteries/drug effects , Cell Line , Disks Large Homolog 4 Protein/metabolism , Female , Humans , Ligands , Mice , Mice, Inbred C57BL , Mice, Knockout , Muscle Relaxation/drug effects , PDZ Domains/genetics , Receptors, Estrogen/drug effects , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/drug effects , Receptors, G-Protein-Coupled/genetics , Saccharomyces cerevisiae/genetics
8.
J Biol Chem ; 295(7): 1992-2000, 2020 02 14.
Article in English | MEDLINE | ID: mdl-31831623

ABSTRACT

Cell scaffolding and signaling are governed by protein-protein interactions. Although a particular interaction is often defined by two specific domains binding to each other, this interaction often occurs in the context of other domains in multidomain proteins. How such adjacent domains form supertertiary structures and modulate protein-protein interactions has only recently been addressed and is incompletely understood. The postsynaptic density protein PSD-95 contains a three-domain supramodule, denoted PSG, which consists of PDZ, Src homology 3 (SH3), and guanylate kinase-like domains. The PDZ domain binds to the C terminus of its proposed natural ligand, CXXC repeat-containing interactor of PDZ3 domain (CRIPT), and results from previous experiments using only the isolated PDZ domain are consistent with the simplest scenario for a protein-protein interaction; namely, a two-state mechanism. Here we analyzed the binding kinetics of the PSG supramodule with CRIPT. We show that PSG binds CRIPT via a more complex mechanism involving two conformational states interconverting on the second timescale. Both conformational states bound a CRIPT peptide with similar affinities but with different rates, and the distribution of the two conformational states was slightly shifted upon CRIPT binding. Our results are consistent with recent structural findings of conformational changes in PSD-95 and demonstrate how conformational transitions in supertertiary structures can shape the ligand-binding energy landscape and modulate protein-protein interactions.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Disks Large Homolog 4 Protein/genetics , Protein Domains/genetics , Protein Interaction Domains and Motifs/genetics , Amino Acid Sequence , Binding Sites , Disks Large Homolog 4 Protein/chemistry , Disks Large Homolog 4 Protein/ultrastructure , Guanylate Kinases/genetics , Humans , Kinetics , Ligands , Models, Molecular , Molecular Conformation , PDZ Domains/genetics , Protein Binding/genetics , Signal Transduction/genetics , src Homology Domains/genetics
9.
Biochem J ; 477(2): 407-429, 2020 01 31.
Article in English | MEDLINE | ID: mdl-31899476

ABSTRACT

Human HtrA3 (high-temperature requirement protease A3) is a trimeric multitasking propapoptotic serine protease associated with critical cellular functions and pathogenicity. Implicated in diseases including cancer and pre-eclampsia, its role as a tumor suppressor and potential therapeutic target cannot be ignored. Therefore, elucidating its mode of activation and regulatory switch becomes indispensable towards modulating its functions with desired effects for disease intervention. Using computational, biochemical and biophysical tools, we delineated the role of all domains, their combinations and the critical phenylalanine residues in regulating HtrA3 activity, oligomerization and specificity. Our findings underline the crucial roles of the N-terminus as well as the PDZ domain in oligomerization and formation of a catalytically competent enzyme, thus providing new insights into its structure-function coordination. Our study also reports an intricate ligand-induced allosteric switch, which redefines the existing hypothesis of HtrA3 activation besides opening up avenues for modulating protease activity favorably through suitable effector molecules.


Subject(s)
Protein Conformation , Serine Endopeptidases/genetics , Serine Proteases/genetics , Structure-Activity Relationship , Allosteric Regulation/genetics , Amino Acid Sequence/genetics , Catalysis , Gene Expression Regulation, Enzymologic/genetics , Humans , PDZ Domains/genetics , Protein Multimerization/genetics , Serine Endopeptidases/chemistry , Serine Endopeptidases/ultrastructure , Serine Proteases/chemistry , Serine Proteases/ultrastructure
10.
Proc Natl Acad Sci U S A ; 114(2): 370-375, 2017 01 10.
Article in English | MEDLINE | ID: mdl-28011764

ABSTRACT

Glioblastoma multiforme (GBM) is an intractable tumor despite therapeutic advances, principally because of its invasive properties. Radiation is a staple in therapeutic regimens, although cells surviving radiation can become more aggressive and invasive. Subtraction hybridization identified melanoma differentiation-associated gene 9 [MDA-9/Syntenin; syndecan-binding protein (SDCBP)] as a differentially regulated gene associated with aggressive cancer phenotypes in melanoma. MDA-9/Syntenin, a highly conserved double-PDZ domain-containing scaffolding protein, is robustly expressed in human-derived GBM cell lines and patient samples, with expression increasing with tumor grade and correlating with shorter survival times and poorer response to radiotherapy. Knockdown of MDA-9/Syntenin sensitizes GBM cells to radiation, reducing postradiation invasion gains. Radiation induces Src and EGFRvIII signaling, which is abrogated through MDA-9/Syntenin down-regulation. A specific inhibitor of MDA-9/Syntenin activity, PDZ1i (113B7), identified through NMR-guided fragment-based drug design, inhibited MDA-9/Syntenin binding to EGFRvIII, which increased following radiation. Both genetic (shmda-9) and pharmacological (PDZ1i) targeting of MDA-9/Syntenin reduced invasion gains in GBM cells following radiation. Although not affecting normal astrocyte survival when combined with radiation, PDZ1i radiosensitized GBM cells. PDZ1i inhibited crucial GBM signaling involving FAK and mutant EGFR, EGFRvIII, and abrogated gains in secreted proteases, MMP-2 and MMP-9, following radiation. In an in vivo glioma model, PDZ1i resulted in smaller, less invasive tumors and enhanced survival. When combined with radiation, survival gains exceeded radiotherapy alone. MDA-9/Syntenin (SDCBP) provides a direct target for therapy of aggressive cancers such as GBM, and defined small-molecule inhibitors such as PDZ1i hold promise to advance targeted brain cancer therapy.


Subject(s)
Glioblastoma/genetics , Neoplasm Invasiveness/genetics , Syntenins/genetics , Animals , Brain Neoplasms/genetics , Cell Line, Tumor , Cell Movement/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Glioma/genetics , Humans , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Melanoma/genetics , Mice , Mice, Nude , PDZ Domains/genetics , Signal Transduction/genetics , src-Family Kinases/genetics
11.
Int J Mol Sci ; 21(21)2020 Nov 06.
Article in English | MEDLINE | ID: mdl-33172212

ABSTRACT

PDZ domains are abundant interaction hubs found in a number of different proteins and they exhibit characteristic differences in their structure and ligand specificity. Their internal dynamics have been proposed to contribute to their biological activity via changes in conformational entropy upon ligand binding and allosteric modulation. Here we investigate dynamic structural ensembles of PDZ3 of the postsynaptic protein PSD-95, calculated based on previously published backbone and side-chain S2 order parameters. We show that there are distinct but interdependent structural rearrangements in PDZ3 upon ligand binding and the presence of the intramolecular allosteric modulator helix α3. We have also compared these rearrangements in PDZ1-2 of PSD-95 and the conformational diversity of an extended set of PDZ domains available in the PDB database. We conclude that although the opening-closing rearrangement, occurring upon ligand binding, is likely a general feature for all PDZ domains, the conformer redistribution upon ligand binding along this mode is domain-dependent. Our findings suggest that the structural and functional diversity of PDZ domains is accompanied by a diversity of internal motional modes and their interdependence.


Subject(s)
Disks Large Homolog 4 Protein/metabolism , PDZ Domains/genetics , PDZ Domains/physiology , Amino Acid Sequence/genetics , Animals , Binding Sites/genetics , Disks Large Homolog 4 Protein/genetics , Disks Large Homolog 4 Protein/ultrastructure , Entropy , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Ligands , Membrane Proteins/metabolism , Models, Molecular , Molecular Conformation , Molecular Dynamics Simulation , Protein Binding/genetics
12.
Glia ; 67(5): 884-895, 2019 05.
Article in English | MEDLINE | ID: mdl-30585357

ABSTRACT

Interactions between axons and Schwann cells are essential for the acquisition of Schwann cell radial and longitudinal polarity and myelin sheath assembly. In the internode, the largest of these longitudinal domains, axon-Schwann cell interactions are mediated by the Nectin-like (Necl) cell adhesion proteins, also known as SynCAMs or Cadms. In particular, Necl-1/Cadm3 expressed on the axon surface binds to Necl-4/Cadm4 expressed along the adaxonal membrane of myelinating Schwann cells. Necl-4 promotes myelination in vitro and is required for the timely onset of myelination and the fidelity of the organization of the myelin sheath and the internode in vivo. A key question is the identity of the downstream effectors of Necl-4 that mediate its effects. The cytoplasmic terminal region (CTR) of Necl-4 contains a PDZ-domain binding motif. Accordingly, we used the CTR of Necl-4 in an unbiased proteomic screen of PDZ-domain proteins. We identify Par-3, a multi-PDZ domain containing protein of the Par-aPKC polarity complex previously implicated in myelination, as an interacting protein. Necl-4 and Par-3 are colocalized along the inner Schwann cell membrane and coprecipitate from Schwann cell lysates. The CTR of Necl-4 binds to the first PDZ domain of Par-3 thereby recruiting Par-3 to sites of Necl-4/Necl-1 interaction. Knockdown of Necl-4 perturbs Par-3 localization to the inner membrane of Schwann cells in myelinating co-cultures. These findings implicate interactions of Necl-1/Necl-4 in the recruitment of Par-3 to the Schwann cell adaxonal membrane and the establishment of Schwann cell radial polarity.


Subject(s)
Axons/metabolism , Cell Adhesion Molecules/metabolism , Cell Membrane/metabolism , Immunoglobulins/metabolism , PDZ Domains/physiology , Schwann Cells/cytology , Adaptor Proteins, Signal Transducing , Animals , CHO Cells , Cell Adhesion Molecules/genetics , Cell Cycle Proteins , Cell Membrane/genetics , Coculture Techniques , Cricetulus , Embryo, Mammalian , Ganglia, Spinal/cytology , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , Immunoglobulins/genetics , Immunoprecipitation , In Vitro Techniques , Mice , Neurons , PDZ Domains/genetics , Rats , Sciatic Nerve/cytology , Transfection
13.
Hum Mol Genet ; 26(3): 624-636, 2017 02 01.
Article in English | MEDLINE | ID: mdl-28031293

ABSTRACT

Usher syndrome (USH) is the most common cause of inherited deaf-blindness, manifested as USH1, USH2 and USH3 clinical types. The protein products of USH2 causative and modifier genes, USH2A, ADGRV1, WHRN and PDZD7, interact to assemble a multiprotein complex at the ankle link region of the mechanosensitive stereociliary bundle in hair cells. Defects in this complex cause stereociliary bundle disorganization and hearing loss. The four USH2 proteins also interact in vitro with USH1 proteins including myosin VIIa, USH1G (SANS), CIB2 and harmonin. However, it is unclear whether the interactions between USH1 and USH2 proteins occur in vivo and whether USH1 proteins play a role in USH2 complex assembly in hair cells. In this study, we identified a novel interaction between myosin VIIa and PDZD7 by FLAG pull-down assay. We further investigated the role of the above-mentioned four USH1 proteins in the cochlear USH2 complex assembly using USH1 mutant mice. We showed that only myosin VIIa is indispensable for USH2 complex assembly at ankle links, indicating the potential transport and/or anchoring role of myosin VIIa for USH2 proteins in hair cells. However, myosin VIIa is not required for USH2 complex assembly in photoreceptors. We further showed that, while PDZ protein harmonin is not involved, its paralogous USH2 proteins, PDZD7 and whirlin, function synergistically in USH2 complex assembly in cochlear hair cells. In summary, our studies provide novel insight into the functional relationship between USH1 and USH2 proteins in the cochlea and the retina as well as the disease mechanisms underlying USH1 and USH2.


Subject(s)
Carrier Proteins/genetics , Extracellular Matrix Proteins/genetics , Myosins/genetics , Usher Syndromes/genetics , Animals , Carrier Proteins/chemistry , Cell Cycle Proteins , Cytoskeletal Proteins , Extracellular Matrix Proteins/chemistry , Hair Cells, Auditory/pathology , Humans , Mice , Multiprotein Complexes/chemistry , Multiprotein Complexes/genetics , Myosin VIIa , Myosins/chemistry , PDZ Domains/genetics , Retina/metabolism , Retina/pathology , Stereocilia/genetics , Stereocilia/metabolism , Stereocilia/pathology , Usher Syndromes/pathology
14.
Mol Syst Biol ; 14(8): e8129, 2018 08 20.
Article in English | MEDLINE | ID: mdl-30126976

ABSTRACT

A key function of reversible protein phosphorylation is to regulate protein-protein interactions, many of which involve short linear motifs (3-12 amino acids). Motif-based interactions are difficult to capture because of their often low-to-moderate affinities. Here, we describe phosphomimetic proteomic peptide-phage display, a powerful method for simultaneously finding motif-based interaction and pinpointing phosphorylation switches. We computationally designed an oligonucleotide library encoding human C-terminal peptides containing known or predicted Ser/Thr phosphosites and phosphomimetic variants thereof. We incorporated these oligonucleotides into a phage library and screened the PDZ (PSD-95/Dlg/ZO-1) domains of Scribble and DLG1 for interactions potentially enabled or disabled by ligand phosphorylation. We identified known and novel binders and characterized selected interactions through microscale thermophoresis, isothermal titration calorimetry, and NMR We uncover site-specific phospho-regulation of PDZ domain interactions, provide a structural framework for how PDZ domains accomplish phosphopeptide binding, and discuss ligand phosphorylation as a switching mechanism of PDZ domain interactions. The approach is readily scalable and can be used to explore the potential phospho-regulation of motif-based interactions on a large scale.


Subject(s)
PDZ Domains/genetics , Peptides/genetics , Protein Interaction Maps/genetics , Proteome/genetics , Amino Acid Sequence/genetics , Binding Sites , Disks Large Homolog 4 Protein/genetics , Humans , Ligands , Oligonucleotides/genetics , Peptide Library , Phosphorylation , Protein Binding/genetics , Protein Interaction Mapping , Zonula Occludens-1 Protein/genetics
15.
Proc Natl Acad Sci U S A ; 113(22): E3081-90, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27185935

ABSTRACT

Shank and SAPAP (synapse-associated protein 90/postsynaptic density-95-associated protein) are two highly abundant scaffold proteins that directly interact with each other to regulate excitatory synapse development and plasticity. Mutations of SAPAP, but not other reported Shank PDZ domain binders, share a significant overlap on behavioral abnormalities with the mutations of Shank both in patients and in animal models. The molecular mechanism governing the exquisite specificity of the Shank/SAPAP interaction is not clear, however. Here we report that a sequence preceding the canonical PDZ domain of Shank, together with the elongated PDZ BC loop, form another binding site for a sequence upstream of the SAPAP PDZ-binding motif, leading to a several hundred-fold increase in the affinity of the Shank/SAPAP interaction. We provide evidence that the specific interaction afforded by this newly identified site is required for Shank synaptic targeting and the Shank-induced synaptic activity increase. Our study provides a molecular explanation of how Shank and SAPAP dosage changes due to their gene copy number variations can contribute to different psychiatric disorders.


Subject(s)
Hippocampus/metabolism , Nerve Tissue Proteins/metabolism , Neurons/metabolism , PDZ Domains/genetics , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , DNA Copy Number Variations , Female , Hippocampus/cytology , Humans , Immunoprecipitation , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Neurons/cytology , Protein Binding , Protein Conformation , SAP90-PSD95 Associated Proteins , Synapses/physiology
16.
PLoS Genet ; 12(10): e1006400, 2016 Oct.
Article in English | MEDLINE | ID: mdl-27783625

ABSTRACT

Z-discs are organizing centers that establish and maintain myofibril structure and function. Important Z-disc proteins are α-actinin, which cross-links actin thin filaments at the Z-disc and Zasp PDZ domain proteins, which directly interact with α-actinin. Here we investigate the biochemical and genetic nature of this interaction in more detail. Zasp52 is the major Drosophila Zasp PDZ domain protein, and is required for myofibril assembly and maintenance. We show by in vitro biochemistry that the PDZ domain plus a C-terminal extension is the only area of Zasp52 involved in the interaction with α-actinin. In addition, site-directed mutagenesis of 5 amino acid residues in the N-terminal part of the PDZ domain, within the PWGFRL motif, abolish binding to α-actinin, demonstrating the importance of this motif for α-actinin binding. Rescue assays of a novel Zasp52 allele demonstrate the crucial importance of the PDZ domain for Zasp52 function. Flight assays also show that a Zasp52 mutant suppresses the α-actinin mutant phenotype, indicating that both proteins are core structural Z-disc proteins required for optimal Z-disc function.


Subject(s)
Actinin/genetics , Drosophila Proteins/genetics , LIM Domain Proteins/genetics , Muscle Proteins/genetics , Myofibrils/genetics , Actin Cytoskeleton/genetics , Actin Cytoskeleton/metabolism , Actinin/metabolism , Amino Acid Motifs/genetics , Animals , Binding Sites , Carrier Proteins , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Flight, Animal , LIM Domain Proteins/metabolism , Muscle Proteins/metabolism , Muscle, Skeletal/metabolism , Myofibrils/metabolism , PDZ Domains/genetics , Protein Binding , Sarcomeres/genetics , Sarcomeres/metabolism
17.
Hum Genet ; 137(3): 231-246, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29426960

ABSTRACT

Deletion and truncation mutations in the X-linked gene CASK are associated with severe intellectual disability (ID), microcephaly and pontine and cerebellar hypoplasia in girls (MICPCH). The molecular origin of CASK-linked MICPCH is presumed to be due to disruption of the CASK-Tbr-1 interaction. This hypothesis, however, has not been directly tested. Missense variants in CASK are typically asymptomatic in girls. We report three severely affected girls with heterozygous CASK missense mutations (M519T (2), G659D (1)) who exhibit ID, microcephaly, and hindbrain hypoplasia. The mutation M519T results in the replacement of an evolutionarily invariant methionine located in the PDZ signaling domain known to be critical for the CASK-neurexin interaction. CASKM519T is incapable of binding to neurexin, suggesting a critically important role for the CASK-neurexin interaction. The mutation G659D is in the SH3 (Src homology 3) domain of CASK, replacing a semi-conserved glycine with aspartate. We demonstrate that the CASKG659D mutation affects the CASK protein in two independent ways: (1) it increases the protein's propensity to aggregate; and (2) it disrupts the interface between CASK's PDZ (PSD95, Dlg, ZO-1) and SH3 domains, inhibiting the CASK-neurexin interaction despite residing outside of the domain deemed critical for neurexin interaction. Since heterozygosity of other aggregation-inducing mutations (e.g., CASKW919R) does not produce MICPCH, we suggest that the G659D mutation produces microcephaly by disrupting the CASK-neurexin interaction. Our results suggest that disruption of the CASK-neurexin interaction, not the CASK-Tbr-1 interaction, produces microcephaly and cerebellar hypoplasia. These findings underscore the importance of functional validation for variant classification.


Subject(s)
Cell Adhesion Molecules, Neuronal/genetics , Cerebellum/abnormalities , Genetic Diseases, X-Linked/genetics , Guanylate Kinases/genetics , Microcephaly/genetics , Nerve Tissue Proteins/genetics , Nervous System Malformations/genetics , Calcium-Binding Proteins , Cell Adhesion Molecules, Neuronal/chemistry , Cerebellum/diagnostic imaging , Cerebellum/physiopathology , Child , Child, Preschool , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Genetic Diseases, X-Linked/physiopathology , Guanylate Kinases/chemistry , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Microcephaly/diagnostic imaging , Microcephaly/physiopathology , Mutation, Missense/genetics , Nerve Tissue Proteins/chemistry , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/physiopathology , Neural Cell Adhesion Molecules , PDZ Domains/genetics , Phenotype , Protein Aggregates/genetics , Protein Binding , Protein Interaction Maps/genetics , T-Box Domain Proteins/genetics , src Homology Domains/genetics
18.
J Mol Recognit ; 31(6): e2698, 2018 06.
Article in English | MEDLINE | ID: mdl-29266444

ABSTRACT

The human HtrA family of serine proteases (HtrA1, HtrA2, HtrA3, and HtrA4) are the key enzymes associated with pregnancy and closely related to the development and progression of many pathological events. Previously, it was found that halogen substitution at the indole moiety of peptide Trp-1 residue can form a geometrically satisfactory halogen bond with the Drosophila discs large, zona occludens-1 (PDZ) domain of HtrA proteases. Here, we attempt to systematically investigate the effect of substitution with 4 halogen types and 2 indole positions on the binding affinity and specificity of peptide ligands to the 4 HtrA PDZ domains. The complex structures, interaction energies, halogen-bonding strength, and binding affinity of domain-peptide systems were modeled, analyzed, and measured via computational modeling and fluorescence-based assay. It is revealed that there is a compromise between the local rearrangement of halogen bond involving different halogen atoms and the global optimization of domain-peptide interaction; the substitution position is fundamentally important for peptide-binding affinity, while the halogen type can effectively shift peptide selectivity between the 4 domains. The HtrA1-PDZ and HtrA4-PDZ as well as HtrA2-PDZ and HtrA3-PDZ respond similarly to different halogen substitutions of peptide; -Br substitution at R2-position and -I substitution at R4-position are most effective in improving peptide selectivity for HtrA1-PDZ/HtrA4-PDZ and HtrA2-PDZ/HtrA3-PDZ, respectively; -F substitution would not address substantial effect on peptide selectivity for all the 4 domains. Consequently, the binding affinities of a native peptide ligand DSRIWWV-COOH as well as its 4 R2-halogenated counterparts were determined as 1.9, 1.4, 0.5, 0.27, and 0.92 µM, which are basically consistent with computational analysis. This study would help to rationally design selective peptide inhibitors of HtrA family members by using different halogen substitutions.


Subject(s)
Peptides/genetics , Crystallography, X-Ray , Female , Halogens/chemistry , High-Temperature Requirement A Serine Peptidase 1/chemistry , High-Temperature Requirement A Serine Peptidase 1/genetics , High-Temperature Requirement A Serine Peptidase 2/chemistry , High-Temperature Requirement A Serine Peptidase 2/genetics , Humans , Indoles/chemistry , Ligands , PDZ Domains/genetics , Peptides/chemistry , Pregnancy , Protein Binding , Quantum Theory , Serine Endopeptidases/chemistry , Serine Endopeptidases/genetics , Serine Proteases/chemistry , Serine Proteases/genetics
19.
BMC Med Genet ; 19(1): 34, 2018 03 02.
Article in English | MEDLINE | ID: mdl-29499638

ABSTRACT

BACKGROUND: Congenital hydrocephalus (CH) results from the accumulation of excessive amounts of cerebrospinal fluid (CSF) in the brain, often leading to severe neurological impairments. However, the adverse effects of CH can be reduced if the condition is detected and treated early. Earlier reports demonstrated that some CH cases are caused by mutations in L1CAM gene encoding the neural cell adhesion molecule L1. On the other hand, recent studies have implicated the multiple PDZ domain (MPDZ) gene in some severe forms of CH, inherited in an autosomal recessive pattern. METHODS: In this study, whole-exome and Sanger sequencing were performed on a 9 months old Emirati child clinically diagnosed by CH. In addition, in silico, cellular, and molecular assays have been conducted to confirm pathogenicity of the identified variants and to establish disease mechanism. RESULTS: Whole exome sequencing revealed two compound heterozygous novel variants (c.394G > A and c.1744C > G) in the affected child within the MPDZ gene. Segregation analysis revealed that each of the parents is heterozygous for one of the two variants and therefore passed that variant to their child. The outcome of the in silico and bioinformatics analyses came in line with the experimental data, suggesting that the two variants are most likely disease causing. CONCLUSIONS: The compound heterozygous variants identified in this study are the most likely cause of CH in the affected child. The study further confirms MPDZ as a gene underlying some CH cases.


Subject(s)
Heterozygote , Hydrocephalus/diagnostic imaging , Hydrocephalus/genetics , Neural Cell Adhesion Molecule L1/genetics , PDZ Domains/genetics , Amino Acid Sequence , Brain/metabolism , Cell Adhesion , Genes, Recessive , Genetic Variation , HEK293 Cells , HeLa Cells , Humans , Infant , Male , Mutation , Neurons/cytology , Neurons/drug effects , Pedigree , Protein Conformation , Sequence Analysis, DNA , Exome Sequencing
20.
Nature ; 491(7422): 138-42, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-23041932

ABSTRACT

Statistical analysis of protein evolution suggests a design for natural proteins in which sparse networks of coevolving amino acids (termed sectors) comprise the essence of three-dimensional structure and function. However, proteins are also subject to pressures deriving from the dynamics of the evolutionary process itself--the ability to tolerate mutation and to be adaptive to changing selection pressures. To understand the relationship of the sector architecture to these properties, we developed a high-throughput quantitative method for a comprehensive single-mutation study in which every position is substituted individually to every other amino acid. Using a PDZ domain (PSD95(pdz3)) model system, we show that sector positions are functionally sensitive to mutation, whereas non-sector positions are more tolerant to substitution. In addition, we find that adaptation to a new binding specificity initiates exclusively through variation within sector residues. A combination of just two sector mutations located near and away from the ligand-binding site suffices to switch the binding specificity of PSD95(pdz3) quantitatively towards a class-switching ligand. The localization of functional constraint and adaptive variation within the sector has important implications for understanding and engineering proteins.


Subject(s)
Adaptation, Physiological , Amino Acid Substitution , Mutant Proteins/chemistry , PDZ Domains/genetics , PDZ Domains/physiology , Proteins/chemistry , Proteins/metabolism , Adaptation, Physiological/genetics , Adaptation, Physiological/physiology , Amino Acid Sequence , Binding Sites/genetics , Evolution, Molecular , Ligands , Models, Molecular , Molecular Sequence Data , Mutant Proteins/genetics , Mutant Proteins/metabolism , Mutation , Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL