Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 294
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Am Chem Soc ; 146(20): 13709-13713, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38738955

ABSTRACT

G-Quadruplex (G4) structures formed by guanine-rich DNA and RNA sequences are implicated in various biological processes. Understanding the mechanisms by which proteins recognize G4 structures is crucial for elucidating their functional roles. Here we present the X-ray crystal structure of an ankyrin protein bound to a parallel G4 structure. Our findings reveal a new specific recognition mode in which a bundle of α-helices and loops of the ankyrin form a flat surface to stack on the G-tetrad core. The protein employs a combination of hydrogen bonds and hydrophobic contacts to interact with the G4, and electrostatic interaction is used to enhance the binding affinity. This binding mechanism provides valuable insights into understanding G4 recognition by proteins.


Subject(s)
Ankyrins , G-Quadruplexes , Models, Molecular , Ankyrins/chemistry , Crystallography, X-Ray , Humans , Protein Binding , Hydrogen Bonding
2.
Biochem Biophys Res Commun ; 733: 150573, 2024 Nov 12.
Article in English | MEDLINE | ID: mdl-39208644

ABSTRACT

In bacteria, the ankyrin-like protein AnkB helps overcome stress by regulating catalase activity when expressed under stressful conditions. As the structural properties of AnkB are largely unexplored, our understanding of various AnkB-mediated functions in bacteria remains limited. In the present study, we describe the structure of AnkB from Acinetobacter baumannii, hereafter referred to as "AbAnkB," which has a unique tertiary configuration compared with that of other ankyrin domain-containing proteins. Structural analysis revealed that AbAnkB has a relatively long loop between AKR3 and AKR4 and an oppositely positioned α8 helix. Based on amino acid conservation and protein surface analyses, we identified a hydrophobic patch that might be critical for the function of AbAnkB. To the best of our knowledge, our study is the first to report the structure of a bacterial AnkB protein; our findings will markedly enhance our understanding of its functions in bacteria.


Subject(s)
Acinetobacter baumannii , Ankyrins , Bacterial Proteins , Acinetobacter baumannii/metabolism , Acinetobacter baumannii/chemistry , Bacterial Proteins/metabolism , Bacterial Proteins/chemistry , Ankyrins/metabolism , Ankyrins/chemistry , Models, Molecular , Amino Acid Sequence , Protein Conformation
3.
J Biol Chem ; 298(9): 102272, 2022 09.
Article in English | MEDLINE | ID: mdl-35850303

ABSTRACT

The axon initial segment (AIS) has characteristically dense clustering of voltage-gated sodium channels (Nav), cell adhesion molecule Neurofascin 186 (Nfasc), and neuronal scaffold protein Ankyrin-G (AnkG) in neurons, which facilitates generation of an action potential and maintenance of axonal polarity. However, the mechanisms underlying AIS assembly, maintenance, and plasticity remain poorly understood. Here, we report the high-resolution crystal structure of the AnkG ankyrin repeat (ANK repeat) domain in complex with its binding site in the Nfasc cytoplasmic tail that shows, in conjunction with binding affinity assays with serial truncation variants, the molecular basis of AnkG-Nfasc binding. We confirm AnkG interacts with the FIGQY motif in Nfasc, and we identify another region required for their high affinity binding. Our structural analysis revealed that ANK repeats form 4 hydrophobic or hydrophilic layers in the AnkG inner groove that coordinate interactions with essential Nfasc residues, including F1202, E1204, and Y1212. Moreover, we show disruption of the AnkG-Nfasc complex abolishes Nfasc enrichment at the AIS in cultured mouse hippocampal neurons. Finally, our structural and biochemical analysis indicated that L1 syndrome-associated mutations in L1CAM, a member of the L1 immunoglobulin family proteins including Nfasc, L1CAM, NrCAM, and CHL1, compromise binding with ankyrins. Taken together, these results define the mechanisms underlying AnkG-Nfasc complex formation and show that AnkG-dependent clustering of Nfasc is required for AIS integrity.


Subject(s)
Ankyrin Repeat , Ankyrins , Axon Initial Segment , Cell Adhesion Molecules , Nerve Growth Factors , Animals , Ankyrins/chemistry , Axon Initial Segment/chemistry , Binding Sites , Cell Adhesion Molecules/chemistry , Cell Adhesion Molecules/genetics , Crystallography, X-Ray , Mice , Nerve Growth Factors/chemistry , Nerve Growth Factors/genetics , Neural Cell Adhesion Molecule L1/genetics , Neural Cell Adhesion Molecule L1/metabolism , Protein Domains
4.
Cell ; 135(7): 1189-200, 2008 Dec 26.
Article in English | MEDLINE | ID: mdl-19109891

ABSTRACT

beta-dystroglycan (DG) and the dystrophin-glycoprotein complex (DGC) are localized at costameres and neuromuscular junctions in the sarcolemma of skeletal muscle. We present evidence for an ankyrin-based mechanism for sarcolemmal localization of dystrophin and beta-DG. Dystrophin binds ankyrin-B and ankyrin-G, while beta-DG binds ankyrin-G. Dystrophin and beta-DG require ankyrin-G for retention at costameres but not delivery to the sarcolemma. Dystrophin and beta-DG remain intracellular in ankyrin-B-depleted muscle, where beta-DG accumulates in a juxta-TGN compartment. The neuromuscular junction requires ankyrin-B for localization of dystrophin/utrophin and beta-DG and for maintenance of its postnatal morphology. A Becker muscular dystrophy mutation reduces ankyrin binding and impairs sarcolemmal localization of dystrophin-Dp71. Ankyrin-B also binds to dynactin-4, a dynactin subunit. Dynactin-4 and a subset of microtubules disappear from sarcolemmal sites in ankyrin-B-depleted muscle. Ankyrin-B thus is an adaptor required for sarcolemmal localization of dystrophin, as well as dynactin-4.


Subject(s)
Ankyrins/metabolism , Costameres/metabolism , Dystroglycans/metabolism , Dystrophin/metabolism , Neuromuscular Junction/metabolism , Amino Acid Sequence , Animals , Ankyrins/chemistry , Ankyrins/genetics , Dynactin Complex , Dystrophin/genetics , Mice , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Molecular Sequence Data , Muscle, Skeletal/metabolism , Muscular Dystrophy, Duchenne/metabolism , Sarcolemma/metabolism , Sequence Alignment
5.
Proc Natl Acad Sci U S A ; 117(2): 1207-1215, 2020 01 14.
Article in English | MEDLINE | ID: mdl-31889000

ABSTRACT

Ankyrin-G (AnkG), a highly enriched scaffold protein in the axon initial segment (AIS) of neurons, functions to maintain axonal polarity and the integrity of the AIS. At the AIS, AnkG regulates selective intracellular cargo trafficking between soma and axons via interaction with the dynein regulator protein Ndel1, but the molecular mechanism underlying this binding remains elusive. Here we report that Ndel1's C-terminal coiled-coil region (CT-CC) binds to giant neuron-specific insertion regions present in both AnkG and AnkB with 2:1 stoichiometry. The high-resolution crystal structure of AnkB in complex with Ndel1 CT-CC revealed the detailed molecular basis governing the AnkB/Ndel1 complex formation. Mechanistically, AnkB binds with Ndel1 by forming a stable 5-helix bundle dominated by hydrophobic interactions spread across 6 distinct interaction layers. Moreover, we found that AnkG is essential for Ndel1 accumulation at the AIS. Finally, we found that cargo sorting at the AIS can be disrupted by blocking the AnkG/Ndel1 complex formation using a peptide designed based on our structural data. Collectively, the atomic structure of the AnkB/Ndel1 complex together with studies of cargo sorting through the AIS establish the mechanistic basis for AnkG/Ndel1 complex formation and for the maintenance of axonal polarity. Our study will also be valuable for future studies of the interaction between AnkB and Ndel1 perhaps at distal axonal cargo transport.


Subject(s)
Ankyrins/metabolism , Carrier Proteins/metabolism , Cell Polarity/physiology , Dyneins/metabolism , Neurons/metabolism , Ankyrins/chemistry , Axon Initial Segment , Axons/metabolism , Carrier Proteins/chemistry , Dyneins/chemistry , Microtubule-Associated Proteins , Models, Molecular , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Protein Transport
6.
J Mol Cell Cardiol ; 168: 44-57, 2022 07.
Article in English | MEDLINE | ID: mdl-35447147

ABSTRACT

Obscurin is a large scaffolding protein in striated muscle that maintains sarcolemmal integrity and aligns the sarcoplasmic reticulum with the underlying contractile machinery. Ankyrins are a family of adaptor proteins with some isoforms that interact with obscurin. Previous studies have examined obscurin interacting with individual ankyrins. In this study, we demonstrate that two different ankyrins interact with obscurin's carboxyl terminus via independent ankyrin-binding domains (ABDs). Using in-vitro binding assays, co-precipitation assays, and FLIM-FRET analysis, we show that obscurin interacts with small ankyrin 1.5 (sAnk1.5) and the muscle-specific ankyrin-G isoform (AnkG107). While there is no direct interaction between sAnk1.5 and AnkG107, obscurin connects the two ankyrins both in vitro and in cells. Moreover, AnkG107 recruits ß-spectrin to this macromolecular protein complex and mutating obscurin's ABDs disrupts complex formation. To further characterize AnkG107 interaction with obscurin, we measure obscurin-binding to different AnkG107 isoforms expressed in the heart and find that the first obscurin-binding domain in AnkG107 principally mediates this interaction. We also find that AnkG107 does not bind to filamin-C and displays minimal binding to plectin-1 compared to obscurin. Finally, both sAnk1.5-GFP and AnkG107-CTD-RFP are targeted to the M-lines of ventricular cardiomyocytes and mutating their obscurin-binding domains disrupts the M-line localization of these ankyrin constructs. Altogether, these findings support a model in which obscurin can interact via independent binding domains with two different ankyrin protein complexes to target them to the sarcomeric M-line of ventricular cardiomyocytes.


Subject(s)
Ankyrins , Muscle Proteins , Ankyrins/chemistry , Muscle Proteins/metabolism , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases , Rho Guanine Nucleotide Exchange Factors/genetics
7.
Cell Commun Signal ; 20(1): 66, 2022 05 18.
Article in English | MEDLINE | ID: mdl-35585601

ABSTRACT

Notch signalling pathway plays a key role in metazoan biology by contributing to resolution of binary decisions in the life cycle of cells during development. Outcomes such as proliferation/differentiation dichotomy are resolved by transcriptional remodelling that follows a switch from Notchon to Notchoff state, characterised by dissociation of Notch intracellular domain (NICD) from DNA-bound RBPJ. Here we provide evidence that transitioning to the Notchoff state is regulated by heat flux, a phenomenon that aligns resolution of fate dichotomies to mitochondrial activity. A combination of phylogenetic analysis and computational biochemistry was utilised to disclose structural adaptations of Notch1 ankyrin domain that enabled function as a sensor of heat flux. We then employed DNA-based micro-thermography to measure heat flux during brain development, followed by analysis in vitro of the temperature-dependent behaviour of Notch1 in mouse neural progenitor cells. The structural capacity of NICD to operate as a thermodynamic sensor in metazoans stems from characteristic enrichment of charged acidic amino acids in ß-hairpins of the ankyrin domain that amplify destabilising inter-residue electrostatic interactions and render the domain thermolabile. The instability emerges upon mitochondrial activity which raises the perinuclear and nuclear temperatures to 50 °C and 39 °C, respectively, leading to destabilization of Notch1 transcriptional complex and transitioning to the Notchoff state. Notch1 functions a metazoan thermodynamic sensor that is switched on by intercellular contacts, inputs heat flux as a proxy for mitochondrial activity in the Notchon state via the ankyrin domain and is eventually switched off in a temperature-dependent manner. Video abstract.


Subject(s)
Ankyrins , Neural Stem Cells , Receptors, Notch , Animals , Ankyrins/chemistry , Ankyrins/metabolism , Mice , Neural Stem Cells/chemistry , Neural Stem Cells/metabolism , Phylogeny , Protein Domains , Receptors, Notch/chemistry , Receptors, Notch/metabolism , Signal Transduction , Thermodynamics
8.
Molecules ; 27(2)2022 Jan 10.
Article in English | MEDLINE | ID: mdl-35056738

ABSTRACT

Ankyrin is one of the most abundant protein repeat families found across all forms of life. It is found in a variety of multi-domain and single domain proteins in humans with diverse number of repeating units. They are observed to occur in several functionally diverse proteins, such as transcriptional initiators, cell cycle regulators, cytoskeletal organizers, ion transporters, signal transducers, developmental regulators, and toxins, and, consequently, defects in ankyrin repeat proteins have been associated with a number of human diseases. In this study, we have classified the human ankyrin proteins into clusters based on the sequence similarity in their ankyrin repeat domains. We analyzed the amino acid compositional bias and consensus ankyrin motif sequence of the clusters to understand the diversity of the human ankyrin proteins. We carried out network-based structural analysis of human ankyrin proteins across different clusters and showed the association of conserved residues with topologically important residues identified by network centrality measures. The analysis of conserved and structurally important residues helps in understanding their role in structural stability and function of these proteins. In this paper, we also discuss the significance of these conserved residues in disease association across the human ankyrin protein clusters.


Subject(s)
Ankyrin Repeat , Ankyrins/chemistry , Databases, Protein , Humans
9.
Parasitol Res ; 120(3): 1067-1076, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33515065

ABSTRACT

Giardia intestinalis is a human parasite that causes a diarrheal disease in developing countries. G. intestinalis has a cytoskeleton (CSK) composed of microtubules and microfilaments, and the Giardia genome does not code for the canonical CSK-binding proteins described in other eukaryotic cells. To identify candidate actin and tubulin cross-linking proteins, we performed a BLAST analysis of the Giardia genome using a spectraplakins consensus sequence as a query. Based on the highest BLAST score, we selected a 259-kDa sequence designated as a cytoskeleton linker protein (CLP259). The sequence was cloned in three fragments and characterized by immunoprecipitation, confocal microscopy, and mass spectrometry (MS). CLP259 was located in the cytoplasm in the form of clusters of thick rods and colocalized with actin at numerous sites and with tubulin in the median body. Immunoprecipitation followed by mass spectrometry revealed that CLP259 interacts with structural proteins such as giardins, SALP-1, axonemal, and eight coiled-coils. The vesicular traffic proteins detected were Mu adaptin, Vacuolar ATP synthase subunit B, Bip, Sec61 alpha, NSF, AP complex subunit beta, and dynamin. These results indicate that CLP259 in trophozoites is a CSK linker protein for actin and tubulin and could act as a scaffold protein driving vesicular traffic.


Subject(s)
Actins/metabolism , Giardia lamblia/metabolism , Plakins/metabolism , Tubulin/metabolism , Actins/chemistry , Amino Acid Sequence , Animals , Ankyrins/chemistry , Base Sequence , Blotting, Western , Computational Biology , Consensus Sequence , Cytoplasm/chemistry , Cytoskeleton/chemistry , Cytoskeleton/physiology , Cytoskeleton/ultrastructure , Dynamins/analysis , Female , Fluorescent Antibody Technique , Giardia lamblia/chemistry , Giardia lamblia/ultrastructure , Humans , Immunoprecipitation , Mice , Mice, Inbred BALB C , Microscopy, Confocal , Plakins/chemistry , Sequence Alignment , Tubulin/chemistry
10.
Molecules ; 26(15)2021 Jul 27.
Article in English | MEDLINE | ID: mdl-34361694

ABSTRACT

Extracellular signal-regulated kinases 1 and 2 (ERK1/2) play key roles in promoting cell survival and proliferation through the phosphorylation of various substrates. Remarkable antitumour activity is found in many inhibitors that act upstream of the ERK pathway. However, drug-resistant tumour cells invariably emerge after their use due to the reactivation of ERK1/2 signalling. ERK1/2 inhibitors have shown clinical efficacy as a therapeutic strategy for the treatment of tumours with mitogen-activated protein kinase (MAPK) upstream target mutations. These inhibitors may be used as a possible strategy to overcome acquired resistance to MAPK inhibitors. Here, we report a class of repeat proteins-designed ankyrin repeat protein (DARPin) macromolecules targeting ERK2 as inhibitors. The structural basis of ERK2-DARPin interactions based on molecular dynamics (MD) simulations was studied. The information was then used to predict stabilizing mutations employing a web-based algorithm, MAESTRO. To evaluate whether these design strategies were successfully deployed, we performed all-atom, explicit-solvent molecular dynamics (MD) simulations. Two mutations, Ala → Asp and Ser → Leu, were found to perform better than the original sequence (DARPin E40) based on the associated energy and key residues involved in protein-protein interaction. MD simulations and analysis of the data obtained on these mutations supported our predictions.


Subject(s)
Ankyrins/metabolism , Drug Design , MAP Kinase Signaling System/drug effects , Mitogen-Activated Protein Kinase 1/metabolism , Molecular Dynamics Simulation , Protein Kinase Inhibitors/metabolism , Algorithms , Ankyrins/chemistry , Ankyrins/genetics , Humans , Hydrogen Bonding , Ligands , MAP Kinase Signaling System/genetics , Mitogen-Activated Protein Kinase 3/metabolism , Mutation , Neoplasms/genetics , Neoplasms/metabolism , Phosphorylation/drug effects , Protein Binding , Protein Conformation, alpha-Helical , Protein Stability
11.
J Mol Cell Cardiol ; 139: 225-237, 2020 02.
Article in English | MEDLINE | ID: mdl-32035138

ABSTRACT

In striated muscles, the large scaffolding protein obscurin and a small SR-integral membrane protein sAnk1.5 control the retention of longitudinal SR across the sarcomere. How a complex of these proteins facilitates localization of longitudinal SR has yet to be resolved, but we hypothesize that obscurin interacts with a complex of sAnk1.5 proteins. To begin to address this hypothesis, we demonstrate that sAnk1.5 interacts with itself and identify two domains mediating self-association. Specifically, we show by co-precipitation and FLIM-FRET analysis that sAnk1.5 and another small AnkR isoform (sAnk1.6) interact with themselves and each other. We demonstrate that obscurin interacts with a complex of sAnk1.5 proteins and that this complex formation is enhanced by obscurin-binding. Using FLIM-FRET analysis, we show that obscurin interacts with sAnk1.5 alone and with sAnk1.6 in the presence of sAnk1.5. We find that sAnk1.5 self-association is disrupted by mutagenesis of residues Arg64-Arg69, residues previously associated with obscurin-binding. Molecular modeling of two interacting sAnk1.5 monomers facilitated the identification of Gly31-Val36 as an additional site of interaction, which was subsequently corroborated by co-precipitation and FLIM-FRET analysis. In closing, these results support a model in which sAnk1.5 forms large oligomers that interact with obscurin to facilitate the retention of longitudinal SR throughout skeletal and cardiac myocytes.


Subject(s)
Ankyrins/chemistry , Amino Acid Sequence , Animals , Ankyrins/metabolism , Binding Sites , Fluorescence Resonance Energy Transfer , HEK293 Cells , Humans , Protein Binding , Protein Domains , Protein Isoforms/chemistry , Protein Isoforms/metabolism , Protein Serine-Threonine Kinases/metabolism , Rats , Rho Guanine Nucleotide Exchange Factors/metabolism
12.
Biochem Biophys Res Commun ; 523(1): 263-266, 2020 02 26.
Article in English | MEDLINE | ID: mdl-31864703

ABSTRACT

RFX7 is an important member in the RFX family of DNA binding proteins and plays critical roles in natural killer cell-mediated immunity and neuron development. Our previous work identified ANKRA2 and RFXANK as the potential binding partners of RFX7. Here we present two structures of a RFX7 fragment, with one bound with the ANKRA2 ankyrin domain and the other bound to the RFXANK ankyrin domain. Our structural analysis reveals that both ANKRA2 and RFXANK recognize the PXLPXL motif of RFX7 and its flanking sequences via extensive hydrophobic interactions. Detailed structural comparison also provides an explanation for the different RFX7 binding affinities of ANKRA2 and RFXANK. Thus our work would provide clue to the understanding the roles of ANKRA2 and RFXANK in the RFX7-associated signaling pathway.


Subject(s)
Ankyrins/chemistry , Ankyrins/metabolism , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Regulatory Factor X Transcription Factors/chemistry , Regulatory Factor X Transcription Factors/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism , Crystallography, X-Ray , Humans , Models, Molecular , Protein Conformation , Regulatory Factor X Transcription Factors/genetics , Signal Transduction
13.
Nat Chem Biol ; 14(8): 778-787, 2018 08.
Article in English | MEDLINE | ID: mdl-29867141

ABSTRACT

The mammalian Atg8 family proteins are central drivers of autophagy and contain six members, classified into the LC3 and GABARAP subfamilies. Due to their high sequence similarity and consequent functional overlaps, it is difficult to delineate specific functions of Atg8 proteins in autophagy. Here we discover a super-strong GABARAP-selective inhibitory peptide harbored in 270/480 kDa ankyrin-G and a super-potent pan-Atg8 inhibitory peptide from 440 kDa ankyrin-B. Structural studies elucidate the mechanism governing the Atg8 binding potency and selectivity of the peptides, reveal a general Atg8-binding sequence motif, and allow development of a more GABARAP-selective inhibitory peptide. These peptides effectively blocked autophagy when expressed in cultured cells. Expression of these ankyrin-derived peptides in Caenorhabditis elegans also inhibited autophagy, causing accumulation of the p62 homolog SQST-1, delayed development and shortened life span. Thus, these genetically encodable autophagy inhibitory peptides can be used to occlude autophagy spatiotemporally in living animals.


Subject(s)
Ankyrins/chemistry , Autophagy-Related Protein 8 Family/antagonists & inhibitors , Autophagy/drug effects , Peptides/pharmacology , Animals , Autophagy-Related Protein 8 Family/metabolism , COS Cells , Cells, Cultured , Chlorocebus aethiops , Peptides/chemistry
14.
J Chem Inf Model ; 60(2): 805-820, 2020 02 24.
Article in English | MEDLINE | ID: mdl-31804821

ABSTRACT

4-HNE-modified ankyrins have been described in diseases such as diabetes, renal failure, G6PD deficient, sickle cell trait, and P. falciparum infected erythrocytes with different AB0 blood groups. However, effects at the atomic level of this carbonylation on structure and function of modified protein are not yet fully understood. We present a study based on molecular dynamics simulations of nine 4-HNE modified residues of the ZU5-ANK ankyrin domain with ß-spectrin and their binding energy profiles. Results show that 4-HNE induced local conformational changes over all protein systems evaluated, increased mobility in the modification sites, and localized structural changes between the positively charged patch of the ZU5-ANK domain. Carbonylation with 4-HNE on lysine residues decreased the affinity between ZU5-ANK and the 14-ß-spectrin repeat by reducing electrostatic and van der Waals interactions. The presented work provides further insight into understanding the loss of human erythrocyte deformation capacity under conditions of oxidative stress in different diseases.


Subject(s)
Aldehydes/chemistry , Ankyrins/chemistry , Ankyrins/metabolism , Molecular Dynamics Simulation , Spectrin/metabolism , Erythrocytes/metabolism , Humans , Oxidative Stress , Protein Binding , Protein Domains
15.
Proc Natl Acad Sci U S A ; 114(48): 12743-12748, 2017 11 28.
Article in English | MEDLINE | ID: mdl-29133412

ABSTRACT

Obesity typically is linked to caloric imbalance as a result of overnutrition. Here we propose a cell-autonomous mechanism for adiposity as a result of persistent cell surface glucose transporter type 4 (GLUT4) in adipocytes resulting from impaired function of ankyrin-B (AnkB) in coupling GLUT4 to clathrin-mediated endocytosis. Adipose tissue-specific AnkB-KO mice develop obesity and progressive pancreatic islet dysfunction with age or high-fat diet (HFD). AnkB-deficient adipocytes exhibit increased lipid accumulation associated with increased glucose uptake and impaired endocytosis of GLUT4. AnkB binds directly to GLUT4 and clathrin and promotes their association in adipocytes. AnkB variants that fail to restore normal lipid accumulation and GLUT4 localization in adipocytes are present in 1.3% of European Americans and 8.4% of African Americans, and are candidates to contribute to obesity susceptibility in humans.


Subject(s)
Adipocytes/metabolism , Adiposity/genetics , Ankyrins/genetics , Glucose Transporter Type 4/genetics , Glucose/metabolism , Obesity/genetics , Adipocytes/pathology , Animals , Ankyrins/chemistry , Ankyrins/metabolism , Biological Transport , Black People , Clathrin/genetics , Clathrin/metabolism , Diet, High-Fat/adverse effects , Endocytosis , Gene Expression Regulation , Glucose Transporter Type 4/metabolism , Humans , Islets of Langerhans/metabolism , Islets of Langerhans/pathology , Male , Mice , Mice, Knockout , Models, Molecular , Mutation , Obesity/etiology , Obesity/metabolism , Obesity/pathology , Protein Binding , Protein Structure, Secondary , Signal Transduction , White People
16.
J Cell Mol Med ; 23(6): 4454-4463, 2019 06.
Article in English | MEDLINE | ID: mdl-31016877

ABSTRACT

Hereditary spherocytosis (HS) is the most common inherited haemolytic anaemia disorder. ANK1 mutations account for most HS cases, but pathogenicity analysis and functional research have not been widely performed for these mutations. In this study, in order to confirm diagnosis, gene mutation was screened in two unrelated Chinese families with HS by a next-generation sequencing (NGS) panel and then confirmed by Sanger sequencing. Two novel heterozygous mutations (c.C841T, p.R281X and c.T290G, p.L97R) of the ANK1 gene were identified in the two families respectively. Then, the pathogenicity of the two new mutations and two previously reported ANK1 mutations (c.C648G, p.Y216X and c.G424T, p.E142X) were studied by in vitro experiments. The four mutations increased the osmotic fragility of cells, reduced the stabilities of ANK1 proteins and prevented the protein from localizing to the plasma membrane and interacting with SPTB and SLC4A1. We classified these four mutations into disease-causing mutations for HS. Thus, conducting the same mutation test and providing genetic counselling for the two families were meaningful and significant. Moreover, the identification of two novel mutations enriches the ANK1 mutation database, especially in China.


Subject(s)
Ankyrins/genetics , Ankyrins/metabolism , Asian People/genetics , Loss of Function Mutation , Spherocytosis, Hereditary/genetics , Spherocytosis, Hereditary/pathology , Amino Acid Sequence , Anion Exchange Protein 1, Erythrocyte/metabolism , Ankyrins/chemistry , Child , Child, Preschool , Female , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Infant , Infant, Newborn , Male , Pedigree , Protein Conformation , Protein Stability , Sequence Homology , Spectrin/metabolism , Spherocytosis, Hereditary/metabolism
17.
Circulation ; 138(23): 2682-2697, 2018 12 04.
Article in English | MEDLINE | ID: mdl-30571258

ABSTRACT

BACKGROUND: Human loss-of-function variants of ANK2 (ankyrin-B) are linked to arrhythmias and sudden cardiac death. However, their in vivo effects and specific arrhythmogenic pathways have not been fully elucidated. METHODS: We identified new ANK2 variants in 25 unrelated Han Chinese probands with ventricular tachycardia by whole-exome sequencing. The potential pathogenic variants were validated by Sanger sequencing. We performed functional and mechanistic experiments in ankyrin-B knockin (KI) mouse models and in single myocytes isolated from KI hearts. RESULTS: We detected a rare, heterozygous ANK2 variant (p.Q1283H) in a proband with recurrent ventricular tachycardia. This variant was localized to the ZU5C region of ANK2, where no variants have been previously reported. KI mice harboring the p.Q1283H variant exhibited an increased predisposition to ventricular arrhythmias after catecholaminergic stress in the absence of cardiac structural abnormalities. Functional studies illustrated an increased frequency of delayed afterdepolarizations and Ca2+ waves and sparks accompanied by decreased sarcoplasmic reticulum Ca2+ content in KI cardiomyocytes on isoproterenol stimulation. The immunoblotting results showed increased levels of phosphorylated ryanodine receptor Ser2814 in the KI hearts, which was further amplified on isoproterenol stimulation. Coimmunoprecipitation experiments demonstrated dissociation of protein phosphatase 2A from ryanodine receptor in the KI hearts, which was accompanied by a decreased binding of ankyrin-B to protein phosphatase 2A regulatory subunit B56α. Finally, the administration of metoprolol or flecainide decreased the incidence of stress-induced ventricular arrhythmias in the KI mice. CONCLUSIONS: ANK2 p.Q1283H is a disease-associated variant that confers susceptibility to stress-induced arrhythmias, which may be prevented by the administration of metoprolol or flecainide. This variant is associated with the loss of protein phosphatase 2A activity, increased phosphorylation of ryanodine receptor, exaggerated delayed afterdepolarization-mediated trigger activity, and arrhythmogenesis.


Subject(s)
Ankyrins/genetics , Arrhythmias, Cardiac/pathology , Protein Phosphatase 2/metabolism , Ryanodine Receptor Calcium Release Channel/metabolism , Action Potentials/drug effects , Animals , Ankyrins/chemistry , Arrhythmias, Cardiac/metabolism , Calcium/metabolism , Disease Models, Animal , Electrocardiography , Female , Humans , Isoproterenol/pharmacology , Mice , Middle Aged , Myocytes, Cardiac/cytology , Myocytes, Cardiac/metabolism , Phosphorylation , Polymorphism, Single Nucleotide , Ryanodine/pharmacology , Sarcoplasmic Reticulum/metabolism
18.
Biochem Biophys Res Commun ; 495(1): 1002-1007, 2018 01 01.
Article in English | MEDLINE | ID: mdl-29175332

ABSTRACT

Legionella pneumophila is a pathogen, causing severe pneumonia in humans called Legionnaires' disease. AnkC (LegA12) is a poorly characterized 495-residue effector protein conserved in multiple Legionella species. Here, we report the crystal structure of a C-terminally truncated AnkC (2-384) at 3.2 Å resolution. The structure shows seven ankyrin repeats (ARs) with unique structural features. AnkC forms a dimer along the outer surface of loops between ARs. The dimer exists both in the crystal form and in solution, as shown by analytical ultracentrifugation. This is the first example of ARs as a dimerization module as opposed to solely a protein interaction domain. In addition, a novel α-helix insert between AR3-AR4 is positioned across the surface opposite the ankyrin groove. Sequence conservation suggests that the ankyrin groove of AnkC is a functional site that interacts with binding targets. This ankyrin domain structure is an important step towards a functional characterization of AnkC.


Subject(s)
Ankyrin Repeat , Ankyrins/chemistry , Ankyrins/ultrastructure , Models, Chemical , Models, Molecular , Protein Multimerization , Amino Acid Sequence , Binding Sites , Computer Simulation , Conserved Sequence , Legionella pneumophila/metabolism , Molecular Sequence Data , Protein Binding , Protein Conformation
19.
Asian Pac J Allergy Immunol ; 36(2): 126-135, 2018 06.
Article in English | MEDLINE | ID: mdl-28802032

ABSTRACT

BACKGROUND: AnkGAG1D4 is an artificial ankyrin repeat protein which recognizes the capsid protein (CA) of the human immunodeficiency virus type 1 (HIV-1) and exhibits the intracellular antiviral activity on the viral assembly process. Improving the binding affinity of AnkGAG1D4 would potentially enhance the AnkGAG1D4-mediated antiviral activity. OBJECTIVE: To augment the affinity of AnkGAG1D4 scaffold towards its CA target, through computational predictions and experimental designs. METHOD: Three dimensional structure of the binary complex formed by AnkGAG1D4 docked to the CA was used as a model for van der Waals (vdW) binding energy calculation. The results generated a simple guideline to select the amino acids for modifications. Following the predictions, modified AnkGAG1D4 proteins were produced and further evaluated for their CA-binding activity, using ELISA-modified method and bio-layer interferometry (BLI). RESULTS: Tyrosine at position 56 (Y56) in AnkGAG1D4 was experimentally identified as the most critical residue for CA binding. Rational substitutions of this residue diminished the binding affinity. However, vdW calculation preconized to substitute serine for tyrosine at position 45. Remarkably, the affinity for the viral CA was significantly enhanced in AnkGAG1D4-S45Y mutant, with no alteration of the target specificity. CONCLUSIONS: The S-to-Y mutation at position 45, based on the prediction of interacting amino acids and on vdW binding energy calculation, resulted in a significant enhancement of the affinity of AnkGAG1D4 ankyrin for its CA target. AnkGAG1D4-S45Y mutant represented the starting point for further construction of variants with even higher affinity towards the viral CA, and higher therapeutic potential in the future.


Subject(s)
Antiviral Agents/chemistry , Antiviral Agents/pharmacology , HIV-1/drug effects , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacology , Amino Acid Sequence , Amino Acids , Ankyrins/chemistry , Ankyrins/metabolism , Ankyrins/pharmacology , Antiviral Agents/metabolism , Capsid Proteins/metabolism , Humans , Protein Binding , Recombinant Fusion Proteins/metabolism , Structure-Activity Relationship
20.
Biophys J ; 113(10): 2249-2260, 2017 Nov 21.
Article in English | MEDLINE | ID: mdl-28988699

ABSTRACT

The Na+,K+-ATPase is a plasma membrane ion transporter of high physiological importance for ion homeostasis and cellular excitability in electrically active tissues. Mutations in the genes coding for Na+,K+-ATPase α-subunit isoforms lead to severe human pathologies including Familial Hemiplegic Migraine type 2, Alternating Hemiplegia of Childhood, Rapid-onset Dystonia Parkinsonism, or epilepsy. Many of the reported mutations lead to change- or loss-of-function effects, whereas others do not alter the functional properties, but lead to, e.g., reduced protein stability, reduced protein expression, or defective plasma membrane targeting. Na+,K+-ATPase frequently assembles with other membrane transporters or cellular matrix proteins in specialized plasma membrane microdomains, but the effects of these interactions on targeting or protein mobility are elusive so far. Mutation of established interaction motifs of the Na+,K+-ATPase with ankyrin B and caveolin-1 are expected to result in changes in plasma membrane targeting, changes of the localization pattern, and of the diffusion behavior of the enzyme. We studied the consequences of mutations in these binding sites by monitoring diffusion of eGFP-labeled Na+,K+-ATPase constructs in the plasma membrane of HEK293T cells by fluorescence correlation spectroscopy as well as fluorescence recovery after photobleaching or photoswitching, and observed significant differences compared to the wild-type enzyme, with synergistic effects for combinations of interaction site mutations. These measurements expand the possibilities to study the consequences of Na+,K+-ATPase mutations and provide information about the interaction of Na+,K+-ATPase α-isoforms with cellular matrix proteins, the cytoskeleton, or other membrane protein complexes.


Subject(s)
Ankyrins/chemistry , Ankyrins/metabolism , Caveolin 1/chemistry , Caveolin 1/metabolism , Cell Membrane/metabolism , Mutation , Sodium-Potassium-Exchanging ATPase/metabolism , Amino Acid Sequence , Animals , Biological Transport/genetics , Diffusion , HEK293 Cells , Humans , Models, Molecular , Oocytes/metabolism , Protein Binding/genetics , Protein Domains , Rubidium/metabolism , Sodium-Potassium-Exchanging ATPase/chemistry , Sodium-Potassium-Exchanging ATPase/genetics , Xenopus laevis/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL