Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 587
Filter
1.
Biomed Pharmacother ; 148: 112756, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35228064

ABSTRACT

The 2019 corona virus disease (COVID-19) has caused a global chaos, where a novel Omicron variant has challenged the healthcare system, followed by which it has been referred to as a variant of concern (VOC) by the World Health Organization (WHO), owing to its alarming transmission and infectivity rate. The large number of mutations in the receptor binding domain (RBD) of the spike protein is responsible for strengthening of the spike-angiotensin-converting enzyme 2 (ACE2) interaction, thereby explaining the elevated threat. This is supplemented by enhanced resistance of the variant towards pre-existing antibodies approved for the COVID-19 therapy. The manuscript brings into light failure of existing therapies to provide the desired effect, however simultaneously discussing the novel possibilities on the verge of establishing suitable treatment portfolio. The authors entail the risks associated with omicron resistance against antibodies and vaccine ineffectiveness on one side, and novel approaches and targets - kinase inhibitors, viral protease inhibitors, phytoconstituents, entry pathways - on the other. The manuscript aims to provide a holistic picture about the Omicron variant, by providing comprehensive discussions related to multiple aspects of the mutated spike variant, which might aid the global researchers and healthcare experts in finding an optimised solution to this pandemic.


Subject(s)
COVID-19/physiopathology , SARS-CoV-2/genetics , SARS-CoV-2/metabolism , Angiotensin-Converting Enzyme 2/metabolism , Animals , COVID-19/immunology , COVID-19 Vaccines/immunology , Cathepsins/metabolism , ErbB Receptors/antagonists & inhibitors , Humans , Immunization Schedule , Immunization, Secondary , Phytotherapy/methods , Plants, Medicinal , Protein Binding/physiology , Protein Interaction Domains and Motifs/physiology , Protein Structural Elements/physiology , Spike Glycoprotein, Coronavirus/metabolism , Viral Protease Inhibitors/pharmacology , Viral Protease Inhibitors/therapeutic use
2.
Nat Struct Mol Biol ; 28(11): 909-922, 2021 11.
Article in English | MEDLINE | ID: mdl-34759380

ABSTRACT

Carboxysomes in cyanobacteria enclose the enzymes Rubisco and carbonic anhydrase to optimize photosynthetic carbon fixation. Understanding carboxysome assembly has implications in agricultural biotechnology. Here we analyzed the role of the scaffolding protein CcmM of the ß-cyanobacterium Synechococcus elongatus PCC 7942 in sequestrating the hexadecameric Rubisco and the tetrameric carbonic anhydrase, CcaA. We find that the trimeric CcmM, consisting of γCAL oligomerization domains and linked small subunit-like (SSUL) modules, plays a central role in mediation of pre-carboxysome condensate formation through multivalent, cooperative interactions. The γCAL domains interact with the C-terminal tails of the CcaA subunits and additionally mediate a head-to-head association of CcmM trimers. Interestingly, SSUL modules, besides their known function in recruiting Rubisco, also participate in intermolecular interactions with the γCAL domains, providing further valency for network formation. Our findings reveal the mechanism by which CcmM functions as a central organizer of the pre-carboxysome multiprotein matrix, concentrating the core components Rubisco and CcaA before ß-carboxysome shell formation.


Subject(s)
Bacterial Proteins/metabolism , Carbonic Anhydrases/metabolism , Protein Interaction Domains and Motifs/physiology , Ribulose-Bisphosphate Carboxylase/metabolism , Synechococcus/metabolism , Biomolecular Condensates/physiology , Cryoelectron Microscopy , Photosynthesis/physiology , Protein Conformation , Synechococcus/genetics
3.
Int J Mol Sci ; 22(21)2021 Oct 22.
Article in English | MEDLINE | ID: mdl-34768862

ABSTRACT

The most common genetic cause of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) is the presence of poly-PR/GR dipeptide repeats, which are encoded by the chromosome 9 open reading frame 72 (C9orf72) gene. Recently, it was shown that poly-PR/GR alters chromatin accessibility, which results in the stabilization and enhancement of transcriptional activity of the tumor suppressor p53 in several neurodegenerative disease models. A reduction in p53 protein levels protects against poly-PR and partially against poly-GR neurotoxicity in cells. Moreover, in model organisms, a reduction of p53 protein levels protects against neurotoxicity of poly-PR. Here, we aimed to study the detailed molecular mechanisms of how p53 contributes to poly-PR/GR-mediated neurodegeneration. Using a combination of biophysical techniques such as nuclear magnetic resonance (NMR) spectroscopy, fluorescence polarization, turbidity assays, and differential interference contrast (DIC) microscopy, we found that p53 physically interacts with poly-PR/GR and triggers liquid-liquid phase separation of p53. We identified the p53 transactivation domain 2 (TAD2) as the main binding site for PR25/GR25 and showed that binding of poly-PR/GR to p53 is mediated by a network of electrostatic and/or hydrophobic interactions. Our findings might help to understand the mechanistic role of p53 in poly-PR/GR-associated neurodegeneration.


Subject(s)
Amyotrophic Lateral Sclerosis/pathology , C9orf72 Protein/metabolism , Dipeptides/metabolism , Frontotemporal Dementia/pathology , Tumor Suppressor Protein p53/metabolism , Amyotrophic Lateral Sclerosis/genetics , Binding Sites , C9orf72 Protein/genetics , Fluorescence Polarization , Frontotemporal Dementia/genetics , Humans , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Interaction Domains and Motifs/physiology , Transcriptional Activation/genetics , Tumor Suppressor Protein p53/genetics
4.
Molecules ; 26(20)2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34684701

ABSTRACT

14-3-3 proteins are abundant, intramolecular proteins that play a pivotal role in cellular signal transduction by interacting with phosphorylated ligands. In addition, they are molecular chaperones that prevent protein unfolding and aggregation under cellular stress conditions in a similar manner to the unrelated small heat-shock proteins. In vivo, amyloid ß (Aß) and α-synuclein (α-syn) form amyloid fibrils in Alzheimer's and Parkinson's diseases, respectively, a process that is intimately linked to the diseases' progression. The 14-3-3ζ isoform potently inhibited in vitro fibril formation of the 40-amino acid form of Aß (Aß40) but had little effect on α-syn aggregation. Solution-phase NMR spectroscopy of 15N-labeled Aß40 and A53T α-syn determined that unlabeled 14-3-3ζ interacted preferentially with hydrophobic regions of Aß40 (L11-H21 and G29-V40) and α-syn (V3-K10 and V40-K60). In both proteins, these regions adopt ß-strands within the core of the amyloid fibrils prepared in vitro as well as those isolated from the inclusions of diseased individuals. The interaction with 14-3-3ζ is transient and occurs at the early stages of the fibrillar aggregation pathway to maintain the native, monomeric, and unfolded structure of Aß40 and α-syn. The N-terminal regions of α-syn interacting with 14-3-3ζ correspond with those that interact with other molecular chaperones as monitored by in-cell NMR spectroscopy.


Subject(s)
14-3-3 Proteins/metabolism , Amyloid beta-Peptides/metabolism , alpha-Synuclein/metabolism , 14-3-3 Proteins/physiology , Amyloid/metabolism , Amyloid/physiology , Amyloid beta-Peptides/physiology , Humans , Molecular Chaperones/physiology , Protein Aggregates , Protein Binding/physiology , Protein Conformation , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs/physiology , Protein Unfolding , alpha-Synuclein/physiology
5.
Molecules ; 26(20)2021 Oct 11.
Article in English | MEDLINE | ID: mdl-34684708

ABSTRACT

Elk-1 is a transcription factor that binds together with a dimer of the serum response factor (SRF) to the serum-response element (SRE), a genetic element that connects cellular stimulation with gene transcription. Elk-1 plays an important role in the regulation of cellular proliferation and apoptosis, thymocyte development, glucose homeostasis and brain function. The biological function of Elk-1 relies essentially on the interaction with other proteins. Elk-1 binds to SRF and generates a functional ternary complex that is required to activate SRE-mediated gene transcription. Elk-1 is kept in an inactive state under basal conditions via binding of a SUMO-histone deacetylase complex. Phosphorylation by extracellular signal-regulated protein kinase, c-Jun N-terminal protein kinase or p38 upregulates the transcriptional activity of Elk-1, mediated by binding to the mediator of RNA polymerase II transcription (Mediator) and the transcriptional coactivator p300. Strong and extended phosphorylation of Elk-1 attenuates Mediator and p300 recruitment and allows the binding of the mSin3A-histone deacetylase corepressor complex. The subsequent dephosphorylation of Elk-1, catalyzed by the protein phosphatase calcineurin, facilitates the re-SUMOylation of Elk-1, transforming Elk-1 back to a transcriptionally inactive state. Thus, numerous protein-protein interactions control the activation cycle of Elk-1 and are essential for its biological function.


Subject(s)
ets-Domain Protein Elk-1/metabolism , ets-Domain Protein Elk-1/physiology , Animals , Extracellular Signal-Regulated MAP Kinases/metabolism , Gene Expression/genetics , Gene Expression Regulation/genetics , Mice , Nuclear Proteins/metabolism , Phosphorylation , Protein Interaction Domains and Motifs/physiology , Protein Interaction Mapping/methods , Protein Interaction Maps/physiology , Proto-Oncogene Proteins/metabolism , Serum Response Factor/metabolism , Transcription Factors/metabolism , Transcription, Genetic/genetics , Transcriptional Activation/genetics , ets-Domain Protein Elk-1/genetics
6.
PLoS One ; 16(9): e0257905, 2021.
Article in English | MEDLINE | ID: mdl-34582502

ABSTRACT

SARS-CoV-2 virus, the causative agent of Covid-19, has fired up a global pandemic. The virus interacts with the human receptor angiotensin-converting enzyme 2 (ACE2) for an invasion via receptor binding domain (RBD) on its spike protein. To provide a deeper understanding of this interaction, we performed microsecond simulations of the RBD-ACE2 complex for SARS-CoV-2 and compared it with the closely related SARS-CoV discovered in 2003. We show residues in the RBD of SARS-CoV-2 that were mutated from SARS-CoV, collectively help make the RBD anchor much stronger to the N-terminal part of ACE2 than the corresponding residues on RBD of SARS-CoV. This would result in a reduced dissociation rate of SARS-CoV-2 from human receptor protein compared to SARS-CoV. The phenomenon was consistently observed in simulations beyond 500 ns and was reproducible across different force fields. Altogether, our study adds more insight into the critical dynamics of the key residues at the virus spike and human receptor binding interface and potentially aids the development of diagnostics and therapeutics to combat the pandemic efficiently.


Subject(s)
Angiotensin-Converting Enzyme 2/metabolism , COVID-19/metabolism , SARS-CoV-2/genetics , Angiotensin-Converting Enzyme 2/genetics , Binding Sites , COVID-19/genetics , Humans , Models, Theoretical , Molecular Dynamics Simulation , Pandemics , Peptidyl-Dipeptidase A/metabolism , Protein Binding , Protein Domains , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , SARS-CoV-2/pathogenicity , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/metabolism
7.
Am J Pathol ; 191(12): 2219-2230, 2021 12.
Article in English | MEDLINE | ID: mdl-34428424

ABSTRACT

Transdifferentiation (or activation) of hepatic stellate cells (HSCs) to myofibroblasts is a key event in liver fibrosis. Activated HSCs in the tumor microenvironment reportedly promote tumor progression. This study analyzed the effect of an inhibitor of HSC activation, retinol-binding protein-albumin domain III fusion protein (R-III), on protumorigenic functions of HSCs. Although conditioned medium collected from activated HSCs enhanced the migration, invasion, and proliferation of the hepatocellular carcinoma cell line Hepa-1c1c7, this effect was not observed in Hepa-1c1c7 cells treated with conditioned medium from R-III-exposed HSCs. In a subcutaneous tumor model, larger tumors with increased vascular density were formed in mice transplanted with Hepa-1c1c7+HSC than in mice transplanted with Hepa-1c1c7 cells alone. Intriguingly, when Hepa-1c1c7+HSC-transplanted mice were injected intravenously with R-III, a reduction in vascular density and extended tumor necrosis were observed. In an orthotopic tumor model, co-transplantation of HSCs enhanced tumor growth, angiogenesis, and regional metastasis accompanied by increased peritumoral lymphatic vessel density, which was abolished by R-III. In vitro study showed that R-III treatment affected the synthesis of pro-angiogenic and anti-angiogenic factors in activated HSCs, which might be the potential mechanism underlying the R-III effect. These findings suggest that the inhibition of HSC activation abrogates HSC-induced tumor angiogenesis and growth, which represents an attractive therapeutic strategy.


Subject(s)
Carcinoma, Hepatocellular/pathology , Hepatic Stellate Cells/drug effects , Liver Neoplasms/pathology , Recombinant Fusion Proteins/pharmacology , Albumins/chemistry , Albumins/pharmacology , Albumins/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Hepatocellular/blood supply , Carcinoma, Hepatocellular/therapy , Cell Transformation, Neoplastic/drug effects , Cells, Cultured , Down-Regulation/drug effects , Female , Hepatic Stellate Cells/physiology , Liver Neoplasms/blood supply , Liver Neoplasms/therapy , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/prevention & control , Protein Interaction Domains and Motifs/physiology , Recombinant Fusion Proteins/therapeutic use , Retinol-Binding Proteins/pharmacology , Retinol-Binding Proteins/therapeutic use , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
8.
Int J Mol Sci ; 22(16)2021 Aug 11.
Article in English | MEDLINE | ID: mdl-34445335

ABSTRACT

Protein inhibition is a natural regulatory process to control cellular metabolic fluxes. PII-family signal-transducing effectors are in this matter key regulators of the nitrogen metabolism. Their interaction with their various targets is governed by the cellular nitrogen level and the energy charge. Structural studies on GlnK, a PII-family inhibitor of the ammonium transporters (Amt), showed that the T-loops responsible for channel obstruction are displaced upon the binding of 2-oxoglutarate, magnesium and ATP in a conserved cleft. However, GlnK from Methanocaldococcus jannaschii was shown to bind 2-oxoglutarate on the tip of its T-loop, causing a moderate disruption to GlnK-Amt interaction, raising the question if methanogenic archaea use a singular adaptive strategy. Here we show that membrane fractions of Methanothermococcus thermolithotrophicus released GlnKs only in the presence of Mg-ATP and 2-oxoglutarate. This observation led us to structurally characterize the two GlnK isoforms apo or in complex with ligands. Together, our results show that the 2-oxoglutarate binding interface is conserved in GlnKs from Methanococcales, including Methanocaldococcus jannaschii, emphasizing the importance of a free carboxy-terminal group to facilitate ligand binding and to provoke the shift of the T-loop positions.


Subject(s)
Ammonium Compounds/metabolism , Ketoglutaric Acids/metabolism , Methanococcales/metabolism , PII Nitrogen Regulatory Proteins , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Binding Sites , Conserved Sequence , Crystallography, X-Ray , Ion Transport , Metabolic Networks and Pathways , Models, Molecular , Nitrogen/metabolism , PII Nitrogen Regulatory Proteins/chemistry , PII Nitrogen Regulatory Proteins/metabolism , Protein Conformation , Protein Interaction Domains and Motifs/physiology , Sequence Analysis, Protein
9.
PLoS One ; 16(6): e0229879, 2021.
Article in English | MEDLINE | ID: mdl-34181638

ABSTRACT

Δ9-tetrahydrocannabinol (Δ9-THC), the main active ingredient of Cannabis sativa (marijuana), interacts with the human brain cannabinoid (CB1) receptor and mimics pharmacological effects of endocannabinoids (eCBs) like N-arachidonylethanolamide (AEA). Due to its flexible nature of AEA structure with more than 15 rotatable bonds, establishing its binding mode to the CB1 receptor is elusive. The aim of the present study was to explore possible binding conformations of AEA within the binding pocket of the CB1 receptor confirmed in the recently available X-ray crystal structures of the CB1 receptor and predict essential AEA binding domains. We performed long time molecular dynamics (MD) simulations of plausible AEA docking poses until its receptor binding interactions became optimally established. Our simulation results revealed that AEA favors to bind to the hydrophobic channel (HC) of the CB1 receptor, suggesting that HC holds essential significance in AEA binding to the CB1 receptor. Our results also suggest that the Helix 2 (H2)/H3 region of the CB1 receptor is an AEA binding subsite privileged over the H7 region.


Subject(s)
Arachidonic Acids/chemistry , Endocannabinoids/chemistry , Polyunsaturated Alkamides/chemistry , Receptor, Cannabinoid, CB1/ultrastructure , Animals , Arachidonic Acids/metabolism , Brain/metabolism , Cannabinoid Receptor Modulators/chemistry , Cannabinoids/pharmacology , Endocannabinoids/metabolism , Endocannabinoids/pharmacology , Humans , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Polyunsaturated Alkamides/metabolism , Protein Conformation , Protein Interaction Domains and Motifs/physiology , Receptor, Cannabinoid, CB1/chemistry , Receptor, Cannabinoid, CB1/metabolism
10.
Mol Immunol ; 136: 16-25, 2021 08.
Article in English | MEDLINE | ID: mdl-34052579

ABSTRACT

Human Leukocyte Antigen (HLA) complexes are critical cell-surface protein assemblies that facilitate T-cell surveillance of almost all cell types in the body. While T-cell receptor binding to HLA class I and class II complexes is well-described with detailed structural information, the nature of cis HLA interactions within the plasma membrane of the surveyed cells remains to be better characterized, as protein-protein interactions in the membrane environment are technically challenging to profile. Here we performed extracellular chemical crosslinking on intact antigen presenting cells to specifically elucidate protein-protein interactions present in the external plasma membrane. We found that the crosslink dataset was dominated by inter- and intra-protein crosslinks involving HLA molecules, which enabled not only the construction of an HLA-centric plasma membrane protein interaction map, but also revealed multiple modes of HLA class I - HLA class II interactions with further structural modeling based on crosslinker distance restraints. Collectively, our data demonstrate that HLA molecules colocalize and can be densely packed on the plasma membrane.


Subject(s)
Cell Membrane/metabolism , Histocompatibility Antigens Class II/metabolism , Histocompatibility Antigens Class I/metabolism , Protein Interaction Domains and Motifs/physiology , Antigen-Presenting Cells/immunology , Cell Line, Transformed , Humans , Mass Spectrometry , Molecular Docking Simulation , Protein Binding/physiology , Receptors, Antigen, T-Cell/immunology
11.
Mol Microbiol ; 116(2): 648-662, 2021 08.
Article in English | MEDLINE | ID: mdl-34028100

ABSTRACT

Group I biotin protein ligases (BPLs) catalyze the covalent attachment of biotin to its cognate acceptor proteins. In contrast, Group II BPLs have an additional N-terminal DNA-binding domain and function not only in biotinylation but also in transcriptional regulation of genes of biotin biosynthesis and transport. Most bacteria contain only a single biotin protein ligase, whereas Clostridium acetobutylicum contains two biotin protein ligase homologs: BplA and BirA'. Sequence alignments showed that BplA is a typical group I BPL, whereas BirA' lacked the C-terminal domain conserved throughout extant BPL proteins. This raised the questions of why two BPL homologs are needed and why the apparently defective BirA' has been retained. We have used in vivo and in vitro assays to show that BplA is a functional BPL whereas BirA' acts as a biotin sensor involved in transcriptional regulation of biotin transport. We also successfully converted BirA' into a functional biotin protein ligase with regulatory activity by fusing it to the C-terminal domain from BplA. Finally, we provide evidence that BplA and BirA' interact in vivo.


Subject(s)
Biotin/metabolism , Biotinylation/physiology , Carbon-Nitrogen Ligases/metabolism , Clostridium acetobutylicum/metabolism , Transcription, Genetic/genetics , Biotin/biosynthesis , Carbon-Nitrogen Ligases/genetics , Clostridium acetobutylicum/genetics , Gene Expression Regulation, Bacterial/genetics , Protein Interaction Domains and Motifs/physiology
12.
Mol Cell ; 81(12): 2549-2565.e8, 2021 06 17.
Article in English | MEDLINE | ID: mdl-33957083

ABSTRACT

Hsp70s comprise a deeply conserved chaperone family that has a central role in maintaining protein homeostasis. In humans, Hsp70 client specificity is provided by 49 different co-factors known as J domain proteins (JDPs). However, the cellular function and client specificity of JDPs have largely remained elusive. We have combined affinity purification-mass spectrometry (AP-MS) and proximity-dependent biotinylation (BioID) to characterize the interactome of all human JDPs and Hsp70s. The resulting network suggests specific functions for many uncharacterized JDPs, and we establish a role of conserved JDPs DNAJC9 and DNAJC27 in histone chaperoning and ciliogenesis, respectively. Unexpectedly, we find that the J domain of DNAJC27 but not of other JDPs can fully replace the function of endogenous DNAJC27, suggesting a previously unappreciated role for J domains themselves in JDP specificity. More broadly, our work expands the role of the Hsp70-regulated proteostasis network and provides a platform for further discovery of JDP-dependent functions.


Subject(s)
HSP40 Heat-Shock Proteins/physiology , HSP70 Heat-Shock Proteins/physiology , Protein Interaction Domains and Motifs/physiology , HEK293 Cells , HSP40 Heat-Shock Proteins/metabolism , HSP70 Heat-Shock Proteins/metabolism , HeLa Cells , Humans , Molecular Chaperones/metabolism , Protein Binding , Protein Domains , rab GTP-Binding Proteins/metabolism
13.
Proc Natl Acad Sci U S A ; 118(16)2021 04 20.
Article in English | MEDLINE | ID: mdl-33846262

ABSTRACT

Enzymes in multistep metabolic pathways utilize an array of regulatory mechanisms to maintain a delicate homeostasis [K. Magnuson, S. Jackowski, C. O. Rock, J. E. Cronan, Jr, Microbiol. Rev. 57, 522-542 (1993)]. Carrier proteins in particular play an essential role in shuttling substrates between appropriate enzymes in metabolic pathways. Although hypothesized [E. Ploskon et al., Chem. Biol. 17, 776-785 (2010)], allosteric regulation of substrate delivery has never before been demonstrated for any acyl carrier protein (ACP)-dependent pathway. Studying these mechanisms has remained challenging due to the transient and dynamic nature of protein-protein interactions, the vast diversity of substrates, and substrate instability [K. Finzel, D. J. Lee, M. D. Burkart, ChemBioChem 16, 528-547 (2015)]. Here we demonstrate a unique communication mechanism between the ACP and partner enzymes using solution NMR spectroscopy and molecular dynamics to elucidate allostery that is dependent on fatty acid chain length. We demonstrate that partner enzymes can allosterically distinguish between chain lengths via protein-protein interactions as structural features of substrate sequestration are translated from within the ACP four-helical bundle to the protein surface, without the need for stochastic chain flipping. These results illuminate details of cargo communication by the ACP that can serve as a foundation for engineering carrier protein-dependent pathways for specific, desired products.


Subject(s)
Acyl Carrier Protein/metabolism , Acyl Carrier Protein/ultrastructure , Allosteric Regulation/physiology , Acyl Carrier Protein/physiology , Amino Acid Sequence , Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Magnetic Resonance Spectroscopy/methods , Molecular Docking Simulation/methods , Molecular Dynamics Simulation , Protein Conformation , Protein Interaction Domains and Motifs/physiology , Protein Interaction Maps/physiology
14.
PLoS Biol ; 19(4): e3001198, 2021 04.
Article in English | MEDLINE | ID: mdl-33909608

ABSTRACT

Transactive response DNA-binding Protein of 43 kDa (TDP-43) assembles various aggregate forms, including biomolecular condensates or functional and pathological amyloids, with roles in disparate scenarios (e.g., muscle regeneration versus neurodegeneration). The link between condensates and fibrils remains unclear, just as the factors controlling conformational transitions within these aggregate species: Salt- or RNA-induced droplets may evolve into fibrils or remain in the droplet form, suggesting distinct end point species of different aggregation pathways. Using microscopy and NMR methods, we unexpectedly observed in vitro droplet formation in the absence of salts or RNAs and provided visual evidence for fibrillization at the droplet surface/solvent interface but not the droplet interior. Our NMR analyses unambiguously uncovered a distinct amyloid conformation in which Phe-Gly motifs are key elements of the reconstituted fibril form, suggesting a pivotal role for these residues in creating the fibril core. This contrasts the minor participation of Phe-Gly motifs in initiation of the droplet form. Our results point to an intrinsic (i.e., non-induced) aggregation pathway that may exist over a broad range of conditions and illustrate structural features that distinguishes between aggregate forms.


Subject(s)
DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Dipeptides/chemistry , Protein Aggregates , Amino Acid Sequence , Amyloid/chemistry , Amyloid/metabolism , Chemical Precipitation , Dipeptides/physiology , Humans , Hydrogen-Ion Concentration , Protein Aggregation, Pathological/metabolism , Protein Aggregation, Pathological/pathology , Protein Interaction Domains and Motifs/physiology , Solvents/chemistry , Solvents/pharmacology
15.
Viruses ; 13(2)2021 02 08.
Article in English | MEDLINE | ID: mdl-33567674

ABSTRACT

Nucleolin is an essential cellular receptor to human respiratory syncytial virus (RSV). Pharmacological targeting of the nucleolin RNA binding domain RBD1,2 can inhibit RSV infections in vitro and in vivo; however, the site(s) on RBD1,2 which interact with RSV are not known. We undertook a series of experiments designed to: document RSV-nucleolin co-localization on the surface of polarized MDCK cells using immunogold electron microscopy, to identify domains on nucleolin that physically interact with RSV using biochemical methods and determine their biological effects on RSV infection in vitro, and to carry out structural analysis toward informing future RSV drug development. Results of immunogold transmission and scanning electron microscopy showed RSV-nucleolin co-localization on the cell surface, as would be expected for a viral receptor. RSV, through its fusion protein (RSV-F), physically interacts with RBD1,2 and these interactions can be competitively inhibited by treatment with Palivizumab or recombinant RBD1,2. Treatment with synthetic peptides derived from two 12-mer domains of RBD1,2 inhibited RSV infection in vitro, with structural analysis suggesting these domains are potentially feasible for targeting in drug development. In conclusion, the identification and characterization of domains of nucleolin that interact with RSV provide the essential groundwork toward informing design of novel nucleolin-targeting compounds in RSV drug development.


Subject(s)
Phosphoproteins/metabolism , Protein Interaction Domains and Motifs/physiology , RNA-Binding Proteins/metabolism , Receptors, Virus/metabolism , Respiratory Syncytial Viruses/metabolism , Animals , Antiviral Agents/pharmacology , Cell Line , Dogs , Humans , Immunohistochemistry , Madin Darby Canine Kidney Cells , Microscopy, Electron , Palivizumab/pharmacology , Nucleolin
16.
Biochem Soc Trans ; 49(1): 79-91, 2021 02 26.
Article in English | MEDLINE | ID: mdl-33599752

ABSTRACT

Primary cilia are hair-like projections of the cell membrane supported by an inner microtubule scaffold, the axoneme, which polymerizes out of a membrane-docked centriole at the ciliary base. By working as specialized signaling compartments, primary cilia provide an optimal environment for many G protein-coupled receptors (GPCRs) and their effectors to efficiently transmit their signals to the rest of the cell. For this to occur, however, all necessary receptors and signal transducers must first accumulate at the ciliary membrane. Serotonin receptor 6 (HTR6) and Somatostatin receptor 3 (SSTR3) are two GPCRs whose signaling in brain neuronal cilia affects cognition and is implicated in psychiatric, neurodegenerative, and oncologic diseases. Over a decade ago, the third intracellular loops (IC3s) of HTR6 and SSTR3 were shown to contain ciliary localization sequences (CLSs) that, when grafted onto non-ciliary GPCRs, could drive their ciliary accumulation. Nevertheless, these CLSs were dispensable for ciliary targeting of HTR6 and SSTR3, suggesting the presence of additional CLSs, which we have recently identified in their C-terminal tails. Herein, we review the discovery and mapping of these CLSs, as well as the state of the art regarding how these CLSs may orchestrate ciliary accumulation of these GPCRs by controlling when and where they interact with the ciliary entry and exit machinery via adaptors such as TULP3, RABL2 and the BBSome.


Subject(s)
Cilia/metabolism , Receptors, Serotonin/metabolism , Receptors, Somatostatin/metabolism , Animals , Humans , Protein Interaction Domains and Motifs/physiology , Protein Sorting Signals/genetics , Protein Transport/physiology , Receptors, Serotonin/chemistry , Receptors, Serotonin/genetics , Receptors, Somatostatin/chemistry , Receptors, Somatostatin/genetics
17.
J Biol Chem ; 296: 100432, 2021.
Article in English | MEDLINE | ID: mdl-33610550

ABSTRACT

Nonribosomal peptide synthetases (NRPSs) are multimodular enzymes that produce a wide range of bioactive peptides, such as siderophores, toxins, and antibacterial and insecticidal agents. NRPSs are dynamic proteins characterized by extensive interdomain communications as a consequence of their assembly-line mode of synthesis. Hence, crystal structures of multidomain fragments of NRPSs have aided in elucidating crucial interdomain interactions that occur during different steps of the NRPS catalytic cycle. One crucial yet unexplored interaction is that between the reductase (R) domain and the peptide carrier protein (PCP) domain. R domains are members of the short-chain dehydrogenase/reductase family and function as termination domains that catalyze the reductive release of the final peptide product from the terminal PCP domain of the NRPS. Here, we report the crystal structure of an archaeal NRPS PCP-R didomain construct. This is the first NRPS R domain structure to be determined together with the upstream PCP domain and is also the first structure of an archaeal NRPS to be reported. The structure reveals that a novel helix-turn-helix motif, found in NRPS R domains but not in other short-chain dehydrogenase/reductase family members, plays a major role in the interface between the PCP and R domains. The information derived from the described PCP-R interface will aid in gaining further mechanistic insights into the peptide termination reaction catalyzed by the R domain and may have implications in engineering NRPSs to synthesize novel peptide products.


Subject(s)
Peptide Synthases/metabolism , Peptide Synthases/ultrastructure , Archaea/metabolism , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Carrier Proteins/metabolism , Catalytic Domain/genetics , Gene Expression Regulation, Archaeal/genetics , Models, Molecular , Oxidoreductases/metabolism , Oxidoreductases/ultrastructure , Peptide Biosynthesis, Nucleic Acid-Independent/genetics , Peptide Biosynthesis, Nucleic Acid-Independent/physiology , Peptide Synthases/chemistry , Peptide Synthases/physiology , Peptides/chemistry , Protein Domains/physiology , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology
18.
Mol Pharm ; 18(3): 1285-1292, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33555888

ABSTRACT

Isotonic concentrations of inert cosolutes or excipients are routinely used in protein therapeutic formulations to minimize physical instabilities including aggregation, particulation, and precipitation that are often manifested during drug substance/product manufacture and long-term storage. Despite their prevalent use within the biopharmaceutical industry, a more detailed understanding for how excipients modulate the specific protein-protein interactions responsible for these instabilities is still needed so that informed formulation decisions can be made at the earliest stages of development when protein supply and time are limited. In the present report, subisotonic concentrations of the five common formulation excipients, sucrose, proline, sorbitol, glycerol, arginine hydrochloride, and the denaturant urea, were studied for their effect on the room temperature liquid-liquid phase separation of a model monoclonal antibody (mAb-B). Although each excipient lowered the onset temperatures of mAb-B liquid-liquid phase separation to different extents, all six were found to be preferentially excluded from the native state monomer by vapor pressure osmometry, and no apparent correlations to the excipient dependence of mAb-B melting temperatures were observed. These results and those of the effects of solution pH, addition of salt, and impact of a small number of charge mutations were most consistent with a mechanism of local excipient accumulation, to an extent dependent on their type, with the specific residues that mediate mAb-B electrostatic protein-protein interactions. These findings suggest that selection of excipients on the basis of their interaction with the solvent exposed residues of the native state may at times be a more effective strategy for limiting protein-protein interactions at pharmaceutically relevant storage conditions than choosing those that are excluded from the residues of the native state interior.


Subject(s)
Antibodies, Monoclonal/chemistry , Excipients/chemistry , Chemistry, Pharmaceutical/methods , Hydrogen-Ion Concentration , Protein Interaction Domains and Motifs/physiology , Static Electricity
19.
Sci Signal ; 14(665)2021 01 12.
Article in English | MEDLINE | ID: mdl-33436497

ABSTRACT

The first reported receptor for SARS-CoV-2 on host cells was the angiotensin-converting enzyme 2 (ACE2). However, the viral spike protein also has an RGD motif, suggesting that cell surface integrins may be co-receptors. We examined the sequences of ACE2 and integrins with the Eukaryotic Linear Motif (ELM) resource and identified candidate short linear motifs (SLiMs) in their short, unstructured, cytosolic tails with potential roles in endocytosis, membrane dynamics, autophagy, cytoskeleton, and cell signaling. These SLiM candidates are highly conserved in vertebrates and may interact with the µ2 subunit of the endocytosis-associated AP2 adaptor complex, as well as with various protein domains (namely, I-BAR, LC3, PDZ, PTB, and SH2) found in human signaling and regulatory proteins. Several motifs overlap in the tail sequences, suggesting that they may act as molecular switches, such as in response to tyrosine phosphorylation status. Candidate LC3-interacting region (LIR) motifs are present in the tails of integrin ß3 and ACE2, suggesting that these proteins could directly recruit autophagy components. Our findings identify several molecular links and testable hypotheses that could uncover mechanisms of SARS-CoV-2 attachment, entry, and replication against which it may be possible to develop host-directed therapies that dampen viral infection and disease progression. Several of these SLiMs have now been validated to mediate the predicted peptide interactions.


Subject(s)
COVID-19/virology , Host Microbial Interactions/physiology , SARS-CoV-2/physiology , SARS-CoV-2/pathogenicity , Virus Internalization , Amino Acid Sequence , Angiotensin-Converting Enzyme 2/chemistry , Angiotensin-Converting Enzyme 2/genetics , Angiotensin-Converting Enzyme 2/physiology , Animals , COVID-19/therapy , Conserved Sequence , Host Microbial Interactions/genetics , Humans , Integrins/chemistry , Integrins/genetics , Integrins/physiology , Intrinsically Disordered Proteins/chemistry , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/physiology , Models, Biological , Models, Molecular , Oligopeptides/chemistry , Oligopeptides/genetics , Oligopeptides/physiology , Protein Interaction Domains and Motifs/genetics , Protein Interaction Domains and Motifs/physiology , Protein Sorting Signals/genetics , Protein Sorting Signals/physiology , Receptors, Virus/chemistry , Receptors, Virus/genetics , Receptors, Virus/physiology , SARS-CoV-2/genetics , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/physiology
20.
Methods Mol Biol ; 2251: 133-142, 2021.
Article in English | MEDLINE | ID: mdl-33481236

ABSTRACT

Proximity ligation assay (PLA) is a well-established method for detecting in situ interactions between two epitopes with high resolution and specificity. Notably, PLA is not only a robust method for studying protein-protein interaction but also an efficient approach to characterize and validate protein posttranslational modifications (PTM) using one antibody against the core protein and one against the PTM residue. Therefore, it could be applied as a powerful approach to detect specific interactions of endogenous phosphoinositides and their binding proteins within cells. Importantly, we have specifically detected the PLA signal between PtdIns(4,5)P2 and its binding effector p53 in the nucleus. This cutting-edge method fully complements other conventional approaches for studying phosphoinositide-protein interactions and provides important localization signals and robust quantitation of the detected interactions. Here, we present the PLA fluorescence protocol for detecting in situ phosphoinositide-protein interactions in cultured cells and is semiquantitative for interactions that are regulated by cellular signaling.


Subject(s)
Phosphatidylinositol Phosphates/analysis , Protein Interaction Domains and Motifs/physiology , Protein Interaction Mapping/methods , Animals , Antibodies/chemistry , Cell Line , Cells, Cultured , Fluorescence , Humans , Phosphatidylinositol Phosphates/chemistry , Phosphatidylinositol Phosphates/metabolism , Phosphatidylinositols/metabolism , Protein Binding/physiology , Protein Processing, Post-Translational/genetics , Protein Transport , Proteins/genetics , Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...