Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
J Biol Chem ; 300(6): 107393, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38777143

ABSTRACT

Protein tyrosine phosphatase nonreceptor type 22 (PTPN22) is encoded by a major autoimmunity gene and is a known inhibitor of T cell receptor (TCR) signaling and drug target for cancer immunotherapy. However, little is known about PTPN22 posttranslational regulation. Here, we characterize a phosphorylation site at Ser325 situated C terminal to the catalytic domain of PTPN22 and its roles in altering protein function. In human T cells, Ser325 is phosphorylated by glycogen synthase kinase-3 (GSK3) following TCR stimulation, which promotes its TCR-inhibitory activity. Signaling through the major TCR-dependent pathway under PTPN22 control was enhanced by CRISPR/Cas9-mediated suppression of Ser325 phosphorylation and inhibited by mimicking it via glutamic acid substitution. Global phospho-mass spectrometry showed Ser325 phosphorylation state alters downstream transcriptional activity through enrichment of Swi3p, Rsc8p, and Moira domain binding proteins, and next-generation sequencing revealed it differentially regulates the expression of chemokines and T cell activation pathways. Moreover, in vitro kinetic data suggest the modulation of activity depends on a cellular context. Finally, we begin to address the structural and mechanistic basis for the influence of Ser325 phosphorylation on the protein's properties by deuterium exchange mass spectrometry and NMR spectroscopy. In conclusion, this study explores the function of a novel phosphorylation site of PTPN22 that is involved in complex regulation of TCR signaling and provides details that might inform the future development of allosteric modulators of PTPN22.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 22 , Receptors, Antigen, T-Cell , Signal Transduction , Humans , Phosphorylation , Receptors, Antigen, T-Cell/metabolism , Receptors, Antigen, T-Cell/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Gain of Function Mutation , T-Lymphocytes/metabolism , T-Lymphocytes/immunology , Jurkat Cells , HEK293 Cells
2.
Sci Adv ; 10(5): eadg7887, 2024 Feb 02.
Article in English | MEDLINE | ID: mdl-38295166

ABSTRACT

Protein tyrosine phosphatases (PTPs) play major roles in cancer and are emerging as therapeutic targets. Recent reports suggest low-molecular weight PTP (LMPTP)-encoded by the ACP1 gene-is overexpressed in prostate tumors. We found ACP1 up-regulated in human prostate tumors and ACP1 expression inversely correlated with overall survival. Using CRISPR-Cas9-generated LMPTP knockout C4-2B and MyC-CaP cells, we identified LMPTP as a critical promoter of prostate cancer (PCa) growth and bone metastasis. Through metabolomics, we found that LMPTP promotes PCa cell glutathione synthesis by dephosphorylating glutathione synthetase on inhibitory Tyr270. PCa cells lacking LMPTP showed reduced glutathione, enhanced activation of eukaryotic initiation factor 2-mediated stress response, and enhanced reactive oxygen species after exposure to taxane drugs. LMPTP inhibition slowed primary and bone metastatic prostate tumor growth in mice. These findings reveal a role for LMPTP as a critical promoter of PCa growth and metastasis and validate LMPTP inhibition as a therapeutic strategy for treating PCa through sensitization to oxidative stress.


Subject(s)
Prostatic Neoplasms , Male , Humans , Mice , Animals , Molecular Weight , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Tyrosine , Protein Tyrosine Phosphatases/metabolism
3.
Sci Signal ; 16(792): eabn8668, 2023 07 04.
Article in English | MEDLINE | ID: mdl-37402225

ABSTRACT

Receptor-type protein phosphatase α (RPTPα) promotes fibroblast-dependent arthritis and fibrosis, in part, by enhancing the activation of the kinase SRC. Synovial fibroblasts lining joint tissue mediate inflammation and tissue damage, and their infiltration into adjacent tissues promotes disease progression. RPTPα includes an ectodomain and two intracellular catalytic domains (D1 and D2) and, in cancer cells, undergoes inhibitory homodimerization, which is dependent on a D1 wedge motif. Through single-molecule localization and labeled molecule interaction microscopy of migrating synovial fibroblasts, we investigated the role of RPTPα dimerization in the activation of SRC, the migration of synovial fibroblasts, and joint damage in a mouse model of arthritis. RPTPα clustered with other RPTPα and with SRC molecules in the context of actin-rich structures. A known dimerization-impairing mutation in the wedge motif (P210L/P211L) and the deletion of the D2 domain reduced RPTPα-RPTPα clustering; however, it also unexpectedly reduced RPTPα-SRC association. The same mutations also reduced recruitment of RPTPα to actin-rich structures and inhibited SRC activation and cellular migration. An antibody against the RPTPα ectodomain that prevented the clustering of RPTPα also inhibited RPTPα-SRC association and SRC activation and attenuated fibroblast migration and joint damage in arthritic mice. A catalytically inactivating RPTPα-C469S mutation protected mice from arthritis and reduced SRC activation in synovial fibroblasts. We conclude that RPTPα clustering retains it to actin-rich structures to promote SRC-mediated fibroblast migration and can be modulated through the extracellular domain.


Subject(s)
Actins , Arthritis , Animals , Mice , Cluster Analysis , Fibroblasts/metabolism , Phosphoprotein Phosphatases , Protein Tyrosine Phosphatases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/genetics , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism
4.
J Biol Chem ; 298(12): 102655, 2022 12.
Article in English | MEDLINE | ID: mdl-36328244

ABSTRACT

T-cell protein tyrosine phosphatase (TC-PTP) is a negative regulator of T-cell receptor and oncogenic receptor tyrosine kinase signaling and implicated in cancer and autoimmune disease. TC-PTP activity is modulated by an intrinsically disordered C-terminal region (IDR) and suppressed in cells under basal conditions. In vitro structural studies have shown that the dynamic reorganization of IDR around the catalytic domain, driven by electrostatic interactions, can lead to TC-PTP activity inhibition; however, the process has not been studied in cells. Here, by assessing a mutant (378KRKRPR383 mutated into 378EAAAPE383, called TC45E/A) with impaired tail-PTP domain interaction, we obtained evidence that the downmodulation of TC-PTP enzymatic activity by the IDR occurs in cells. However, we found that the regulation of TC-PTP by the IDR is only recapitulated in vitro when crowding polymers that mimic the intracellular environment are present in kinetic assays using a physiological phosphopeptide. Our FRET-based assays in vitro and in cells confirmed that the effect of the mutant correlates with an impairment of the intramolecular inhibitory remodeling of TC-PTP by the IDR. This work presents an early example of the allosteric regulation of a protein tyrosine phosphatase being controlled by the cellular environment and provides a framework for future studies and targeting of TC-PTP function.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 2 , Signal Transduction , Protein Tyrosine Phosphatase, Non-Receptor Type 2/metabolism , Allosteric Regulation , Signal Transduction/physiology , Phosphorylation
5.
JCI Insight ; 7(8)2022 04 22.
Article in English | MEDLINE | ID: mdl-35451370

ABSTRACT

Systemic sclerosis (SSc) is a fibrotic autoimmune disease characterized by pathogenic activation of fibroblasts enhanced by local oxidative stress. The tyrosine phosphatase PTP4A1 was identified as a critical promoter of TGF-ß signaling in SSc. Oxidative stress is known to functionally inactivate tyrosine phosphatases. Here, we assessed whether oxidation of PTP4A1 modulates its profibrotic action and found that PTP4A1 forms a complex with the kinase SRC in scleroderma fibroblasts, but surprisingly, oxidative stress enhanced rather than reduced PTP4A1's association with SRC and its profibrotic action. Through structural assessment of the oxo-PTP4A1-SRC complex, we unraveled an unexpected mechanism whereby oxidation of a tyrosine phosphatase promotes its function through modification of its protein complex. Considering the importance of oxidative stress in the pathogenesis of SSc and fibrosis, our findings suggest routes for leveraging PTP4A1 oxidation as a potential strategy for developing antifibrotic agents.


Subject(s)
Scleroderma, Systemic , Fibroblasts/metabolism , Fibrosis , Humans , Oxidative Stress , Scleroderma, Systemic/pathology , Tyrosine/metabolism
6.
J Med Chem ; 64(9): 5645-5653, 2021 05 13.
Article in English | MEDLINE | ID: mdl-33914534

ABSTRACT

Obesity-associated insulin resistance plays a central role in the pathogenesis of type 2 diabetes. A promising approach to decrease insulin resistance in obesity is to inhibit the protein tyrosine phosphatases that negatively regulate insulin receptor signaling. The low-molecular-weight protein tyrosine phosphatase (LMPTP) acts as a critical promoter of insulin resistance in obesity by inhibiting phosphorylation of the liver insulin receptor activation motif. Here, we report development of a novel purine-based chemical series of LMPTP inhibitors. These compounds inhibit LMPTP with an uncompetitive mechanism and are highly selective for LMPTP over other protein tyrosine phosphatases. We also report the generation of a highly orally bioavailable purine-based analogue that reverses obesity-induced diabetes in mice.


Subject(s)
Enzyme Inhibitors/chemistry , Protein Tyrosine Phosphatases/antagonists & inhibitors , Purines/chemistry , Administration, Oral , Animals , Binding Sites , Crystallography, X-Ray , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/etiology , Disease Models, Animal , Drug Evaluation, Preclinical , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Half-Life , Humans , Insulin Resistance , Kinetics , Molecular Dynamics Simulation , Obesity/complications , Obesity/pathology , Phosphorylation/drug effects , Protein Tyrosine Phosphatases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Purines/metabolism , Purines/pharmacology , Purines/therapeutic use , Signal Transduction/drug effects , Structure-Activity Relationship
7.
Sci Adv ; 6(26): eaba4353, 2020 06.
Article in English | MEDLINE | ID: mdl-32637608

ABSTRACT

Fibroblast-like synoviocytes (FLS) are joint-lining cells that promote rheumatoid arthritis (RA) pathology. Current disease-modifying antirheumatic agents (DMARDs) operate through systemic immunosuppression. FLS-targeted approaches could potentially be combined with DMARDs to improve control of RA without increasing immunosuppression. Here, we assessed the potential of immunoglobulin-like domains 1 and 2 (Ig1&2), a decoy protein that activates the receptor tyrosine phosphatase sigma (PTPRS) on FLS, for RA therapy. We report that PTPRS expression is enriched in synovial lining RA FLS and that Ig1&2 reduces migration of RA but not osteoarthritis FLS. Administration of an Fc-fusion Ig1&2 attenuated arthritis in mice without affecting innate or adaptive immunity. Furthermore, PTPRS was down-regulated in FLS by tumor necrosis factor (TNF) via a phosphatidylinositol 3-kinase-mediated pathway, and TNF inhibition enhanced PTPRS expression in arthritic joints. Combination of ineffective doses of TNF inhibitor and Fc-Ig1&2 reversed arthritis in mice, providing an example of synergy between FLS-targeted and immunosuppressive DMARD therapies.


Subject(s)
Antirheumatic Agents , Arthritis, Rheumatoid , Synoviocytes , Animals , Antirheumatic Agents/therapeutic use , Cells, Cultured , Fibroblasts/metabolism , Mice , Synoviocytes/metabolism , Synoviocytes/pathology , Tumor Necrosis Factor-alpha/metabolism
8.
Sci Signal ; 13(623)2020 03 17.
Article in English | MEDLINE | ID: mdl-32184287

ABSTRACT

The hematopoietic-specific protein tyrosine phosphatase nonreceptor type 22 (PTPN22) is encoded by a major autoimmunity risk gene. PTPN22 inhibits T cell activation by dephosphorylating substrates involved in proximal T cell receptor (TCR) signaling. Here, we found by mass spectrometry that PTPN22 was phosphorylated at Ser751 by PKCα in Jurkat and primary human T cells activated with phorbol ester/ionomycin or antibodies against CD3/CD28. The phosphorylation of PTPN22 at Ser751 prolonged its half-life by inhibiting K48-linked ubiquitination and impairing recruitment of the phosphatase to the plasma membrane, which is necessary to inhibit proximal TCR signaling. Additionally, the phosphorylation of PTPN22 at Ser751 enhanced the interaction of PTPN22 with the carboxyl-terminal Src kinase (CSK), an interaction that is impaired by the PTPN22 R620W variant associated with autoimmune disease. The phosphorylation of Ser751 did not affect the recruitment of PTPN22 R620W to the plasma membrane but protected this mutant from degradation. Together, out data indicate that phosphorylation at Ser751 mediates a reciprocal regulation of PTPN22 stability versus translocation to TCR signaling complexes by CSK-dependent and CSK-independent mechanisms.


Subject(s)
Protein Tyrosine Phosphatase, Non-Receptor Type 22/metabolism , Receptors, Antigen, T-Cell/metabolism , Serine/metabolism , Signal Transduction , Autoimmune Diseases/genetics , Autoimmune Diseases/metabolism , CSK Tyrosine-Protein Kinase/metabolism , Cells, Cultured , HEK293 Cells , Humans , Jurkat Cells , Mass Spectrometry/methods , Mutation, Missense , Phosphorylation , Protein Tyrosine Phosphatase, Non-Receptor Type 22/genetics , Serine/genetics , T-Lymphocytes/metabolism
9.
J Biol Chem ; 295(15): 4923-4936, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32139509

ABSTRACT

Receptor-type protein tyrosine phosphatase α (RPTPα) is an important positive regulator of SRC kinase activation and a known promoter of cancer growth, fibrosis, and arthritis. The domain structure of RPTPs comprises an extracellular region, a transmembrane helix, and two tandem intracellular catalytic domains referred to as D1 and D2. The D2 domain of RPTPs is believed to mostly play a regulatory function; however, no regulatory model has been established for RPTPα-D2 or other RPTP-D2 domains. Here, we solved the 1.8 Å resolution crystal structure of the cytoplasmic region of RPTPα, encompassing D1 and D2, trapped in a conformation that revealed a possible mechanism through which D2 can allosterically inhibit D1 activity. Using a D2-truncation RPTPα variant and mutational analysis of the D1/D2 interfaces, we show that D2 inhibits RPTPα phosphatase activity and identified a 405PFTP408 motif in D1 that mediates the inhibitory effect of D2. Expression of the gain-of-function F406A/T407A RPTPα variant in HEK293T cells enhanced SRC activation, supporting the relevance of our proposed D2-mediated regulation mechanism in cell signaling. There is emerging interest in the development of allosteric inhibitors of RPTPs but a scarcity of validated allosteric sites for RPTPs. The results of our study not only shed light on the regulatory role of RPTP-D2 domains, but also provide a potentially useful tool for the discovery of chemical probes targeting RPTPα and other RPTPs.


Subject(s)
Cell Membrane/metabolism , Protein Tyrosine Phosphatases/metabolism , Receptor-Like Protein Tyrosine Phosphatases, Class 4/chemistry , Receptor-Like Protein Tyrosine Phosphatases, Class 4/metabolism , Allosteric Regulation , Amino Acid Sequence , Catalytic Domain , Crystallography, X-Ray , Humans , Models, Molecular , Protein Binding , Protein Conformation , Protein Tyrosine Phosphatases/chemistry , Sequence Homology
10.
J Clin Invest ; 129(3): 1193-1210, 2019 03 01.
Article in English | MEDLINE | ID: mdl-30620725

ABSTRACT

Genetic variants at the PTPN2 locus, which encodes the tyrosine phosphatase PTPN2, cause reduced gene expression and are linked to rheumatoid arthritis (RA) and other autoimmune diseases. PTPN2 inhibits signaling through the T cell and cytokine receptors, and loss of PTPN2 promotes T cell expansion and CD4- and CD8-driven autoimmunity. However, it remains unknown whether loss of PTPN2 in FoxP3+ regulatory T cells (Tregs) plays a role in autoimmunity. Here we aimed to model human autoimmune-predisposing PTPN2 variants, the presence of which results in a partial loss of PTPN2 expression, in mouse models of RA. We identified that reduced expression of Ptpn2 enhanced the severity of autoimmune arthritis in the T cell-dependent SKG mouse model and demonstrated that this phenotype was mediated through a Treg-intrinsic mechanism. Mechanistically, we found that through dephosphorylation of STAT3, PTPN2 inhibits IL-6-driven pathogenic loss of FoxP3 after Tregs have acquired RORγt expression, at a stage when chromatin accessibility for STAT3-targeted IL-17-associated transcription factors is maximized. We conclude that PTPN2 promotes FoxP3 stability in mouse RORγt+ Tregs and that loss of function of PTPN2 in Tregs contributes to the association between PTPN2 and autoimmunity.


Subject(s)
Arthritis, Rheumatoid/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/pathology , Disease Models, Animal , Female , Forkhead Transcription Factors/genetics , Forkhead Transcription Factors/immunology , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-6/genetics , Interleukin-6/immunology , Mice , Mice, Inbred BALB C , Mice, Knockout , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Nuclear Receptor Subfamily 1, Group F, Member 3/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/immunology , T-Lymphocytes, Regulatory/pathology
11.
Nat Commun ; 8(1): 1060, 2017 10 20.
Article in English | MEDLINE | ID: mdl-29057934

ABSTRACT

Systemic sclerosis (SSc) is an autoimmune disease characterized by fibrosis of skin and internal organs. Protein tyrosine phosphatases have received little attention in the study of SSc or fibrosis. Here, we show that the tyrosine phosphatase PTP4A1 is highly expressed in fibroblasts from patients with SSc. PTP4A1 and its close homolog PTP4A2 are critical promoters of TGFß signaling in primary dermal fibroblasts and of bleomycin-induced fibrosis in vivo. PTP4A1 promotes TGFß signaling in human fibroblasts through enhancement of ERK activity, which stimulates SMAD3 expression and nuclear translocation. Upstream from ERK, we show that PTP4A1 directly interacts with SRC and inhibits SRC basal activation independently of its phosphatase activity. Unexpectedly, PTP4A2 minimally interacts with SRC and does not promote the SRC-ERK-SMAD3 pathway. Thus, in addition to defining PTP4A1 as a molecule of interest for TGFß-dependent fibrosis, our study provides information regarding the functional specificity of different members of the PTP4A subclass of phosphatases.


Subject(s)
Immediate-Early Proteins/metabolism , MAP Kinase Signaling System , Protein Tyrosine Phosphatases/metabolism , Scleroderma, Systemic/enzymology , Transforming Growth Factor beta/physiology , Animals , Cell Line , Dermis/pathology , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblasts/enzymology , Fibroblasts/metabolism , Humans , Immediate-Early Proteins/antagonists & inhibitors , Immediate-Early Proteins/genetics , Immediate-Early Proteins/physiology , Mice, Inbred C57BL , Mice, Knockout , Protein Tyrosine Phosphatases/antagonists & inhibitors , Protein Tyrosine Phosphatases/genetics , Protein Tyrosine Phosphatases/physiology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/pathology , Smad3 Protein/metabolism
12.
J Gen Physiol ; 148(4): 313-24, 2016 10.
Article in English | MEDLINE | ID: mdl-27670897

ABSTRACT

Anthrax toxin comprises three soluble proteins: protective antigen (PA), lethal factor (LF), and edema factor (EF). PA must be cleaved by host proteases before it oligomerizes and forms a prepore, to which LF and EF bind. After endocytosis of this tripartite complex, the prepore transforms into a narrow transmembrane pore that delivers unfolded LF and EF into the host cytosol. Here, we find that translocation of multiple 90-kD LF molecules is rapid and efficient. To probe the molecular basis of this translocation, we calculated a three-dimensional map of the fully loaded (PA63)7-(LF)3 prepore complex by cryo-electron microscopy (cryo-EM). The map shows three LFs bound in a similar way to one another, via their N-terminal domains, to the surface of the PA heptamer. The model also reveals contacts between the N- and C-terminal domains of adjacent LF molecules. We propose that this molecular arrangement plays an important role in the maintenance of translocation efficiency through the narrow PA pore.


Subject(s)
Antigens, Bacterial/chemistry , Bacterial Toxins/chemistry , Biological Transport , Escherichia coli , Models, Molecular , Protein Conformation , Protein Folding , Structure-Activity Relationship
13.
Chem Biol ; 20(8): 973-82, 2013 Aug 22.
Article in English | MEDLINE | ID: mdl-23891150

ABSTRACT

The E3 ubiquitin ligase Siah regulates key cellular events that are central to cancer development and progression. A promising route to Siah inhibition is disrupting its interactions with adaptor proteins. However, typical of protein-protein interactions, traditional unbiased approaches to ligand discovery did not produce viable hits against this target, despite considerable effort and a multitude of approaches. Ultimately, a rational structure-based design strategy was successful for the identification of Siah inhibitors in which peptide binding drives specific covalent bond formation with the target. X-ray crystallography, mass spectrometry, and functional data demonstrate that these peptide mimetics are efficient covalent inhibitors of Siah and antagonize Siah-dependent regulation of Erk and Hif signaling in the cell. The proposed strategy may result useful as a general approach to the design of peptide-based inhibitors of other protein-protein interactions.


Subject(s)
Drug Design , Enzyme Inhibitors/chemistry , Nuclear Proteins/antagonists & inhibitors , Peptides/chemistry , Peptidomimetics/chemistry , Ubiquitin-Protein Ligases/antagonists & inhibitors , Amino Acid Sequence , Animals , Binding Sites , Crystallography, X-Ray , Enzyme Inhibitors/pharmacology , Humans , Models, Molecular , Molecular Sequence Data , Nuclear Proteins/chemistry , Nuclear Proteins/metabolism , Peptides/pharmacology , Peptidomimetics/pharmacology , Protein Interaction Maps/drug effects , Signal Transduction/drug effects , Ubiquitin-Protein Ligases/chemistry , Ubiquitin-Protein Ligases/metabolism
14.
J Biol Chem ; 286(10): 8448-8458, 2011 Mar 11.
Article in English | MEDLINE | ID: mdl-21216948

ABSTRACT

Pathogenesis by Bacillus anthracis requires coordination between two distinct activities: plasmid-encoded virulence factor expression (which protects vegetative cells from immune surveillance during outgrowth and replication) and chromosomally encoded sporulation (required only during the final stages of infection). Sporulation is regulated by at least five sensor histidine kinases that are activated in response to various environmental cues. One of these kinases, BA2291, harbors a sensor domain that has ∼35% sequence identity with two plasmid proteins, pXO1-118 and pXO2-61. Because overexpression of pXO2-61 (or pXO1-118) inhibits sporulation of B. anthracis in a BA2291-dependent manner, and pXO2-61 expression is strongly up-regulated by the major virulence gene regulator, AtxA, it was suggested that their function is to titrate out an environmental signal that would otherwise promote untimely sporulation. To explore this hypothesis, we determined crystal structures of both plasmid-encoded proteins. We found that they adopt a dimeric globin fold but, most unusually, do not bind heme. Instead, they house a hydrophobic tunnel and hydrophilic chamber that are occupied by fatty acid, which engages a conserved arginine and chloride ion via its carboxyl head group. In vivo, these domains may therefore recognize changes in fatty acid synthesis, chloride ion concentration, and/or pH. Structure-based comparisons with BA2291 suggest that it binds ligand and dimerizes in an analogous fashion, consistent with the titration hypothesis. Analysis of newly sequenced bacterial genomes points to the existence of a much broader family of non-heme, globin-based sensor domains, with related but distinct functionalities, that may have evolved from an ancestral heme-linked globin.


Subject(s)
Bacillus anthracis/chemistry , Bacterial Proteins/chemistry , Protein Folding , Protein Multimerization/physiology , Trans-Activators/chemistry , Virulence Factors/chemistry , Bacillus anthracis/genetics , Bacillus anthracis/metabolism , Bacillus anthracis/pathogenicity , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Crystallography, X-Ray , Protein Structure, Quaternary , Protein Structure, Tertiary , Trans-Activators/genetics , Trans-Activators/metabolism , Virulence Factors/genetics , Virulence Factors/metabolism
15.
J Nanosci Nanotechnol ; 10(8): 5293-301, 2010 Aug.
Article in English | MEDLINE | ID: mdl-21125885

ABSTRACT

Nanotechnology-introduced materials have promising applications as nanocarriers for drugs, peptides, proteins and nucleic acids. Several studies showed that the geometry (shape and size) and chemical properties of nanoparticles affect the kinetics and pathways of cellular uptake and their intracellular trafficking and signaling. Accurate physico-chemical characterization of nanoparticles customarily precedes their use in cell biology and in vivo experiments. However, a fact that is easily overlooked is that nanomaterials decorated with organic matter or resuspended in aqueous buffers can be theoretically contaminated by fungal and bacterial microorganisms. While investigating the effects of extensively characterized PEGylated carbon nanotubes (PNTs) on T lymphocyte activation, we demonstrated bacterial contamination of PNTs, which correlated with low reproducibility and artifacts in cell signaling assays. Contamination and artifacts were easily eliminated by preparing the materials in sterile conditions. We propose that simple sterile preparation procedures should be adopted and sterility evaluation of nanoparticles should be customarily performed, prior to assessing nanoparticle intracellular internalization, trafficking and their effects on cells and entire organisms.


Subject(s)
Drug Carriers , Drug Contamination , Nanoconjugates/microbiology , Nanotubes, Carbon/microbiology , Artifacts , Endocytosis/drug effects , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Interleukin-2/genetics , Interleukin-2/metabolism , Jurkat Cells , Microscopy, Atomic Force , Microscopy, Confocal , Nanoconjugates/chemistry , Nanotubes, Carbon/chemistry , Particle Size , Polyethylene Glycols , Reproducibility of Results , T-Lymphocytes/drug effects , T-Lymphocytes/metabolism , Toll-Like Receptors/metabolism
16.
Protein Sci ; 19(1): 155-61, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19845004

ABSTRACT

Plants use a highly evolved immune system to exhibit defense response against microbial infections. The plant TIR domain, together with the nucleotide-binding (NB) domain and/or a LRR region, forms a type of molecule, named resistance (R) proteins, that interact with microbial effector proteins and elicit hypersensitive responses against infection. Here, we report the first crystal structure of a plant TIR domain from Arabidopsis thaliana (AtTIR) solved at a resolution of 2.0 A. The structure consists of five beta-strands forming a parallel beta-sheet at the core of the protein. The beta-strands are connected by a series of alpha-helices and the overall fold mimics closely that of other mammalian and bacterial TIR domains. However, the region of the alphaD-helix reveals significant differences when compared with other TIR structures, especially the alphaD3-helix that corresponds to an insertion only present in plant TIR domains. Available mutagenesis data suggest that several conserved and exposed residues in this region are involved in the plant TIR signaling function.


Subject(s)
Arabidopsis Proteins/chemistry , Arabidopsis/chemistry , Amino Acid Sequence , Arabidopsis/metabolism , Arabidopsis Proteins/genetics , Arabidopsis Proteins/metabolism , Conserved Sequence , Crystallography, X-Ray/methods , Escherichia coli/genetics , Models, Molecular , Molecular Sequence Data , Protein Interaction Mapping , Protein Structure, Secondary , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Sequence Alignment
17.
J Biol Chem ; 284(32): 21386-92, 2009 Aug 07.
Article in English | MEDLINE | ID: mdl-19535337

ABSTRACT

Macrophages detect pathogen infection via the activation of their plasma membrane-bound Toll-like receptor proteins (TLRs). The heterotypic interaction between the Toll/interleukin-1 receptor (TIR) domains of TLRs and adaptor proteins, like Myeloid differentiation primary response gene 88 (MyD88), is the first intracellular step in the signaling pathway of the mammalian innate immune response. The hetero-oligomerization of the TIRs of the receptor and adaptor brings about the activation of the transcription factor NF-kappaB, which regulates the synthesis of pro-inflammatory cytokines. Here, we report the first crystal structure of a bacterial TIR domain solved at 2.5 A resolution. The three-dimensional fold of Paracoccus denitrificans TIR is identical to that observed for the TIR of human TLRs and MyD88 proteins. The structure shows a unique dimerization interface involving the DD-loop and EE-loop residues, whereas leaving the BB-loop highly exposed. Peptide amide hydrogen-deuterium exchange mass spectrometry also reveals that the same region is used for dimerization in solution and in the context of the full-length protein. These results, together with a functional interaction between P. denitrificans TIR and MyD88 visualized in a co-immunoprecipitation assay, further substantiate the model that bacterial TIR proteins adopt structural mimicry of the host active receptor TIR domains to interfere with the signaling of TLRs and their adaptors to decrease the inflammatory response.


Subject(s)
Bacterial Proteins/chemistry , Escherichia coli/metabolism , Myeloid Differentiation Factor 88/physiology , Paracoccus denitrificans/metabolism , Crystallography, X-Ray/methods , Cytokines/metabolism , Dimerization , Humans , Immunoprecipitation , Inflammation , Models, Biological , Models, Molecular , Molecular Conformation , Myeloid Differentiation Factor 88/chemistry , Protein Structure, Tertiary , Toll-Like Receptors/chemistry
18.
Nat Struct Mol Biol ; 16(3): 265-73, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19234466

ABSTRACT

Influenza virus remains a serious health threat, owing to its ability to evade immune surveillance through rapid genetic drift and reassortment. Here we used a human non-immune antibody phage-display library and the H5 hemagglutinin ectodomain to select ten neutralizing antibodies (nAbs) that were effective against all group 1 influenza viruses tested, including H5N1 'bird flu' and the H1N1 'Spanish flu'. The crystal structure of one such nAb bound to H5 shows that it blocks infection by inserting its heavy chain into a conserved pocket in the stem region, thus preventing membrane fusion. Nine of the nAbs employ the germline gene VH1-69, and all seem to use the same neutralizing mechanism. Our data further suggest that this region is recalcitrant to neutralization escape and that nAb-based immunotherapy is a promising strategy for broad-spectrum protection against seasonal and pandemic influenza viruses.


Subject(s)
Antibodies, Viral/chemistry , Antibodies, Viral/immunology , Hemagglutinin Glycoproteins, Influenza Virus/chemistry , Influenza A Virus, H1N1 Subtype/immunology , Influenza A Virus, H5N1 Subtype/immunology , Virus Internalization/drug effects , Amino Acid Sequence , Animals , Antibodies, Viral/therapeutic use , Crystallography, X-Ray , HeLa Cells , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Humans , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Neutralization Tests , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Peptide Library , Protein Binding , Protein Structure, Quaternary , Sequence Alignment , Survival Analysis
19.
J Biol Chem ; 283(4): 2098-107, 2008 Jan 25.
Article in English | MEDLINE | ID: mdl-18006495

ABSTRACT

The transthyretin amyloidoses appear to be caused by rate-limiting tetramer dissociation and partial monomer unfolding of the human serum protein transthyretin, resulting in aggregation and extracellular deposition of amorphous aggregates and amyloid fibrils. Mice transgenic for few copies of amyloid-prone human transthyretin variants, including the aggressive L55P mutant, failed to develop deposits. Silencing the murine transthyretin gene in the presence of the L55P human gene resulted in enhanced tissue deposition. To test the hypothesis that the murine protein interacted with human transthyretin, preventing the dissociation and partial unfolding required for amyloidogenesis, we produced recombinant murine transthyretin and human/murine transthyretin heterotetramers and compared their structures and biophysical properties to recombinant human transthyretin. We found no significant differences between the crystal structures of murine and human homotetramers. Murine transthyretin is not amyloidogenic because the native homotetramer is kinetically stable under physiologic conditions and cannot dissociate into partially unfolded monomers, the misfolding and aggregation precursor. Heterotetramers composed of murine and human subunits are also kinetically stable. These observations explain the lack of transthyretin deposition in transgenics carrying a low copy number of human transthyretin genes. The incorporation of mouse subunits into tetramers otherwise composed of human amyloid-prone transthyretin subunits imposes kinetic stability, preventing dissociation and subsequent amyloidogenesis.


Subject(s)
Amyloid/chemistry , Prealbumin/chemistry , Protein Folding , Amyloid/genetics , Amyloid/metabolism , Animals , Crystallography, X-Ray , Humans , Kinetics , Mice , Prealbumin/genetics , Prealbumin/metabolism , Protein Structure, Quaternary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics , Recombinant Proteins/metabolism , Species Specificity
20.
PLoS One ; 2(10): e1044, 2007 Oct 17.
Article in English | MEDLINE | ID: mdl-17940608

ABSTRACT

B. anthracis is the causative agent of anthrax. Pathogenesis is primarily mediated through the exotoxins lethal factor and edema factor, which bind protective antigen (PA) to gain entry into the host cell. The current anthrax vaccine (AVA, Biothrax) consists of aluminum-adsorbed cell-free filtrates of unencapsulated B. anthracis, wherein PA is thought to be the principle target of neutralization. In this study, we evaluated the efficacy of the natural adjuvant, C3d, versus alum in eliciting an anti-PA humoral response and found that C3d conjugation to PA and emulsion in incomplete Freund's adjuvant (IFA) imparted superior protection from anthrax challenge relative to PA in IFA or PA adsorbed to alum. Relative to alum-PA, immunization of mice with C3d-PA/IFA augmented both the onset and sustained production of PA-specific antibodies, including neutralizing antibodies to the receptor-binding portion (domain 4) of PA. C3d-PA/IFA was efficacious when administered either i.p. or s.c., and in adolescent mice lacking a fully mature B cell compartment. Induction of PA-specific antibodies by C3d-PA/IFA correlated with increased efficiency of germinal center formation and plasma cell generation. Importantly, C3d-PA immunization effectively protected mice from intranasal challenge with B. anthracis spores, and was approximately 10-fold more effective than alum-PA immunization or PA/IFA based on dose challenge. These data suggest that incorporation of C3d as an adjuvant may overcome shortcomings of the currently licensed aluminum-based vaccine, and may confer protection in the early days following acute anthrax exposure.


Subject(s)
Antigens, Bacterial/chemistry , Bacillus anthracis/metabolism , Bacterial Toxins/chemistry , Complement C3d/chemistry , Adsorption , Aluminum/chemistry , Animals , Anthrax Vaccines/chemistry , Antibodies, Bacterial , Antibody Formation , Cell-Free System , Freund's Adjuvant/chemistry , Mice , Mice, Inbred BALB C , Mice, Inbred C3H
SELECTION OF CITATIONS
SEARCH DETAIL
...