Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 74
Filter
1.
Appl Environ Microbiol ; 89(5): e0033523, 2023 05 31.
Article in English | MEDLINE | ID: mdl-37078870

ABSTRACT

Small proteins perform a diverse array of functions, from microbial competition, to endocrine signaling, to building biomaterials. Microbial systems that can produce recombinant small proteins enable discovery of new effectors, exploration of sequence activity relationships, and have the potential for in vivo delivery. However, we lack simple systems for controlling small-protein secretion from Gram-negative bacteria. Microcins are small-protein antibiotics secreted by Gram-negative bacteria that inhibit the growth of neighboring microbes. They are exported from the cytosol to the environment in a one-step process through a specific class of type I secretion systems (T1SSs). However, relatively little is known about substrate requirements for small proteins exported through microcin T1SSs. Here, we investigate the prototypic microcin V T1SS from Escherichia coli and show that it can export a remarkably wide range of natural and synthetic small proteins. We demonstrate that secretion is largely independent of the cargo protein's chemical properties and appears to be constrained only by protein length. We show that a varied range of bioactive sequences, including an antibacterial protein, a microbial signaling factor, a protease inhibitor, and a human hormone, can all be secreted and elicit their intended biological effect. Secretion through this system is not limited to E. coli, and we demonstrate its function in additional Gram-negative species that can inhabit the gastrointestinal tract. Our findings uncover the highly promiscuous nature of small-protein export through the microcin V T1SS, which has implications for native-cargo capacity and the use of this system in Gram-negative bacteria for small-protein research and delivery. IMPORTANCE Type I secretion systems for microcin export in Gram-negative bacteria transport small antibacterial proteins from the cytoplasm to the extracellular environment in a single step. In nature, each secretion system is generally paired with a specific small protein. We know little about the export capacity of these transporters and how cargo sequence influences secretion. Here, we investigate the microcin V type I system. Remarkably, our studies show that this system can export small proteins of diverse sequence composition and is only limited by protein length. Furthermore, we demonstrate that a wide range of bioactive small proteins can be secreted and that this system can be used in Gram-negative species that colonize the gastrointestinal tract. These findings expand our understanding of secretion through type I systems and their potential uses in a variety of small-protein applications.


Subject(s)
Escherichia coli , Type I Secretion Systems , Humans , Escherichia coli/genetics , Escherichia coli/metabolism , Membrane Transport Proteins/metabolism , Gram-Negative Bacteria/metabolism , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/genetics , Bacterial Proteins/metabolism
2.
bioRxiv ; 2023 Jan 26.
Article in English | MEDLINE | ID: mdl-36747863

ABSTRACT

Microcins are peptide antibiotics secreted by Gram-negative bacteria that inhibit the growth of neighboring microbes. They are exported from the cytosol to the environment in a one-step process through a specific type I secretion system (T1SS). While the rules governing export of natural or non-native substrates have been resolved for T1SSs that secrete large proteins, relatively little is known about substrate requirements for peptides exported through T1SSs that secrete microcins. Here, we investigate the prototypic microcin V T1SS from Escherichia coli and show it can export a remarkably wide range of natural and synthetic peptides. We demonstrate that secretion through this system is not affected by peptide charge or hydrophobicity and appears only constrained by peptide length. A varied range of bioactive peptides, including an antibacterial peptide, a microbial signaling factor, a protease inhibitor, and a human hormone, can all be secreted and elicit their intended biological effect. Secretion through this system is not limited to E. coli , and we demonstrate its function in additional Gram-negative species that can inhabit the gastrointestinal tract. Our findings uncover the highly promiscuous nature of peptide export thorough the microcin V T1SS, which has implications for native cargo capacity and use of Gram-negative bacteria for peptide research and delivery. Importance: Microcin type I secretion systems in Gram-negative bacteria transport antibacterial peptides from the cytoplasm to the extracellular environment in single step. In nature, each microcin secretion system is generally paired with a specific peptide. We know little about the export capacity of these transporters and how peptide sequence influences secretion. Here, we investigate the microcin V type I secretion system. Remarkably, our studies show this system can export diverse peptides and is only limited by peptide length. Furthermore, we demonstrate that various bioactive peptides can be secreted, and this system can be used in Gram-negative species that colonize the gastrointestinal tract. These finding expand our understanding of secretion through type I systems and their potential uses in peptide applications.

3.
Biochem J ; 479(24): 2465-2475, 2022 12 19.
Article in English | MEDLINE | ID: mdl-36416757

ABSTRACT

The Epidermal Growth Factor Receptor (EGFR) is a Receptor Tyrosine Kinase that mediates cell proliferation and differentiation events during development and maintenance of complex organisms. Formation of specific, ligand-dependent EGFR dimers is a key step in stimulating EGFR signaling, and crystal structures of active, dimeric forms of isolated EGFR extracellular regions and kinase domains have revealed much about how dimer interactions regulate EGFR activity. The nature and role of the transmembrane region in regulating EGFR activity remains less clear, however. Proposed roles for the transmembrane region range from nonspecific but energetically favorable interactions to specific transmembrane dimer conformations being associated with active, inactive, or activity-modulated states of EGFR. To investigate the role of specific transmembrane dimers in modulating EGFR activity we generated thirteen EGFR variants with altered transmembrane sequences designed to favor or disfavor specific types of transmembrane region interactions. We show using FRET microscopy that EGFR transmembrane regions have an intrinsic propensity to associate in mammalian cell membranes that is counteracted by the extracellular region. We show using cell-based assays that each of the EGFR transmembrane variants except the Neu variant, which results in constitutive receptor phosphorylation, is able to autophosphorylate and stimulate phosphorylation of downstream effectors Erk and Akt. Our results indicate that many transmembrane sequences, including polyleucine, are compatible with EGFR activity and provide no evidence for specific transmembrane dimers regulating EGFR function.


Subject(s)
ErbB Receptors , Signal Transduction , Animals , Phosphorylation , ErbB Receptors/metabolism , Signal Transduction/physiology , Cell Membrane/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Mammals/metabolism
4.
Front Sports Act Living ; 4: 961957, 2022.
Article in English | MEDLINE | ID: mdl-35992158

ABSTRACT

Background: Many adults do not reach the recommended exercise participation guidelines, often citing lack of time as a barrier. Reduced exertion high-intensity training (REHIT) is a mode of exercise that takes as few as 10 min and has been shown to be as effective as other modalities. The Fitness Fatness Index (FFI) is a recently developed index that is used to predict cardiovascular disease (CVD) risk. The aim of this study was to determine the efficacy of a REHIT vs. a traditional moderate-intensity continuous training (MICT) on FFI in physically inactive adults. Methods: Thirty-two participants were randomized into one of two 8-week exercise intervention groups: (i) REHIT (n = 16); (ii) MICT (n = 16). The REHIT group performed 10 min of individualized cycling intervals on 2-4 days of the week. The MICT group were prescribed aerobic exercise at 50-65% of their heart rate reserve (HRR) on 3-5 days of the week. FFI was recorded at baseline and post 8-weeks, with FFI being calculated as cardiorespiratory fitness (CRF) (expressed as metabolic equivalents) divided by waist to height ratio (WtHR). A 1-unit increase in FFI was recognized as a clinically significant change in FFI. Results: The REHIT group showed significantly greater (+1.95, ±0.63) improvements in FFI compared to those in the MICT (+0.99, ±0.47) group (between group difference, p < 0.001). Furthermore, there was a greater proportion of participants who achieved a clinically significant change in FFI in the REHIT group (12/12, 100%) than in the MICT group (8/15, 53%) (between group difference, p = 0.01). Conclusion: This study suggests that REHIT may be a more efficacious exercise modality to increase FFI than MICT. This outcome is beneficial as the clinician can prescribe REHIT to physically inactive adults who cite lack of time as a barrier to physical activity participation and achieve significant reductions in CVD risk.

5.
Biochem J ; 477(21): 4207-4220, 2020 11 13.
Article in English | MEDLINE | ID: mdl-33043983

ABSTRACT

Receptor Tyrosine Kinases (RTKs) comprise a diverse group of cell-surface receptors that mediate key signaling events during animal development and are frequently activated in cancer. We show here that deletion of the extracellular regions of 10 RTKs representing 7 RTK classes or their substitution with the dimeric immunoglobulin Fc region results in constitutive receptor phosphorylation but fails to result in phosphorylation of downstream signaling effectors Erk or Akt. Conversely, substitution of RTK extracellular regions with the extracellular region of the Epidermal Growth Factor Receptor (EGFR) results in increases in effector phosphorylation in response to EGF. These results indicate that the activation signal generated by the EGFR extracellular region is capable of activating at least seven different RTK classes. Failure of phosphorylated Fc-RTK chimeras or RTKs with deleted extracellular regions to stimulate phosphorylation of downstream effectors indicates that either dimerization and receptor phosphorylation per se are insufficient to activate signaling or constitutive dimerization leads to pathway inhibition.


Subject(s)
ErbB Receptors/metabolism , Receptor Protein-Tyrosine Kinases/metabolism , Animals , ErbB Receptors/genetics , Humans , Phosphorylation/genetics , Phosphorylation/physiology , Receptor Protein-Tyrosine Kinases/genetics , Signal Transduction/genetics , Signal Transduction/physiology
6.
J Biol Chem ; 295(38): 13353-13362, 2020 09 18.
Article in English | MEDLINE | ID: mdl-32727847

ABSTRACT

The human epidermal growth factor receptor (EGFR/ERBB1) is a receptor tyrosine kinase (RTK) that forms activated oligomers in response to ligand. Much evidence indicates that EGFR/ERBB1 also forms oligomers in the absence of ligand, but the structure and physiological role of these ligand-independent oligomers remain unclear. To examine these features, we use fluorescence microscopy to measure the oligomer stability and FRET efficiency for homo- and hetero-oligomers of fluorescent protein-labeled forms of EGFR and its paralog, human epidermal growth factor receptor 2 (HER2/ERBB2) in vesicles derived from mammalian cell membranes. We observe that both receptors form ligand-independent oligomers at physiological plasma membrane concentrations. Mutations introduced in the kinase region at the active state asymmetric kinase dimer interface do not affect the stability of ligand-independent EGFR oligomers. These results indicate that ligand-independent EGFR oligomers form using interactions that are distinct from the EGFR active state.


Subject(s)
Protein Multimerization , Animals , CHO Cells , Cricetulus , ErbB Receptors/genetics , ErbB Receptors/metabolism , Fluorescence Resonance Energy Transfer , Humans , Mutation , Protein Domains , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism
7.
Structure ; 28(3): 355-362.e4, 2020 03 03.
Article in English | MEDLINE | ID: mdl-31995743

ABSTRACT

Intermediate filaments (IFs) provide vital mechanical support in a broad array of cell types. Interference with this role causes cell fragility and accounts for a large number of human diseases. Gaining an understanding of the structure of IFs is paramount to understanding their function and designing therapeutic agents for relevant diseases. Here, we report the 2.6-Å resolution crystal structure of a complex of interacting 2B domains of keratin 5 (K5) and K14. K5 and K14 form a long-range, left-handed coiled coil, with participating α helices aligned in parallel and in register. Follow-up mutagenesis revealed that specific contacts between interacting 2B domains play a crucial role during 10-nm IF assembly, likely at the step of octamer-octamer association. The resulting structural model represents an atomic-resolution visualization of 2B-2B interactions important to filament assembly and provides insight into the defects introduced by mutations in IF genes associated with human skin diseases.


Subject(s)
Keratin-14/chemistry , Keratin-14/metabolism , Keratin-5/chemistry , Keratin-5/metabolism , Mutation , Animals , Crystallography, X-Ray , Humans , Intermediate Filaments/metabolism , Keratin-14/genetics , Keratin-5/genetics , Mice , Models, Molecular , Mutagenesis, Site-Directed , NIH 3T3 Cells , Protein Domains , Protein Multimerization , Protein Structure, Secondary
8.
Mol Cell ; 75(1): 13-25.e5, 2019 07 11.
Article in English | MEDLINE | ID: mdl-31151856

ABSTRACT

Arc is a synaptic protein essential for memory consolidation. Recent studies indicate that Arc originates in evolution from a Ty3-Gypsy retrotransposon GAG domain. The N-lobe of Arc GAG domain acquired a hydrophobic binding pocket in higher vertebrates that is essential for Arc's canonical function to weaken excitatory synapses. Here, we report that Arc GAG also acquired phosphorylation sites that can acutely regulate its synaptic function. CaMKII phosphorylates the N-lobe of the Arc GAG domain and disrupts an interaction surface essential for high-order oligomerization. In Purkinje neurons, CaMKII phosphorylation acutely reverses Arc's synaptic action. Mutant Arc that cannot be phosphorylated by CaMKII enhances metabotropic receptor-dependent depression in the hippocampus but does not alter baseline synaptic transmission or long-term potentiation. Behavioral studies indicate that hippocampus- and amygdala-dependent learning requires Arc GAG domain phosphorylation. These studies provide an atomic model for dynamic and local control of Arc function underlying synaptic plasticity and memory.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cytoskeletal Proteins/metabolism , Long-Term Potentiation/physiology , Memory/physiology , Nerve Tissue Proteins/metabolism , Purkinje Cells/metabolism , Amino Acid Sequence , Amygdala/cytology , Amygdala/metabolism , Animals , Binding Sites , Calcium-Calmodulin-Dependent Protein Kinase Type 2/chemistry , Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Cytoskeletal Proteins/chemistry , Cytoskeletal Proteins/genetics , Gene Knock-In Techniques , HEK293 Cells , Hippocampus/cytology , Hippocampus/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Models, Molecular , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/genetics , Phosphorylation , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Protein Multimerization , Purkinje Cells/cytology , Sequence Alignment , Sequence Homology, Amino Acid , Synapses/physiology , Synaptic Transmission
9.
Cell ; 171(3): 683-695.e18, 2017 Oct 19.
Article in English | MEDLINE | ID: mdl-28988771

ABSTRACT

Epidermal growth factor receptor (EGFR) regulates many crucial cellular programs, with seven different activating ligands shaping cell signaling in distinct ways. Using crystallography and other approaches, we show how the EGFR ligands epiregulin (EREG) and epigen (EPGN) stabilize different dimeric conformations of the EGFR extracellular region. As a consequence, EREG or EPGN induce less stable EGFR dimers than EGF-making them partial agonists of EGFR dimerization. Unexpectedly, this weakened dimerization elicits more sustained EGFR signaling than seen with EGF, provoking responses in breast cancer cells associated with differentiation rather than proliferation. Our results reveal how responses to different EGFR ligands are defined by receptor dimerization strength and signaling dynamics. These findings have broad implications for understanding receptor tyrosine kinase (RTK) signaling specificity. Our results also suggest parallels between partial and/or biased agonism in RTKs and G-protein-coupled receptors, as well as new therapeutic opportunities for correcting RTK signaling output.


Subject(s)
Epigen/chemistry , Epiregulin/chemistry , ErbB Receptors/chemistry , ErbB Receptors/metabolism , Crystallography, X-Ray , Epigen/metabolism , Epiregulin/metabolism , Fluorescence Resonance Energy Transfer , Humans , Kinetics , Ligands , Models, Molecular , Protein Multimerization
10.
Hum Mutat ; 38(11): 1464-1470, 2017 11.
Article in English | MEDLINE | ID: mdl-28677295

ABSTRACT

Holoprosencephaly (HPE), a common developmental defect of the forebrain and midface, has a complex etiology. Heterozygous, loss-of-function mutations in the sonic hedgehog (SHH) pathway are associated with HPE. However, mutation carriers display highly variable clinical presentation, leading to an "autosomal dominant with modifier" model, in which the penetrance and expressivity of a predisposing mutation is graded by genetic or environmental modifiers. Such modifiers have not been identified. Boc encodes a SHH coreceptor and is a silent HPE modifier gene in mice. Here, we report the identification of missense BOC variants in HPE patients. Consistent with these alleles functioning as HPE modifiers, individual variant BOC proteins had either loss- or gain-of-function properties in cell-based SHH signaling assays. Therefore, in addition to heterozygous loss-of-function mutations in specific SHH pathway genes and an ill-defined environmental component, our findings identify a third variable in HPE: low-frequency modifier genes, BOC being the first identified.


Subject(s)
Genes, Modifier , Holoprosencephaly/genetics , Immunoglobulin G/genetics , Receptors, Cell Surface/genetics , Animals , Gene Expression , Genetic Variation , Holoprosencephaly/metabolism , Humans , Immunoglobulin G/chemistry , Immunoglobulin G/metabolism , Mice , Models, Molecular , Mutation , Protein Conformation , Protein Domains , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism
11.
Nat Commun ; 7: 10339, 2016 Feb 04.
Article in English | MEDLINE | ID: mdl-26842342

ABSTRACT

Two-pore domain potassium (K2P) channels act to maintain cell resting membrane potential--a prerequisite for many biological processes. KCNK9, a member of K2P family, is implicated in cancer, owing to its overexpression in human tumours and its ability to promote neoplastic cell survival and growth. However, KCNK9's underlying contributions to malignancy remain elusive due to the absence of specific modulators. Here we describe the development of monoclonal antibodies against the KCNK9 extracellular domain and their functional effects. We show that one antibody (Y4) with the highest affinity binding induces channel internalization. The addition of Y4 to KCNK9-expressing carcinoma cells reduces cell viability and increases cell death. Systemic administration of Y4 effectively inhibits growth of human lung cancer xenografts and murine breast cancer metastasis in mice. Evidence for Y4-mediated carcinoma cell autonomous and immune-dependent cytotoxicity is presented. Our study reveals that antibody-based KCNK9 targeting is a promising therapeutic strategy in KCNK9-expressing malignancies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Neoplasms/metabolism , Potassium Channels, Tandem Pore Domain/antagonists & inhibitors , Animals , Blotting, Western , COS Cells , Cell Cycle , Cell Line, Tumor , Cell Survival/drug effects , Chlorocebus aethiops , Drug Screening Assays, Antitumor , Flow Cytometry , HEK293 Cells , Humans , Immunohistochemistry , In Vitro Techniques , Membrane Potentials/drug effects , Mice, Inbred BALB C , Mice, Nude , Microscopy, Confocal , Neoplasm Metastasis , Neoplasm Transplantation , Neoplasms/genetics , Neoplasms/pathology , Potassium Channels, Tandem Pore Domain/genetics , Potassium Channels, Tandem Pore Domain/metabolism , Protein Structure, Tertiary , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Xenograft Model Antitumor Assays
12.
J Biol Chem ; 290(31): 18904-13, 2015 Jul 31.
Article in English | MEDLINE | ID: mdl-26067272

ABSTRACT

During systemic RNA interference (RNAi) in Caenorhabditis elegans, RNA spreads across different cells and tissues in a process that requires the systemic RNA interference deficient-1 (sid-1) gene, which encodes an integral membrane protein. SID-1 acts cell-autonomously and is required for cellular import of interfering RNAs. Heterologous expression of SID-1 in Drosophila Schneider 2 cells enables passive uptake of dsRNA and subsequent soaking RNAi. Previous studies have suggested that SID-1 may serve as an RNA channel, but its precise molecular role remains unclear. To test the hypothesis that SID-1 mediates a direct biochemical recognition of RNA molecule and subsequent permeation, we expressed the extracellular domain (ECD) of SID-1 and purified it to near homogeneity. Recombinant purified SID-1 ECD selectively binds dsRNA but not dsDNA in a length-dependent and sequence-independent manner. Genetic missense mutations in SID-1 ECD causal for deficient systemic RNAi resulted in significant reduction in its affinity for dsRNA. Furthermore, full-length proteins with these mutations decrease SID-1-mediated RNA transport efficiency, providing evidence that dsRNA binding to SID-1 ECD is related to RNA transport. To examine the functional similarity of mammalian homologs of SID-1 (SIDT1 and SIDT2), we expressed and purified mouse SIDT1 and SIDT2 ECDs. We show that they bind long dsRNA in vitro, supportive of dsRNA recognition. In summary, our study illustrates the functional importance of SID-1 ECD as a dsRNA binding domain that contributes to RNA transport.


Subject(s)
Caenorhabditis elegans Proteins/genetics , Caenorhabditis elegans/metabolism , Membrane Proteins/genetics , RNA, Double-Stranded/metabolism , Amino Acid Sequence , Animals , Binding, Competitive , Caenorhabditis elegans Proteins/chemistry , Caenorhabditis elegans Proteins/metabolism , DNA/chemistry , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Mice , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary , RNA Interference , RNA Transport , RNA, Double-Stranded/chemistry
13.
Methods Enzymol ; 559: 27-36, 2015.
Article in English | MEDLINE | ID: mdl-26096501

ABSTRACT

This protocol entails a single-step, high-affinity purification of proteins using an immobilized antibody column.


Subject(s)
Antibodies/chemistry , Chromatography, Affinity/methods , Proteins/isolation & purification , Biochemistry/methods , Buffers , Chromatography, Affinity/instrumentation , Hydrogen-Ion Concentration , Proteins/chemistry , Reducing Agents/chemistry
14.
Neuron ; 86(2): 490-500, 2015 Apr 22.
Article in English | MEDLINE | ID: mdl-25864631

ABSTRACT

Arc is a cellular immediate-early gene (IEG) that functions at excitatory synapses and is required for learning and memory. We report crystal structures of Arc subdomains that form a bi-lobar architecture remarkably similar to the capsid domain of human immunodeficiency virus (HIV) gag protein. Analysis indicates Arc originated from the Ty3/Gypsy retrotransposon family and was "domesticated" in higher vertebrates for synaptic functions. The Arc N-terminal lobe evolved a unique hydrophobic pocket that mediates intermolecular binding with synaptic proteins as resolved in complexes with TARPγ2 (Stargazin) and CaMKII peptides and is essential for Arc's synaptic function. A consensus sequence for Arc binding identifies several additional partners that include genes implicated in schizophrenia. Arc N-lobe binding is inhibited by small chemicals suggesting Arc's synaptic action may be druggable. These studies reveal the remarkable evolutionary origin of Arc and provide a structural basis for understanding Arc's contribution to neural plasticity and disease.


Subject(s)
Cognition Disorders/metabolism , Genes, Immediate-Early/physiology , Neuronal Plasticity/physiology , Retroelements/physiology , Synapses/metabolism , Amino Acid Sequence , Animals , Binding Sites , Calcium Channels/chemistry , Calcium Channels/metabolism , Calcium-Calmodulin-Dependent Protein Kinase Type 2/metabolism , Cognition/physiology , Homeostasis/physiology , Mice , Mice, Inbred C57BL , Models, Molecular , Receptors, AMPA/metabolism
15.
J Biol Chem ; 290(6): 3500-7, 2015 Feb 06.
Article in English | MEDLINE | ID: mdl-25519909

ABSTRACT

A general aim of studies of signal transduction is to identify mediators of specific signals, order them into pathways, and understand the nature of interactions between individual components and how these interactions alter pathway behavior. Despite years of intensive study and its central importance to animal development and human health, our understanding of the Hedgehog (Hh) signaling pathway remains riddled with gaps, question marks, assumptions, and poorly understood connections. In particular, understanding how interactions between Hh and Patched (Ptc), a 12-pass integral membrane protein, lead to modulation of the function of Smoothened (Smo), a 7-pass integral membrane protein, has defied standard biochemical characterization. Recent structural and biochemical characterizations of Smoothened domains have begun to unlock this riddle, however, and lay the groundwork for improved cancer therapies.


Subject(s)
Hedgehog Proteins/metabolism , Receptors, G-Protein-Coupled/metabolism , Signal Transduction , Amino Acid Sequence , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Hedgehog Proteins/genetics , Humans , Molecular Sequence Data , Neoplasms/drug therapy , Neoplasms/metabolism , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry
16.
J Biol Chem ; 290(3): 1570-9, 2015 Jan 16.
Article in English | MEDLINE | ID: mdl-25468910

ABSTRACT

The EGF receptor (EGFR) family comprises four homologs in humans collectively known as the ErbB or HER proteins. ErbB proteins are receptor tyrosine kinases that become activated when ligands bind to their extracellular regions and promote formation of specific homo- and heterodimers with enhanced tyrosine kinase activity. An essential feature of ErbB activation is formation of an asymmetric kinase dimer in which the C-terminal lobe of one kinase serves as the activator or donor kinase by binding the N-terminal lobe of a receiver or acceptor kinase and stabilizing its active conformation. ErbB extracellular regions are also thought to form active asymmetric dimers in which only one subunit binds ligand. The observation that the unliganded ErbB2 kinase preferentially serves as the activator kinase when paired with EGFR/ErbB1 implied that extracellular asymmetry in ErbB proteins might be coupled to intracellular asymmetry with unliganded partners favoring the activator kinase position. Using cell-based stimulation assays and chimeric ErbB proteins, we show that extracellular asymmetry is not coupled to intracellular asymmetry and that ErbB intracellular regions are sufficient to determine relative kinase activator-receiver orientation. We further show a hierarchy of activator-receiver preferences among ErbB proteins, with EGFR/ErbB1 being the strongest receiver, followed by ErbB2 and then ErbB4, and that cis-phosphorylation of EGFR and ErbB2 appears to be negligible. This hierarchy shapes the nature of signaling responses to different ligands in cells expressing multiple ErbB proteins.


Subject(s)
Receptor, ErbB-2/metabolism , Receptor, ErbB-4/metabolism , Amino Acid Sequence , Animals , Cell Line , Cell Membrane/metabolism , Drosophila melanogaster , ErbB Receptors/metabolism , Humans , Ligands , Molecular Sequence Data , Phosphorylation , Plasmids/metabolism , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Rats , Sequence Homology, Amino Acid , Signal Transduction
17.
J Biol Chem ; 289(45): 31361-72, 2014 Nov 07.
Article in English | MEDLINE | ID: mdl-25248746

ABSTRACT

S-Adenosylhomocysteine hydrolase (SAHH) is an NAD(+)-dependent tetrameric enzyme that catalyzes the breakdown of S-adenosylhomocysteine to adenosine and homocysteine and is important in cell growth and the regulation of gene expression. Loss of SAHH function can result in global inhibition of cellular methyltransferase enzymes because of high levels of S-adenosylhomocysteine. Prior proteomics studies have identified two SAHH acetylation sites at Lys(401) and Lys(408) but the impact of these post-translational modifications has not yet been determined. Here we use expressed protein ligation to produce semisynthetic SAHH acetylated at Lys(401) and Lys(408) and show that modification of either position negatively impacts the catalytic activity of SAHH. X-ray crystal structures of 408-acetylated SAHH and dually acetylated SAHH have been determined and reveal perturbations in the C-terminal hydrogen bonding patterns, a region of the protein important for NAD(+) binding. These crystal structures along with mutagenesis data suggest that such hydrogen bond perturbations are responsible for SAHH catalytic inhibition by acetylation. These results suggest how increased acetylation of SAHH may globally influence cellular methylation patterns.


Subject(s)
Adenosylhomocysteinase/metabolism , Lysine/metabolism , Acetylation , Amino Acid Sequence , Catalysis , Crystallography, X-Ray , Humans , Hydrogen Bonding , Methylation , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , NAD/metabolism , Plasmids/metabolism , Protein Binding , Protein Processing, Post-Translational , Protein Structure, Tertiary , Recombinant Proteins/metabolism , Sequence Homology, Amino Acid , Structure-Activity Relationship
18.
Protein Expr Purif ; 104: 92-102, 2014 12.
Article in English | MEDLINE | ID: mdl-25261717

ABSTRACT

Patched (Ptc) is a twelve-pass transmembrane protein that functions as a receptor for the Hedgehog (Hh) family of morphogens. In addition to Ptc, several accessory proteins including the CDO/Ihog family of co-receptors are necessary for proper Hh signaling. Structures of Hh proteins bound to members of the CDO/Ihog family are known, but the nature of the full Hh receptor complex is not well understood. We have expressed the Drosophila Patched and Mouse Patched-1 proteins in Sf9 cells and find that Sonic Hedgehog will bind to Mouse Patched-1 in isolated Sf9 cell membranes but that purified, detergent-solubilized Ptc proteins do not interact strongly with cognate Hh and CDO/Ihog homologs. These results may reflect a nonnative conformation of detergent-solubilized Ptc or that an additional factor or factors lost during purification are required for high-affinity Ptc binding to Hh.


Subject(s)
Detergents/chemistry , Drosophila Proteins/chemistry , Drosophila Proteins/genetics , Hedgehog Proteins/chemistry , Membrane Glycoproteins/chemistry , Patched-1 Receptor/genetics , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/genetics , Animals , Baculoviridae/genetics , Drosophila Proteins/isolation & purification , Mice , Patched-1 Receptor/chemistry , Patched-1 Receptor/isolation & purification , Protein Binding , Protein Conformation , Receptors, Cell Surface/isolation & purification , Sf9 Cells , Solubility
19.
Elife ; 32014 Sep 25.
Article in English | MEDLINE | ID: mdl-25255214

ABSTRACT

The type I insulin-like growth factor receptor (IGF1R) is involved in growth and survival of normal and neoplastic cells. A ligand-dependent conformational change is thought to regulate IGF1R activity, but the nature of this change is unclear. We point out an underappreciated dimer in the crystal structure of the related Insulin Receptor (IR) with Insulin bound that allows direct comparison with unliganded IR and suggests a mechanism by which ligand regulates IR/IGF1R activity. We test this mechanism in a series of biochemical and biophysical assays and find the IGF1R ectodomain maintains an autoinhibited state in which the TMs are held apart. Ligand binding releases this constraint, allowing TM association and unleashing an intrinsic propensity of the intracellular regions to autophosphorylate. Enzymatic studies of full-length and kinase-containing fragments show phosphorylated IGF1R is fully active independent of ligand and the extracellular-TM regions. The key step triggered by ligand binding is thus autophosphorylation.


Subject(s)
Insulin-Like Growth Factor I/metabolism , Receptor, IGF Type 1/metabolism , Amino Acid Sequence , Animals , Conserved Sequence , HEK293 Cells , Humans , Ligands , Mice , Models, Molecular , Molecular Sequence Data , Mutation/genetics , Phosphorylation , Protein Binding , Protein Multimerization , Protein Structure, Tertiary , Receptor, IGF Type 1/chemistry , Receptor, IGF Type 1/genetics , Receptor, Insulin/chemistry , Receptor, Insulin/metabolism
20.
Cancer Res ; 74(18): 5206-17, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25060518

ABSTRACT

There have been a number of clinical trials testing the efficacy of FMS-like tyrosine kinase-3 (FLT3) tyrosine kinase inhibitors (TKI) in patients with acute myeloid leukemia (AML) harboring a constitutively activating mutation in FLT3. However, there has been limited efficacy, most often because of inadequate achievement of FLT3 inhibition through a variety of mechanisms. In a previous study, TTT-3002 was identified as a novel FLT3 inhibitor with the most potent activity to date against FLT3 internal tandem duplication (FLT3/ITD) mutations. Here, the activity of TTT-3002 is demonstrated against a broad spectrum of FLT3-activating point mutations, including the most frequently occurring D835 mutations. The compound is also active against a number of point mutations selected for in FLT3/ITD alleles that confer resistance to other TKIs, including the F691L gatekeeper mutation. TTT-3002 maintains activity against patients with relapsed AML samples that are resistant to sorafenib and AC220. Studies utilizing human plasma samples from healthy donors and patients with AML indicate that TTT-3002 is only moderately protein bound compared with several other TKIs currently in clinical trials. Tumor burden of mice in a FLT3 TKI-resistant transplant model is significantly improved by oral dosing of TTT-3002. Therefore, TTT-3002 has demonstrated preclinical potential as a promising new FLT3 TKI that may overcome some of the limitations of other TKIs in the treatment of FLT3-mutant AML. Cancer Res; 74(18); 5206-17. ©2014 AACR.


Subject(s)
Carbazoles/pharmacology , Indoles/pharmacology , Leukemia, Myeloid, Acute/genetics , Protein Kinase Inhibitors/pharmacology , fms-Like Tyrosine Kinase 3/antagonists & inhibitors , Animals , Cell Line, Tumor , Drug Resistance , Drug Resistance, Neoplasm , Female , Humans , Mice , Mice, Inbred BALB C , Mutation
SELECTION OF CITATIONS
SEARCH DETAIL