Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 27
Filter
1.
Biochemistry ; 58(39): 4017-4027, 2019 10 01.
Article in English | MEDLINE | ID: mdl-31365236

ABSTRACT

The human voltage-gated proton channel [Hv1(1) or VSDO(2)] plays an important role in the human innate immune system. Its structure differs considerably from those of other cation channels. It is built solely of a voltage-sensing domain and thus lacks the central pore domain, which is essential for other cation channels. Here, we determined the solution structure of an N- and C-terminally truncated human Hv1 (Δ-Hv1) in the resting state by nuclear magnetic resonance (NMR) spectroscopy. Δ-Hv1 comprises the typical voltage-sensing antiparallel four-helix bundle (S1-S4) preceded by an amphipathic helix (S0). The solution structure corresponds to an intermediate state between resting and activated forms of voltage-sensing domains. Furthermore, Zn2+-induced closing of proton channel Δ-Hv1 was studied with two-dimensional NMR spectroscopy, which showed that characteristic large scale dynamics of open Δ-Hv1 are absent in the closed state of the channel. Additionally, pH titration studies demonstrated that a higher H+ concentration is required for the protonation of side chains in the Zn2+-induced closed state than in the open state. These observations demonstrate both structural and dynamical changes involved in the process of voltage gating of the Hv1 channel and, in the future, may help to explain the unique properties of unidirectional conductance and the exceptional ion selectivity of the channel.


Subject(s)
Ion Channel Gating , Ion Channels/chemistry , Magnetic Resonance Spectroscopy/methods , Basic-Leucine Zipper Transcription Factors/chemistry , Crystallization , Crystallography, X-Ray , Escherichia coli/genetics , Escherichia coli/metabolism , Humans , Hydrogen Bonding , Hydrogen-Ion Concentration , Ion Channels/genetics , Kinetics , Models, Molecular , Phosphoric Monoester Hydrolases/chemistry , Protein Binding , Protein Structure, Secondary , Protons , Saccharomyces cerevisiae Proteins/chemistry , Zinc/chemistry
2.
J Am Chem Soc ; 141(18): 7391-7398, 2019 05 08.
Article in English | MEDLINE | ID: mdl-30973010

ABSTRACT

In potassium (K+) channels, permeation, selectivity, and gating at the selectivity filter are all governed by the thermodynamics and kinetics of the ion-protein interactions. Specific contacts between the carbonyl groups from the Thr-Val-Gly-Tyr-Gly signature filter sequence and the permeant ions generate four equidistant K+ binding sites, thereby defining the high ion selectivity and controlling the transport rate of K+ channels. Here, we used 15N-labeled ammonium (15NH4+) as a proxy for K+ to study ion interaction with the selectivity filter of the prototypical full-length K+ channel KcsA by solution state NMR spectroscopy in order to obtain detailed insights into the physicochemical basis of K+ gating. We found that in the closed inactive state of KcsA (at pH 7) four K+ binding sites are occupied over a wide range of 15NH4+ concentrations, while in intermediate closed-open conformations (at pH ∼6) the number and occupancy of K+ binding sites are reduced to two. However, in the presence of the scorpion toxin agitoxin II a total loss of 15NH4+ binding is observed. 15NH4+ titration studies allowed us to determine the dissociation constants of the four binding sites with values around 10 mM in the closed state of KcsA. Moreover, kinetic NMR experiments measured in the steady state equilibrium detected an off- and on-rate for 15NH4+ of ca. 102 s-1 and 103 s-1 between KcsA-bound 15NH4+ and the bulk. These findings reveal both the thermodynamics and kinetics of the ion binding sites and thus contribute to our understanding of the action of K+ channels.


Subject(s)
Ammonium Compounds/chemistry , Bacterial Proteins/chemistry , Potassium Channels/chemistry , Binding Sites , Ions/chemistry , Kinetics , Models, Molecular , Nitrogen Isotopes , Thermodynamics
3.
Bioconjug Chem ; 30(12): 3098-3106, 2019 12 18.
Article in English | MEDLINE | ID: mdl-31715102

ABSTRACT

In this study, we have designed and synthesized two novel peptide-drug conjugates (PDCs) where the drug, doxorubicin (Dox), is linked to the peptide via a succinimidyl thioether bond or a hydrazone linker. A highly specific and proteolytically stable breast cancer cell targeting peptide (WxEAAYQrFL) is conjugated to Dox to synthesize peptide-Dox thioether (1) or hydrazone (2) conjugate. The evaluation of the stability in water, media, and human serum showed that the conjugate 1 with the succinimidyl thioether linkage is more stable compared to the acid-sensitive hydrazone containing conjugate 2. The cytotoxicity studies showed that the two PDCs were as toxic as free Dox toward the triple negative breast cancer (TNBC) cells and were 7-30 times less toxic (IC50 1.2-4.7 µM for TNBC cells versus 15-39 µM for noncancerous cells) toward the noncancerous breast cells compared to the free doxorubicin (IC50 0.35-1.5 µM for TNBC cells versus 0.24 µM for noncancerous cells). The results from the comparative study of the two PDCs suggest that both may have translational potential for TNBC treatment.


Subject(s)
Doxorubicin/chemistry , Peptides/chemistry , Triple Negative Breast Neoplasms/drug therapy , Cross-Linking Reagents/chemistry , Cross-Linking Reagents/pharmacology , Cytotoxins/chemistry , Cytotoxins/toxicity , Humans , Hydrazones , Sulfides
4.
J Biol Chem ; 291(16): 8516-27, 2016 Apr 15.
Article in English | MEDLINE | ID: mdl-26846854

ABSTRACT

Multiple neurodegenerative diseases are caused by the aggregation of the human α-Synuclein (α-Syn) protein. α-Syn possesses high structural plasticity and the capability of interacting with membranes. Both features are not only essential for its physiological function but also play a role in the aggregation process. Recently it has been proposed that α-Syn is able to form lipid-protein particles reminiscent of high-density lipoproteins. Here, we present a method to obtain a stable and homogeneous population of nanometer-sized particles composed of α-Syn and anionic phospholipids. These particles are called α-Syn lipoprotein (nano)particles to indicate their relationship to high-density lipoproteins formed by human apolipoproteins in vivo and of in vitro self-assembling phospholipid bilayer nanodiscs. Structural investigations of the α-Syn lipoprotein particles by circular dichroism (CD) and magic angle solid-state nuclear magnetic resonance (MAS SS-NMR) spectroscopy establish that α-Syn adopts a helical secondary structure within these particles. Based on cryo-electron microscopy (cryo-EM) and dynamic light scattering (DLS) α-Syn lipoprotein particles have a defined size with a diameter of ∼23 nm. Chemical cross-linking in combination with solution-state NMR and multiangle static light scattering (MALS) of α-Syn particles reveal a high-order protein-lipid entity composed of ∼8-10 α-Syn molecules. The close resemblance in size between cross-linked in vitro-derived α-Syn lipoprotein particles and a cross-linked species of endogenous α-Syn from SH-SY5Y human neuroblastoma cells indicates a potential functional relevance of α-Syn lipoprotein nanoparticles.


Subject(s)
Lipoproteins, HDL/chemistry , Nanoparticles/chemistry , Phospholipids/chemistry , alpha-Synuclein/chemistry , Cell Line, Tumor , Humans , Nuclear Magnetic Resonance, Biomolecular
5.
J Biol Chem ; 289(34): 23482-503, 2014 Aug 22.
Article in English | MEDLINE | ID: mdl-24958726

ABSTRACT

The solution NMR structure of the α-helical integral membrane protein YgaP from Escherichia coli in mixed 1,2-diheptanoyl-sn-glycerol-3-phosphocholine/1-myristoyl-2-hydroxy-sn-glycero-3-phospho-(1'-rac-glycerol) micelles is presented. In these micelles, YgaP forms a homodimer with the two transmembrane helices being the dimer interface, whereas the N-terminal cytoplasmic domain includes a rhodanese-fold in accordance to its sequence homology to the rhodanese family of sulfurtransferases. The enzymatic sulfur transfer activity of full-length YgaP as well as of the N-terminal rhodanese domain only was investigated performing a series of titrations with sodium thiosulfate and potassium cyanide monitored by NMR and EPR. The data indicate the thiosulfate concentration-dependent addition of several sulfur atoms to the catalytic Cys-63, which process can be reversed by the addition of potassium cyanide. The catalytic reaction induces thereby conformational changes within the rhodanese domain, as well as on the transmembrane α-helices of YgaP. These results provide insights into a potential mechanism of YgaP during the catalytic thiosulfate activity in vivo.


Subject(s)
Escherichia coli Proteins/chemistry , Escherichia coli Proteins/physiology , Escherichia coli/chemistry , Membrane Proteins/chemistry , Membrane Proteins/physiology , Nuclear Magnetic Resonance, Biomolecular/methods , Models, Molecular , Protein Conformation , Structure-Activity Relationship , Thiosulfate Sulfurtransferase/chemistry
6.
J Am Chem Soc ; 137(35): 11218-21, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26301538

ABSTRACT

Optical modulation of proteins provides superior spatiotemporal resolution for understanding biological processes, and photoswitches built on light-sensitive proteins have been significantly advancing neuronal and cellular studies. Small molecule photoswitches could complement protein-based switches by mitigating potential interference and affording high specificity for modulation sites. However, genetic encodability and responsiveness to nonultraviolet light, two desired properties possessed by protein photoswitches, are challenging to be engineered into small molecule photoswitches. Here we developed a small molecule photoswitch that can be genetically installed onto proteins in situ and controlled by visible light. A pentafluoro azobenzene-based photoswitchable click amino acid (F-PSCaa) was designed to isomerize in response to visible light. After genetic incorporation into proteins via the expansion of the genetic code, F-PSCaa reacts with a nearby cysteine within the protein generating an azo bridge in situ. The resultant bridge is switchable by visible light and allows conformation and binding of CaM to be regulated by such light. This photoswitch should prove valuable in optobiology for its minimal interference, site flexibility, genetic encodability, and response to the more biocompatible visible light.


Subject(s)
Azo Compounds/chemistry , Light , Optogenetics/methods , Proteins/chemistry , Proteins/genetics , Amino Acids/chemistry , Models, Molecular , Protein Conformation , Stereoisomerism
7.
Nat Methods ; 9(8): 834-9, 2012 May 20.
Article in English | MEDLINE | ID: mdl-22609626

ABSTRACT

Although nearly half of today's major pharmaceutical drugs target human integral membrane proteins (hIMPs), only 30 hIMP structures are currently available in the Protein Data Bank, largely owing to inefficiencies in protein production. Here we describe a strategy for the rapid structure determination of hIMPs, using solution NMR spectroscopy with systematically labeled proteins produced via cell-free expression. We report new backbone structures of six hIMPs, solved in only 18 months from 15 initial targets. Application of our protocols to an additional 135 hIMPs with molecular weight <30 kDa yielded 38 hIMPs suitable for structural characterization by solution NMR spectroscopy without additional optimization.


Subject(s)
Membrane Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Databases, Protein , Humans , Models, Molecular , Molecular Weight , Protein Conformation
8.
Biochem Biophys Res Commun ; 445(4): 724-33, 2014 Mar 21.
Article in English | MEDLINE | ID: mdl-24525125

ABSTRACT

About 8000 genes encode membrane proteins in the human genome. The information about their druggability will be very useful to facilitate drug discovery and development. The main problem, however, consists of limited structural and functional information about these proteins because they are difficult to produce biochemically and to study. In this paper we describe the strategy that combines Cell-free protein expression, NMR spectroscopy, and molecular DYnamics simulation (CNDY) techniques. Results of a pilot CNDY experiment provide us with a guiding light towards expedited identification of the hit compounds against a new uncharacterized membrane protein as a potentially druggable target. These hits can then be further characterized and optimized to develop the initial lead compound quicker. We illustrate such "omics" approach for drug discovery with the CNDY strategy applied to two example proteins: hypoxia-induced genes HIGD1A and HIGD1B.


Subject(s)
Drug Design , Membrane Proteins/chemistry , Neoplasm Proteins/chemistry , Nuclear Magnetic Resonance, Biomolecular/methods , Binding Sites , Humans , Intracellular Signaling Peptides and Proteins , Ligands , Membrane Proteins/metabolism , Mitochondrial Proteins , Molecular Docking Simulation , Neoplasm Proteins/metabolism , Protein Conformation
9.
Proc Natl Acad Sci U S A ; 108(14): 5831-6, 2011 Apr 05.
Article in English | MEDLINE | ID: mdl-21422294

ABSTRACT

G protein-gated inwardly rectifying potassium (GIRK) channels are important gatekeepers of neuronal excitability. The surface expression of neuronal GIRK channels is regulated by the psychostimulant-sensitive sorting nexin 27 (SNX27) protein through a class I (-X-Ser/Thr-X-Φ, where X is any residue and Φ is a hydrophobic amino acid) PDZ-binding interaction. The G protein-insensitive inward rectifier channel (IRK1) contains the same class I PDZ-binding motif but associates with a different synaptic PDZ protein, postsynaptic density protein 95 (PSD95). The mechanism by which SNX27 and PSD95 discriminate these channels was previously unclear. Using high-resolution structures coupled with biochemical and functional analyses, we identified key amino acids upstream of the channel's canonical PDZ-binding motif that associate electrostatically with a unique structural pocket in the SNX27-PDZ domain. Changing specific charged residues in the channel's carboxyl terminus or in the PDZ domain converts the selective association and functional regulation by SNX27. Elucidation of this unique interaction site between ion channels and PDZ-containing proteins could provide a therapeutic target for treating brain diseases.


Subject(s)
G Protein-Coupled Inwardly-Rectifying Potassium Channels/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Models, Molecular , Neurons/physiology , Sorting Nexins/metabolism , Amino Acid Motifs/physiology , Animals , COS Cells , Chlorocebus aethiops , Crystallization , Disks Large Homolog 4 Protein , Electrophysiology , G Protein-Coupled Inwardly-Rectifying Potassium Channels/chemistry , G Protein-Coupled Inwardly-Rectifying Potassium Channels/genetics , Hippocampus/cytology , Humans , Image Processing, Computer-Assisted , Immunohistochemistry , Intracellular Signaling Peptides and Proteins/chemistry , Intracellular Signaling Peptides and Proteins/genetics , Membrane Proteins/chemistry , Membrane Proteins/genetics , Microscopy, Confocal , Mutagenesis, Site-Directed , Neurons/metabolism , Protein Structure, Tertiary , Rats
10.
Heliyon ; 10(5): e26345, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38468948

ABSTRACT

Ubiquitin-specific protease7 (USP7) regulates the stability of the p53 tumor suppressor protein and several other proteins critical for tumor cell survival. Aberrant expression of USP7 facilitates human malignancies by altering the activity of proto-oncogenes/proteins, and tumor suppressor genes. Therefore, USP7 is a validated anti-cancer drug target. In this study, a drug repurposing approach was used to identify new hits against the USP7 enzyme. It is one of the most strategic approaches to find new uses for drugs in a cost- and time-effective way. Nuclear Magnetic Resonance-based screening of 172 drugs identified 11 compounds that bind to the catalytic domain of USP7 with dissociation constant (Kd) values in the range of 0.6-1.49 mM. These 11 compounds could thermally destabilize the USP7 enzyme by decreasing its melting temperature up to 9 °C. Molecular docking and simulation studies provided structural insights into the ligand-protein complexes, suggesting that these compounds bind to the putative substrate binding pocket of USP7, and interact with its catalytically important residues. Among the identified 11 hits, compound 6 (oxybutynin), 7 (ketotifen), 10 (pantoprazole sodium), and 11 (escitalopram) also showed anti-cancer activity with an effect on the expression of proto-oncogenes and tumor-suppressor gene at mRNA level in HCT116 cells. The compounds identified in this study can serve as potential leads for further studies.

11.
Eur J Pharm Sci ; 197: 106776, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38663759

ABSTRACT

The emergence of multidrug-resistant (MDR) strains causes severe problems in the treatment of microbial infections owing to limited treatment options. Antimicrobial peptides (AMPs) are drawing considerable attention as promising antibiotic alternative candidates to combat MDR bacterial and fungal infections. Herein, we present a series of small amphiphilic membrane-active cyclic peptides composed, in part, of various nongenetically encoded hydrophilic and hydrophobic amino acids. Notably, lead cyclic peptides 3b and 4b showed broad-spectrum activity against drug-resistant Gram-positive (MIC = 1.5-6.2 µg/mL) and Gram-negative (MIC = 12.5-25 µg/mL) bacteria, and fungi (MIC = 3.1-12.5 µg/mL). Furthermore, lead peptides displayed substantial antibiofilm action comparable to standard antibiotics. Hemolysis (HC50 = 230 µg/mL) and cytotoxicity (>70 % cell viability against four different mammalian cells at 100 µg/mL) assay results demonstrated the selective lethal action of 3b against microbes over mammalian cells. A calcein dye leakage experiment substantiated the membranolytic effect of 3b and 4b, which was further confirmed by scanning electron microscopy. The behavior of 3b and 4b in aqueous solution and interaction with phospholipid bilayers were assessed by employing nuclear magnetic resonance (NMR) spectroscopy in conjunction with molecular dynamics (MD) simulations, providing a solid structural basis for understanding their membranolytic action. Moreover, 3b exhibited stability in human blood plasma (t1/2 = 13 h) and demonstrated no signs of resistance development against antibiotic-resistant S. aureus and E. coli. These findings underscore the potential of these newly designed amphiphilic cyclic peptides as promising anti-infective agents, especially against Gram-positive bacteria.


Subject(s)
Biofilms , Drug Resistance, Multiple, Bacterial , Hemolysis , Microbial Sensitivity Tests , Humans , Drug Resistance, Multiple, Bacterial/drug effects , Biofilms/drug effects , Hemolysis/drug effects , Peptides, Cyclic/pharmacology , Peptides, Cyclic/chemistry , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Antimicrobial Cationic Peptides/pharmacology , Antimicrobial Cationic Peptides/chemistry , Animals , Fungi/drug effects , Cell Survival/drug effects , Antifungal Agents/pharmacology , Antifungal Agents/chemistry , Gram-Negative Bacteria/drug effects
12.
Proteomics Clin Appl ; : e2300128, 2024 Mar 05.
Article in English | MEDLINE | ID: mdl-38444254

ABSTRACT

PURPOSE: Micropeptides are an emerging class of proteins that play critical roles in cell signaling. Here, we describe the discovery of a novel micropeptide, dubbed slitharin (Slt), in conditioned media from Cardiosphere-derived cells (CDCs), a therapeutic cardiac stromal cell type. EXPERIMENTAL DESIGN: We performed mass spectrometry of peptide-enriched fractions from the conditioned media of CDCs and a therapeutically inert cell type (human dermal fibrobasts). We then evaluated the therapeutic capacity of the candidate peptide using an in vitro model of cardiomyocyte injury and a rat model of myocardial infarction. RESULTS: We identified a novel 24-amino acid micropeptide (dubbed Slitharin [Slt]) with a non-canonical leucine start codon, arising from long intergenic non-coding (LINC) RNA 2099. Neonatal rat ventricular myocytes (NRVMs) exposed to Slt were protected from hypoxic injury in vitro compared to a vehicle or scrambled control. Transcriptomic analysis of cardiomyocytes exposed to Slt reveals cytoprotective capacity, putatively through regulation of stress-induced MAPK-ERK. Slt also exerted cardioprotective effects in rats with myocardial infarction as shown by reduced infarct size 48 h post-injury. Conclusions and clinical relavance: Thus, Slt is a non-coding RNA-derived micropeptide, identified in the extracellular space, with a potential cardioprotective function.

13.
Proc Natl Acad Sci U S A ; 107(24): 10902-7, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20498088

ABSTRACT

NMR structural studies of membrane proteins (MP) are hampered by complications in MP expression, technical difficulties associated with the slow process of NMR spectral peak assignment, and limited distance information obtainable for transmembrane (TM) helices. To overcome the inherent challenges in the determination of MP structures, we have developed a rapid and cost-efficient strategy that combines cell-free (CF) protein synthesis, optimized combinatorial dual-isotope labeling for nearly instant resonance assignment, and fast acquisition of long-distance information using paramagnetic probes. Here we report three backbone structures for the TM domains of the three classes of Escherichia coli histidine kinase receptors (HKRs). The ArcB and QseC TM domains are both two-helical motifs, whereas the KdpD TM domain comprises a four-helical bundle with shorter second and third helices. The interhelical distances (up to 12 A) reveal weak interactions within the TM domains of all three receptors. Determined consecutively within 8 months, these structures offer insight into the abundant and underrepresented in the Protein Data Bank class of 2-4 TM crossers and demonstrate the efficiency of our CF combinatorial dual-labeling strategy, which can be applied to solve MP structures in high numbers and at a high speed. Our results greatly expand the current knowledge of HKR structure, opening the doors to studies on their widespread and pharmaceutically important bacterial signaling mechanism.


Subject(s)
Escherichia coli Proteins/chemistry , Membrane Proteins/chemistry , Protein Kinases/chemistry , Amino Acid Sequence , Bacteriological Techniques , Carbon Isotopes , Escherichia coli/chemistry , Escherichia coli/genetics , Escherichia coli Proteins/genetics , Histidine Kinase , Membrane Proteins/genetics , Models, Molecular , Molecular Sequence Data , Multiprotein Complexes/chemistry , Nitrogen Isotopes , Nuclear Magnetic Resonance, Biomolecular , Protein Kinases/genetics , Protein Structure, Tertiary , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
14.
J Med Chem ; 66(1): 855-874, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36574364

ABSTRACT

A series of small (7-12 mer) amphipathic cationic peptides were designed and synthesized to create short helical peptides with broad-range bactericidal activity and selectivity toward the bacterial cells. The analysis identified a lead 12-mer peptide 8b with broad-spectrum activity against Gram-positive (MIC = 3.1-6.2 µg/mL) and Gram-negative (MIC = 6.2-12.5 µg/mL) bacteria and selectivity toward prokaryotic versus eukaryotic cells (HC50 = 280 µg/mL, >75% cell viability at 150 µg/mL). The rapid membranolytic action of 8b was demonstrated by a calcein dye leakage assay and confirmed using scanning electron microscopy. According to circular dichroism and NMR spectroscopy, the peptides have an irregular spatial structure in water. A lipid bilayer induced an amphipathic helix only in 12-mer peptides, including 8b. Molecular dynamics simulations provided detailed information about the interaction of 8b and its closest analogues with bacterial and mammalian membranes and revealed the roles of particular amino acids in the activity and selectivity of peptides.


Subject(s)
Anti-Bacterial Agents , Antimicrobial Cationic Peptides , Animals , Antimicrobial Cationic Peptides/chemistry , Anti-Bacterial Agents/chemistry , Lipid Bilayers/metabolism , Protein Conformation, alpha-Helical , Bacteria/metabolism , Circular Dichroism , Microbial Sensitivity Tests , Mammals/metabolism
15.
Med Chem ; 19(10): 996-1001, 2023.
Article in English | MEDLINE | ID: mdl-37005533

ABSTRACT

INTRODUCTION: Breast cancer is the most common cancer affecting women worldwide, including Pakistan. More than half of breast cancer patients have hormone-dependent breast cancer, which is developed due to the over-production of estrogen (the main hormone in breast cancer). METHOD: The biosynthesis of estrogen is catalyzed by the aromatase enzyme, which thus serves as a target for the treatment of breast cancer. During the current study, biochemical, computational, and STD-NMR methods were employed to identify new aromatase inhibitors. A series of phenyl-3- butene-2-one derivatives 1-9 were synthesized and evaluated for human placental aromatase inhibitory activity. Among them, four compounds 2, 3, 4, and 8 showed a moderate to weak inhibitory activity (IC50 = 22.6 - 47.9 µM), as compared to standard aromatase inhibitory drugs, letrozole (IC50 = 0.0147 ± 1.45 µM), anastrozole (IC50 = 0.0094 ± 0.91 µM), and exemestane (IC50 = 0.2 ± 0.032 µM). Kinetic studies on two moderate inhibitors, 4 and 8, revealed a competitive- and mixed-type of inhibition, respectively. RESULT: Docking studies on all active compounds indicated their binding adjacent to the heme group and interaction with Met374, a critical residue of aromatase. STD-NMR further highlighted the interactions of these ligands with the aromatase enzyme. CONCLUSION: STD-NMR-based epitope mapping indicated close proximity of the alkyl chain followed by an aromatic ring with the receptor (aromatase). These compounds were also found to be non-cytotoxic against human fibroblast cells (BJ cells). Thus, the current study has identified new aromatase inhibitors (compounds 4, and 8) for further pre-clinical and clinical research.


Subject(s)
Aromatase Inhibitors , Breast Neoplasms , Pregnancy , Female , Humans , Aromatase Inhibitors/pharmacology , Aromatase Inhibitors/chemistry , Aromatase Inhibitors/therapeutic use , Aromatase/chemistry , Aromatase/metabolism , Aromatase/therapeutic use , Kinetics , Placenta/metabolism , Breast Neoplasms/drug therapy , Estrogens/therapeutic use , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use
16.
J Med Chem ; 65(1): 665-687, 2022 01 13.
Article in English | MEDLINE | ID: mdl-34978443

ABSTRACT

We report the synthesis and antibacterial activities of a series of amphiphilic membrane-active peptides composed, in part, of various nongenetically coded hydrophobic amino acids. The lead cyclic peptides, 8C and 9C, showed broad-spectrum activity against drug-resistant Gram-positive (minimum inhibitory concentration (MIC) = 1.5-6.2 µg/mL) and Gram-negative (MIC = 12.5-25 µg/mL) bacteria. The cytotoxicity study showed the predominant lethal action of the peptides against bacteria as compared with mammalian cells. A plasma stability study revealed approximately 2-fold higher stability of lead cyclic peptides as compared to their linear counterparts after 24 h of incubation. A calcein dye leakage experiment revealed the membranolytic effect of the cyclic peptides. Nuclear magnetic resonance spectroscopy and molecular dynamics simulation studies of the interaction of the peptides with the phospholipid bilayer provided a solid structural basis to explain the membranolytic action of the peptides with atomistic details. These results highlight the potential of newly designed amphiphilic peptides as the next generation of peptide-based antibiotics.


Subject(s)
Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Cell Membrane/drug effects , Peptides/chemical synthesis , Peptides/pharmacology , Antimicrobial Cationic Peptides , Cell Survival/drug effects , Drug Design , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , HEK293 Cells , Hemolysis/drug effects , Humans , Magnetic Resonance Spectroscopy , Microbial Sensitivity Tests , Molecular Dynamics Simulation
17.
PLoS Biol ; 6(10): e223, 2008 Oct 28.
Article in English | MEDLINE | ID: mdl-18959476

ABSTRACT

The RCK-containing MthK channel undergoes two inactivation processes: activation-coupled desensitization and acid-induced inactivation. The acid inactivation is mediated by the C-terminal RCK domain assembly. Here, we report that the desensitization gating is governed by a desensitization domain (DD) of the cytoplasmic N-terminal 17 residues. Deletion of DD completely removes the desensitization, and the process can be fully restored by a synthetic DD peptide added in trans. Mutagenesis analyses reveal a sequence-specific determinant for desensitization within the initial hydrophobic segment of DD. Proton nuclear magnetic resonance ((1)H NMR) spectroscopy analyses with synthetic peptides and isolated RCK show interactions between the two terminal domains. Additionally, we show that deletion of DD does not affect the acid-induced inactivation, indicating that the two inactivation processes are mutually independent. Our results demonstrate that the short N-terminal DD of MthK functions as a complete moveable module responsible for the desensitization. Its interaction with the C-terminal RCK domain may play a role in the gating process.


Subject(s)
Archaeal Proteins/physiology , Peptides/physiology , Potassium Channels/physiology , Amino Acid Sequence , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Chromatography, Gel , Cytoplasm/metabolism , Electrophysiology , Escherichia coli/metabolism , Escherichia coli/physiology , Hydrogen-Ion Concentration , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Molecular Sequence Data , Mutation , Peptides/genetics , Peptides/metabolism , Potassium Channels/chemistry , Potassium Channels/genetics , Spheroplasts/metabolism , Spheroplasts/physiology
18.
J Struct Funct Genomics ; 10(1): 25-35, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19214777

ABSTRACT

Bottlenecks in expression, solubilization, purification and crystallization hamper the structural study of integral membrane proteins (IMPs). Successful crystallization is critically dependent on the purity, stability and oligomeric homogeneity of an IMP sample. These characteristics are in turn strongly influenced by the type and concentration of the detergents used in IMP preparation. By utilizing the techniques and analytical tools we earlier developed for the characterization of protein-detergent complexes (PDCs) [21], we demonstrate that for successful protein extraction from E. coli membrane fractions, the solubilizing detergent associates preferentially to IMPs rather than to membrane lipids. Notably, this result is contrary to the generally accepted mechanism of detergent-mediated IMP solubilization. We find that for one particular member of the family of proteins studied (E. coli receptor kinases, which is purified in mixed multimeric states and oligomerizes through its transmembrane region), the protein oligomeric composition is largely unaffected by a 10-fold increase in protein concentration, by alteration of micelle properties through addition of other detergents to the PDC sample, or by a 20-fold variation in the detergent concentration used for solubilization of the IMP from the membrane. We observed that the conditions used for expression of the IMP, which impact protein density in the membrane, has the greatest influence on the IMP oligomeric structure. Finally, we argue that for concentrating PDCs smaller than 30 kDa, stirred concentration cells are less prone to over-concentration of detergent and are therefore more effective than centrifugal ultrafiltration devices.


Subject(s)
Detergents/chemistry , Magnetic Resonance Spectroscopy/methods , Membrane Proteins/chemistry , Ultracentrifugation/methods , Light , Membrane Proteins/metabolism , Scattering, Radiation , Surface Properties
19.
J Mol Biol ; 431(17): 3146-3156, 2019 08 09.
Article in English | MEDLINE | ID: mdl-31247202

ABSTRACT

Although the Ub-binding domain in ABIN proteins and NEMO (UBAN) is highly conserved, UBAN-containing proteins exhibit different Ub-binding properties, resulting in their diverse biological roles. Post-translational modifications further control UBAN domain specificity for poly-Ub chains. However, precisely, how the UBAN domain structurally confers such functional diversity remains poorly understood. Here we report crystal structures of ABIN-1 alone and in complex with one or two M1-linked di-Ub chains. ABIN-1 UBAN forms a homo-dimer that provides two symmetrical Ub-binding sites on either side of the coiled-coil structure. Moreover, crystal structures of ABIN1 UBAN in complex with di-Ub chains reveal a concentration-dependency of UBAN/di-Ub binding stoichiometry. Analysis of UBAN/M1-linked di-Ub binding characteristics indicates that phosphorylated S473 in OPTN and its corresponding phospho-mimetic residue in ABIN-1 (E484) are essential for high affinity interactions with M1-linked Ub chains. Also, a phospho-mimetic mutation of A303 in NEMO, corresponding to S473 of OPTN, increases binding affinity for M1-linked Ub chains. These findings are in line with the diverse physiological roles of UBAN domains, as phosphorylation of OPTN UBAN is required to enhance its binding to Ub during mitophagy.


Subject(s)
DNA-Binding Proteins/chemistry , I-kappa B Kinase/chemistry , Ubiquitin/chemistry , Ubiquitin/metabolism , Binding Sites , Crystallography, X-Ray , Humans , I-kappa B Kinase/genetics , Mitophagy , Models, Molecular , Phosphorylation , Protein Binding , Protein Conformation , Protein Interaction Domains and Motifs , Sequence Analysis, Protein , Ubiquitination , X-Ray Diffraction
20.
BMC Struct Biol ; 7: 74, 2007 Nov 08.
Article in English | MEDLINE | ID: mdl-17988403

ABSTRACT

BACKGROUND: Structural studies of integral membrane proteins (IMPs) are hampered by inherent difficulties in their heterologous expression and in the purification of solubilized protein-detergent complexes (PDCs). The choice and concentrations of detergents used in an IMP preparation play a critical role in protein homogeneity and are thus important for successful crystallization. RESULTS: Seeking an effective and standardized means applicable to genomic approaches for the characterization of PDCs, we chose 1D-NMR spectroscopic analysis to monitor the detergent content throughout their purification: protein extraction, detergent exchange, and sample concentration. We demonstrate that a single NMR measurement combined with a SDS-PAGE of a detergent extracted sample provides a useful gauge of the detergent's extraction potential for a given protein. Furthermore, careful monitoring of the detergent content during the process of IMP production allows for a high level of reproducibility. We also show that in many cases a simple sedimentation velocity measurement provides sufficient data to estimate both the oligomeric state and the detergent-to-protein ratio in PDCs, as well as to evaluate the homogeneity of the samples prior to crystallization screening. CONCLUSION: The techniques presented here facilitate the screening and selection of the extraction detergent, as well as help to maintain reproducibility in the detergent exchange and PDC concentration procedures. Such reproducibility is particularly important for the optimization of initial crystallization conditions, for which multiple purifications are routinely required.


Subject(s)
Detergents/chemistry , Membrane Proteins/chemistry , Cell Membrane/chemistry , Crystallography, X-Ray , Electrophoresis, Polyacrylamide Gel , Gene Expression , Magnetic Resonance Spectroscopy , Membrane Proteins/genetics , Reproducibility of Results , Solubility , Ultracentrifugation
SELECTION OF CITATIONS
SEARCH DETAIL