Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 1.934
Filter
1.
J Transl Med ; 22(1): 641, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38982548

ABSTRACT

BACKGROUND: Trastuzumab and pertuzumab combination has been approved for the treatment of patients with HER2-positive metastatic breast cancer. However, trastuzumab and pertuzumab combination did not show improvement in overall survival in patients with HER2-positive metastatic gastric cancer. METHODS: We developed a new HER2-targeted monoclonal antibody, HLX22, targeting HER2 subdomain IV as trastuzumab but with non-overlapping epitopes. We examined the antitumor effects of this novel HER2-antibody in gastric cell lines and cell line-derived xenograft (CDX) and patient-derived xenograft (PDX) models. RESULTS: HLX22 in combination with HLX02 (trastuzumab biosimilar) induced enhancement of HER2/HER2 homodimers and HER2/EGFR heterodimers internalization, which ultimately led to the reduction in signal transductions involving STAT3, P70 S6, and AKT; gene expressions of FGF-FGFR-PI3K-MTOR, EGF-EGFR-RAS, TGF-ß-SMAD, PLCG and cell cycle progression related pathways that favor tumor development, proliferation, progression, migration and survival in gastric cancer cell line NCI-N87 were also reduced. These differing but complementary actions contributed to the synergistic antitumor efficacy of the HLX22 and HLX02 combination in gastric cancer cell lines, CDX and PDX. In addition, HLX22 in combination with HLX02 demonstrated stronger antitumor efficacy than HLX02 and HLX11 (a potential pertuzumab biosimilar) combination treatment both in vitro and in vivo. CONCLUSIONS: These results suggested that the application of non-competing antibodies HLX22 and HLX02 targeting HER2 subdomain IV together may be of substantial benefit to gastric cancer patients who currently respond suboptimal to trastuzumab therapy.


Subject(s)
Epitopes , ErbB Receptors , Receptor, ErbB-2 , Stomach Neoplasms , Xenograft Model Antitumor Assays , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/metabolism , Humans , Receptor, ErbB-2/metabolism , Cell Line, Tumor , Animals , ErbB Receptors/metabolism , Protein Multimerization/drug effects , Signal Transduction/drug effects , Cell Proliferation/drug effects , Protein Domains , Female , Mice , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use
2.
Int J Biol Macromol ; 276(Pt 2): 133955, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39025177

ABSTRACT

Coronaviruses (CoV) are highly pathogenic single-strand RNA viruses. CoV infections cause fatal respiratory symptoms and lung injuries in humans and significant economic losses in livestock. Since the SARS-2 outbreak in 2019, the highly conserved main protease (Mpro), also termed 3-chymotrypsin-like protease (3CLpro), has been considered an attractive drug target for treating CoV infections. Mpro mediates the proteolytic cleavage of eleven sites in viral polypeptides necessary for virus replication. Here, we report that disulfiram, an FDA-approved drug for alcoholic treatment, exhibits a broad-spectrum inhibitory effect on CoV Mpros. Analytical ultracentrifugation and circular dichroism analyses indicated that disulfiram treatment blocks the dimeric formation of SARS and PEDV Mpros and decreases the thermostability of SARS, SARS-2, and PEDV Mpros, whereas it facilitates the dimerization and stability of MERS Mpro. Furthermore, mass spectrometry and structural alignment revealed that disulfiram targets the Cys44 residue of Mpros, which is located at the substrate entrance and close to the catalytic His41. In addition, molecular docking analysis suggests that disulfiram conjugation interferes with substrate entry to the catalytic center. In agreement, mutation of Cys44 modulates the disulfiram sensitivity of CoV Mpros. Our study suggests a broad-spectrum inhibitory function of disulfiram against CoV Mpros.


Subject(s)
Coronavirus 3C Proteases , Disulfiram , Molecular Docking Simulation , Disulfiram/pharmacology , Disulfiram/chemistry , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , SARS-CoV-2/drug effects , SARS-CoV-2/enzymology , Humans , Catalytic Domain , Substrate Specificity , Protein Multimerization/drug effects , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry , Antiviral Agents/pharmacology , Antiviral Agents/chemistry
4.
J Phys Chem Lett ; 15(24): 6375-6382, 2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38857530

ABSTRACT

The effects of two macromolecular cosolutes, specifically the polysaccharide dextran-20 and the protein lysozyme, on the aggregation kinetics of a pathogenic huntingtin exon-1 protein (hhtex1) with a 35 polyglutamine repeat, httex1Q35, are described. A unified kinetic model that establishes a direct connection between reversible tetramerization occurring on the microsecond time scale and irreversible fibril formation on a time scale of hours/days forms the basis for quantitative analysis of httex1Q35 aggregation, monitored by measuring cross-peak intensities in a series of 2D 1H-15N NMR correlation spectra acquired during the course of aggregation. The primary effects of the two cosolutes are associated with shifts in the prenucleation tetramerization equilibrium resulting in substantial changes in concentration of "preformed" httex1Q35 tetramers. Similar effects of the two cosolutes on the tetramerization equilibrium observed for a shorter, nonaggregating huntingtin variant with a 7-glutamine repeat, httex1Q7, lend confidence to the conclusions drawn from the fits to the httex1Q35 aggregation kinetics.


Subject(s)
Huntingtin Protein , Muramidase , Huntingtin Protein/chemistry , Huntingtin Protein/genetics , Huntingtin Protein/metabolism , Kinetics , Muramidase/chemistry , Muramidase/metabolism , Humans , Dextrans/chemistry , Peptides/chemistry , Nuclear Magnetic Resonance, Biomolecular , Protein Aggregates/drug effects , Macromolecular Substances/chemistry , Protein Multimerization/drug effects , Magnetic Resonance Spectroscopy
5.
Chem Commun (Camb) ; 60(53): 6717-6727, 2024 Jun 27.
Article in English | MEDLINE | ID: mdl-38835221

ABSTRACT

Fibril formation is a key feature in neurodegenerative diseases like Alzheimer's, Parkinson's, and systemic amyloidosis. Polyphenols, found in plant-based foods, show promise in inhibiting fibril formation and disrupting disease progression. The ability of polyphenols to break the amyloid fibrils of many disease-linked proteins has been tested in numerous studies. Polyphenols have their distinctive mechanism of action. They behave differently on various events in the aggregation pathway. Their action also differs for different proteins. Some polyphenols only inhibit the formation of fibrils whereas others break the preformed fibrils. Some break the fibrils into smaller species, and some change them to other morphologies. This article delves into the intricate molecular mechanisms underlying the inhibitory effects of polyphenols on fibrillogenesis, shedding light on their interactions with amyloidogenic proteins and the disruption of fibril assembly pathways. However, addressing the challenges associated with solubility, stability, and bioavailability of polyphenols is crucial. The current strategies involve nanotechnology to improve the solubility and bioavailability, thus showing the potential to enhance the efficacy of polyphenols as therapeutics. Advancements in structural biology, computational modeling, and biophysics have provided insights into polyphenol-fibril interactions, offering hope for novel therapies for neurodegenerative diseases and amyloidosis.


Subject(s)
Polyphenols , Protein Multimerization/drug effects , Polyphenols/chemistry , Polyphenols/pharmacology , Ligands , Protein Conformation , Models, Molecular , Amyloid/chemistry , Kinetics
6.
Biomolecules ; 14(6)2024 Jun 05.
Article in English | MEDLINE | ID: mdl-38927062

ABSTRACT

Rattusin, an α-defensin-related antimicrobial peptide isolated from the small intestine of rats, has been previously characterized through NMR spectroscopy to elucidate its three-dimensional structure, revealing a C2 homodimeric scaffold stabilized by five disulfide bonds. This study aimed to identify the functional region of rattusin by designing and synthesizing various short analogs, subsequently leading to the development of novel peptide-based antibiotics. The analogs, designated as F1, F2, F3, and F4, were constructed based on the three-dimensional configuration of rattusin, among which F2 is the shortest peptide and exhibited superior antimicrobial efficacy compared to the wild-type peptide. The central cysteine residue of F2 prompted an investigation into its potential to form a dimer at neutral pH, which is critical for its antimicrobial function. This activity was abolished upon the substitution of the cysteine residue with serine, indicating the necessity of dimerization for antimicrobial action. Further, we synthesized ß-hairpin-like analogs, both parallel and antiparallel, based on the dimeric structure of F2, which maintained comparable antimicrobial potency. In contrast to rattusin, which acts by disrupting bacterial membranes, the F2 dimer binds directly to DNA, as evidenced by fluorescence assays and DNA retardation experiments. Importantly, F2 exhibited negligible cytotoxicity up to 515 µg/mL, assessed via hemolysis and MTT assays, underscoring its potential as a lead compound for novel peptide-based antibiotic development.


Subject(s)
alpha-Defensins , Animals , alpha-Defensins/chemistry , alpha-Defensins/pharmacology , alpha-Defensins/chemical synthesis , Microbial Sensitivity Tests , Rats , Antimicrobial Peptides/chemistry , Antimicrobial Peptides/pharmacology , Antimicrobial Peptides/chemical synthesis , Protein Multimerization/drug effects , DNA/metabolism , DNA/chemistry , Hemolysis/drug effects , Anti-Infective Agents/pharmacology , Anti-Infective Agents/chemistry , Humans , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/chemical synthesis , Amino Acid Sequence
7.
Antiviral Res ; 228: 105949, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38942150

ABSTRACT

The SARS-CoV-2 Spike glycoprotein (S) utilizes a unique trimeric conformation to interact with the ACE2 receptor on host cells, making it a prime target for inhibitors that block viral entry. We have previously identified a novel proteinaceous cavity within the Spike protein homotrimer that could serve as a binding site for small molecules. However, it is not known whether these molecules would inhibit, stimulate, or have no effect on viral replication. To address this, we employed structural-based screening to identify small molecules that dock into the trimer cavity and assessed their impact on viral replication. Our findings show that a cohort of identified small molecules binding to the Spike trimer cavity effectively reduces the replication of various SARS-CoV-2 variants. These molecules exhibited inhibitory effects on B.1 (European original, D614G, EDB2) and B.1.617.2 (δ) variants, while showing moderate activity against the B.1.1.7 (α) variant. We further categorized these molecules into distinct groups based on their structural similarities. Our experiments demonstrated a dose-dependent viral replication inhibitory activity of these compounds, with some, like BCC0040453 exhibiting no adverse effects on cell viability even at high concentrations. Further investigation revealed that pre-incubating virions with compounds like BCC0031216 at different temperatures significantly inhibited viral replication, suggesting their specificity towards the S protein. Overall, our study highlights the inhibitory impact of a diverse set of chemical molecules on the biological activity of the Spike protein. These findings provide valuable insights into the role of the trimer cavity in the viral replication cycle and aid drug discovery programs aimed at targeting the coronavirus family.


Subject(s)
Antiviral Agents , SARS-CoV-2 , Spike Glycoprotein, Coronavirus , Virus Replication , Spike Glycoprotein, Coronavirus/metabolism , Spike Glycoprotein, Coronavirus/chemistry , Spike Glycoprotein, Coronavirus/genetics , Spike Glycoprotein, Coronavirus/antagonists & inhibitors , SARS-CoV-2/drug effects , SARS-CoV-2/physiology , Virus Replication/drug effects , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , Chlorocebus aethiops , Vero Cells , Animals , Binding Sites , Virus Internalization/drug effects , COVID-19/virology , Protein Multimerization/drug effects , COVID-19 Drug Treatment , Small Molecule Libraries/pharmacology
8.
Anal Chem ; 96(27): 10962-10968, 2024 07 09.
Article in English | MEDLINE | ID: mdl-38925633

ABSTRACT

Overexpression of receptor tyrosine kinases (RTKs) or binding to ligands can lead to the formation of specific unliganded and liganded RTK dimers, and these two RTK dimers are potential targets for preventing tumor metastasis. Traditional RTK dimer inhibitor analysis was mostly based on end point assays, which required cumbersome cell handling and behavior monitoring. There are still challenges in developing intuitive process-based analytical methods to study RTK dimer inhibitors, especially those used to visually distinguish between unliganded and liganded RTK dimer inhibitors. Herein, taking the mesenchymal-epithelial transition factor (MET) receptor, an intuitive method for evaluating MET inhibitors has been developed based on atomic force microscopy (AFM) lifetime analysis. The time interval between the start of the force and the bond break point was regarded as the bond lifetime, which could reflect the stability of the MET dimer. The results showed that there was a significant difference in the lifetime (τ) of unliganded MET dimers (τ1 = 207.87 ± 4.69 ms) and liganded MET dimers (τ2 = 330.58 ± 15.60 ms) induced by the hepatocyte growth factor, and aptamer SL1 could decrease τ1 and τ2, suggesting that SL1 could inhibit both unliganded and liganded MET dimers. However, heparin only decreased τ2, suggesting that it could inhibit only the liganded MET dimer. AFM-based lifetime analysis methods could monitor RTK dimer status rather than provide overall average results, allowing for intuitive process-based analysis and evaluation of RTK dimers and related inhibitors at the single-molecule level. This study provides a novel complementary strategy for simple and intuitive RTK inhibitor research.


Subject(s)
Microscopy, Atomic Force , Protein Kinase Inhibitors , Proto-Oncogene Proteins c-met , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/chemistry , Humans , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Proto-Oncogene Proteins c-met/metabolism , Protein Multimerization/drug effects , Ligands , Hepatocyte Growth Factor/metabolism , Aptamers, Nucleotide/chemistry , Aptamers, Nucleotide/metabolism
9.
Chembiochem ; 25(13): e202400001, 2024 Jul 02.
Article in English | MEDLINE | ID: mdl-38720172

ABSTRACT

Coronavirus (CoV) infections have caused contagious and fatal respiratory diseases in humans worldwide. CoV 3-chymotrypsin-like proteases (3CLpro or Mpro) play an important role in viral maturation, and maintenance of their dimeric conformation is crucial for viral activity. Therefore, allosterically regulated dimerization of 3CLpro can be employed as a drug development target. Here, we investigated the allosteric regulatory mechanism of 3CLpro dimerization by using hydrogen/deuterium exchange coupled with mass spectrometry (HDX-MS) technology. We found that the FLAG tag directly coupled to the N-finger of 3CLpro significantly increased HDX kinetics at the dimer interface, and 3CLpro transformed from a dimer to a monomer. The 3CLpro mutants of SARS-CoV-2, which are monomeric, also exhibited increased deuterium exchange. Binding of the allosteric inhibitor Gastrodenol to most betacoronavirus 3CLpros led to increased allosteric deuterium exchange, resulting in the monomeric conformation of the CoV 3CLpro upon binding. Molecular dynamics (MD) simulation analysis further indicated the molecular mechanism of action of Gastrodenol on CoV 3CLpro: binding of Gastrodenol to SARS-CoV-2 3CLpro destroyed the hydrogen bond in the dimer interface. These results suggest that Gastrodenol may be a potential broad-spectrum anti-betacoronavirus drug.


Subject(s)
Coronavirus 3C Proteases , Hydrogen Deuterium Exchange-Mass Spectrometry , Molecular Dynamics Simulation , SARS-CoV-2 , Allosteric Regulation/drug effects , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/metabolism , Coronavirus 3C Proteases/chemistry , SARS-CoV-2/enzymology , SARS-CoV-2/drug effects , Humans , Protein Multimerization/drug effects , Kinetics , Deuterium Exchange Measurement
10.
Elife ; 132024 May 14.
Article in English | MEDLINE | ID: mdl-38742856

ABSTRACT

The type II class of RAF inhibitors currently in clinical trials paradoxically activate BRAF at subsaturating concentrations. Activation is mediated by induction of BRAF dimers, but why activation rather than inhibition occurs remains unclear. Using biophysical methods tracking BRAF dimerization and conformation, we built an allosteric model of inhibitor-induced dimerization that resolves the allosteric contributions of inhibitor binding to the two active sites of the dimer, revealing key differences between type I and type II RAF inhibitors. For type II inhibitors the allosteric coupling between inhibitor binding and BRAF dimerization is distributed asymmetrically across the two dimer binding sites, with binding to the first site dominating the allostery. This asymmetry results in efficient and selective induction of dimers with one inhibited and one catalytically active subunit. Our allosteric models quantitatively account for paradoxical activation data measured for 11 RAF inhibitors. Unlike type II inhibitors, type I inhibitors lack allosteric asymmetry and do not activate BRAF homodimers. Finally, NMR data reveal that BRAF homodimers are dynamically asymmetric with only one of the subunits locked in the active αC-in state. This provides a structural mechanism for how binding of only a single αC-in inhibitor molecule can induce potent BRAF dimerization and activation.


Subject(s)
Protein Kinase Inhibitors , Protein Multimerization , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Proto-Oncogene Proteins B-raf/chemistry , Allosteric Regulation/drug effects , Protein Kinase Inhibitors/pharmacology , Protein Kinase Inhibitors/metabolism , Protein Multimerization/drug effects , Humans , Protein Conformation , Protein Binding , Models, Molecular
11.
Chem Commun (Camb) ; 60(37): 4910-4913, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38623638

ABSTRACT

Several natural cytotoxic C2-symmetric bis-lactones, such as swinholide A and rhizopodin, sequester actin dimer from the actin network and potently inhibit actin dynamics. To develop new protein-protein interaction (PPI) modulators, we synthesized structurally simplified actin-binding side-chain dimers of antitumor macrolide aplyronine A. By fixing the two side-chains closer than those of rhizopodin, the C4 linker analog depolymerized filamentous actin more potently than natural aplyronines. Cross-link experiments revealed that actin dimer was formed by treatment with the C4 linker analog. Molecular dynamics simulations showed that this analog significantly changed the interaction and spatial arrangement of the two actins compared to those in rhizopodin to provide a highly distorted and twisted orientation in the complex. Our study may promote the development of PPI-based anticancer and other drug leads related to cytoskeletal dynamics.


Subject(s)
Actins , Macrolides , Protein Multimerization , Actin Depolymerizing Factors/chemistry , Actin Depolymerizing Factors/pharmacology , Actins/metabolism , Actins/chemistry , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/chemical synthesis , Dimerization , Macrolides/chemistry , Macrolides/pharmacology , Macrolides/chemical synthesis , Molecular Dynamics Simulation , Protein Multimerization/drug effects
12.
J Phys Chem B ; 128(18): 4354-4366, 2024 May 09.
Article in English | MEDLINE | ID: mdl-38683784

ABSTRACT

G protein-coupled receptors (GPCRs) are a major gateway to cellular signaling, which respond to ligands binding at extracellular sites through allosteric conformational changes that modulate their interactions with G proteins and arrestins at intracellular sites. High-resolution structures in different ligand states, together with spectroscopic studies and molecular dynamics simulations, have revealed a rich conformational landscape of GPCRs. However, their supramolecular structure and spatiotemporal distribution is also thought to play a significant role in receptor activation and signaling bias within the native cell membrane environment. Here, we applied single-molecule fluorescence techniques, including single-particle tracking, single-molecule photobleaching, and fluorescence correlation spectroscopy, to characterize the diffusion and oligomerization behavior of the muscarinic M1 receptor (M1R) in live cells. Control samples included the monomeric protein CD86 and fixed cells, and experiments performed in the presence of different orthosteric M1R ligands and of several compounds known to change the fluidity and organization of the lipid bilayer. M1 receptors exhibit Brownian diffusion characterized by three diffusion constants: confined/immobile (∼0.01 µm2/s), slow (∼0.04 µm2/s), and fast (∼0.14 µm2/s), whose populations were found to be modulated by both orthosteric ligands and membrane disruptors. The lipid raft disruptor C6 ceramide led to significant changes for CD86, while the diffusion of M1R remained unchanged, indicating that M1 receptors do not partition in lipid rafts. The extent of receptor oligomerization was found to be promoted by increasing the level of expression and the binding of orthosteric ligands; in particular, the agonist carbachol elicited a large increase in the fraction of M1R oligomers. This study provides new insights into the balance between conformational and environmental factors that define the movement and oligomerization states of GPCRs in live cells under close-to-native conditions.


Subject(s)
Receptor, Muscarinic M1 , Ligands , Receptor, Muscarinic M1/metabolism , Receptor, Muscarinic M1/chemistry , Diffusion , Humans , Cell Membrane/metabolism , Cell Membrane/chemistry , Protein Multimerization/drug effects , Animals , Spectrometry, Fluorescence , Molecular Dynamics Simulation , Lipid Bilayers/chemistry , Lipid Bilayers/metabolism
13.
Int J Biol Macromol ; 267(Pt 1): 131392, 2024 May.
Article in English | MEDLINE | ID: mdl-38582483

ABSTRACT

The main protease (Mpro) of SARS-CoV-2 is critical in the virus's replication cycle, facilitating the maturation of polyproteins into functional units. Due to its conservation across taxa, Mpro is a promising target for broad-spectrum antiviral drugs. Targeting Mpro with small molecule inhibitors, such as nirmatrelvir combined with ritonavir (Paxlovid™), which the FDA has approved for post-exposure treatment and prophylaxis, can effectively interrupt the replication process of the virus. A key aspect of Mpro's function is its ability to form a functional dimer. However, the mechanics of dimerization and its influence on proteolytic activity remain less understood. In this study, we utilized biochemical, structural, and molecular modelling approaches to explore Mpro dimerization. We evaluated critical residues, specifically Arg4 and Arg298, that are essential for dimerization. Our results show that changes in the oligomerization state of Mpro directly affect its enzymatic activity and dimerization propensity. We discovered a synergistic relationship influencing dimer formation, involving both intra- and intermolecular interactions. These findings highlight the potential for developing allosteric inhibitors targeting Mpro, offering promising new directions for therapeutic strategies.


Subject(s)
Antiviral Agents , Coronavirus 3C Proteases , Protein Multimerization , SARS-CoV-2 , SARS-CoV-2/drug effects , Protein Multimerization/drug effects , Coronavirus 3C Proteases/antagonists & inhibitors , Coronavirus 3C Proteases/chemistry , Coronavirus 3C Proteases/metabolism , Humans , Antiviral Agents/pharmacology , Antiviral Agents/chemistry , COVID-19 Drug Treatment , Models, Molecular , COVID-19/virology , Protease Inhibitors/pharmacology , Protease Inhibitors/chemistry
14.
Nature ; 621(7977): 206-214, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37648856

ABSTRACT

Transient receptor potential (TRP) channels are a large, eukaryotic ion channel superfamily that control diverse physiological functions, and therefore are attractive drug targets1-5. More than 210 structures from more than 20 different TRP channels have been determined, and all are tetramers4. Despite this wealth of structures, many aspects concerning TRPV channels remain poorly understood, including the pore-dilation phenomenon, whereby prolonged activation leads to increased conductance, permeability to large ions and loss of rectification6,7. Here, we used high-speed atomic force microscopy (HS-AFM) to analyse membrane-embedded TRPV3 at the single-molecule level and discovered a pentameric state. HS-AFM dynamic imaging revealed transience and reversibility of the pentamer in dynamic equilibrium with the canonical tetramer through membrane diffusive protomer exchange. The pentamer population increased upon diphenylboronic anhydride (DPBA) addition, an agonist that has been shown to induce TRPV3 pore dilation. On the basis of these findings, we designed a protein production and data analysis pipeline that resulted in a cryogenic-electron microscopy structure of the TRPV3 pentamer, showing an enlarged pore compared to the tetramer. The slow kinetics to enter and exit the pentameric state, the increased pentamer formation upon DPBA addition and the enlarged pore indicate that the pentamer represents the structural correlate of pore dilation. We thus show membrane diffusive protomer exchange as an additional mechanism for structural changes and conformational variability. Overall, we provide structural evidence for a non-canonical pentameric TRP-channel assembly, laying the foundation for new directions in TRP channel research.


Subject(s)
Protein Multimerization , TRPV Cation Channels , Anhydrides/chemistry , Anhydrides/pharmacology , Data Analysis , Diffusion , Protein Subunits/chemistry , Protein Subunits/drug effects , Protein Subunits/metabolism , TRPV Cation Channels/chemistry , TRPV Cation Channels/drug effects , TRPV Cation Channels/metabolism , TRPV Cation Channels/ultrastructure , Microscopy, Atomic Force , Molecular Targeted Therapy , Cryoelectron Microscopy , Protein Structure, Quaternary/drug effects , Protein Multimerization/drug effects
15.
Chem Biol Interact ; 378: 110489, 2023 Jun 01.
Article in English | MEDLINE | ID: mdl-37059213

ABSTRACT

We assessed the mechanism of human androgen receptor-mediated endocrine-disrupting effect by a triazole fungicide, metconazole in this study. The internationally validated stably transfected transactivation (STTA) in vitro assay, which was established for determination of a human androgen receptor (AR) agonist/antagonist by using 22Rv1/MMTV_GR-KO cell line, alongside an in vitro reporter-gene assay to confirm AR homodimerization was used. The STTA in vitro assay results showed that metconazole is a true AR antagonist. Furthermore, the results from the in vitro reporter-gene assay and western blotting showed that metconazole blocks the nuclear transfer of cytoplasmic AR proteins by suppressing the homodimerization of AR. These results suggest that metconazole can be considered to have an AR-mediated endocrine-disrupting effect. Additionally, the evidence from this study might help identify the endocrine-disrupting mechanism of triazole fungicides containing a phenyl ring.


Subject(s)
Androgen Receptor Antagonists , Endocrine Disruptors , Fungicides, Industrial , Protein Multimerization , Receptors, Androgen , Transcriptional Activation , Triazoles , Triazoles/chemistry , Triazoles/toxicity , Fungicides, Industrial/chemistry , Fungicides, Industrial/toxicity , Protein Multimerization/drug effects , Humans , Receptors, Androgen/chemistry , Receptors, Androgen/metabolism , Endocrine Disruptors/chemistry , Endocrine Disruptors/pharmacology , Androgen Receptor Antagonists/chemistry , Androgen Receptor Antagonists/toxicity , Cell Line, Tumor , Transcriptional Activation/drug effects , Cytotoxins/chemistry , Cytotoxins/toxicity
16.
Proc Natl Acad Sci U S A ; 119(35): e2201204119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994658

ABSTRACT

Bacteria utilize two-component system (TCS) signal transduction pathways to sense and adapt to changing environments. In a typical TCS, a stimulus induces a sensor histidine kinase (SHK) to phosphorylate a response regulator (RR), which then dimerizes and activates a transcriptional response. Here, we demonstrate that oligomerization-dependent depolarization of excitation light by fused mNeonGreen fluorescent protein probes enables real-time monitoring of RR dimerization dynamics in live bacteria. Using inducible promoters to independently express SHKs and RRs, we detect RR dimerization within seconds of stimulus addition in several model pathways. We go on to combine experiments with mathematical modeling to reveal that TCS phosphosignaling accelerates with SHK expression but decelerates with RR expression and SHK phosphatase activity. We further observe pulsatile activation of the SHK NarX in response to addition and depletion of the extracellular electron acceptor nitrate when the corresponding TCS is expressed from both inducible systems and the native chromosomal operon. Finally, we combine our method with polarized light microscopy to enable single-cell measurements of RR dimerization under changing stimulus conditions. Direct in vivo characterization of RR oligomerization dynamics should enable insights into the regulation of bacterial physiology.


Subject(s)
Bacteria , Bacterial Proteins , Histidine Kinase , Microbial Viability , Bacteria/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/radiation effects , Electrons , Histidine Kinase/genetics , Histidine Kinase/metabolism , Microscopy, Polarization , Nitrates , Operon/genetics , Phosphorylation , Promoter Regions, Genetic , Protein Multimerization/drug effects , Single-Cell Analysis , Time Factors
17.
Molecules ; 27(4)2022 Feb 18.
Article in English | MEDLINE | ID: mdl-35209177

ABSTRACT

Alzheimer's disease displays aggregates of the amyloid-beta (Aß) peptide in the brain, and there is increasing evidence that cholesterol may contribute to the pathogenesis of the disease. Though many experimental and theoretical studies have focused on the interactions of Aß oligomers with membrane models containing cholesterol, an understanding of the effect of free cholesterol on small Aß42 oligomers is not fully established. To address this question, we report on replica exchange with a solute tempering simulation of an Aß42 trimer with cholesterol and compare it with a previous replica exchange molecular dynamics simulation. We show that the binding hot spots of cholesterol are rather complex, involving hydrophobic residues L17-F20 and L30-M35 with a non-negligible contribution of loop residues D22-K28 and N-terminus residues. We also examine the effects of cholesterol on the trimers of the disease-causing A21G and disease-protective A2T mutations by molecular dynamics simulations. We show that these two mutations moderately impact cholesterol-binding modes. In our REST2 simulations, we find that cholesterol is rarely inserted into aggregates but rather attached as dimers and trimers at the surface of Aß42 oligomers. We propose that cholesterol acts as a glue to speed up the formation of larger aggregates; this provides a mechanistic link between cholesterol and Alzheimer's disease.


Subject(s)
Amyloid beta-Peptides/chemistry , Cholesterol/chemistry , Mutant Proteins/chemistry , Peptide Fragments/chemistry , Protein Multimerization , Amino Acid Sequence , Cholesterol/pharmacology , Hydrogen-Ion Concentration , Molecular Conformation , Molecular Docking Simulation , Molecular Dynamics Simulation , Protein Aggregates , Protein Aggregation, Pathological , Protein Binding , Protein Multimerization/drug effects , Structure-Activity Relationship
18.
Int J Mol Sci ; 23(3)2022 Jan 31.
Article in English | MEDLINE | ID: mdl-35163573

ABSTRACT

Inflammasomes are multiprotein complexes that represent critical elements of the inflammatory response. The dysregulation of the best-characterized complex, the NLRP3 inflammasome, has been linked to the pathogenesis of diseases such as multiple sclerosis, type 2 diabetes mellitus, Alzheimer's disease, and cancer. While there exist molecular inhibitors specific for the various components of inflammasome complexes, no currently reported inhibitors specifically target NLRP3PYD homo-oligomerization. In the present study, we describe the identification of QM380 and QM381 as NLRP3PYD homo-oligomerization inhibitors after screening small molecules from the MyriaScreen library using a split-luciferase complementation assay. Our results demonstrate that these NLRP3PYD inhibitors interfere with ASC speck formation, inhibit pro-inflammatory cytokine IL1-ß release, and decrease pyroptotic cell death. We employed spectroscopic techniques and computational docking analyses with QM380 and QM381 and the PYD domain to confirm the experimental results and predict possible mechanisms underlying the inhibition of NLRP3PYD homo-interactions.


Subject(s)
Anti-Inflammatory Agents , NLR Family, Pyrin Domain-Containing 3 Protein , Protein Multimerization/drug effects , Pyroptosis/drug effects , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacology , HEK293 Cells , Humans , NLR Family, Pyrin Domain-Containing 3 Protein/antagonists & inhibitors , NLR Family, Pyrin Domain-Containing 3 Protein/chemistry , NLR Family, Pyrin Domain-Containing 3 Protein/genetics , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism
19.
Molecules ; 27(1)2022 Jan 05.
Article in English | MEDLINE | ID: mdl-35011550

ABSTRACT

A series of eleven 4-substituted 5,6,7,8-tetrahydrobenzo[4,5]thieno[2,3-d]pyrimidines were designed and synthesized and their biological activities were evaluated. Synthesis involved the Gewald reaction to synthesize ethyl 2-amino-4,5,6,7-tetrahydrobenzo[b]thiophene-3-carboxylate ring, and SNAr reactions. Compound 4 was 1.6- and ~7-fold more potent than the lead compound 1 in cell proliferation and microtubule depolymerization assays, respectively. Compounds 4, 5 and 7 showed the most potent antiproliferative effects (IC50 values < 40 nM), while compounds 6, 8, 10, 12 and 13 had lower antiproliferative potencies (IC50 values of 53-125 nM). Additionally, compounds 4-8, 10 and 12-13 circumvented Pgp and ßIII-tubulin mediated drug resistance, mechanisms that diminish the clinical efficacy of paclitaxel (PTX). In the NCI-60 cell line panel, compound 4 exhibited an average GI50 of ~10 nM in the 40 most sensitive cell lines. Compound 4 demonstrated statistically significant antitumor effects in a murine MDA-MB-435 xenograft model.


Subject(s)
Chemistry Techniques, Synthetic , Drug Design , Pyrimidines/chemistry , Pyrimidines/pharmacology , Tubulin Modulators/chemistry , Tubulin Modulators/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm , Humans , Models, Molecular , Molecular Conformation , Molecular Structure , Protein Multimerization/drug effects , Pyrimidines/chemical synthesis , Structure-Activity Relationship , Tubulin/chemistry , Tubulin/metabolism , Tubulin Modulators/chemical synthesis
20.
Int J Mol Sci ; 23(2)2022 Jan 14.
Article in English | MEDLINE | ID: mdl-35055076

ABSTRACT

The most common type of dementia, Alzheimer's disease, is associated with senile plaques formed by the filamentous aggregation of hydrophobic amyloid-ß (Aß) in the brains of patients. Small oligomeric assemblies also occur and drugs and chemical compounds that can interact with such assemblies have attracted much attention. However, these compounds need to be solubilized in appropriate solvents, such as ethanol, which may also destabilize their protein structures. As the impact of ethanol on oligomeric Aß assembly is unknown, we investigated the effect of various concentrations of ethanol (0 to 7.2 M) on Aß pentameric assemblies (Aßp) by combining blue native-PAGE (BN-PAGE) and ambient air atomic force microscopy (AFM). This approach was proven to be very convenient and reliable for the quantitative analysis of Aß assembly. The Gaussian analysis of the height histogram obtained from the AFM images was correlated with band intensity on BN-PAGE for the quantitative estimation of Aßp. Our observations indicated up to 1.4 M (8.3%) of added ethanol can be used as a solvent/vehicle without quantitatively affecting Aß pentamer stability. Higher concentration induced significant destabilization of Aßp and eventually resulted in the complete disassembly of Aßp.


Subject(s)
Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/metabolism , Ethanol/chemistry , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Protein Aggregates/drug effects , Protein Multimerization/drug effects , Electrophoresis , Ethanol/pharmacology , Humans , Microscopy, Atomic Force , Protein Aggregation, Pathological
SELECTION OF CITATIONS
SEARCH DETAIL