Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 420
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nucleic Acids Res ; 50(D1): D518-D525, 2022 01 07.
Article in English | MEDLINE | ID: mdl-34570219

ABSTRACT

Two-thirds of signaling substances, several sensory stimuli and over one-third of drugs act via receptors coupling to G proteins. Here, we present an online platform for G protein research with reference data and tools for analysis, visualization and design of scientific studies across disciplines and areas. This platform may help translate new pharmacological, structural and genomic data into insights on G protein signaling vital for human physiology and medicine. The G protein database is accessible at https://gproteindb.org.


Subject(s)
Databases, Protein , GTP-Binding Proteins/metabolism , Prescription Drugs/chemistry , Receptors, G-Protein-Coupled/metabolism , Small Molecule Libraries/chemistry , Software , Amino Acid Sequence , Binding Sites , Eukaryotic Cells/cytology , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , GTP-Binding Proteins/antagonists & inhibitors , GTP-Binding Proteins/chemistry , GTP-Binding Proteins/genetics , Gene Expression Regulation , Humans , Models, Molecular , Molecular Sequence Annotation , Mutation , Prescription Drugs/pharmacology , Protein Binding , Protein Conformation, alpha-Helical , Protein Conformation, beta-Strand , Protein Interaction Domains and Motifs , Receptors, G-Protein-Coupled/antagonists & inhibitors , Receptors, G-Protein-Coupled/chemistry , Receptors, G-Protein-Coupled/genetics , Sequence Alignment , Sequence Homology, Amino Acid , Signal Transduction , Small Molecule Libraries/pharmacology , Structure-Activity Relationship
2.
J Antimicrob Chemother ; 78(2): 512-520, 2023 02 01.
Article in English | MEDLINE | ID: mdl-36512374

ABSTRACT

BACKGROUND: Following the invasion of eukaryotic cells, Salmonella enterica serovar Typhimurium replaces PBP2/PBP3, main targets of ß-lactam antibiotics, with PBP2SAL/PBP3SAL, two homologue peptidoglycan synthases absent in Escherichia coli. PBP3SAL promotes pathogen cell division in acidic environments independently of PBP3 and shows low affinity for ß-lactams that bind to PBP3 such as aztreonam, cefepime, cefotaxime, ceftazidime, ceftriaxone, cefuroxime and cefalotin. OBJECTIVES: To find compounds with high affinity for PBP3SAL to control Salmonella intracellular infections. METHODS: An S. Typhimurium ΔPBP3 mutant that divides using PBP3SAL and its parental wild-type strain, were exposed to a library of 1520 approved drugs in acidified (pH 4.6) nutrient-rich LB medium. Changes in optical density associated with cell filamentation, a read-out of blockage in cell division, were monitored. Compounds causing filamentation in the ΔPBP3 mutant but not in wild-type strain-the latter strain expressing both PBP3 and PBP3SAL in LB pH 4.6-were selected for further study. The bactericidal effect due to PBP3SAL inhibition was evaluated in vitro using a bacterial infection model of cultured fibroblasts. RESULTS: The cephalosporin cefotiam exhibited higher affinity for PBP3SAL than for PBP3 in bacteria growing in acidified LB pH 4.6 medium. Cefotiam also proved to be effective against intracellular Salmonella in a PBP3SAL-dependent manner. Conversely, cefuroxime, which has higher affinity for PBP3, showed decreased effectiveness in killing intracellular Salmonella. CONCLUSIONS: Antibiotics with affinity for PBP3SAL, like the cephalosporin cefotiam, have therapeutic value for treating Salmonella intracellular infections.


Subject(s)
Anti-Bacterial Agents , Bacterial Proteins , Cefuroxime , Eukaryotic Cells , Penicillin-Binding Proteins , Salmonella typhimurium , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/metabolism , Bacterial Proteins/metabolism , Cefotiam/metabolism , Cefotiam/pharmacology , Ceftazidime/pharmacology , Cefuroxime/pharmacology , Cephalosporins/pharmacology , Cephalosporins/metabolism , Escherichia coli , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Monobactams/pharmacology , Penicillin-Binding Proteins/genetics , Penicillin-Binding Proteins/metabolism , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism
3.
Int J Mol Sci ; 22(11)2021 May 31.
Article in English | MEDLINE | ID: mdl-34072929

ABSTRACT

Metalloid tellurium is characterized as a chemical element belonging to the chalcogen group without known biological function. However, its compounds, especially the oxyanions, exert numerous negative effects on both prokaryotic and eukaryotic organisms. Recent evidence suggests that increasing environmental pollution with tellurium has a causal link to autoimmune, neurodegenerative and oncological diseases. In this review, we provide an overview about the current knowledge on the mechanisms of tellurium compounds' toxicity in bacteria and humans and we summarise the various ways organisms cope and detoxify these compounds. Over the last decades, several gene clusters conferring resistance to tellurium compounds have been identified in a variety of bacterial species and strains. These genetic determinants exhibit great genetic and functional diversity. Besides the existence of specific resistance mechanisms, tellurium and its toxic compounds interact with molecular systems, mediating general detoxification and mitigation of oxidative stress. We also discuss the similarity of tellurium and selenium biochemistry and the impact of their compounds on humans.


Subject(s)
Eukaryotic Cells/drug effects , Oxidative Stress/drug effects , Prokaryotic Cells/drug effects , Tellurium/adverse effects , Anions/adverse effects , Bacteria/drug effects , Environmental Pollution/analysis , Humans , Selenium/chemistry , Tellurium/chemistry , Tellurium/toxicity
4.
Int J Mol Sci ; 22(22)2021 Nov 16.
Article in English | MEDLINE | ID: mdl-34830229

ABSTRACT

The decellularization of plant-based biomaterials to generate tissue-engineered substitutes or in vitro cellular models has significantly increased in recent years. These vegetal tissues can be sourced from plant leaves and stems or fruits and vegetables, making them a low-cost, accessible, and sustainable resource from which to generate three-dimensional scaffolds. Each construct is distinct, representing a wide range of architectural and mechanical properties as well as innate vasculature networks. Based on the rapid rise in interest, this review aims to detail the current state of the art and presents the future challenges and perspectives of these unique biomaterials. First, we consider the different existing decellularization techniques, including chemical, detergent-free, enzymatic, and supercritical fluid approaches that are used to generate such scaffolds and examine how these protocols can be selected based on plant cellularity. We next examine strategies for cell seeding onto the plant-derived constructs and the importance of the different functionalization methods used to assist in cell adhesion and promote cell viability. Finally, we discuss how their structural features, such as inherent vasculature, porosity, morphology, and mechanical properties (i.e., stiffness, elasticity, etc.) position plant-based scaffolds as a unique biomaterial and drive their use for specific downstream applications. The main challenges in the field are presented throughout the discussion, and future directions are proposed to help improve the development and use of vegetal constructs in biomedical research.


Subject(s)
Biocompatible Materials/chemistry , Cellulose/chemistry , Extracellular Matrix/chemistry , Plant Leaves/chemistry , Tissue Engineering/methods , Tissue Scaffolds/chemistry , Animals , Biocompatible Materials/pharmacology , Biomechanical Phenomena , Cell Adhesion , Cell Survival , Cellulose/pharmacology , Detergents/chemistry , Elastic Modulus , Eukaryotic Cells/cytology , Eukaryotic Cells/drug effects , Eukaryotic Cells/physiology , Humans , Plant Cells/chemistry , Plant Leaves/anatomy & histology , Plant Stems/anatomy & histology , Plant Stems/chemistry , Plants/anatomy & histology , Plants/chemistry , Solvents/chemistry
5.
Molecules ; 26(24)2021 Dec 07.
Article in English | MEDLINE | ID: mdl-34946504

ABSTRACT

Imaging-guided delivery is developed for hydrophobic drugs, and to a much lesser extent, hydrophilic ones. In this work we have designed a novel strategy for real-time monitoring of hydrophilic drug delivery. Traditionally, the drug and the dye are covalently attached to a nanocarrier or are electrostatically adsorbed. Recently, we found an efficient way to bind the drug by ion-paring with an appropriate counter-ion to form the aggregate that embeds a hydrophobic dye with a considerable fluorescence enhancement. We synthesized a series of carbocyanine dyes of hydrophobicity sufficient for solubilization in hydrophobic ion pairs, which restores their emission in the near-infrared (NIR) region upon the formation of the ternary aggregates. To avoid using toxic surfactants, we applied an amphiphilic polymer-oligomer poly(hexamethylene guanidine) (PHMG) as a counter-ion. Сeftriaxone was used as a model hydrophilic drug ensuring the highest fluorescent signal. The so-formed drug-counter-ion-dye aggregates were encapsulated into a cross-linked maleated chitosan carrier. Confocal laser scanning microscopy (CLSM) studies have demonstrated internalization of the encapsulated model drug by breast adenocarcinoma cells at 40 min after treatment. These results suggest the potential application of hydrophobic ion pairs containing an NIR dye in imaging-guided delivery of hydrophilic compounds.


Subject(s)
Carbocyanines/chemistry , Ceftriaxone/pharmacology , Chitosan/chemistry , Drug Delivery Systems , Eukaryotic Cells/drug effects , Guanidines/chemistry , Carbocyanines/chemical synthesis , Ceftriaxone/chemistry , Drug Carriers/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Ions/chemistry , Molecular Structure
6.
Nat Prod Rep ; 37(5): 677-702, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32022056

ABSTRACT

Covering: up to 2019Nature furnishes bioactive compounds (natural products) with complex chemical structures, yet with simple, sophisticated molecular mechanisms. When natural products exhibit their activities in cells or bodies, they first have to bind or react with a target molecule in/on the cell. The cell membrane is a major target for bioactive compounds. Recently, our understanding of the molecular mechanism of interactions between natural products and membrane lipids progressed with the aid of newly-developed analytical methods. New technology reconnects old compounds with membrane lipids, while new membrane-targeting molecules are being discovered through the screening for antimicrobial potential of natural products. This review article focuses on natural products that bind to eukaryotic membrane lipids, and includes clinically important molecules and key research tools. The chemical diversity of membrane-targeting natural products and the molecular basis of lipid recognition are described. The history of how their mechanism was unveiled, and how these natural products are used in research are also mentioned.


Subject(s)
Biological Products/chemistry , Biological Products/pharmacology , Cell Membrane/drug effects , Membrane Lipids/metabolism , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Eukaryotic Cells/drug effects , Humans , Membrane Lipids/chemistry
7.
Nat Prod Rep ; 37(5): 717-736, 2020 05 01.
Article in English | MEDLINE | ID: mdl-32067014

ABSTRACT

Covering: up to the end of 2019Diverse natural product small molecules have allowed critical insights into processes that govern eukaryotic cells' ability to secrete cytosolically synthesized secretory proteins into their surroundings or to insert newly synthesized integral membrane proteins into the lipid bilayer of the endoplasmic reticulum. In addition, many components of the endoplasmic reticulum, required for protein homeostasis or other processes such as lipid metabolism or maintenance of calcium homeostasis, are being investigated for their potential in modulating human disease conditions such as cancer, neurodegenerative conditions and diabetes. In this review, we cover recent findings up to the end of 2019 on natural products that influence protein secretion or impact ER protein homeostasis, and serve as powerful chemical tools to understand protein flux through the mammalian secretory pathway and as leads for the discovery of new therapeutics.


Subject(s)
Biological Products/pharmacology , Eukaryotic Cells/drug effects , Proteins/metabolism , Animals , Biological Products/chemistry , Calcium/metabolism , Endoplasmic Reticulum Stress/drug effects , Endoplasmic Reticulum Stress/physiology , Eukaryotic Cells/metabolism , Humans , Protein Transport/drug effects , RNA Splicing/drug effects , RNA Splicing/physiology
8.
Nat Prod Rep ; 37(5): 634-652, 2020 05 01.
Article in English | MEDLINE | ID: mdl-31764930

ABSTRACT

Covering: 2014-2019We review recent progress on natural products that target cytoskeletal components, including microtubules, actin, intermediate filaments, and septins and highlight their demonstrated and potential utility in the treatment of human disease. The anticancer efficacy of microtubule targeted agents identified from plants, microbes, and marine organisms is well documented. We highlight new microtubule targeted agents currently in clinical evaluations for the treatment of drug resistant cancers and the accumulating evidence that the anticancer efficacy of these agents is not solely due to their antimitotic effects. Indeed, the effects of microtubule targeted agents on interphase microtubules are leading to their potential for more mechanistically guided use in cancers as well as neurological disease. The discussion of these agents as more targeted drugs also prompts a reevaluation of our thinking about natural products that target other components of the cytoskeleton. For instance, actin active natural products are largely considered chemical probes and non-selective toxins. However, studies utilizing these probes have uncovered aspects of actin biology that can be more specifically targeted to potentially treat cancer, neurological disorders, and infectious disease. Compounds that target intermediate filaments and septins are understudied, but their continued discovery and mechanistic evaluations have implications for numerous therapeutic indications.


Subject(s)
Actins/metabolism , Biological Products/pharmacology , Cytoskeleton/drug effects , Microtubules/drug effects , Animals , Biological Products/chemistry , Colchicine/chemistry , Colchicine/metabolism , Colchicine/pharmacology , Cytoskeleton/metabolism , Drug Resistance, Neoplasm/drug effects , Eukaryotic Cells/cytology , Eukaryotic Cells/drug effects , Genome , Humans , Maytansine/chemistry , Maytansine/metabolism , Maytansine/pharmacology , Microtubules/metabolism , Neoplasms/drug therapy , Neoplasms/genetics , Nervous System Diseases/drug therapy , Nervous System Diseases/pathology , Taxoids/chemistry , Taxoids/pharmacology
9.
Mol Cell Probes ; 49: 101493, 2020 02.
Article in English | MEDLINE | ID: mdl-31816413

ABSTRACT

Autophagy plays a crucial role in eliminating protein aggregates, damaged organelles and invading pathogens. Genetically engineered cell line stably expressing green fluorescent protein (GFP)-tagged microtubule-associated protein light chain 3 (LC3) is extensively used to test autophagy through observing GFP puncta formation in the cells by fluorescence imaging. However, canine LC3 (cLC3) gene has not been cloned, therefore, GFP-tagged canine LC3 (GFP-cLC3) detection system has not been established. To generate GFP-cLC3 stably expressing canine-derived macrophages, the cLC3 cDNA was first amplified by RT-PCR and inserted into pEGFP-C1 plasmid to create GFP-cLC3 gene fusion. This genetic element was then transducted into canine macrophages mediated by lentivirus vector to generate the canine macrophages stably expressing fusion protein. Results showed that the sequence of cLC3 cloned in this study is highly homologous with other animals (80-95% homology). Phenotypic and functional analysis of these engineered cells revealed that GFP-cLC3 was indeed stably expressed and rapamycin or starvation can effectively induce GFP puncta formation in the cells, indicative of autophagosome formation. These GFP-cLC3-expressing cells may thus be useful to study autophagy in canine.


Subject(s)
Autophagy , Dogs/metabolism , Green Fluorescent Proteins/metabolism , Macrophages/cytology , Macrophages/metabolism , Microtubule-Associated Proteins/metabolism , Amino Acid Sequence , Animals , Autophagy/drug effects , Base Sequence , DNA, Complementary/genetics , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Gene Amplification , Genetic Vectors/metabolism , Genome , Lentivirus/genetics , Macrophages/drug effects , Madin Darby Canine Kidney Cells , Microtubule-Associated Proteins/chemistry , Microtubule-Associated Proteins/genetics , Recombinant Fusion Proteins/metabolism , Recombination, Genetic/genetics , Sirolimus/pharmacology
10.
Nature ; 513(7519): 517-22, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25209664

ABSTRACT

The ribosome is a molecular machine responsible for protein synthesis and a major target for small-molecule inhibitors. Compared to the wealth of structural information available on ribosome-targeting antibiotics in bacteria, our understanding of the binding mode of ribosome inhibitors in eukaryotes is currently limited. Here we used X-ray crystallography to determine 16 high-resolution structures of 80S ribosomes from Saccharomyces cerevisiae in complexes with 12 eukaryote-specific and 4 broad-spectrum inhibitors. All inhibitors were found associated with messenger RNA and transfer RNA binding sites. In combination with kinetic experiments, the structures suggest a model for the action of cycloheximide and lactimidomycin, which explains why lactimidomycin, the larger compound, specifically targets the first elongation cycle. The study defines common principles of targeting and resistance, provides insights into translation inhibitor mode of action and reveals the structural determinants responsible for species selectivity which could guide future drug development.


Subject(s)
Eukaryotic Cells/chemistry , Protein Synthesis Inhibitors/chemistry , Protein Synthesis Inhibitors/pharmacology , Ribosomes/chemistry , Ribosomes/drug effects , Saccharomyces cerevisiae/chemistry , Base Sequence , Binding Sites/drug effects , Crystallography, X-Ray , Cycloheximide/pharmacology , Drug Resistance/drug effects , Eukaryotic Cells/drug effects , Eukaryotic Cells/enzymology , Kinetics , Macrolides/pharmacology , Models, Molecular , Molecular Targeted Therapy , Molecular Weight , Peptide Chain Elongation, Translational/drug effects , Peptidyl Transferases/chemistry , Peptidyl Transferases/metabolism , Piperidones/pharmacology , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Transfer/genetics , RNA, Transfer/metabolism , Ribosome Subunits, Large, Eukaryotic/chemistry , Ribosome Subunits, Large, Eukaryotic/drug effects , Ribosome Subunits, Large, Eukaryotic/metabolism , Ribosomes/metabolism , Species Specificity , Substrate Specificity
11.
Biometals ; 32(3): 469-489, 2019 06.
Article in English | MEDLINE | ID: mdl-30701379

ABSTRACT

Cadmium (Cd2+) is a toxic and non-essential divalent metal ion in eukaryotic cells. Cells can only be targeted by Cd2+ if it hijacks physiological high-affinity entry pathways, which transport essential divalent metal ions in a process termed "ionic and molecular mimicry". Hence, "free" Cd2+ ions and Cd2+ complexed with small organic molecules are transported across cellular membranes via ion channels, carriers and ATP hydrolyzing pumps, whereas receptor-mediated endocytosis (RME) internalizes Cd2+-protein complexes. Only Cd2+ transport pathways validated by stringent methodology, namely electrophysiology, 109Cd2+ tracer studies, inductively coupled plasma mass spectrometry, atomic absorption spectroscopy, Cd2+-sensitive fluorescent dyes, or specific ligand binding and internalization assays for RME are reviewed whereas indirect correlative studies are excluded. At toxicologically relevant concentrations in the submicromolar range, Cd2+ permeates voltage-dependent Ca2+ channels ("T-type" CaV3.1, CatSper), transient receptor potential (TRP) channels (TRPA1, TRPV5/6, TRPML1), solute carriers (SLCs) (DMT1/SLC11A2, ZIP8/SLC39A8, ZIP14/SLC39A14), amino acid/cystine transporters (SLC7A9/SLC3A1, SLC7A9/SLC7A13), and Cd2+-protein complexes are endocytosed by the lipocalin-2/NGAL receptor SLC22A17. Cd2+ transport via the mitochondrial Ca2+ uniporter, ATPases ABCC1/2/5 and transferrin receptor 1 is likely but requires further evidence. Cd2+ flux occurs through the influx carrier OCT2/SLC22A2, efflux MATE proteins SLC47A1/A2, the efflux ATPase ABCB1, and RME of Cd2+-metallothionein by the receptor megalin (low density lipoprotein receptor-related protein 2, LRP2):cubilin albeit at high concentrations thus questioning their relevance in Cd2+ loading. Which Cd2+-protein complexes are internalized by megalin:cubilin in vivo still needs to be determined. A stringent conservative and reductionist approach is mandatory to verify relevance of transport pathways for Cd2+ toxicity and to overcome dissemination of unsubstantiated conjectures.


Subject(s)
Amino Acid Transport Systems/metabolism , Cadmium/metabolism , Coordination Complexes/metabolism , Eukaryotic Cells/metabolism , Ion Channels/metabolism , Receptors, Cell Surface/metabolism , Cadmium/pharmacology , Cell Membrane/drug effects , Cell Membrane/metabolism , Coordination Complexes/pharmacology , Eukaryotic Cells/drug effects , Humans
12.
Angew Chem Int Ed Engl ; 58(16): 5387-5391, 2019 04 08.
Article in English | MEDLINE | ID: mdl-30802354

ABSTRACT

Cycloheximide (CHX) is an inhibitor of eukaryotic translation elongation that has played an essential role in the study of protein synthesis. Despite its ubiquity, few studies have been directed towards accessing synthetic CHX derivatives, even though such efforts may lead to protein synthesis inhibitors with improved or alternate properties. Described here is the total synthesis of CHX and analogues, and the establishment of structure-activity relationships (SAR) responsible for translation inhibition. The SAR studies aided the design of more potent compounds, one of which irreversibly blocks ribosomal elongation, preserves polysome profiles, and may be a broadly useful tool for investigating protein synthesis.


Subject(s)
Biological Products/pharmacology , Cycloheximide/pharmacology , Eukaryotic Cells/drug effects , Ribosomes/drug effects , Biological Products/chemical synthesis , Biological Products/chemistry , Cycloheximide/chemical synthesis , Cycloheximide/chemistry , Dose-Response Relationship, Drug , Eukaryotic Cells/metabolism , Molecular Conformation , Protein Biosynthesis/drug effects , Ribosomes/metabolism , Structure-Activity Relationship
13.
J Biol Chem ; 292(36): 15159-15160, 2017 09 08.
Article in English | MEDLINE | ID: mdl-28887436

ABSTRACT

Bacterial toxins introduce protein modifications such as ADP-ribosylation to manipulate host cell signaling and physiology. Several general mechanisms for toxin function have been established, but the extent to which previously uncharacterized toxins utilize these mechanisms is unknown. A study of an Escherichia coli pertussis-like toxin demonstrates that this protein acts on a known toxin substrate but displays distinct and dual chemoselectivity, suggesting this E. coli pertussis-like toxin may serve as a unique tool to study G-protein signaling in eukaryotic cells.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/pharmacology , Escherichia coli/chemistry , Heterotrimeric GTP-Binding Proteins/antagonists & inhibitors , Pertussis Toxin/chemistry , Animals , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Heterotrimeric GTP-Binding Proteins/metabolism , Humans , Models, Molecular , Signal Transduction/drug effects
14.
Curr Top Microbiol Immunol ; 406: 229-256, 2017.
Article in English | MEDLINE | ID: mdl-28674946

ABSTRACT

A-B types of toxins are among the most potent bacterial protein toxins produced by gram-positive bacteria. Prominent examples are the tripartite anthrax toxin of Bacillus anthracis and the different A-B type clostridial toxins that are the causative agents of severe human and animal diseases and could serve as biological weapons. The components of all these toxins comprise one binding/transport (B) subunit and one or two separate, non-linked enzymatically active (A) subunits. The A and B subunits are separately produced and secreted by the pathogenic gram-positive bacteria and must assemble on the surface of eukaryotic target cells to form biologically active toxin complexes. The B components are cleaved by proteases to generate the biologically active species that binds to receptors on the surface of the target cells and form there oligomers which bind the A subunits. The AB complexes are internalized by receptor-mediated endocytosis and reach early or late endosomes that become acidified. Subsequently, the B components form channels in endosomal membranes that are indispensable for the transport of the enzymatic subunits across these membranes into the cytosol of target cells via the trans-membrane channels. In addition to the channels formed by the B components, host cell factors including chaperones and further folding helper enzymes are involved in the import of the enzymatic subunits into the cytosol of eukaryotic cells. Positively charged heterocyclic molecules, such as chloroquine and related aminoquinolinium and azolopyridinium salts have been shown in recent years to bind with high affinity to the channels formed by the B components of binary toxins. Since binding to the B components is also a prerequisite for transport of the A components across the endosomal membranes the channel blockers also prevent transport of the A subunits into the host cell cytosol. The inhibition of toxin uptake into cells by such pharmacological compounds should also be of clinically interest because the toxins are the major virulence factors causing anthrax on the one hand and severe enteric disease on the other hand. Therefore, the novel toxin inhibitors should be attractive compounds for an application in combination with antibiotics to prevent or treat the diseases associated with binary toxins. Here the different processes involved in channel block in vitro and inhibition of intoxication of living target cells are reviewed in some detail.


Subject(s)
Bacterial Toxins/metabolism , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Animals , Biological Transport/drug effects , Humans
15.
Biochim Biophys Acta Gen Subj ; 1862(7): 1584-1591, 2018 Jul.
Article in English | MEDLINE | ID: mdl-29631058

ABSTRACT

BACKGROUND: Metal-responsive transcription factor 1 (MTF-1) induces the expression of metallothioneins (MTs) which bind and sequester labile metal ions. While MTF-1 primarily responds to excess metal exposure, additional stress response mechanisms are activated by excess metals. Evidence suggests potential crosstalk between responses mediated by MTF-1 and stress signaling enhances cellular tolerance to metal exposure. SCOPE OF REVIEW: This review aims to summarize the current understanding of interaction between the stress response mediated by MTF-1 and other cellular mechanisms, notably the nuclear factor κB (NF-κB) and heat shock response (HSR). MAJOR CONCLUSIONS: Crosstalk between MTF-1 mediated metal response and NF-κB signaling or HSR can modulate expression of stress proteins in response to metal exposure via effects on precursor signals or direct interaction of transcriptional activators. The interaction between stress signaling pathways can enhance cell survival and tolerance through a unified response system. GENERAL SIGNIFICANCE: Elucidating the interactions between MTF-1 and cell stress response mechanisms is critical to a comprehensive understanding of metal-based cellular effects. Co-activation of HSR and NF-κB signaling allows the cell to detect metal contamination in the environment and improve survival outcomes.


Subject(s)
DNA-Binding Proteins/physiology , Eukaryotic Cells/drug effects , Metals, Heavy/pharmacology , Transcription Factors/physiology , Animals , Cytokines/physiology , Cytosol/metabolism , Drug Synergism , Gene Expression Regulation/drug effects , Heat-Shock Proteins/physiology , Heat-Shock Response/physiology , Humans , NF-kappa B/physiology , Promoter Regions, Genetic , Signal Transduction/physiology , Transcription Factor MTF-1
16.
Biochem J ; 474(7): 1109-1125, 2017 03 15.
Article in English | MEDLINE | ID: mdl-28298556

ABSTRACT

Macrocyclic compounds such as cyclic peptides have emerged as a new and exciting class of drug candidates for inhibition of intracellular protein-protein interactions, which are challenging targets for conventional drug modalities (i.e. small molecules and proteins). Over the past decade, several complementary technologies have been developed to synthesize macrocycle libraries and screen them for binding to therapeutically relevant targets. Two different approaches have also been explored to increase the membrane permeability of cyclic peptides. In this review, we discuss these methods and their applications in the discovery of macrocyclic compounds against protein-protein interactions.


Subject(s)
Peptide Library , Peptides, Cyclic/pharmacology , Protein Interaction Domains and Motifs/drug effects , Proteins/antagonists & inhibitors , Small Molecule Libraries/pharmacology , Animals , Biological Products/chemical synthesis , Biological Products/isolation & purification , Biological Products/pharmacology , Biological Transport , Cell Membrane Permeability/drug effects , Diffusion , Drug Discovery , Eukaryotic Cells/cytology , Eukaryotic Cells/drug effects , Eukaryotic Cells/metabolism , Humans , Inteins/drug effects , Peptides, Cyclic/chemical synthesis , Protein Binding/drug effects , Proteins/chemistry , Small Molecule Libraries/chemical synthesis
17.
Biochim Biophys Acta Biomembr ; 1859(5): 982-992, 2017 May.
Article in English | MEDLINE | ID: mdl-28173991

ABSTRACT

Pore-forming toxins (PFTs) form holes in membranes causing one of the most catastrophic damages to a target cell. Target organisms have evolved a regulated response against PFTs damage including cell membrane repair. This ability of cells strongly depends on the toxin concentration and the properties of the pores. It has been hypothesized that there is an inverse correlation between the size of the pores and the time required to repair the membrane, which has been for long a non-intuitive concept and far to be completely understood. Moreover, there is a lack of information about how cells react to the injury triggered by eukaryotic PFTs. Here, we investigated some molecular events related with eukaryotic cells response against the membrane damage caused by sticholysin II (StII), a eukaryotic PFT produced by a sea anemone. We evaluated the change in the cytoplasmic potassium, identified the main MAPK pathways activated after pore-formation by StII, and compared its effect with those from two well-studied bacterial PFTs: aerolysin and listeriolysin O (LLO). Strikingly, we found that membrane recovery upon StII damage takes place in a time scale similar to LLO in spite of the fact that they form pores by far different in size. Furthermore, our data support a common role of the potassium ion, as well as MAPKs in the mechanism that cells use to cope with these toxins injury.


Subject(s)
Cnidarian Venoms/toxicity , Eukaryotic Cells/drug effects , Pore Forming Cytotoxic Proteins/toxicity , Potassium/metabolism , Sea Anemones/pathogenicity , Animals , Cells, Cultured , Cricetinae , Eukaryotic Cells/metabolism , Extracellular Signal-Regulated MAP Kinases/physiology , MAP Kinase Signaling System/drug effects , p38 Mitogen-Activated Protein Kinases/physiology
18.
J Theor Biol ; 425: 103-112, 2017 07 21.
Article in English | MEDLINE | ID: mdl-28501636

ABSTRACT

Chemotaxis, the motion of cells directed by a gradient of chemoattractant molecules, guides cells in immune response, development, wound healing, and cancer. Unfortunately, this process is difficult to distinguish from chemokinesis, i.e., stimulated random cell motion. Chemotaxis is frequently inferred by determining how many cells cross a boundary in a chemotaxis assay, for example how many cells crawl into a chemoattractant-infused filter, or how many cells enter a defined region in an under-agarose assay or agarose spot assay. To mitigate possible ambiguity in whether motion observed in these assays is directed by the chemoattractant gradient or by chemokinesis, we developed a mathematical model to determine when such methods indeed indicate directed motion of cells. In contrast to previous analyses of chemotaxis assays, we report not just the gradients that arise in the assays but also resulting cell motion. We applied the model to data obtained from rigorous measurements and show, as examples, that MDA-MB-231 breast-cancer cells are at least 20 times less sensitive to gradients of EGF or CXCL12 than neutrophils are to formyl peptides; we then used this information to determine the extent to which gradient sensing increases the rate of boundary crossing relative to a random-motility control. Results show, for example, that in the filter assay, 2-4 times as many neutrophils pass through the filter when exposed to a gradient as when the gradient is absent. However, in the other combinations of cells and assays we considered, only 10-20% more cells are counted as having migrated in a directed, rather than random, motility condition. We also discuss the design of appropriate controls for these assays, which is difficult for the under-agarose and agarose spot assays. Moreover, although straightforward to perform with the filter assay, reliable controls are often not done. Consequently, we infer that chemotaxis is frequently over-reported, especially for cells like MDA-MB-231 cells, which move slowly and are relatively insensitive to gradients. Such results provide insights into the use of chemotaxis assays, particularly if one wants to acquire and analyze quantitative data.


Subject(s)
Chemotaxis/physiology , Eukaryotic Cells/physiology , Models, Biological , Breast Neoplasms/pathology , Cell Movement/drug effects , Cell Movement/physiology , Chemokine CXCL12/pharmacology , Chemotactic Factors/pharmacology , Chemotaxis/drug effects , Chemotaxis, Leukocyte/drug effects , Epidermal Growth Factor/pharmacology , Eukaryotic Cells/drug effects , Female , Humans , Neutrophils/drug effects , Neutrophils/physiology , Sepharose
19.
Glycobiology ; 26(8): 789-96, 2016 08.
Article in English | MEDLINE | ID: mdl-27066802

ABSTRACT

Cell surface glycans are critical mediators of cell-cell, cell-ligand, and cell-pathogen interactions. By controlling the set of glycans displayed on the surface of a cell, it is possible to gain insight into the biological functions of glycans. Moreover, control of glycan expression can be used to direct cellular behavior. While genetic approaches to manipulate glycosyltransferase gene expression are available, their utility in glycan engineering has limitations due to the combinatorial nature of glycan biosynthesis and the functional redundancy of glycosyltransferase genes. Biochemical and chemical strategies offer valuable complements to these genetic approaches, notably by enabling introduction of unnatural functionalities, such as fluorophores, into cell surface glycans. Here, we describe some of the most recent developments in glycoengineering of cell surfaces, with an emphasis on strategies that employ novel chemical reagents. We highlight key examples of how these advances in cell surface glycan engineering enable study of cell surface glycans and their function. Exciting new technologies include synthetic lipid-glycans, new chemical reporters for metabolic oligosaccharide engineering to allow tandem and in vivo labeling of glycans, improved chemical and enzymatic methods for glycoproteomics, and metabolic glycosyltransferase inhibitors. Many chemical and biochemical reagents for glycan engineering are commercially available, facilitating their adoption by the biological community.


Subject(s)
Cell Engineering/methods , Cell Membrane/chemistry , Glycosyltransferases/chemistry , Monosaccharides/chemistry , Polysaccharides/chemistry , Animals , Carbohydrate Sequence , Cell Membrane/drug effects , Cell Membrane/enzymology , Enzyme Inhibitors/pharmacology , Eukaryotic Cells/chemistry , Eukaryotic Cells/cytology , Eukaryotic Cells/drug effects , Eukaryotic Cells/enzymology , Fluorescent Dyes/chemistry , Glycomics/methods , Glycosylation , Glycosyltransferases/antagonists & inhibitors , Humans , Proteomics/methods , Staining and Labeling/methods
20.
J Antimicrob Chemother ; 71(6): 1547-55, 2016 06.
Article in English | MEDLINE | ID: mdl-26945708

ABSTRACT

OBJECTIVES: The objectives of this study were to: (i) determine the in vitro activities of a series of di-, tri- and tetra-nuclear ruthenium complexes (Rubbn, Rubbn-tri and Rubbn-tetra) against a range of Gram-positive and -negative bacteria and compare the antimicrobial activities with the corresponding toxicities against eukaryotic cells; and (ii) compare MIC values with achievable in vivo serum concentrations for the least toxic ruthenium complex. METHODS: The in vitro activities were determined by MIC assays and time-kill curve experiments, while the toxicities of the ruthenium complexes were determined using the Alamar blue cytotoxicity assay. A preliminary pharmacokinetic study was undertaken to determine the Rubb12 serum concentration in mice as a function of time after administration. RESULTS: Rubb12, Rubb12-tri and Rubb12-tetra are highly active, with MIC values of 1-2 mg/L (0.5-1.5 µM) for a range of Gram-positive strains, but showed variable activities against a panel of Gram-negative bacteria. Time-kill experiments indicated that Rubb12, Rubb12-tri and Rubb12-tetra are bactericidal and kill bacteria within 3-8 h. The di-, tri- and tetra-nuclear complexes were ∼50 times more toxic to Gram-positive bacteria and 25 times more toxic to Gram-negative strains, classified as susceptible, than to liver and kidney cells. Preliminary pharmacokinetic experiments established that serum concentrations higher than MIC values can be obtained for Rubb12 with an administered dose of 32 mg/kg. CONCLUSIONS: The ruthenium complexes, particularly Rubb12, have potential as new antimicrobial agents. The structure of the dinuclear ruthenium complex can be readily further modified in order to increase the selectivity for bacteria over eukaryotic cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacteria/drug effects , Eukaryotic Cells/drug effects , Organometallic Compounds/pharmacology , Ruthenium/pharmacology , Animals , Anti-Bacterial Agents/pharmacokinetics , Anti-Bacterial Agents/toxicity , Cell Survival/drug effects , Colorimetry/methods , Female , Male , Mice, Inbred BALB C , Microbial Sensitivity Tests , Microbial Viability/drug effects , Organometallic Compounds/pharmacokinetics , Organometallic Compounds/toxicity , Oxazines/analysis , Ruthenium/pharmacokinetics , Ruthenium/toxicity , Serum/chemistry , Xanthenes/analysis
SELECTION OF CITATIONS
SEARCH DETAIL