Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
FEBS Open Bio ; 9(3): 420-427, 2019 03.
Article in English | MEDLINE | ID: mdl-30868050

ABSTRACT

The base excision repair pathway plays an important role in correcting damage induced by either physiological or external effects. This repair pathway removes incorrect bases from the DNA. The uracil base is among the most frequently occurring erroneous bases in DNA, and is cut out from the phosphodiester backbone via the catalytic action of uracil-DNA glycosylase. Uracil excision repair is an evolutionarily highly conserved pathway and can be specifically inhibited by a protein inhibitor of uracil-DNA glycosylase. Interestingly, both uracil-DNA glycosylase (Staphylococcus aureus uracil-DNA glycosylase; SAUDG) and its inhibitor (S. aureus uracil-DNA glycosylase inhibitor; SAUGI) are present in the staphylococcal cell. The interaction of these two proteins effectively decreases the efficiency of uracil-DNA excision repair. The physiological relevance of this complexation has not yet been addressed in detailed; however, numerous mutations have been identified within SAUGI. Here, we investigated whether these mutations drastically perturb the interaction with SAUDG. To perform quantitative analysis of the macromolecular interactions, we applied native mass spectrometry and demonstrated that this is a highly efficient and specific method for determination of dissociation constants. Our results indicate that several naturally occurring mutations of SAUGI do indeed lead to appreciable changes in the dissociation constants for complex formation. However, all of these Kd values remain in the nanomolar range and therefore the association of these two proteins is preserved. We conclude that complexation is most likely preserved even with the naturally occurring mutant uracil-DNA glycosylase inhibitor proteins.


Subject(s)
Macromolecular Substances/analysis , Mutation , Staphylococcus aureus/enzymology , Uracil-DNA Glycosidase/chemistry , Enzyme Inhibitors/pharmacology , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/metabolism , Mass Spectrometry , Models, Molecular , Uracil-DNA Glycosidase/antagonists & inhibitors , Uracil-DNA Glycosidase/metabolism
2.
Methods Mol Biol ; 1517: 239-249, 2017.
Article in English | MEDLINE | ID: mdl-27924487

ABSTRACT

microRNAs (miRNAs) have been identified as high-value drug targets. A widely applied strategy in miRNA inhibition is the use of antisense agents. However, it has been shown that oligonucleotides are poorly cell permeable because of their complex chemical structure and due to their negatively charged backbone. Consequently, the general application of oligonucleotides in therapy is limited. Since miRNAs' functions are executed exclusively by the Argonaute 2 protein, we therefore describe a protocol for the design of a novel miRNA inhibitor class: antagonists of the miRNA-Argonaute 2 protein complex, so-called anti-miR-AGOs, that not only block the crucial binding site of the target miRNA but also bind to the protein's active site. Due to their lower molecular weight and, thus, more drug-like chemical structure, the novel inhibitor class may show better pharmacokinetic properties than reported oligonucleotide inhibitors, enabling them for potential therapeutic use.


Subject(s)
Argonaute Proteins/antagonists & inhibitors , MicroRNAs/antagonists & inhibitors , Molecular Biology/methods , Oligonucleotides, Antisense/chemistry , Antagomirs/chemistry , Antagomirs/therapeutic use , Argonaute Proteins/chemistry , Argonaute Proteins/genetics , Binding Sites , Humans , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/chemistry , MicroRNAs/chemistry , MicroRNAs/genetics , Oligonucleotides, Antisense/therapeutic use , Protein Binding , RNA, Messenger/antagonists & inhibitors
3.
Article in English | MEDLINE | ID: mdl-27048277

ABSTRACT

BACKGROUND: Hyperstructures are large assemblies of molecules and macromolecules that perform functions such as metabolism (including RNA and protein synthesis and degradation), transport, DNA replication, cell division, signalling and chemotaxis. METHODS: Such hyperstructures might be manipulated by hybrid metabolites or hybolites made by a pairwise, covalently linked combination of the thousands of small molecules involved in metabolism and signalling. RESULTS: Here, we review recent evidence for hyperstructures in prokaryotes and for interactions between hyperstructures as a determinant of the phenotype. We also mention extending hybolite therapy to eukaryotes, consider new designs for hybolites, and discuss relevant patents.


Subject(s)
Anti-Bacterial Agents/chemistry , Bacteria/drug effects , Drug Discovery/methods , Macromolecular Substances/antagonists & inhibitors , Animals , Anti-Bacterial Agents/therapeutic use , Bacteria/genetics , Bacteria/metabolism , Bacteria/pathogenicity , Drug Resistance, Bacterial , Energy Metabolism/drug effects , Gene Expression Regulation, Bacterial/drug effects , Humans , Macromolecular Substances/metabolism , Molecular Structure , Molecular Targeted Therapy , Structure-Activity Relationship , Virulence/drug effects
4.
Curr Top Med Chem ; 15(18): 1801-13, 2015.
Article in English | MEDLINE | ID: mdl-25961517

ABSTRACT

Drug discovery is aimed at finding therapeutic agents for the treatment of many diverse diseases and infections. However, this is a very slow an expensive process, and for this reason, in silico approaches are needed to rationalize the search for new molecular entities with desired biological profiles. Models focused on quantitative structure-activity relationships (QSAR) have constituted useful complementary tools in medicinal chemistry, allowing the virtual predictions of dissimilar pharmacological activities of compounds. In the last 10 years, multi-target (mt) QSAR models have been reported, representing great advances with respect to those models generated from classical approaches. Thus, mt- QSAR models can simultaneously predict activities against different biological targets (proteins, microorganisms, cell lines, etc.) by using large and heterogeneous datasets of chemicals. The present review is devoted to discuss the most promising mt-QSAR models, particularly those developed for the prediction of protein inhibitors. We also report the first multi-tasking QSAR (mtk-QSAR) model for simultaneous prediction of inhibitors against biomacromolecules (specifically proteins) present in Gram-negative bacteria. This model allowed us to consider both different proteins and multiple experimental conditions under which the inhibitory activities of the chemicals were determined. The mtk-QSAR model exhibited accuracies higher than 98% in both training and prediction sets, also displaying a very good performance in the classification of active and inactive cases that depended on the specific elements of the experimental conditions. The physicochemical interpretations of the molecular descriptors were also analyzed, providing important insights regarding the molecular patterns associated with the appearance/enhancement of the inhibitory potency.


Subject(s)
Gram-Negative Bacteria/chemistry , Proteins/antagonists & inhibitors , Quantitative Structure-Activity Relationship , Drug Discovery , Macromolecular Substances/antagonists & inhibitors , Models, Molecular
5.
Appl Microbiol Biotechnol ; 98(14): 6205-13, 2014.
Article in English | MEDLINE | ID: mdl-24841116

ABSTRACT

Plasma is ionized gas, which is found in various forms in nature and can also be generated artificially. A variety of cold atmospheric-pressure plasmas are currently being investigated for their clinical utility, and first studies reporting on the treatment of patients showed that plasma treatment may support the wound healing process. One of the benefits of plasma treatment is the effective inactivation of bacteria including tenacious pathogens such as Pseudomonas aeruginosa or multiresistant Staphylococcus aureus (MRSA). Neither the molecular mechanisms promoting wound healing nor those underlying bacterial inactivation are fully understood yet. The review has a focus on plasma jets, a particular type of cold atmospheric-pressure plasma sources featuring an indirect treatment whereby the treated substrates do not come into contact with the plasma directly but are exposed to the plasma-emitted reactive species and photons. Such plasma jets are being employed as tools in basic research regarding the effects of plasmas on biological samples. This review provides a brief overview on the recent clinical investigations into the benefits of cold atmospheric-pressure plasmas. It then describes our current understanding of the mechanisms leading to bacterial inactivation and inactivation of biomacromolecules gained by employing plasma jets.


Subject(s)
Bacteria/drug effects , Disinfectants/pharmacology , Macromolecular Substances/antagonists & inhibitors , Plasma Gases/pharmacology , Atmospheric Pressure , Microbial Viability/drug effects
6.
Nat Rev Drug Discov ; 11(1): 25-36, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22173432

ABSTRACT

Interfacial inhibitors belong to a broad class of natural products and synthetic drugs that are commonly used to treat cancers as well as bacterial and HIV infections. They bind selectively to interfaces as macromolecular machines assemble and are set in motion. The bound drugs transiently arrest the targeted molecular machines, which can initiate allosteric effects, or desynchronize macromolecular machines that normally function in concert. Here, we review five archetypical examples of interfacial inhibitors: the camptothecins, etoposide, the quinolone antibiotics, the vinca alkaloids and the novel anti-HIV inhibitor raltegravir. We discuss the common and diverging elements between interfacial and allosteric inhibitors and give a perspective for the rationale and methods used to discover novel interfacial inhibitors.


Subject(s)
Drug Delivery Systems/methods , Macromolecular Substances/metabolism , Pharmaceutical Preparations/metabolism , Animals , Drug Delivery Systems/trends , Humans , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/chemistry , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/chemistry
7.
J Steroid Biochem Mol Biol ; 126(3-5): 104-12, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21569845

ABSTRACT

The ability of inhibitors of transcription and translation to prevent glucocorticoid-induced apoptosis has been interpreted to indicate that the cell death machinery requires de novo protein synthesis. The transcriptional inhibitors actinomycin D (Act D) and DRB as well as the translational inhibitors CHX and puromycin inhibited early loss of mitochondrial membrane integrity in a dose-dependent manner. This effect was not observed with the transcriptional inhibitor α-amanitin suggesting they may have additional effects. Their role in the glucocorticoid receptor (GR) intracellular trafficking was therefore investigated. Here, we show that Act D and CHX reduced glucocorticoid binding, GR turnover and impaired GR nuclear translocation. We performed the same experiments in different thymocyte subpopulations of Balb/c mice. At the highest dose tested, actinomycin D and cycloheximide abolished glucocorticoid-induced cell death of CD4+CD8+ and CD4+CD8-. In all subsets, Act D, DRB, as well as CHX and puromycin prevented receptor nuclear translocation, indicating a general alteration of GR trafficking. Overall, our data support a direct effect of macromolecular inhibitors on GR activation and trafficking. Finally, direct alterations of the functional properties of the glucocorticoid receptor might be responsible for cell death prevention by actinomycin D, DRB, cycloheximide and puromycin.


Subject(s)
Nucleic Acid Synthesis Inhibitors/pharmacology , Protein Synthesis Inhibitors/pharmacology , Receptors, Glucocorticoid/metabolism , Alpha-Amanitin/pharmacology , Animals , Cells, Cultured , Cycloheximide/pharmacology , Dactinomycin/pharmacology , Glucocorticoids/metabolism , HeLa Cells , Humans , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/metabolism , Mice , Mice, Inbred BALB C , Protein Binding , Protein Transport/drug effects , Puromycin/pharmacology , Thymus Gland/cytology , Thymus Gland/drug effects , Thymus Gland/metabolism
8.
Biochem J ; 435(2): 451-62, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21299497

ABSTRACT

CFTR (cystic fibrosis transmembrane conductance regulator) has been shown to form multiple protein macromolecular complexes with its interacting partners at discrete subcellular microdomains to modulate trafficking, transport and signalling in cells. Targeting protein-protein interactions within these macromolecular complexes would affect the expression or function of the CFTR channel. We specifically targeted the PDZ domain-based LPA2 (type 2 lysophosphatidic acid receptor)-NHERF2 (Na+/H+ exchanger regulatory factor-2) interaction within the CFTR-NHERF2-LPA2-containing macromolecular complexes in airway epithelia and tested its regulatory role on CFTR channel function. We identified a cell-permeable small-molecule compound that preferentially inhibits the LPA2-NHERF2 interaction. We show that this compound can disrupt the LPA2-NHERF2 interaction in cells and thus compromises the integrity of macromolecular complexes. Functionally, it elevates cAMP levels in proximity to CFTR and upregulates its channel activity. The results of the present study demonstrate that CFTR Cl- channel function can be finely tuned by modulating PDZ domain-based protein-protein interactions within the CFTR-containing macromolecular complexes. The present study might help to identify novel therapeutic targets to treat diseases associated with dysfunctional CFTR Cl- channels.


Subject(s)
Cystic Fibrosis Transmembrane Conductance Regulator/antagonists & inhibitors , Drug Delivery Systems/methods , Macromolecular Substances/antagonists & inhibitors , Animals , Cells, Cultured , Cystic Fibrosis Transmembrane Conductance Regulator/metabolism , Cystic Fibrosis Transmembrane Conductance Regulator/physiology , Drug Discovery/methods , Drug Evaluation, Preclinical , High-Throughput Screening Assays/methods , Humans , Indoles/pharmacology , Macromolecular Substances/metabolism , Models, Biological , Phenylpropionates/pharmacology , Phosphoproteins/antagonists & inhibitors , Phosphoproteins/metabolism , Phosphoproteins/physiology , Protein Binding/drug effects , Receptors, Lysophosphatidic Acid/antagonists & inhibitors , Receptors, Lysophosphatidic Acid/metabolism , Receptors, Lysophosphatidic Acid/physiology , Small Molecule Libraries/pharmacology , Sodium-Hydrogen Exchangers/antagonists & inhibitors , Sodium-Hydrogen Exchangers/metabolism , Sodium-Hydrogen Exchangers/physiology , Swine
9.
Biochemistry ; 50(4): 451-7, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21110513

ABSTRACT

Fluorescence resonance energy transfer (FRET) is a powerful tool for studying macromolecular assemblies in vitro under near-physiological conditions. Here we present a new type of one-sample FRET (OS-FRET) method employing a novel, nonfluorescent methanethiosulfonate-linked acceptor that can be reversibly coupled to a target sulfhydryl residue via a disulfide bond. After the quenched donor emission is quantitated, the acceptor is removed by reduction, allowing measurement of unquenched donor emission in the same sample. Previous one-sample methods provide distinct advantages in specific FRET applications. The new OS-FRET method is a generalizable spectrochemical approach that can be applied to macromolecular systems lacking essential disulfide bonds and eliminates the potential systematic errors of some earlier one-sample methods. In addition, OS-FRET enables quantitative FRET measurements in virtually any fluorescence spectrometer or detection device. Compared to conventional multisample FRET methods, OS-FRET conserves sample, increases the precision of data, and shortens the time per measurement. The utility of the method is illustrated by its application to a protein complex of known structure formed by CheW and the P4-P5 fragment of CheA, both from Thermotoga maritima. The findings confirm the practicality and advantages of OS-FRET. Anticipated applications of OS-FRET include analysis of macromolecular structure, binding and conformational dynamics, and high-throughput screening for interactions and inhibitors.


Subject(s)
Fluorescence Resonance Energy Transfer/methods , Thermotoga maritima/enzymology , Bacterial Proteins/antagonists & inhibitors , Bacterial Proteins/chemistry , Bacterial Proteins/genetics , Chemotaxis/genetics , Crystallography, X-Ray , Fluorescence Resonance Energy Transfer/standards , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/chemistry , Membrane Proteins/genetics , Methyl-Accepting Chemotaxis Proteins , Mutagenesis, Site-Directed , Oxidation-Reduction , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/genetics , Protein Binding/genetics , Protein Conformation , Protein Interaction Mapping/methods , Reproducibility of Results , Spectrometry, Fluorescence/methods , Spectrometry, Fluorescence/standards , Thermotoga maritima/genetics , Thermotoga maritima/metabolism
10.
J Immunol ; 183(1): 543-51, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19542465

ABSTRACT

There is an abundance of antimicrobial peptides in cystic fibrosis (CF) lungs. Despite this, individuals with CF are susceptible to microbial colonization and infection. In this study, we investigated the antimicrobial response within the CF lung, focusing on the human cathelicidin LL-37. We demonstrate the presence of the LL-37 precursor, human cathelicidin precursor protein designated 18-kDa cationic antimicrobial protein, in the CF lung along with evidence that it is processed to active LL-37 by proteinase-3. We demonstrate that despite supranormal levels of LL-37, the lung fluid from CF patients exhibits no demonstrable antimicrobial activity. Furthermore Pseudomonas killing by physiological concentrations of exogenous LL-37 is inhibited by CF bronchoalveolar lavage (BAL) fluid due to proteolytic degradation of LL-37 by neutrophil elastase and cathepsin D. The endogenous LL-37 in CF BAL fluid is protected from this proteolysis by interactions with glycosaminoglycans, but while this protects LL-37 from proteolysis it results in inactivation of LL-37 antimicrobial activity. By digesting glycosaminoglycans in CF BAL fluid, endogenous LL-37 is liberated and the antimicrobial properties of CF BAL fluid restored. High sodium concentrations also liberate LL-37 in CF BAL fluid in vitro. This is also seen in vivo in CF sputum where LL-37 is complexed to glycosaminoglycans but is liberated following nebulized hypertonic saline resulting in increased antimicrobial effect. These data suggest glycosaminoglycan-LL-37 complexes to be potential therapeutic targets. Factors that disrupt glycosaminoglycan-LL-37 aggregates promote the antimicrobial effects of LL-37 with the caveat that concomitant administration of antiproteases may be needed to protect the now liberated LL-37 from proteolytic cleavage.


Subject(s)
Antimicrobial Cationic Peptides/antagonists & inhibitors , Antimicrobial Cationic Peptides/metabolism , Cystic Fibrosis/immunology , Cystic Fibrosis/metabolism , Glycosaminoglycans/metabolism , Lung/immunology , Lung/metabolism , Saline Solution, Hypertonic/pharmacology , Adjuvants, Immunologic/physiology , Adolescent , Antibody Specificity/physiology , Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/physiology , Cathepsin D/physiology , Child , Cystic Fibrosis/enzymology , Cystic Fibrosis/microbiology , Glycosaminoglycans/physiology , Humans , Hydrolysis , Leukocyte Elastase/physiology , Lung/enzymology , Lung/microbiology , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/immunology , Macromolecular Substances/metabolism , Molecular Weight , Myeloblastin/metabolism , Nebulizers and Vaporizers , Protein Precursors/metabolism , Protein Processing, Post-Translational , Saline Solution, Hypertonic/administration & dosage , Solubility , Sputum/enzymology , Sputum/immunology , Sputum/microbiology , Cathelicidins
11.
Biochem Biophys Res Commun ; 378(2): 313-8, 2009 Jan 09.
Article in English | MEDLINE | ID: mdl-19032949

ABSTRACT

The toxicity of acrolein was compared with that of reactive oxygen species using a mouse mammary carcinoma FM3A cell culture system. Complete inhibition of cell growth was accomplished with 10 microM acrolein, 100 microM H(2)O(2), and 20 microM H(2)O(2) plus 1mM vitamin C, which produce ()OH, suggesting that toxicity of acrolein is more severe than H(2)O(2) and nearly equal to that of ()OH, when these compounds were added extracellularly. Acrolein toxicity was prevented by N-acetyl-l-cysteine and N-benzylhydroxylamine, and attenuated by putrescine and spermidine. Toxicity of H(2)O(2) was prevented by glutathione peroxidase plus N-acetyl-l-cysteine, pyruvate, catalase, and reduced by polyphenol, and toxicity of ()OH was prevented by glutathione peroxidase plus N-acetyl-l-cysteine, pyruvate, catalase and reduced by N-acetyl-l-cysteine. The results indicate that prevention of cell toxicity by N-acetyl-l-cysteine was more effective with acrolein than with ()OH. Protein and DNA synthesis was damaged primarily by acrolein and reactive oxygen species, respectively.


Subject(s)
Acrolein/toxicity , Reactive Oxygen Species/toxicity , Acetylcysteine/pharmacology , Acrolein/antagonists & inhibitors , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Hydrogen Peroxide/antagonists & inhibitors , Hydrogen Peroxide/toxicity , Hydroxylamines/pharmacology , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/metabolism , Mice , Oxidative Stress , Protein Biosynthesis/drug effects , Putrescine/pharmacology , Reactive Oxygen Species/antagonists & inhibitors , Singlet Oxygen/metabolism , Spermidine/pharmacology
13.
Physiology (Bethesda) ; 22: 342-50, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17928548

ABSTRACT

Many cardiac ion channels exist within macromolecular signaling complexes, comprised of pore-forming subunits that associate with auxiliary subunits, regulatory enzymes, and targeting proteins. This complex protein assembly ensures proper modulation of channel activity and ion homeostasis. The association of genetic defects in regulatory and targeting proteins to inherited arrhythmia syndromes has led to a better understanding of the critical role these proteins play in ion channel modulation.


Subject(s)
Arrhythmias, Cardiac , Macromolecular Substances/metabolism , Animals , Ankyrins/genetics , Ankyrins/metabolism , Arrhythmias, Cardiac/etiology , Arrhythmias, Cardiac/genetics , Arrhythmias, Cardiac/metabolism , Caveolins/genetics , Caveolins/metabolism , Humans , Ion Channel Gating/genetics , Macromolecular Substances/antagonists & inhibitors , Mutation/physiology , Myocytes, Cardiac/metabolism , Myocytes, Cardiac/ultrastructure , Protein Subunits/genetics , Protein Subunits/metabolism , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Signal Transduction/genetics
14.
Biochemistry ; 44(18): 6776-87, 2005 May 10.
Article in English | MEDLINE | ID: mdl-15865423

ABSTRACT

In recent studies, the amyloid fibrils produced in vitro from recombinant prion protein encompassing residues 89-230 (rPrP 89-230) were shown to produce transmissible form of prion disease in transgenic mice (Legname et al., (2004) Science 305, 673-676). Long incubation time observed upon inoculation of the amyloid fibrils, however, suggests that the fibrils generated in vitro have low infectivity titers. These results emphasize the need to define optimal conditions for prion conversion in vitro, under which high levels of infectivity can be generated in a cell-free system. Because copper(II) has been implicated in normal and pathological functions of the prion protein, here we investigated the effect of Cu(2+) on cell-free conversion of recombinant PrP. Our results show that at pH 7.2 and at micromolar concentrations, Cu(2+) inhibited conversion of full-length recombinant PrP (rPrP 23-230) into amyloid fibrils. This effect was most pronounced for Cu(2+), and less so for Zn(2+), while Mn(2+) had no effect on the conversion. Cu(2+)-dependent inhibition of the amyloid formation was less effective at pH 6.0, at which rPrP 23-230 displays lower Cu(2+)-binding capacity. Using rPrP 89-230, we found that Cu(2+)-dependent inhibition occurred even in the absence of octarepeat region; however, it was less effective. Our further studies indicated that Cu(2+) inhibited conversion by stabilizing a nonamyloidogenic PK-resistant form of alpha-rPrP. Remarkably, Cu(2+) also had a profound effect on preformed amyloid fibrils. When added to the fibrils, Cu(2+) induced long-range coiling of individual fibrils and enhanced their PK-resistance. It, however, produced only minor changes in their secondary structures. In addition, Cu(2+) induced further aggregation of the amyloid fibrils into large clumps, presumably, through interfibrillar coordination of copper ions by octarepeats. Taken together, our studies suggest that the role of Cu(2+) in the pathogenesis of prion diseases is complex. Because Cu(2+) may inhibit prion replication, while at the same time stabilize disease-specific isoform against proteolytic clearance, the final outcome of copper-induced effect on progression of prion disease may not be straightforward.


Subject(s)
Amyloid/antagonists & inhibitors , Amyloid/metabolism , Copper/chemistry , Prions/antagonists & inhibitors , Prions/metabolism , Amyloid/chemistry , Animals , Binding Sites , Cations, Divalent , Drug Resistance , Endopeptidase K/chemistry , Hydrogen-Ion Concentration , Hydrolysis , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/chemistry , Macromolecular Substances/metabolism , Manganese/chemistry , Mice , Peptide Fragments/antagonists & inhibitors , Peptide Fragments/chemistry , Peptide Fragments/metabolism , Prions/chemistry , Protein Binding , Protein Structure, Secondary , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Repetitive Sequences, Amino Acid , Zinc/chemistry
15.
Trends Pharmacol Sci ; 26(3): 138-45, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15749159

ABSTRACT

One of nature's strategies for interfering with molecular interactions is to trap macromolecules in transition states with their partners in dead-end complexes that are unable to complete their biological function. This type of inhibition, which we refer to as "interfacial inhibition", is illustrated by two natural inhibitors, brefeldin A (BFA) and camptothecin (CPT), whose modes of action have been elucidated fully in structural studies. Interfacial inhibition occurs at the protein-protein interface in the case of BFA and at the protein-DNA interface in the case of CPT. In both systems, the drugs take advantage of transient structural and energetic conditions created by the macromolecular complex, which give rise to "hot-spots" for drug binding. In addition to these examples, several natural compounds such as forskolin, tubulin inhibitors and immunophilins target protein interfaces. We propose that interfacial inhibition is a paradigm for the discovery of drugs that interfere with macromolecular complexes.


Subject(s)
Drug Design , Macromolecular Substances/antagonists & inhibitors , Macromolecular Substances/metabolism , Pharmaceutical Preparations/metabolism , Animals , Drug Interactions/physiology , Humans , Macromolecular Substances/chemistry , Pharmaceutical Preparations/chemistry , Protein Binding/drug effects , Protein Binding/physiology
16.
Mol Cancer ; 3: 8, 2004 Mar 22.
Article in English | MEDLINE | ID: mdl-15035668

ABSTRACT

BACKGROUND: Nuclear factor kappa B (NFkappaB) is a pro-malignant transcription factor with reciprocal effects on pro-metastatic and anti-metastatic gene expression. Interestingly, NFkappaB blockade results in the reciprocal induction of retinoic acid receptors (RARs). Given the established property of RARs as negative regulators of malignant progression, we postulated that reciprocal interactions between NFkappaB and RARs constitute a signaling module in metastatic gene expression and malignant progression. Using Line 1 tumor cells as a model for signal regulation of metastatic gene expression, we investigated the reciprocal interactions between NFkappaB and RARs in response to the pan-RAR agonist, all-trans retinoic acid (at-RA) and the pan-RAR antagonist, AGN193109. RESULTS: At-RA [0.1-1 microM] dose-dependently activated RAR and coordinately trans-repressed NFkappaB, while AGN193109 [1-10 microM] dose-dependently antagonized the effects of at-RA. At-RA and AGN193109 reciprocally regulate pro-metastatic matrix metalloprotease 9 (MMP 9) and its endogenous inhibitor, the tissue inhibitor of metalloprotease 1 (TIMP 1), in a manner consistent with the putative roles of NFkappaB and RAR in malignant progression. Activation of RAR concurs with its ubiquitination and proteosomal degradation. Accordingly, the proteosome inhibitor, MG132 [5 microM], blocked RAR degradation, quelled RAR trans-activation and enhanced RAR trans-repression of NFkappaB. CONCLUSION: We conclude that reciprocal interactions between NFkappaB and RARs constitute a signaling module in metastatic gene expression and malignant progression and propose that the dissociative effect of proteosome inhibitors could be harnessed towards enhancing the anticancer activity of retinoids.


Subject(s)
Leupeptins/pharmacology , NF-kappa B/genetics , Receptors, Retinoic Acid/antagonists & inhibitors , Repressor Proteins/physiology , Transcriptional Activation/drug effects , Active Transport, Cell Nucleus/physiology , Cell Line, Tumor , Cell Nucleus/metabolism , DNA, Neoplasm/metabolism , DNA-Binding Proteins/antagonists & inhibitors , DNA-Binding Proteins/metabolism , Enzyme Activation/physiology , Gene Expression Regulation/physiology , Humans , Ligands , Macromolecular Substances/antagonists & inhibitors , Matrix Metalloproteinase 9/metabolism , NF-kappa B/metabolism , Neoplasm Metastasis/pathology , Neoplasm Proteins , Protein Transport/physiology , Receptors, Retinoic Acid/metabolism , Receptors, Retinoic Acid/physiology , Retinoids , Tissue Inhibitor of Metalloproteinase-1/metabolism
17.
Proc Natl Acad Sci U S A ; 75(5): 2098-102, 1978 May.
Article in English | MEDLINE | ID: mdl-276855

ABSTRACT

Two active components alpha and beta of micrococcus luteus DNA gyrase, of peptide weights of 115,000 and 97,000, respectively, have been purified. Each individual component exhibits little DNA gyrase activity; the ATP-dependent negative supercoiling of a covalently closed circular DNA duplex is catalyzed by a combination of the two. Covalent closure by Escherichia coli ligase of a circular DNA containing single-chain scissions, when carried out in the presence of a combination of the DNA gyrase components alpha and beta, gives a positively supercoiled DNA upon removal of the bound protein molecules. ATP was not present during the ligase treatment; therefore the positive supercoiling of DNA observed is a result of the binding of gyrase molecules, presumably as multi-subunit oligomers, during the ligation step. This is in contrast to the negative supercoiling of DNA catalyzed by gyrase in the presence of ATP. A model in which negative supercoiling of DNA is achieved by ATP-modulated repetitive wrapping of the DNA around gyrase is described. The model also suggests a plausible mode of action by which translocation of a DNA along its helix axis can be actively driven by an ATPase.


Subject(s)
DNA, Bacterial , DNA, Superhelical , Adenosine Triphosphate/metabolism , Anti-Bacterial Agents/pharmacology , DNA, Bacterial/metabolism , DNA, Superhelical/metabolism , Macromolecular Substances/antagonists & inhibitors , Magnesium/metabolism , Micrococcus/enzymology , Models, Chemical , Molecular Weight , Nucleic Acid Conformation , Potassium/metabolism , Spermidine/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...