108 results on '"Tsodikov OV"'
Search Results
2. Structural investigations of two bifunctional co-dependent enzymes, MtmGIV and MtmC, involved in the biosynthesis of mithramycin
- Author
-
Chen, JM, primary, Hou, C, additional, Tsodikov, OV, additional, and Rohr, J, additional
- Published
- 2014
- Full Text
- View/download PDF
3. Association of COVID-19 risk factors with systemic fungal infections in hospitalized patients.
- Author
-
Wilbourn AC, Tsodikov OV, and Garneau-Tsodikova S
- Abstract
Purpose: A new category of systemic co-infections that emerged with the COVID-19 pandemic is known as COVID-19-associated (CA) fungal infections, which include pulmonary aspergillosis (CAPA), candidiasis (CAC), and mucormycosis (CAM). We aimed to study the association between patient characteristics of hospitalized COVID-19 patients, COVID-19 comorbidities, and COVID-19 therapies with secondary non-superficial fungal infections., Methods: We performed descriptive and regression analyses of data from 4,999 hospitalized COVID-19 patients from the University of Kentucky Healthcare (UKHC) system., Results: The patients with secondary systemic fungal infections had a 6-fold higher risk of death than those without such infections. Generally, the risk factors for severe COVID-19 (age, obesity, cardiovascular disease, diabetes, and lack of COVID-19 vaccination) were strong predictors of a secondary fungal infection. However, several characteristics had much higher risks, suggesting that a causative link may be at play: ICU admission, mechanical ventilation, length of hospital stay, and steroid use., Conclusions: In sum, this study found that the known risk factors for severe COVID-19 disease, age, diabetes, cardiovascular disease, obesity, ventilation, and high steroid doses were all predictors of a secondary fungal infection. Steroid therapy may need to be modified to account for a risk or a presence of a fungal infection in vulnerable patients., Competing Interests: Conflicts of interest/Competing interests The authors have no relevant financial or non-financial interests to disclose.
- Published
- 2024
- Full Text
- View/download PDF
4. Transient interactions modulate the affinity of NF-κB transcription factors for DNA.
- Author
-
Li T, Shahabi S, Biswas T, Tsodikov OV, Pan W, Huang DB, Wang VY, Wang Y, and Ghosh G
- Subjects
- Humans, Transcription Factor RelA metabolism, Transcription Factor RelA genetics, Binding Sites, Crystallography, X-Ray, DNA metabolism, Molecular Dynamics Simulation, Protein Binding, NF-kappa B metabolism
- Abstract
The dimeric nuclear factor kappa B (NF-κB) transcription factors (TFs) regulate gene expression by binding to a variety of κB DNA elements with conserved G:C-rich flanking sequences enclosing a degenerate central region. Toward defining mechanistic principles of affinity regulated by degeneracy, we observed an unusual dependence of the affinity of RelA on the identity of the central base pair, which appears to be noncontacted in the complex crystal structures. The affinity of κB sites with A or T at the central position is ~10-fold higher than with G or C. The crystal structures of neither the complexes nor the free κB DNAs could explain the differences in affinity. Interestingly, differential dynamics of several residues were revealed in molecular dynamics simulation studies, where simulation replicates totaling 148 μs were performed on NF-κB:DNA complexes and free κB DNAs. Notably, Arg187 and Arg124 exhibited selectivity in transient interactions that orchestrated a complex interplay among several DNA-interacting residues in the central region. Binding and simulation studies with mutants supported these observations of transient interactions dictating specificity. In combination with published reports, this work provides insights into the nuanced mechanisms governing the discriminatory binding of NF-κB family TFs to κB DNA elements and sheds light on cancer pathogenesis of cRel, a close homolog of RelA., Competing Interests: Competing interests statement:The authors declare no competing interest.
- Published
- 2024
- Full Text
- View/download PDF
5. Structural insights into the diverse prenylating capabilities of DMATS prenyltransferases.
- Author
-
Miller ET, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Prenylation, Fungi metabolism, Dimethylallyltranstransferase chemistry
- Abstract
Covering: 2009 up to August 2023Prenyltransferases (PTs) are involved in the primary and the secondary metabolism of plants, bacteria, and fungi, and they are key enzymes in the biosynthesis of many clinically relevant natural products (NPs). The continued biochemical and structural characterization of the soluble dimethylallyl tryptophan synthase (DMATS) PTs over the past two decades have revealed the significant promise that these enzymes hold as biocatalysts for the chemoenzymatic synthesis of novel drug leads. This is a comprehensive review of DMATSs describing the structure-function relationships that have shaped the mechanistic underpinnings of these enzymes, as well as the application of this knowledge to the engineering of DMATSs. We summarize the key findings and lessons learned from these studies over the past 14 years (2009-2023). In addition, we identify current gaps in our understanding of these fascinating enzymes.
- Published
- 2024
- Full Text
- View/download PDF
6. Discovery and development of novel substituted monohydrazides as potent antifungal agents.
- Author
-
Thamban Chandrika N, Green KD, Spencer AC, Tsodikov OV, and Garneau-Tsodikova S
- Abstract
Novel substituted monohydrazides synthesized for this study displayed broad-spectrum activity against various fungal strains, including a panel of clinically relevant Candida auris strains. The activity of these compounds was either comparable or superior to amphotericin B against most of the fungal strains tested. These compounds possessed fungistatic activity in a time-kill assay and exhibited no mammalian cell toxicity. In addition, they prevented the formation of fungal biofilms. Even after repeated exposures, the Candida albicans ATCC 10231 (strain A) fungal strain did not develop resistance to these monohydrazides., Competing Interests: The authors have no conflict of interest to disclose., (This journal is © The Royal Society of Chemistry.)
- Published
- 2023
- Full Text
- View/download PDF
7. Targeting thiol isomerase activity with zafirlukast to treat ovarian cancer from the bench to clinic.
- Author
-
Gelzinis JA, Szahaj MK, Bekendam RH, Wurl SE, Pantos MM, Verbetsky CA, Dufresne A, Shea M, Howard KC, Tsodikov OV, Garneau-Tsodikova S, Zwicker JI, and Kennedy DR
- Subjects
- Animals, Female, Humans, Mice, Indoles, Phenylcarbamates metabolism, Sulfhydryl Compounds metabolism, Blood Platelets metabolism, Ovarian Neoplasms drug therapy, Ovarian Neoplasms metabolism
- Abstract
Thiol isomerases, including PDI, ERp57, ERp5, and ERp72, play important and distinct roles in cancer progression, cancer cell signaling, and metastasis. We recently discovered that zafirlukast, an FDA-approved medication for asthma, is a pan-thiol isomerase inhibitor. Zafirlukast inhibited the growth of multiple cancer cell lines with an IC
50 in the low micromolar range, while also inhibiting cellular thiol isomerase activity, EGFR activation, and downstream phosphorylation of Gab1. Zafirlukast also blocked the procoagulant activity of OVCAR8 cells by inhibiting tissue factor-dependent Factor Xa generation. In an ovarian cancer xenograft model, statistically significant differences in tumor size between control vs treated groups were observed by Day 18. Zafirlukast also significantly reduced the number and size of metastatic tumors found within the lungs of the mock-treated controls. When added to a chemotherapeutic regimen, zafirlukast significantly reduced growth, by 38% compared with the mice receiving only the chemotherapeutic treatment, and by 83% over untreated controls. Finally, we conducted a pilot clinical trial in women with tumor marker-only (CA-125) relapsed ovarian cancer, where the rate of rise of CA-125 was significantly reduced following treatment with zafirlukast, while no severe adverse events were reported. Thiol isomerase inhibition with zafirlukast represents a novel, well-tolerated therapeutic in the treatment of ovarian cancer., (© 2023 Federation of American Societies for Experimental Biology.)- Published
- 2023
- Full Text
- View/download PDF
8. Identification and analysis of small molecule inhibitors of FosB from Staphylococcus aureus .
- Author
-
Travis S, Green KD, Thamban Chandrika N, Pang AH, Frantom PA, Tsodikov OV, Garneau-Tsodikova S, and Thompson MK
- Abstract
Antimicrobial resistance (AMR) poses a significant threat to human health around the world. Though bacterial pathogens can develop resistance through a variety of mechanisms, one of the most prevalent is the production of antibiotic-modifying enzymes like FosB, a Mn
2+ -dependent l-cysteine or bacillithiol (BSH) transferase that inactivates the antibiotic fosfomycin. FosB enzymes are found in pathogens such as Staphylococcus aureus , one of the leading pathogens in deaths associated with AMR. fosB gene knockout experiments establish FosB as an attractive drug target, showing that the minimum inhibitory concentration (MIC) of fosfomycin is greatly reduced upon removal of the enzyme. Herein, we have identified eight potential inhibitors of the FosB enzyme from S. aureus by applying high-throughput in silico screening of the ZINC15 database with structural similarity to phosphonoformate, a known FosB inhibitor. In addition, we have obtained crystal structures of FosB complexes to each compound. Furthermore, we have kinetically characterized the compounds with respect to inhibition of FosB. Finally, we have performed synergy assays to determine if any of the new compounds lower the MIC of fosfomycin in S. aureus . Our results will inform future studies on inhibitor design for the FosB enzymes., Competing Interests: There are no conflicts of interest to declare., (This journal is © The Royal Society of Chemistry.)- Published
- 2023
- Full Text
- View/download PDF
9. Aromatic hydrazides: A potential solution for Acinetobacter baumannii infections.
- Author
-
Green KD, Thamban Chandrika N, Vu LY, Pang AH, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Animals, Microbial Sensitivity Tests, Anti-Bacterial Agents pharmacology, Drug Resistance, Multiple, Bacterial, Mammals, Acinetobacter baumannii
- Abstract
The emergence of multidrug-resistant bacteria and the poor efficacy of available antibiotics against these infections have led to the urgent need for novel antibiotics. Acinetobacter baumannii is one of high-priority pathogens due to its ability to mount resistance to different classes of antibiotics. In an effort to provide novel agents in the fight against infections caused by A. baumannii, we synthesized a series of 46 aromatic hydrazides as potential treatments. In this series, 34 compounds were found to be low- to sub-μM inhibitors of A. baumannii growth, with MIC values in the range of 8 μg/mL to ≤0.125 μg/mL against a broad set of multidrug-resistant clinical isolates. These compounds were not highly active against other bacteria. We showed that one of the most potent compounds, 3e, was bacteriostatic and inhibitory to biofilm formation, although it did not disrupt the preformed biofilm. Additionally, we found that these compounds lacked mammalian cytotoxicity. The high antibacterial potency and the lack of mammalian cytotoxicity make these compounds a promising lead series for development of a novel selective anti-A. baumannii antibiotic., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Sylvie Garneau-Tsodikova reports a relationship with University of Kentucky that includes a patent “Monohydrazide Compounds with AntiAcinetobacterbaumannii Activity” pending to University of Kentucky., (Copyright © 2023 Elsevier Masson SAS. All rights reserved.)
- Published
- 2023
- Full Text
- View/download PDF
10. Discovery and Mechanistic Analysis of Structurally Diverse Inhibitors of Acetyltransferase Eis among FDA-Approved Drugs.
- Author
-
Pang AH, Green KD, Punetha A, Thamban Chandrika N, Howard KC, Garneau-Tsodikova S, and Tsodikov OV
- Subjects
- Humans, Antitubercular Agents pharmacology, Antitubercular Agents chemistry, Bacterial Proteins antagonists & inhibitors, Kanamycin pharmacology, Kanamycin chemistry, Proguanil metabolism, Acetyltransferases antagonists & inhibitors, Mycobacterium tuberculosis drug effects, Mycobacterium tuberculosis enzymology, Tuberculosis drug therapy
- Abstract
Over one and a half million people die of tuberculosis (TB) each year. Multidrug-resistant TB infections are especially dangerous, and new drugs are needed to combat them. The high cost and complexity of drug development make repositioning of drugs that are already in clinical use for other indications a potentially time- and money-saving avenue. In this study, we identified among existing drugs five compounds: azelastine, venlafaxine, chloroquine, mefloquine, and proguanil as inhibitors of acetyltransferase Eis from Mycobacterium tuberculosis , a causative agent of TB. Eis upregulation is a cause of clinically relevant resistance of TB to kanamycin, which is inactivated by Eis-catalyzed acetylation. Crystal structures of these drugs as well as chlorhexidine in complexes with Eis showed that these inhibitors were bound in the aminoglycoside binding cavity, consistent with their established modes of inhibition with respect to kanamycin. Among three additionally synthesized compounds, a proguanil analogue, designed based on the crystal structure of the Eis-proguanil complex, was 3-fold more potent than proguanil. The crystal structures of these compounds in complexes with Eis explained their inhibitory potencies. These initial efforts in rational drug repositioning can serve as a starting point in further development of Eis inhibitors.
- Published
- 2023
- Full Text
- View/download PDF
11. Mithplatins: Mithramycin SA-Pt(II) Complex Conjugates for the Treatment of Platinum-Resistant Ovarian Cancers.
- Author
-
Bhosale SS, Mandal A, Hou C, McCorkle JR, Schweer D, Hill KS, Subramanian V, Kolesar JM, Tsodikov OV, and Rohr J
- Subjects
- Humans, Female, Cisplatin pharmacology, Cisplatin chemistry, Plicamycin pharmacology, DNA metabolism, Cell Line, Tumor, Drug Resistance, Neoplasm, Antineoplastic Agents pharmacology, Antineoplastic Agents chemistry, Ovarian Neoplasms drug therapy, Ovarian Neoplasms pathology
- Abstract
DNA coordinating platinum (Pt) containing compounds cisplatin and carboplatin have been used for the treatment of ovarian cancer therapy for four decades. However, recurrent Pt-resistant cancers are a major cause of mortality. To combat Pt-resistant ovarian cancers, we designed and synthesized a conjugate of an anticancer drug mithramycin with a reactive Pt(II) bearing moiety, which we termed mithplatin. The conjugates displayed both the Mg
2+ -dependent noncovalent DNA binding characteristic of mithramycin and the covalent crosslinking to DNA of the Pt. The conjugate was three times as potent as cisplatin against ovarian cancer cells. The DNA lesions caused by the conjugate led to the generation of DNA double-strand breaks, as also observed with cisplatin. Nevertheless, the conjugate was highly active against both Pt-sensitive and Pt-resistant ovarian cancer cells. This study paves the way to developing mithplatins to combat Pt-resistant ovarian cancers., (© 2022 Wiley-VCH GmbH.)- Published
- 2023
- Full Text
- View/download PDF
12. Inhibition of Fosfomycin Resistance Protein FosB from Gram-Positive Pathogens by Phosphonoformate.
- Author
-
Travis S, Green KD, Gilbert NC, Tsodikov OV, Garneau-Tsodikova S, and Thompson MK
- Subjects
- Anti-Bacterial Agents chemistry, Foscarnet metabolism, Foscarnet pharmacology, Microbial Sensitivity Tests, Staphylococcus aureus metabolism, Transferases metabolism, Drug Resistance, Bacterial, Bacterial Proteins metabolism, Fosfomycin chemistry
- Abstract
The Gram-positive pathogen Staphylococcus aureus is a leading cause of antimicrobial resistance related deaths worldwide. Like many pathogens with multidrug-resistant strains, S. aureus contains enzymes that confer resistance through antibiotic modification(s). One such enzyme present in S. aureus is FosB, a Mn
2+ -dependent l-cysteine or bacillithiol (BSH) transferase that inactivates the antibiotic fosfomycin. fosB gene knockout experiments show that the minimum inhibitory concentration (MIC) of fosfomycin is significantly reduced when the FosB enzyme is not present. This suggests that inhibition of FosB could be an effective method to restore fosfomycin activity. We used high-throughput in silico -based screening to identify small-molecule analogues of fosfomycin that inhibited thiol transferase activity. Phosphonoformate (PPF) was a top hit from our approach. Herein, we have characterized PPF as a competitive inhibitor of FosB from S. aureus (FosBSa ) and Bacillus cereus (FosBBc ). In addition, we have determined a crystal structure of FosBBc with PPF bound in the active site. Our results will be useful for future structure-based development of FosB inhibitors that can be delivered in combination with fosfomycin in order to increase the efficacy of this antibiotic.- Published
- 2023
- Full Text
- View/download PDF
13. A Colorimetric Assay to Identify and Characterize Bacterial Primase Inhibitors.
- Author
-
Pang AH and Tsodikov OV
- Subjects
- Colorimetry, Biological Assay, DNA, Bacterial, DNA Primase, Mycobacterium tuberculosis
- Abstract
Bacterial DNA primase DnaG is an attractive target for antibiotic discovery since it plays an essential role in DNA replication. Over the last 10 years, we have developed and optimized a robust colorimetric assay that enabled us to identify and validate inhibitors of bacterial primases. Here, we provide a detailed protocol for this colorimetric assay for DnaG from three different pathogenic bacteria (Mycobacterium tuberculosis, Bacillus anthracis, and Staphylococcus aureus), which can be performed in high throughput. We also describe secondary assays to characterize hits from this high-throughput screening assay. These assays are designed to identify inhibitors of the coupled enzyme inorganic pyrophosphatase, DNA binding agents, and elucidate the mode of inhibition of primase inhibitors., (© 2023. The Author(s), under exclusive license to Springer Science+Business Media, LLC, part of Springer Nature.)
- Published
- 2023
- Full Text
- View/download PDF
14. Discovery and development of inhibitors of acetyltransferase Eis to combat Mycobacterium tuberculosis.
- Author
-
Pang AH, Green KD, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Humans, Bacterial Proteins chemistry, Anti-Bacterial Agents pharmacology, Aminoglycosides, Acetyltransferases chemistry, Mycobacterium tuberculosis, Tuberculosis
- Abstract
Aminoglycosides are bactericidal antibiotics with a broad spectrum of activity, used to treat infections caused mostly by Gram-negative pathogens and as a second-line therapy against tuberculosis. A common resistance mechanism to aminoglycosides is bacterial aminoglycoside acetyltransferase enzymes (AACs), which render aminoglycosides inactive by acetylating their amino groups. In Mycobacterium tuberculosis, an AAC called Eis (enhanced intracellular survival) acetylates kanamycin and amikacin. When upregulated as a result of mutations, Eis causes clinically important aminoglycoside resistance; therefore, Eis inhibitors are attractive as potential aminoglycoside adjuvants for treatment of aminoglycoside-resistant tuberculosis. For over a decade, we have studied Eis and discovered several series of Eis inhibitors. Here, we provide a detailed protocol for a colorimetric assay used for high-throughput discovery of Eis inhibitors, their characterization, and testing their selectivity. We describe protocols for in vitro cell culture assays for testing aminoglycoside adjuvant properties of the inhibitors. A procedure for obtaining crystals of Eis-inhibitor complexes and determining their structures is also presented. Finally, we discuss applicability of these methods to discovery and testing of inhibitors of other AACs., (Copyright © 2023. Published by Elsevier Inc.)
- Published
- 2023
- Full Text
- View/download PDF
15. Discovery of substituted benzyloxy-benzylamine inhibitors of acetyltransferase Eis and their anti-mycobacterial activity.
- Author
-
Pang AH, Green KD, Chandrika NT, Garzan A, Punetha A, Holbrook SYL, Willby MJ, Posey JE, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Aminoglycosides pharmacology, Animals, Anti-Bacterial Agents metabolism, Anti-Bacterial Agents pharmacology, Antitubercular Agents chemistry, Bacterial Proteins, Benzylamines pharmacology, Kanamycin chemistry, Kanamycin pharmacology, Mammals metabolism, Acetyltransferases chemistry, Mycobacterium tuberculosis metabolism
- Abstract
A clinically significant mechanism of tuberculosis resistance to the aminoglycoside kanamycin (KAN) is its acetylation catalyzed by upregulated Mycobacterium tuberculosis (Mtb) acetyltransferase Eis. In search for inhibitors of Eis, we discovered an inhibitor with a substituted benzyloxy-benzylamine scaffold. A structure-activity relationship study of 38 compounds in this structural family yielded highly potent (IC
50 ∼ 1 μM) Eis inhibitors, which did not inhibit other acetyltransferases. Crystal structures of Eis in complexes with three of the inhibitors showed that the inhibitors were bound in the aminoglycoside binding site of Eis, consistent with the competitive mode of inhibition, as established by kinetics measurements. When tested in Mtb cultures, two inhibitors (47 and 55) completely abolished resistance to KAN of the highly KAN-resistant strain Mtb mc2 6230 K204, likely due to Eis inhibition as a major mechanism. Thirteen of the compounds were toxic even in the absence of KAN to Mtb and other mycobacteria, but not to non-mycobacteria or to mammalian cells. This, yet unidentified mechanism of toxicity, distinct from Eis inhibition, will merit future studies along with further development of these molecules as anti-mycobacterial agents., Competing Interests: Declaration of competing interest The authors declare the following financial interests/personal relationships which may be considered as potential competing interests: Sylvie Garneau-Tsodikova reports financial support was provided by National Institutes of Health., (Copyright © 2022 Elsevier Masson SAS. All rights reserved.)- Published
- 2022
- Full Text
- View/download PDF
16. Discovery and Optimization of 6-(1-Substituted pyrrole-2-yl)- s -triazine Containing Compounds as Antibacterial Agents.
- Author
-
Green KD, Pang AH, Thamban Chandrika N, Garzan A, Baughn AD, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Animals, Mammals, Microbial Sensitivity Tests, Pyrroles pharmacology, Triazines pharmacology, Anti-Bacterial Agents pharmacology, Methicillin-Resistant Staphylococcus aureus
- Abstract
Antimicrobial drug resistance is a major health issue plaguing healthcare worldwide and leading to hundreds of thousands of deaths globally each year. Tackling this problem requires discovery and development of new antibacterial agents. In this study, we discovered novel 6-(1-substituted pyrrole-2-yl)- s -triazine containing compounds that potently inhibited the growth of Staphylococcus aureus regardless of its methicillin-resistant status, displaying minimum inhibitory concentration (MIC) values as low as 1 μM. The presence of a single imidazole substituent was critical to the antibacterial activity of these compounds. Some of the compounds also inhibited several nontubercular mycobacteria. We have shown that these molecules are potent bacteriostatic agents and that they are nontoxic to mammalian cells at relevant concentrations. Further development of these compounds as novel antimicrobial agents will be aimed at expanding our armamentarium of antibiotics.
- Published
- 2022
- Full Text
- View/download PDF
17. Structure-based design of haloperidol analogues as inhibitors of acetyltransferase Eis from Mycobacterium tuberculosis to overcome kanamycin resistance.
- Author
-
Punetha A, Green KD, Garzan A, Thamban Chandrika N, Willby MJ, Pang AH, Hou C, Holbrook SYL, Krieger K, Posey JE, Parish T, Tsodikov OV, and Garneau-Tsodikova S
- Abstract
Tuberculosis (TB), caused by Mycobacterium tuberculosis ( Mtb ), is a deadly bacterial disease. Drug-resistant strains of Mtb make eradication of TB a daunting task. Overexpression of the enhanced intracellular survival (Eis) protein by Mtb confers resistance to the second-line antibiotic kanamycin (KAN). Eis is an acetyltransferase that acetylates KAN, inactivating its antimicrobial function. Development of Eis inhibitors as KAN adjuvant therapeutics is an attractive path to forestall and overcome KAN resistance. We discovered that an antipsychotic drug, haloperidol (HPD, 1 ), was a potent Eis inhibitor with IC
50 = 0.39 ± 0.08 μM. We determined the crystal structure of the Eis-haloperidol ( 1 ) complex, which guided synthesis of 34 analogues. The structure-activity relationship study showed that in addition to haloperidol ( 1 ), eight analogues, some of which were smaller than 1 , potently inhibited Eis (IC50 ≤ 1 μM). Crystal structures of Eis in complexes with three potent analogues and droperidol (DPD), an antiemetic and antipsychotic, were determined. Three compounds partially restored KAN sensitivity of a KAN-resistant Mtb strain K204 overexpressing Eis. The Eis inhibitors generally did not exhibit cytotoxicity against mammalian cells. All tested compounds were modestly metabolically stable in human liver microsomes, exhibiting 30-60% metabolism over the course of the assay. While direct repurposing of haloperidol as an anti-TB agent is unlikely due to its neurotoxicity, this study reveals potential approaches to modifying this chemical scaffold to minimize toxicity and improve metabolic stability, while preserving potent Eis inhibition., Competing Interests: There is no conflict of interest to declare., (This journal is © The Royal Society of Chemistry.)- Published
- 2021
- Full Text
- View/download PDF
18. Development of Single-Stranded DNA Bisintercalating Inhibitors of Primase DnaG as Antibiotics.
- Author
-
Green KD, Punetha A, Chandrika NT, Hou C, Garneau-Tsodikova S, and Tsodikov OV
- Subjects
- Anti-Bacterial Agents chemical synthesis, Anti-Bacterial Agents chemistry, Cell Line, DNA Primase metabolism, DNA, Single-Stranded chemical synthesis, DNA, Single-Stranded chemistry, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Humans, Microbial Sensitivity Tests, Staphylococcus aureus enzymology, Anti-Bacterial Agents pharmacology, DNA Primase antagonists & inhibitors, DNA, Single-Stranded pharmacology, Drug Development, Enzyme Inhibitors pharmacology, Staphylococcus aureus drug effects
- Abstract
Many essential enzymes in bacteria remain promising potential targets of antibacterial agents. In this study, we discovered that dequalinium, a topical antibacterial agent, is an inhibitor of Staphylococcus aureus primase DnaG (SaDnaG) with low-micromolar minimum inhibitory concentrations against several S. aureus strains, including methicillin-resistant bacteria. Mechanistic studies of dequalinium and a series of nine of its synthesized analogues revealed that these compounds are single-stranded DNA bisintercalators that penetrate a bacterium by compromising its membrane. The best compound of this series likely interacts with DnaG directly, inhibits both staphylococcal cell growth and biofilm formation, and displays no significant hemolytic activity or toxicity to mammalian cells. This compound is an excellent lead for further development of a novel anti-staphylococcal therapeutic., (© 2021 Wiley-VCH GmbH.)
- Published
- 2021
- Full Text
- View/download PDF
19. Allosteric interference in oncogenic FLI1 and ERG transactions by mithramycins.
- Author
-
Hou C, Mandal A, Rohr J, and Tsodikov OV
- Subjects
- Allosteric Regulation drug effects, Antibiotics, Antineoplastic chemistry, Binding Sites, Core Binding Factor Alpha 1 Subunit chemistry, Core Binding Factor Alpha 1 Subunit metabolism, Core Binding Factor beta Subunit chemistry, Core Binding Factor beta Subunit metabolism, Humans, Molecular Docking Simulation, Plicamycin chemistry, Protein Binding, Proto-Oncogene Protein c-fli-1 metabolism, Transcriptional Regulator ERG chemistry, Transcriptional Regulator ERG metabolism, Antibiotics, Antineoplastic pharmacology, Plicamycin pharmacology, Proto-Oncogene Protein c-fli-1 chemistry
- Abstract
ETS family transcription factors of ERG and FLI1 play a key role in oncogenesis of prostate cancer and Ewing sarcoma by binding regulatory DNA sites and interfering with function of other factors. Mithramycin (MTM) is an anti-cancer, DNA binding natural product that functions as a potent antagonist of ERG and FLI1 by an unknown mechanism. We present a series of crystal structures of the DNA binding domain (DBD) of ERG/FLI1 culminating in a structure of a high-order complex of the ERG/FLI1 DBD, transcription factor Runx2, core-binding factor beta (Cbfβ), and MTM on a DNA enhancer site, along with supporting DNA binding studies using MTM and its analogues. Taken together, these data provide insight into allosteric mechanisms underlying ERG and FLI1 transactions and their disruption by MTM analogues., Competing Interests: Declaration of interests The MTM analogues used in this study are described in the US Patent 9,447,135 titled “Semi-synthetic mithramycin derivatives with anti-cancer activity,” where J.R. and O.V.T are inventors., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
20. Mithramycin and Analogs for Overcoming Cisplatin Resistance in Ovarian Cancer.
- Author
-
Schweer D, McCorkle JR, Rohr J, Tsodikov OV, Ueland F, and Kolesar J
- Abstract
Ovarian cancer is a highly deadly malignancy in which recurrence is considered incurable. Resistance to platinum-based chemotherapy bodes a particularly abysmal prognosis, underscoring the need for novel therapeutic agents and strategies. The use of mithramycin, an antineoplastic antibiotic, has been previously limited by its narrow therapeutic window. Recent advances in semisynthetic methods have led to mithramycin analogs with improved pharmacological profiles. Mithramycin inhibits the activity of the transcription factor Sp1, which is closely linked with ovarian tumorigenesis and platinum-resistance. This article summarizes recent clinical developments related to mithramycin and postulates a role for the use of mithramycin, or its analog, in the treatment of platinum-resistant ovarian cancer.
- Published
- 2021
- Full Text
- View/download PDF
21. Mithramycin 2'-Oximes with Improved Selectivity, Pharmacokinetics, and Ewing Sarcoma Antitumor Efficacy.
- Author
-
Liu Y, Eckenrode JM, Zhang Y, Zhang J, Hayden RC, Kyomuhangi A, Ponomareva LV, Cui Z, Rohr J, Tsodikov OV, Van Lanen SG, Shaaban KA, Leggas M, and Thorson JS
- Subjects
- Animals, Antibiotics, Antineoplastic chemistry, Apoptosis, Bone Neoplasms pathology, Cell Proliferation, Female, Humans, Mice, Mice, SCID, Sarcoma, Ewing pathology, Tissue Distribution, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Antibiotics, Antineoplastic pharmacokinetics, Antibiotics, Antineoplastic pharmacology, Bone Neoplasms drug therapy, Oximes chemistry, Plicamycin chemistry, Sarcoma, Ewing drug therapy
- Abstract
Mithramycin A (MTM) inhibits the oncogenic transcription factor EWS-FLI1 in Ewing sarcoma, but poor pharmacokinetics (PK) and toxicity limit its clinical use. To address this limitation, we report an efficient MTM 2'-oxime (MTM
ox ) conjugation strategy for rapid MTM diversification. Comparative cytotoxicity assays of 41 MTMox analogues using E-twenty-six (ETS) fusion-dependent and ETS fusion-independent cancer cell lines revealed improved ETS fusion-independent/dependent selectivity indices for select 2'-conjugated analogues as compared to MTM. Luciferase-based reporter assays demonstrated target engagement at low nM concentrations, and molecular assays revealed that analogues inhibit the transcriptional activity of EWS-FLI1. These in vitro screens identified MTMox 32 E (a Phe-Trp dipeptide-based 2'-conjugate) for in vivo testing. Relative to MTM, MTMox 32 E displayed an 11-fold increase in plasma exposure and improved efficacy in an Ewing sarcoma xenograft. Importantly, these studies are the first to point to simple C3 aliphatic side-chain modification of MTM as an effective strategy to improve PK.- Published
- 2020
- Full Text
- View/download PDF
22. Structural Insight into the DNA Binding Function of Transcription Factor ERF.
- Author
-
Hou C, McCown C, Ivanov DN, and Tsodikov OV
- Abstract
ETS family transcription factors control development of different cell types in humans, whereas deregulation of these proteins leads to severe developmental syndromes and cancers. One of a few members of the ETS family that are known to act solely as repressors, ERF, is required for normal osteogenesis and hematopoiesis. Another important function of ERF is acting as a tumor suppressor by antagonizing oncogenic fusions involving other ETS family factors. The structure of ERF and the DNA binding properties specific to this protein have not been elucidated. In this study, we determined two crystal structures of the complexes of the DNA binding domain of ERF with DNA. In one, ERF is in a distinct dimeric form, with Cys72 in a reduced state. In the other, two dimers of ERF are assembled into a tetramer that is additionally locked by two Cys72-Cys72 disulfide bonds across the dimers. In the tetramer, the ERF molecules are bound to a pseudocontinuous DNA on the same DNA face at two GGAA binding sites on opposite strands. Sedimentation velocity analysis showed that this tetrameric assembly forms on continuous DNA containing such tandem sites spaced by 7 bp. Our bioinformatic analysis of three previously reported sets of ERF binding loci across entire genomes showed that these loci were enriched in such 7 bp spaced tandem sites. Taken together, these results strongly suggest that the observed tetrameric assembly is a functional state of ERF in the human cell.
- Published
- 2020
- Full Text
- View/download PDF
23. Unimodular Methylation by Adenylation-Thiolation Domains Containing an Embedded Methyltransferase.
- Author
-
Mori S, Garneau-Tsodikova S, and Tsodikov OV
- Subjects
- Adenosine Monophosphate metabolism, Depsipeptides chemistry, Echinomycin chemistry, Kinetics, Methylation, Methyltransferases chemistry, Microsporidia metabolism, Peptide Biosynthesis, Nucleic Acid-Independent, Peptide Synthases chemistry, Protein Domains, Streptomyces metabolism, Sulfhydryl Compounds metabolism, Depsipeptides metabolism, Echinomycin metabolism, Methyltransferases metabolism, Microsporidia enzymology, Peptide Synthases metabolism, Streptomyces enzymology
- Abstract
Nonribosomal peptides (NRPs) are natural products that are biosynthesized by large multi-enzyme assembly lines called nonribosomal peptide synthetases (NRPSs). We have previously discovered that backbone or side chain methylation of NRP residues is carried out by an interrupted adenylation (A) domain that contains an internal methyltransferase (M) domain, while maintaining a monolithic AMA fold of the bifunctional enzyme. A key question that has remained unanswered is at which step of the assembly line mechanism the methylation by these embedded M domains takes place. Does the M domain methylate an amino acid residue tethered to a thiolation (T) domain on same NRPS module (in cis), or does it methylate this residue on a nascent peptide tethered to a T domain on another module (in trans)? In this study, we investigated the kinetics of methylation by wild-type AMAT tridomains from two NRPSs involved in biosynthesis of anticancer depsipeptides thiocoraline and echinomycin, and by mutants of these domains, for which methylation can occur only in trans. The analysis of the methylation kinetics unequivocally demonstrated that the wild-type AMATs methylate overwhelmingly in cis, strongly suggesting that this is also the case in the context of the entire NRPS assembly line process. The mechanistic insight gained in this study will facilitate rational genetic engineering of NRPS to generate unnaturally methylated NRPs., (Copyright © 2020 Elsevier Ltd. All rights reserved.)
- Published
- 2020
- Full Text
- View/download PDF
24. Structure-Guided Optimization of Inhibitors of Acetyltransferase Eis from Mycobacterium tuberculosis .
- Author
-
Punetha A, Ngo HX, Holbrook SYL, Green KD, Willby MJ, Bonnett SA, Krieger K, Dennis EK, Posey JE, Parish T, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Drug Design, Kanamycin Resistance drug effects, Microbial Sensitivity Tests, Models, Molecular, Molecular Structure, Mycobacterium tuberculosis drug effects, Structure-Activity Relationship, Acetyltransferases antagonists & inhibitors, Bacterial Proteins antagonists & inhibitors, Enzyme Inhibitors chemistry, Enzyme Inhibitors pharmacology, Mycobacterium tuberculosis enzymology
- Abstract
The enhanced intracellular survival (Eis) protein of Mycobacterium tuberculosis ( Mtb ) is a versatile acetyltransferase that multiacetylates aminoglycoside antibiotics abolishing their binding to the bacterial ribosome. When overexpressed as a result of promoter mutations, Eis causes drug resistance. In an attempt to overcome the Eis-mediated kanamycin resistance of Mtb , we designed and optimized structurally unique thieno[2,3- d ]pyrimidine Eis inhibitors toward effective kanamycin adjuvant combination therapy. We obtained 12 crystal structures of enzyme-inhibitor complexes, which guided our rational structure-based design of 72 thieno[2,3- d ]pyrimidine analogues divided into three families. We evaluated the potency of these inhibitors in vitro as well as their ability to restore the activity of kanamycin in a resistant strain of Mtb , in which Eis was upregulated. Furthermore, we evaluated the metabolic stability of 11 compounds in vitro . This study showcases how structural information can guide Eis inhibitor design.
- Published
- 2020
- Full Text
- View/download PDF
25. Discovery of a Cryptic Intermediate in Late Steps of Mithramycin Biosynthesis.
- Author
-
Wheeler R, Yu X, Hou C, Mitra P, Chen JM, Herkules F, Ivanov DN, Tsodikov OV, and Rohr J
- Subjects
- Catalysis, Biological Products metabolism, Plicamycin biosynthesis
- Abstract
MtmOIV and MtmW catalyze the final two reactions in the mithramycin (MTM) biosynthetic pathway, the Baeyer-Villiger opening of the fourth ring of premithramycin B (PMB), creating the C3 pentyl side chain, strictly followed by reduction of the distal keto group on the new side chain. Unexpectedly this results in a C2 stereoisomer of mithramycin, iso-mithramycin (iso-MTM). Iso-MTM undergoes a non-enzymatic isomerization to MTM catalyzed by Mg
2+ ions. Crystal structures of MtmW and its complexes with co-substrate NADPH and PEG, suggest a catalytic mechanism of MtmW. The structures also show that a tetrameric assembly of this enzyme strikingly resembles the ring-shaped β subunit of a vertebrate ion channel. We show that MtmW and MtmOIV form a complex in the presence of PMB and NADPH, presumably to hand over the unstable MtmOIV product to MtmW, yielding iso-MTM, as a potential self-resistance mechanism against MTM toxicity., (© 2020 Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim.)- Published
- 2020
- Full Text
- View/download PDF
26. Combining Chalcones with Donepezil to Inhibit Both Cholinesterases and Aβ Fibril Assembly.
- Author
-
Thamban Chandrika N, Fosso MY, Tsodikov OV, LeVine Iii H, and Garneau-Tsodikova S
- Subjects
- Acetylcholinesterase metabolism, Amyloid beta-Peptides drug effects, Aniline Compounds chemistry, Butyrylcholinesterase metabolism, Chalcones chemical synthesis, Chalcones chemistry, Donepezil chemical synthesis, Donepezil chemistry, Humans, Models, Molecular, Thiazoles chemistry, Tritium metabolism, Amyloid beta-Peptides metabolism, Chalcones pharmacology, Cholinesterase Inhibitors pharmacology, Donepezil pharmacology
- Abstract
The fact that the number of people with Alzheimer's disease is increasing, combined with the limited availability of drugs for its treatment, emphasize the need for the development of novel effective therapeutics for treating this brain disorder. Herein, we focus on generating 12 chalcone-donepezil hybrids, with the goal of simultaneously targeting amyloid-β (Aβ) peptides as well as cholinesterases (i.e., acetylcholinesterase (AChE) and butyrylcholinesterase (BChE)). We present the design, synthesis, and biochemical evaluation of these two series of novel 1,3-chalcone-donepezil ( 15a - 15f ) or 1,4-chalcone-donepezil ( 16a - 16f ) hybrids. We evaluate the relationship between their structures and their ability to inhibit AChE/BChE activity as well as their ability to bind Aβ peptides. We show that several of these novel chalcone-donepezil hybrids can successfully inhibit AChE/BChE as well as the assembly of N -biotinylated Aβ
(1-42) oligomers. We also demonstrate that the Aβ binding site of these hybrids differs from that of Pittsburgh Compound B (PIB)., Competing Interests: The authors declare no conflict of interest.- Published
- 2019
- Full Text
- View/download PDF
27. Probing the Robustness of Inhibitors of Tuberculosis Aminoglycoside Resistance Enzyme Eis by Mutagenesis.
- Author
-
Green KD, Punetha A, Hou C, Garneau-Tsodikova S, and Tsodikov OV
- Subjects
- Acetylation, Acetyltransferases, Aminoglycosides pharmacology, Antigens, Bacterial metabolism, Antitubercular Agents pharmacology, Bacterial Proteins genetics, Bacterial Proteins metabolism, Binding Sites genetics, Drug Resistance, Bacterial drug effects, Drug Resistance, Bacterial genetics, Enzyme Inhibitors pharmacology, Kanamycin chemistry, Kanamycin pharmacology, Kinetics, Models, Molecular, Mutation, Mycobacterium tuberculosis genetics, Protein Conformation, Tuberculosis drug therapy, Tuberculosis, Multidrug-Resistant, Aminoglycosides metabolism, Antigens, Bacterial drug effects, Antigens, Bacterial genetics, Antitubercular Agents chemistry, Enzyme Inhibitors chemistry, Mutagenesis, Mycobacterium tuberculosis drug effects, Mycobacterium tuberculosis enzymology
- Abstract
Each year, millions of people worldwide contract tuberculosis (TB), the deadliest infection. The spread of infections with drug-resistant strains of Mycobacterium tuberculosis ( Mtb ) that are refractory to treatment poses a major global challenge. A major cause of resistance to antitubercular drugs of last resort, aminoglycosides, is overexpression of the Eis (enhanced intracellular survival) enzyme of Mtb , which inactivates aminoglycosides by acetylating them. We showed previously that this inactivation of aminoglycosides could be overcome by our recently reported Eis inhibitors that are currently in development as potential aminoglycoside adjunctive therapeutics against drug-resistant TB. To interrogate the robustness of the Eis inhibitors, we investigated the enzymatic activity of Eis and its inhibition by Eis inhibitors from three different structural families for nine single-residue mutants of Eis, including those found in the clinic. Three engineered mutations of the substrate binding site, D26A, W36A, and F84A, abolished inhibitor binding while compromising Eis enzymatic activity 2- to 3-fold. All other Eis mutants, including clinically observed ones, were potently inhibited by at least one inhibitor. This study helps position us one step ahead of Mtb resistance to Eis inhibitors as they are being developed for TB therapy.
- Published
- 2019
- Full Text
- View/download PDF
28. Publisher Correction: Unusual substrate and halide versatility of phenolic halogenase PltM.
- Author
-
Mori S, Pang AH, Chandrika NT, Garneau-Tsodikova S, and Tsodikov OV
- Abstract
The original version of this Article contained an error in Fig. 1, in which the labels 'NADP
+ ' and 'NADPH + H+ ' were incorrectly given as 'NADPH' and 'NADPH+ + H+ ', respectively. This has been corrected in both the PDF and HTML versions of the Article.- Published
- 2019
- Full Text
- View/download PDF
29. How mithramycin stereochemistry dictates its structure and DNA binding function.
- Author
-
Hou C, Rohr J, Parkin S, and Tsodikov OV
- Abstract
An aureolic acid natural product mithramycin (MTM) has been known for its potent antineoplastic properties. MTM inhibits cell growth by binding in the minor groove of double-stranded DNA as a dimer, in which the two molecules of MTM are coordinated to each other through a divalent metal ion. A crystal structure of an MTM analogue, MTM SA-Phe, in the active metal ion-coordinated dimeric form demonstrates how the stereochemical features of MTM define the helicity of the dimeric scaffold for its binding to a right-handed DNA double helix. We also show crystallographically and biochemically that MTM, but not MTM SA-Phe, can be inactivated by boric acid through formation of a large macrocyclic species, in which two molecules of MTM are crosslinked to each other through 3-side chain-boron-sugar intermolecular bonds. We discuss these structural and biochemical properties in the context of MTM biosynthesis and the design of MTM analogues as anticancer therapeutics.
- Published
- 2019
- Full Text
- View/download PDF
30. Unusual substrate and halide versatility of phenolic halogenase PltM.
- Author
-
Mori S, Pang AH, Thamban Chandrika N, Garneau-Tsodikova S, and Tsodikov OV
- Subjects
- Bacterial Proteins chemistry, Bacterial Proteins genetics, Binding Sites, Crystallography, X-Ray, Flavin-Adenine Dinucleotide chemistry, Flavins chemistry, Halogenation, Models, Molecular, Mutagenesis, Oxidoreductases genetics, Phloroglucinol chemistry, Phloroglucinol metabolism, Substrate Specificity, Bacterial Proteins metabolism, Flavin-Adenine Dinucleotide metabolism, Oxidoreductases metabolism
- Abstract
Controlled halogenation of chemically versatile substrates is difficult to achieve. Here we describe a unique flavin-dependent halogenase, PltM, which is capable of utilizing a wide range of halides for installation on a diverse array of phenolic compounds, including FDA-approved drugs and natural products, such as terbutaline, fenoterol, resveratrol, and catechin. Crystal structures of PltM in complex with phloroglucinol and FAD in different states yield insight into substrate recognition and the FAD recycling mechanism of this halogenase.
- Published
- 2019
- Full Text
- View/download PDF
31. Utilizing guanine-coordinated Zn 2+ ions to determine DNA crystal structures by single-wavelength anomalous diffraction.
- Author
-
Hou C and Tsodikov OV
- Subjects
- Ions, Plicamycin chemistry, Crystallography, X-Ray methods, DNA chemistry, Guanine chemistry, Oligonucleotides chemistry, Zinc chemistry
- Abstract
The experimental phase determination of crystal structures of nucleic acids and nucleic acid-ligand complexes would benefit from a facile method. Even for double-stranded DNA, software-generated models are generally insufficiently accurate to serve as molecular replacement search models, necessitating experimental phasing. Here, it is demonstrated that Zn
2+ ions coordinated to the N7 atom of guanine bases generate sufficient anomalous signal for single-wavelength anomalous diffraction (SAD) phasing of DNA crystal structures. Using zinc SAD, three crystal structures of double-stranded DNA oligomers, 5'-AGGGATCCCT-3', 5'-GGGATCCC-3' and 5'-GAGGCCTC-3', were determined. By determining the crystal structure of one of these oligomers, GAGGCCTC, in the presence of Mg2+ instead of Zn2+ , it was demonstrated that Zn2+ is not structurally perturbing. These structures allowed the analysis of structural changes in the DNA on the binding of analogues of the natural product mithramycin to two of these oligomers, AGGGATCCCT and GAGGCCTC. Zinc SAD may become a routine approach for determining the crystal structures of nucleic acids and their complexes with small molecules.- Published
- 2019
- Full Text
- View/download PDF
32. A crystal structure of coil 1B of vimentin in the filamentous form provides a model of a high-order assembly of a vimentin filament.
- Author
-
Pang AH, Obiero JM, Kulczyk AW, Sviripa VM, and Tsodikov OV
- Subjects
- Crystallography, X-Ray, Humans, Protein Conformation, Protein Multimerization, Vimentin genetics, Intermediate Filaments metabolism, Vimentin chemistry, Vimentin metabolism
- Abstract
Vimentin is an intermediate filament (IF) protein that is expressed in leukocytes, fibroblasts and endothelial cells of blood vessels. Vimentin filaments contribute to structural stability of the cell membrane, organelle positioning and protein transport. Vimentin self-assembles into a dimer that subsequently forms high-order structures, including tetramers and octamers. The details of IF assembly at crystallographic resolutions are limited to the tetrameric form. We describe a crystal structure of a fragment of a vimentin rod domain (coil 1B) with a dimer of tetramers in the asymmetric unit. Coil 1B in the crystal is in an infinitely high-order filamentous assembly state, in which the tetramers are packed against each other laterally in an antiparallel fashion across the crystal lattice. In one of the directions of lateral packing, the tetramers pack against each other strictly head-to-tail, and in the orthogonal direction the tetramers pack in a staggered manner. This organization of the tetramers of coil 1B in the crystal lattice, together with previously reported biochemical and structural data, yield a model of high-order vimentin filament assembly., Database: Structural data are available in the PDB under the accession number 5WHF., (© 2018 Federation of European Biochemical Societies.)
- Published
- 2018
- Full Text
- View/download PDF
33. Nucleoside triphosphate cosubstrates control the substrate profile and efficiency of aminoglycoside 3'- O -phosphotransferase type IIa.
- Author
-
Holbrook SYL, Gentry MS, Tsodikov OV, and Garneau-Tsodikova S
- Abstract
Aminoglycosides (AGs) are broad-spectrum antibiotics that play an important role in the control and treatment of bacterial infections. Despite the great antibacterial potency of AGs, resistance to these antibiotics has limited their clinical applications. The AG 3'- O -phosphotransferase of type IIa (APH(3')-IIa) encoded by the neo
R gene is a common bacterial AG resistance enzyme that inactivates AG antibiotics. This enzyme is used as a selection marker in molecular biology research. APH(3')-IIa catalyzes the transfer of the γ-phosphoryl group of ATP to an AG at its 3'-OH group. Although APH(3')-IIa has been reported to utilize exclusively ATP as a cosubstrate, we demonstrate that this enzyme can utilize a broad array of NTPs. By substrate profiling, TLC, and enzyme kinetics experiments, we probe AG phosphorylation by APH(3')-IIa with an extensive panel of substrates and cosubstrates (13 AGs and 10 NTPs) for the purpose of gaining a thorough understanding of this resistance enzyme. We find, for the first time, that the identity of the NTP cosubstrate dictates the set of AGs modified by APH(3')-IIa and the phosphorylation efficiency for different AGs.- Published
- 2018
- Full Text
- View/download PDF
34. Potent 1,2,4-Triazino[5,6 b]indole-3-thioether Inhibitors of the Kanamycin Resistance Enzyme Eis from Mycobacterium tuberculosis.
- Author
-
Ngo HX, Green KD, Gajadeera CS, Willby MJ, Holbrook SYL, Hou C, Garzan A, Mayhoub AS, Posey JE, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- A549 Cells, Acetyltransferases metabolism, Antitubercular Agents pharmacology, Bacterial Proteins metabolism, Binding Sites, HEK293 Cells, Humans, Indoles chemical synthesis, Kanamycin pharmacology, Microbial Sensitivity Tests, Mycobacterium tuberculosis drug effects, Protein Binding, Protein Structure, Secondary, Regression Analysis, Sulfides chemistry, Triazines chemistry, Acetyltransferases antagonists & inhibitors, Acetyltransferases chemistry, Bacterial Proteins antagonists & inhibitors, Bacterial Proteins chemistry, Indoles chemistry, Indoles pharmacology, Kanamycin Resistance drug effects, Mycobacterium tuberculosis enzymology
- Abstract
A common cause of resistance to kanamycin (KAN) in tuberculosis is overexpression of the enhanced intracellular survival (Eis) protein. Eis is an acetyltransferase that multiacetylates KAN and other aminoglycosides, rendering them unable to bind the bacterial ribosome. By high-throughput screening, a series of substituted 1,2,4-triazino[5,6 b]indole-3-thioether molecules were identified as effective Eis inhibitors. Herein, we purchased 17 and synthesized 22 new compounds, evaluated their potency, and characterized their steady-state kinetics. Four inhibitors were found not only to inhibit Eis in vitro, but also to act as adjuvants of KAN and partially restore KAN sensitivity in a Mycobacterium tuberculosis KAN-resistant strain in which Eis is upregulated. A crystal structure of Eis in complex with a potent inhibitor and CoA shows that the inhibitors bind in the aminoglycoside binding site snugly inserted into a hydrophobic cavity. These inhibitors will undergo preclinical development as novel KAN adjuvant therapies to treat KAN-resistant tuberculosis.
- Published
- 2018
- Full Text
- View/download PDF
35. Structural basis for backbone N-methylation by an interrupted adenylation domain.
- Author
-
Mori S, Pang AH, Lundy TA, Garzan A, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Adenosine Monophosphate chemistry, Catalysis, Catalytic Domain, Crystallography, X-Ray, Escherichia coli metabolism, Imines chemistry, Kinetics, Peptide Synthases chemistry, Protein Domains, Substrate Specificity, Time Factors, Amino Acids chemistry, Enzymes chemistry, Methylation, Peptides chemistry
- Abstract
Interrupted adenylation domains are enigmatic fusions, in which one enzyme is inserted into another to form a highly unusual bifunctional enzyme. We present the first crystal structure of an interrupted adenylation domain that reveals a unique embedded methyltransferase. The structure and functional data provide insight into how these enzymes N-methylate amino acid precursors en route to nonribosomal peptides.
- Published
- 2018
- Full Text
- View/download PDF
36. Structures of the Catalytic Domain of Bacterial Primase DnaG in Complexes with DNA Provide Insight into Key Priming Events.
- Author
-
Hou C, Biswas T, and Tsodikov OV
- Subjects
- Bacterial Proteins metabolism, DNA Primase metabolism, DNA, Bacterial biosynthesis, Models, Chemical, Protein Domains, Bacterial Proteins chemistry, DNA Primase chemistry, DNA, Bacterial chemistry, Models, Molecular, Mycobacterium tuberculosis enzymology
- Abstract
Bacterial primase DnaG is an essential nucleic acid polymerase that generates primers for replication of chromosomal DNA. The mechanism of DnaG remains unclear due to the paucity of structural information on DnaG in complexes with other replisome components. Here we report the first crystal structures of noncovalent DnaG-DNA complexes, obtained with the RNA polymerase domain of Mycobacterium tuberculosis DnaG and various DNA ligands. One structure, obtained with ds DNA, reveals interactions with DnaG as it slides on ds DNA and suggests how DnaG binds template for primer synthesis. In another structure, DNA in the active site of DnaG mimics the primer, providing insight into mechanisms for the nucleotide transfer and DNA translocation. In conjunction with the recent cryo-EM structure of the bacteriophage T7 replisome, this study yields a model for primer elongation and hand-off to DNA polymerase.
- Published
- 2018
- Full Text
- View/download PDF
37. Acetylation by Eis and Deacetylation by Rv1151c of Mycobacterium tuberculosis HupB: Biochemical and Structural Insight.
- Author
-
Green KD, Biswas T, Pang AH, Willby MJ, Reed MS, Stuchlik O, Pohl J, Posey JE, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Acetylation, Acetyltransferases antagonists & inhibitors, Acetyltransferases genetics, Amino Acid Sequence, Bacterial Proteins antagonists & inhibitors, Bacterial Proteins genetics, Cloning, Molecular, Crystallography, X-Ray, Drug Resistance, Multiple, Bacterial genetics, Gene Expression Regulation, Bacterial drug effects, Gene Expression Regulation, Bacterial physiology, Histone Deacetylases genetics, Histones genetics, Kinetics, Lysine chemistry, Models, Molecular, Mycobacterium tuberculosis genetics, Peptide Fragments metabolism, Protein Conformation, Protein Interaction Mapping, Protein Processing, Post-Translational, Recombinant Proteins metabolism, Sequence Alignment, Sequence Homology, Amino Acid, Tandem Mass Spectrometry, Acetyltransferases metabolism, Bacterial Proteins metabolism, Histone Deacetylases metabolism, Histones metabolism, Mycobacterium tuberculosis metabolism
- Abstract
Bacterial nucleoid-associated proteins (NAPs) are critical to genome integrity and chromosome maintenance. Post-translational modifications of bacterial NAPs appear to function similarly to their better studied mammalian counterparts. The histone-like NAP HupB from Mycobacterium tuberculosis (Mtb) was previously observed to be acetylated by the acetyltransferase Eis, leading to genome reorganization. We report biochemical and structural aspects of acetylation of HupB by Eis. We also found that the SirT-family NAD
+ -dependent deacetylase Rv1151c from Mtb deacetylated HupB in vitro and characterized the deacetylation kinetics. We propose that activities of Eis and Rv1151c could regulate the acetylation status of HupB to remodel the mycobacterial chromosome in response to environmental changes.- Published
- 2018
- Full Text
- View/download PDF
38. Alkylated Piperazines and Piperazine-Azole Hybrids as Antifungal Agents.
- Author
-
Thamban Chandrika N, Shrestha SK, Ngo HX, Tsodikov OV, Howard KC, and Garneau-Tsodikova S
- Subjects
- 14-alpha Demethylase Inhibitors chemistry, 14-alpha Demethylase Inhibitors metabolism, 14-alpha Demethylase Inhibitors pharmacology, 14-alpha Demethylase Inhibitors toxicity, Alkylation, Animals, Antifungal Agents metabolism, Antifungal Agents toxicity, Aspergillus drug effects, Candida albicans drug effects, Cell Line, Hemolysis drug effects, Humans, Mice, Microbial Sensitivity Tests, Molecular Docking Simulation, Piperazines metabolism, Piperazines toxicity, Protein Conformation, Sterol 14-Demethylase chemistry, Sterol 14-Demethylase metabolism, Antifungal Agents chemistry, Antifungal Agents pharmacology, Azoles chemistry, Piperazines chemistry, Piperazines pharmacology
- Abstract
The extensive use of fluconazole (FLC) and other azole drugs has caused the emergence and rise of azole-resistant fungi. The fungistatic nature of FLC in combination with toxicity concerns have resulted in an increased demand for new azole antifungal agents. Herein, we report the synthesis and antifungal activity of novel alkylated piperazines and alkylated piperazine-azole hybrids, their time-kill studies, their hemolytic activity against murine erythrocytes, as well as their cytotoxicity against mammalian cells. Many of these molecules exhibited broad-spectrum activity against all tested fungal strains, with excellent minimum inhibitory concentration (MIC) values against non-albicans Candida and Aspergillus strains. The most promising compounds were found to be less hemolytic than the FDA-approved antifungal agent voriconazole (VOR). Finally, we demonstrate that the synthetic alkylated piperazine-azole hybrids do not function by fungal membrane disruption, but instead by disruption of the ergosterol biosynthetic pathway via inhibition of the 14α-demethylase enzyme present in fungal cells.
- Published
- 2018
- Full Text
- View/download PDF
39. Deciphering Nature's Intricate Way of N,S-Dimethylating l-Cysteine: Sequential Action of Two Bifunctional Adenylation Domains.
- Author
-
Mori S, Garzan A, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Biosynthetic Pathways, Methylation, Peptide Biosynthesis, Nucleic Acid-Independent, Peptide Synthases chemistry, Protein Domains, Cysteine metabolism, Hydroxyquinolines metabolism, Micromonosporaceae enzymology, Micromonosporaceae metabolism, Oligopeptides metabolism, Peptide Synthases metabolism
- Abstract
Dimethylation of amino acids consists of an interesting and puzzling series of events that could be achieved, during nonribosomal peptide biosynthesis, either by a single adenylation (A) domain interrupted by a methyltransferase (M) domain or by the sequential action of two of such independent enzymes. Herein, to establish the method by which Nature N,S-dimethylates l-Cys, we studied its formation during thiochondrilline A biosynthesis by evaluating TioS(A
3a M3S A3b T3 ) and TioN(Aa MN Ab ). This study not only led to identification of the exact pathway followed in Nature by these two enzymes for N,S-dimethylation of l-Cys, but also revealed that a single interrupted A domain can N,N-dimethylate amino acids, a novel phenomenon in the nonribosomal peptide field. These findings offer important and useful insights for the development and engineering of novel interrupted A domain enzymes to serve, in the future, as tools for combinatorial biosynthesis.- Published
- 2017
- Full Text
- View/download PDF
40. Combating Enhanced Intracellular Survival (Eis)-Mediated Kanamycin Resistance of Mycobacterium tuberculosis by Novel Pyrrolo[1,5-a]pyrazine-Based Eis Inhibitors.
- Author
-
Garzan A, Willby MJ, Ngo HX, Gajadeera CS, Green KD, Holbrook SY, Hou C, Posey JE, Tsodikov OV, and Garneau-Tsodikova S
- Subjects
- Acetyltransferases antagonists & inhibitors, Acetyltransferases chemistry, Antitubercular Agents chemistry, Bacterial Proteins antagonists & inhibitors, Bacterial Proteins chemistry, Binding Sites, Enzyme Inhibitors chemistry, Gene Expression Regulation, Bacterial drug effects, Gene Expression Regulation, Enzymologic drug effects, Mycobacterium tuberculosis growth & development, Protein Binding, Pyrazines chemistry, Pyrazines pharmacology, Structure-Activity Relationship, Antitubercular Agents pharmacology, Enzyme Inhibitors pharmacology, Kanamycin Resistance drug effects, Mycobacterium tuberculosis drug effects
- Abstract
Tuberculosis (TB) remains one of the leading causes of mortality worldwide. Hence, the identification of highly effective antitubercular drugs with novel modes of action is crucial. In this paper, we report the discovery and development of pyrrolo[1,5-a]pyrazine-based analogues as highly potent inhibitors of the Mycobacterium tuberculosis (Mtb) acetyltransferase enhanced intracellular survival (Eis), whose up-regulation causes clinically observed resistance to the aminoglycoside (AG) antibiotic kanamycin A (KAN). We performed a structure-activity relationship (SAR) study to optimize these compounds as potent Eis inhibitors both against purified enzyme and in mycobacterial cells. A crystal structure of Eis in complex with one of the most potent inhibitors reveals that the compound is bound to Eis in the AG binding pocket, serving as the structural basis for the SAR. These Eis inhibitors have no observed cytotoxicity to mammalian cells and are promising leads for the development of innovative AG adjuvant therapies against drug-resistant TB.
- Published
- 2017
- Full Text
- View/download PDF
41. In Vitro Assays to Identify Antibiotics Targeting DNA Metabolism.
- Author
-
Pang AH, Garneau-Tsodikova S, and Tsodikov OV
- Subjects
- High-Throughput Screening Assays, Indicators and Reagents, Inorganic Pyrophosphatase metabolism, Mycobacterium tuberculosis enzymology, Rosaniline Dyes chemistry, Anti-Bacterial Agents pharmacology, Biological Assay methods, DNA, Bacterial metabolism
- Abstract
DNA metabolism embodies a number of biochemical pathways, which include targets of clinically used antibiotics as well as those that are only being explored as potential targets for inhibitory compounds. We give an overview of representative cell-based and enzymatic assays suitable for high-throughput-driven search for novel DNA metabolism inhibitors of established and novel DNA metabolism targets in bacteria. The protocol for a colorimetric coupled primase-inorganic pyrophosphatase assay developed by our group is described in detail.
- Published
- 2017
- Full Text
- View/download PDF
42. Sulfonamide-Based Inhibitors of Aminoglycoside Acetyltransferase Eis Abolish Resistance to Kanamycin in Mycobacterium tuberculosis.
- Author
-
Garzan A, Willby MJ, Green KD, Gajadeera CS, Hou C, Tsodikov OV, Posey JE, and Garneau-Tsodikova S
- Subjects
- Acetyltransferases metabolism, Anti-Bacterial Agents pharmacology, Dose-Response Relationship, Drug, Enzyme Inhibitors chemical synthesis, Enzyme Inhibitors chemistry, Humans, Kanamycin pharmacology, Microbial Sensitivity Tests, Models, Molecular, Molecular Structure, Mycobacterium tuberculosis metabolism, Structure-Activity Relationship, Sulfonamides chemical synthesis, Sulfonamides chemistry, Acetyltransferases antagonists & inhibitors, Drug Resistance, Multiple, Bacterial drug effects, Enzyme Inhibitors pharmacology, Mycobacterium tuberculosis drug effects, Sulfonamides pharmacology
- Abstract
A two-drug combination therapy where one drug targets an offending cell and the other targets a resistance mechanism to the first drug is a time-tested, yet underexploited approach to combat or prevent drug resistance. By high-throughput screening, we identified a sulfonamide scaffold that served as a pharmacophore to generate inhibitors of Mycobacterium tuberculosis acetyltransferase Eis, whose upregulation causes resistance to the aminoglycoside (AG) antibiotic kanamycin A (KAN) in Mycobacterium tuberculosis. Rational systematic derivatization of this scaffold to maximize Eis inhibition and abolish the Eis-mediated KAN resistance of M. tuberculosis yielded several highly potent agents. A crystal structure of Eis in complex with one of the most potent inhibitors revealed that the inhibitor bound Eis in the AG-binding pocket held by a conformationally malleable region of Eis (residues 28-37) bearing key hydrophobic residues. These Eis inhibitors are promising leads for preclinical development of innovative AG combination therapies against resistant TB., Competing Interests: The authors declare no competing financial interest.
- Published
- 2016
- Full Text
- View/download PDF
43. Discovery of Allosteric and Selective Inhibitors of Inorganic Pyrophosphatase from Mycobacterium tuberculosis.
- Author
-
Pang AH, Garzan A, Larsen MJ, McQuade TJ, Garneau-Tsodikova S, and Tsodikov OV
- Subjects
- Allosteric Regulation, Crystallography, X-Ray, Drug Discovery, Enzyme Inhibitors chemistry, Inorganic Pyrophosphatase metabolism, Molecular Structure, Enzyme Inhibitors pharmacology, Inorganic Pyrophosphatase antagonists & inhibitors, Mycobacterium tuberculosis enzymology
- Abstract
Inorganic pyrophosphatase (PPiase) is an essential enzyme that hydrolyzes inorganic pyrophosphate (PP
i ), driving numerous metabolic processes. We report a discovery of an allosteric inhibitor (2,4-bis(aziridin-1-yl)-6-(1-phenylpyrrol-2-yl)-s-triazine) of bacterial PPiases. Analogues of this lead compound were synthesized to target specifically Mycobacterium tuberculosis (Mtb) PPiase (MtPPiase). The best analogue (compound 16) with a Ki of 11 μM for MtPPiase is a species-specific inhibitor. Crystal structures of MtPPiase in complex with the lead compound and one of its analogues (compound 6) demonstrate that the inhibitors bind in a nonconserved interface between monomers of the hexameric MtPPiase in a yet unprecedented pairwise manner, while the remote conserved active site of the enzyme is occupied by a bound PPi substrate. Consistent with the structural studies, the kinetic analysis of the most potent inhibitor has indicated that it functions uncompetitively, by binding to the enzyme-substrate complex. The inhibitors appear to allosterically lock the active site in a closed state causing its dysfunctionalization and blocking the hydrolysis. These inhibitors are the first examples of allosteric, species-selective inhibitors of PPiases, serving as a proof-of-principle that PPiases can be selectively targeted.- Published
- 2016
- Full Text
- View/download PDF
44. Structures of mithramycin analogues bound to DNA and implications for targeting transcription factor FLI1.
- Author
-
Hou C, Weidenbach S, Cano KE, Wang Z, Mitra P, Ivanov DN, Rohr J, and Tsodikov OV
- Subjects
- Base Sequence, DNA metabolism, Models, Molecular, Molecular Conformation, Molecular Structure, Oligodeoxyribonucleotides chemistry, Oligodeoxyribonucleotides metabolism, Plicamycin metabolism, Protein Binding, Protein Interaction Domains and Motifs, Proto-Oncogene Protein c-fli-1 metabolism, Structure-Activity Relationship, DNA chemistry, Plicamycin chemistry, Proto-Oncogene Protein c-fli-1 chemistry
- Abstract
Transcription factors have been considered undruggable, but this paradigm has been recently challenged. DNA binding natural product mithramycin (MTM) is a potent antagonist of oncogenic transcription factor EWS-FLI1. Structural details of MTM recognition of DNA, including the FLI1 binding sequence GGA(A/T), are needed to understand how MTM interferes with EWS-FLI1. We report a crystal structure of an MTM analogue MTM SA-Trp bound to a DNA oligomer containing a site GGCC, and two structures of a novel analogue MTM SA-Phe in complex with DNA. MTM SA-Phe is bound to sites AGGG and GGGT on one DNA, and to AGGG and GGGA(T) (a FLI1 binding site) on the other, revealing how MTM recognizes different DNA sequences. Unexpectedly, at sub-micromolar concentrations MTMs stabilize FLI1-DNA complex on GGAA repeats, which are critical for the oncogenic function of EWS-FLI1. We also directly demonstrate by nuclear magnetic resonance formation of a ternary FLI1-DNA-MTM complex on a single GGAA FLI1/MTM binding site. These biochemical and structural data and a new FLI1-DNA structure suggest that MTM binds the minor groove and perturbs FLI1 bound nearby in the major groove. This ternary complex model may lead to development of novel MTM analogues that selectively target EWS-FLI1 or other oncogenic transcription factors, as anti-cancer therapeutics., (© The Author(s) 2016. Published by Oxford University Press on behalf of Nucleic Acids Research.)
- Published
- 2016
- Full Text
- View/download PDF
45. Discovery and Optimization of Two Eis Inhibitor Families as Kanamycin Adjuvants against Drug-Resistant M. tuberculosis .
- Author
-
Garzan A, Willby MJ, Green KD, Tsodikov OV, Posey JE, and Garneau-Tsodikova S
- Abstract
Drug-resistant tuberculosis (TB) is a global threat and innovative approaches such as using adjuvants of anti-TB therapeutics are required to combat it. High-throughput screening yielded two lead scaffolds of inhibitors of Mycobacterium tuberculosis ( Mtb ) acetyltransferase Eis, whose upregulation causes resistance to the anti-TB drug kanamycin (KAN). Chemical optimization on these scaffolds resulted in potent Eis inhibitors. One compound restored the activity of KAN in a KAN-resistant Mtb strain. Model structures of Eis-inhibitor complexes explain the structure-activity relationship.
- Published
- 2016
- Full Text
- View/download PDF
46. Potent Inhibitors of Acetyltransferase Eis Overcome Kanamycin Resistance in Mycobacterium tuberculosis.
- Author
-
Willby MJ, Green KD, Gajadeera CS, Hou C, Tsodikov OV, Posey JE, and Garneau-Tsodikova S
- Subjects
- Antitubercular Agents chemistry, Crystallography, X-Ray, Cyclic S-Oxides chemistry, Drug Design, High-Throughput Screening Assays, Kanamycin metabolism, Kanamycin pharmacology, Mycobacterium tuberculosis enzymology, Staphylococcus aureus drug effects, Structure-Activity Relationship, Thiazoles chemistry, Acetyltransferases antagonists & inhibitors, Antitubercular Agents pharmacology, Bacterial Proteins antagonists & inhibitors, Cyclic S-Oxides pharmacology, Kanamycin Resistance drug effects, Mycobacterium tuberculosis drug effects, Thiazoles pharmacology
- Abstract
A major cause of tuberculosis (TB) resistance to the aminoglycoside kanamycin (KAN) is the Mycobacterium tuberculosis (Mtb) acetyltransferase Eis. Upregulation of this enzyme is responsible for inactivation of KAN through acetylation of its amino groups. A 123 000-compound high-throughput screen (HTS) yielded several small-molecule Eis inhibitors that share an isothiazole S,S-dioxide heterocyclic core. These were investigated for their structure-activity relationships. Crystal structures of Eis in complex with two potent inhibitors show that these molecules are bound in the conformationally adaptable aminoglycoside binding site of the enzyme, thereby obstructing binding of KAN for acetylation. Importantly, we demonstrate that several Eis inhibitors, when used in combination with KAN against resistant Mtb, efficiently overcome KAN resistance. This approach paves the way toward development of novel combination therapies against aminoglycoside-resistant TB.
- Published
- 2016
- Full Text
- View/download PDF
47. Data publication with the structural biology data grid supports live analysis.
- Author
-
Meyer PA, Socias S, Key J, Ransey E, Tjon EC, Buschiazzo A, Lei M, Botka C, Withrow J, Neau D, Rajashankar K, Anderson KS, Baxter RH, Blacklow SC, Boggon TJ, Bonvin AM, Borek D, Brett TJ, Caflisch A, Chang CI, Chazin WJ, Corbett KD, Cosgrove MS, Crosson S, Dhe-Paganon S, Di Cera E, Drennan CL, Eck MJ, Eichman BF, Fan QR, Ferré-D'Amaré AR, Fromme JC, Garcia KC, Gaudet R, Gong P, Harrison SC, Heldwein EE, Jia Z, Keenan RJ, Kruse AC, Kvansakul M, McLellan JS, Modis Y, Nam Y, Otwinowski Z, Pai EF, Pereira PJ, Petosa C, Raman CS, Rapoport TA, Roll-Mecak A, Rosen MK, Rudenko G, Schlessinger J, Schwartz TU, Shamoo Y, Sondermann H, Tao YJ, Tolia NH, Tsodikov OV, Westover KD, Wu H, Foster I, Fraser JS, Maia FR, Gonen T, Kirchhausen T, Diederichs K, Crosas M, and Sliz P
- Subjects
- Crystallography, X-Ray, Internet, Software, Databases, Genetic, Macromolecular Substances chemistry, Publications
- Abstract
Access to experimental X-ray diffraction image data is fundamental for validation and reproduction of macromolecular models and indispensable for development of structural biology processing methods. Here, we established a diffraction data publication and dissemination system, Structural Biology Data Grid (SBDG; data.sbgrid.org), to preserve primary experimental data sets that support scientific publications. Data sets are accessible to researchers through a community driven data grid, which facilitates global data access. Our analysis of a pilot collection of crystallographic data sets demonstrates that the information archived by SBDG is sufficient to reprocess data to statistics that meet or exceed the quality of the original published structures. SBDG has extended its services to the entire community and is used to develop support for other types of biomedical data sets. It is anticipated that access to the experimental data sets will enhance the paradigm shift in the community towards a much more dynamic body of continuously improving data analysis.
- Published
- 2016
- Full Text
- View/download PDF
48. Dimerization and DNA recognition rules of mithramycin and its analogues.
- Author
-
Weidenbach S, Hou C, Chen JM, Tsodikov OV, and Rohr J
- Subjects
- Dimerization, Antibiotics, Antineoplastic chemistry, DNA chemistry, Plicamycin chemistry
- Abstract
The antineoplastic and antibiotic natural product mithramycin (MTM) is used against cancer-related hypercalcemia and, experimentally, against Ewing sarcoma and lung cancers. MTM exerts its cytotoxic effect by binding DNA as a divalent metal ion (Me(2+))-coordinated dimer and disrupting the function of transcription factors. A precise molecular mechanism of action of MTM, needed to develop MTM analogues selective against desired transcription factors, is lacking. Although it is known that MTM binds G/C-rich DNA, the exact DNA recognition rules that would allow one to map MTM binding sites remain incompletely understood. Towards this goal, we quantitatively investigated dimerization of MTM and several of its analogues, MTM SDK (for Short side chain, DiKeto), MTM SA-Trp (for Short side chain and Acid), MTM SA-Ala, and a biosynthetic precursor premithramycin B (PreMTM B), and measured the binding affinities of these molecules to DNA oligomers of different sequences and structural forms at physiological salt concentrations. We show that MTM and its analogues form stable dimers even in the absence of DNA. All molecules, except for PreMTM B, can bind DNA with the following rank order of affinities (strong to weak): MTM=MTM SDK>MTM SA-Trp>MTM SA-Ala. An X(G/C)(G/C)X motif, where X is any base, is necessary and sufficient for MTM binding to DNA, without a strong dependence on DNA conformation. These recognition rules will aid in mapping MTM sites across different promoters towards development of MTM analogues as useful anticancer agents., (Copyright © 2015. Published by Elsevier Inc.)
- Published
- 2016
- Full Text
- View/download PDF
49. Par-4 secretion: stoichiometry of 3-arylquinoline binding to vimentin.
- Author
-
Sviripa VM, Burikhanov R, Obiero JM, Yuan Y, Nickell JR, Dwoskin LP, Zhan CG, Liu C, Tsodikov OV, Rangnekar VM, and Watt DS
- Subjects
- Binding Sites drug effects, Endoplasmic Reticulum Chaperone BiP, Ether-A-Go-Go Potassium Channels metabolism, Humans, Molecular Structure, Quinolones chemistry, Stereoisomerism, Structure-Activity Relationship, Vimentin chemistry, Apoptosis Regulatory Proteins metabolism, Quinolones metabolism, Quinolones pharmacology, Vimentin metabolism
- Abstract
Advanced prostate tumors usually metastasize to the lung, bone, and other vital tissues and are resistant to conventional therapy. Prostate apoptosis response-4 protein (Par-4) is a tumor suppressor that causes apoptosis in therapy-resistant prostate cancer cells by binding specifically to a receptor, Glucose-regulated protein-78 (GRP78), found only on the surface of cancer cells. 3-Arylquinolines or "arylquins" induce normal cells to release Par-4 from the intermediate filament protein, vimentin and promote Par-4 secretion that targets cancer cells in a paracrine manner. A structure-activity study identified arylquins that promote Par-4 secretion, and an evaluation of arylquin binding to the hERG potassium ion channel using a [(3)H]-dofetilide binding assay permitted the identification of structural features that separated this undesired activity from the desired Par-4 secretory activity. A binding study that relied on the natural fluorescence of arylquins and that used the purified rod domain of vimentin (residues 99-411) suggested that the mechanism behind Par-4 release involved arylquin binding to multiple sites in the rod domain.
- Published
- 2016
- Full Text
- View/download PDF
50. Structural Basis for Dimerization and DNA Binding of Transcription Factor FLI1.
- Author
-
Hou C and Tsodikov OV
- Subjects
- Amino Acid Sequence, Crystallography, X-Ray, Humans, Models, Molecular, Molecular Sequence Data, Protein Binding, Protein Conformation, Sequence Homology, Amino Acid, DNA metabolism, Proto-Oncogene Protein c-fli-1 chemistry, Proto-Oncogene Protein c-fli-1 metabolism
- Abstract
FLI1 (Friend leukemia integration 1) is a metazoan transcription factor that is upregulated in a number of cancers. In addition, rearrangements of the fli1 gene cause sarcomas, leukemias, and lymphomas. These rearrangements encode oncogenic transcription factors, in which the DNA binding domain (DBD or ETS domain) of FLI1 on the C-terminal side is fused to a part of an another protein on the N-terminal side. Such abnormal cancer cell-specific fusions retain the DNA binding properties of FLI1 and acquire non-native protein-protein or protein-nucleic acid interactions of the substituted region. As a result, these fusions trigger oncogenic transcriptional reprogramming of the host cell. Interactions of FLI1 fusions with other proteins and with itself play a critical role in the oncogenic regulatory functions, and they are currently under intense scrutiny, mechanistically and as potential novel anticancer drug targets. We report elusive crystal structures of the FLI1 DBD, alone and in complex with cognate DNA containing a GGAA recognition sequence. Both structures reveal a previously unrecognized dimer of this domain, consistent with its dimerization in solution. The homodimerization interface is helix-swapped and dominated by hydrophobic interactions, including those between two interlocking Phe362 residues. A mutation of Phe362 to an alanine disrupted the propensity of this domain to dimerize without perturbing its structure or the DNA binding function, consistent with the structural observations. We propose that FLI1 DBD dimerization plays a role in transcriptional activation and repression by FLI1 and its fusions at promoters containing multiple FLI1 binding sites.
- Published
- 2015
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.