Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
1.
Microbiology (Reading) ; 168(7)2022 07.
Article in English | MEDLINE | ID: mdl-35829699

ABSTRACT

The enterohemorrhagic Escherichia coli pathotype is responsible for severe and dangerous infections in humans. Establishment of the infection requires colonization of the gastro-intestinal tract, which is dependent on the Type III Secretion System. The Type III Secretion System (T3SS) allows attachment of the pathogen to the mammalian host cell and cytoskeletal rearrangements within the host cell. Blocking the functionality of the T3SS is likely to reduce colonization and therefore limit the disease. This route offers an alternative to antibiotics, and problems with the development of antibiotics resistance. Salicylidene acylhydrazides have been shown to have an inhibitory effect on the T3SS in several pathogens. However, the main target of these compounds is still unclear. Past work has identified a number of putative protein targets of these compounds, one of which being WrbA. Whilst WrbA is considered an off-target interaction, this study presents the effect of the salicylidne acylhydrazide compounds on the activity of WrbA, along with crystal structures of WrbA from Yersinia pseudotuberculosis and Salmonella serovar Typhimurium; the latter also containing parts of the compound in the structure. We also present data showing that the original compounds were unstable in acidic conditions, and that later compounds showed improved stability.


Subject(s)
Enterohemorrhagic Escherichia coli , Escherichia coli Proteins , Yersinia pseudotuberculosis , Animals , Anti-Bacterial Agents/metabolism , Enterohemorrhagic Escherichia coli/metabolism , Escherichia coli Proteins/metabolism , Humans , Mammals/metabolism , Repressor Proteins/metabolism , Salmonella typhimurium/metabolism , Type III Secretion Systems/metabolism , Yersinia pseudotuberculosis/metabolism
2.
Cancers (Basel) ; 14(8)2022 Apr 08.
Article in English | MEDLINE | ID: mdl-35454783

ABSTRACT

Sialic acid (SA) is a monosaccharide usually linked to the terminus of glycan chains on the cell surface. It plays a crucial role in many biological processes, and hypersialylation is a common feature in cancer. Lectins are widely used to analyze the cell surface expression of SA. However, these protein molecules are usually expensive and easily denatured, which calls for the development of alternative glycan-specific receptors and cell imaging technologies. In this study, SA-imprinted fluorescent core-shell molecularly imprinted polymer particles (SA-MIPs) were employed to recognize SA on the cell surface of cancer cell lines. The SA-MIPs improved suspensibility and scattering properties compared with previously used core-shell SA-MIPs. Although SA-imprinting was performed using SA without preference for the α2,3- and α2,6-SA forms, we screened the cancer cell lines analyzed using the lectins Maackia Amurensis Lectin I (MAL I, α2,3-SA) and Sambucus Nigra Lectin (SNA, α2,6-SA). Our results show that the selected cancer cell lines in this study presented a varied binding behavior with the SA-MIPs. The binding pattern of the lectins was also demonstrated. Moreover, two different pentavalent SA conjugates were used to inhibit the binding of the SA-MIPs to breast, skin, and lung cancer cell lines, demonstrating the specificity of the SA-MIPs in both flow cytometry and confocal fluorescence microscopy. We concluded that the synthesized SA-MIPs might be a powerful future tool in the diagnostic analysis of various cancer cells.

3.
RSC Adv ; 12(4): 2319-2331, 2022 Jan 12.
Article in English | MEDLINE | ID: mdl-35425270

ABSTRACT

Coxsackievirus A24 variant (CVA24v) is responsible for several outbreaks and two pandemics of the highly contagious eye infection acute hemorrhagic conjunctivitis (AHC). Currently, neither prevention (vaccines) nor treatments (antivirals) are available for combating this disease. CVA24v attaches to cells by binding Neu5Ac-containing glycans on the surface of cells which facilitates entry. Previously, we have demonstrated that pentavalent Neu5Ac conjugates attenuate CVA24v infection of human corneal epithelial (HCE) cells. In this study, we report on the structure-based design of three classes of divalent Neu5Ac conjugates, with varying spacer lengths, and their effect on CVA24v transduction in HCE cells. In relative terms, the most efficient class of divalent Neu5Ac conjugates are more efficient than the pentavalent Neu5Ac conjugates previously reported.

4.
Int J Mol Sci ; 22(16)2021 Aug 05.
Article in English | MEDLINE | ID: mdl-34445134

ABSTRACT

Coxsackievirus A24 variant (CVA24v) is the primary causative agent of the highly contagious eye infection designated acute hemorrhagic conjunctivitis (AHC). It is solely responsible for two pandemics and several recurring outbreaks of the disease over the last decades, thus affecting millions of individuals throughout the world. To date, no antiviral agents or vaccines are available for combating this disease, and treatment is mainly supportive. CVA24v utilizes Neu5Ac-containing glycans as attachment receptors facilitating entry into host cells. We have previously reported that pentavalent Neu5Ac conjugates based on a glucose-scaffold inhibit CVA24v infection of human corneal epithelial cells. In this study, we report on the design and synthesis of scaffold-replaced pentavalent Neu5Ac conjugates and their effect on CVA24v cell transduction and the use of cryogenic electron microscopy (cryo-EM) to study the binding of these multivalent conjugates to CVA24v. The results presented here provide insights into the development of Neu5Ac-based inhibitors of CVA24v and, most significantly, the first application of cryo-EM to study the binding of a multivalent ligand to a lectin.


Subject(s)
Antiviral Agents/pharmacology , Coxsackievirus Infections/diet therapy , Enterovirus C, Human/drug effects , N-Acetylneuraminic Acid/pharmacology , Conjunctivitis, Acute Hemorrhagic/drug therapy , Conjunctivitis, Acute Hemorrhagic/metabolism , Conjunctivitis, Acute Hemorrhagic/virology , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/virology , Glucose/metabolism , Humans , Lectins/metabolism , Ligands , Polysaccharides/metabolism , Receptors, Virus/metabolism
5.
J Org Chem ; 86(13): 9145-9154, 2021 07 02.
Article in English | MEDLINE | ID: mdl-34138565

ABSTRACT

The synthesis of 4-O-alkyl analogues of N-acetylneuraminic acid (Neu5Ac) and the scope of the reaction are described. Activated alkyl halides and sulfonates and primary alkyl iodides give products in useful yields. The utility of the methodology is exemplified using a thiophenyl Neu5Ac building block to synthesize a 4-O-alkyl DANA analogue. These results expand the toolbox of Neu5Ac chemistry with value in drug discovery and for the design of novel tools to study the biology of Neu5Ac lectins.


Subject(s)
N-Acetylneuraminic Acid , Sialic Acids
6.
Biomolecules ; 11(1)2021 01 04.
Article in English | MEDLINE | ID: mdl-33406810

ABSTRACT

Pseudomonas aeruginosa is an opportunistic bacterial pathogen that employs its type III secretion system (T3SS) during the acute phase of infection to translocate cytotoxins into the host cell cytoplasm to evade the immune system. The PcrV protein is located at the tip of the T3SS, facilitates the integration of pore-forming proteins into the eukaryotic cell membrane, and is required for translocation of cytotoxins into the host cell. In this study, we used surface plasmon resonance screening to identify small molecule binders of PcrV. A follow-up structure-activity relationship analysis resulted in PcrV binders that protect macrophages in a P. aeruginosa cell-based infection assay. Treatment of P. aeruginosa infections is challenging due to acquired, intrinsic, and adaptive resistance in addition to a broad arsenal of virulence systems such as the T3SS. Virulence blocking molecules targeting PcrV constitute valuable starting points for development of next generation antibacterials to treat infections caused by P. aeruginosa.


Subject(s)
Antigens, Bacterial/metabolism , Bacterial Proteins/metabolism , Bacterial Toxins/metabolism , Pore Forming Cytotoxic Proteins/metabolism , Pseudomonas aeruginosa/metabolism , Small Molecule Libraries/pharmacology , Type III Secretion Systems/metabolism , Animals , Cell Line , Cell Survival/drug effects , Mice , Protein Binding/drug effects , Proton Magnetic Resonance Spectroscopy , Pseudomonas aeruginosa/drug effects , Pseudomonas aeruginosa/pathogenicity , Reproducibility of Results , Small Molecule Libraries/chemistry , Structure-Activity Relationship , Virulence/drug effects
7.
ACS Chem Biol ; 15(10): 2683-2691, 2020 10 16.
Article in English | MEDLINE | ID: mdl-32845119

ABSTRACT

Coxsackievirus A24 variant (CVA24v) and human adenovirus 37 (HAdV-37) are leading causative agents of the severe and highly contagious ocular infections acute hemorrhagic conjunctivitis and epidemic keratoconjunctivitis, respectively. Currently, neither vaccines nor antiviral agents are available for treating these diseases, which affect millions of individuals worldwide. CVA24v and HAdV-37 utilize sialic acid as attachment receptors facilitating entry into host cells. Previously, we and others have shown that derivatives based on sialic acid are effective in preventing HAdV-37 binding and infection of cells. Here, we designed and synthesized novel pentavalent sialic acid conjugates and studied their inhibitory effect against CVA24v and HAdV-37 binding and infection of human corneal epithelial cells. The pentavalent conjugates are the first reported inhibitors of CVA24v infection and proved efficient in blocking HAdV-37 binding. Taken together, the pentavalent conjugates presented here form a basis for the development of general inhibitors of these highly contagious ocular pathogens.


Subject(s)
Adenoviruses, Human/drug effects , Antiviral Agents/pharmacology , Enterovirus C, Human/drug effects , Sialic Acids/pharmacology , Adenoviruses, Human/chemistry , Binding Sites , Enterovirus C, Human/chemistry , Humans , Virus Attachment/drug effects , Virus Internalization/drug effects
8.
Virulence ; 11(1): 795-804, 2020 12.
Article in English | MEDLINE | ID: mdl-32507000

ABSTRACT

PSEUDOMONAS AERUGINOSA: is an opportunistic pathogen and a major cause of corneal infections worldwide. The bacterium secretes several toxins through its type III secretion system (T3SS) to subvert host immune responses. In addition, it is armed with intrinsic as well as acquired antibiotic resistance mechanisms that make treatment a significant challenge and new therapeutic interventions are needed. Type III secretion inhibitors have been studied as an alternative or in accompaniment to traditional antibiotics to inhibit virulence of bacteria. In this study, INP0341, a T3SS inhibitor, inhibited cytotoxicity by P. aeruginosa toward human corneal epithelial cells (HCEC) at 100 µM without affecting bacterial growth in the liquid media. An increased expression of antimicrobial peptides and reactive oxygen species generation was also observed in cells exposed to P. aeruginosa in the presence of INP0341. Furthermore, INP0341 efficiently attenuated corneal infection by P. aeruginosa in an experimental model of murine keratitis as evident from corneal opacity, clinical score and bacterial load. Thus, INP0341 appears to be a promising candidate to treat corneal infection caused by P. aeruginosa and can be further considered as an alternative therapeutic intervention.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Epithelial Cells/drug effects , Hydrazines/therapeutic use , Keratitis/drug therapy , Pseudomonas Infections/drug therapy , Pseudomonas aeruginosa/drug effects , Animals , Bacterial Load/drug effects , Cell Line , Cornea/cytology , Cornea/drug effects , Cornea/microbiology , Disease Models, Animal , Epithelial Cells/microbiology , Humans , Keratitis/microbiology , Mice , Mice, Inbred C57BL , Pseudomonas aeruginosa/pathogenicity , Type III Secretion Systems/antagonists & inhibitors , Virulence
9.
Viruses ; 12(3)2020 03 22.
Article in English | MEDLINE | ID: mdl-32235763

ABSTRACT

Most flaviviruses are arthropod-borne viruses, transmitted by either ticks or mosquitoes, and cause morbidity and mortality worldwide. They are endemic in many countries and have recently emerged in new regions, such as the Zika virus (ZIKV) in South-and Central America, the West Nile virus (WNV) in North America, and the Yellow fever virus (YFV) in Brazil and many African countries, highlighting the need for preparedness. Currently, there are no antiviral drugs available to treat flavivirus infections. We have previously discovered a broad-spectrum antiviral compound, benzavir-2, with potent antiviral activity against both DNA- and RNA-viruses. Our purpose was to investigate the inhibitory activity of benzavir-2 against flaviviruses. We used a ZIKV ZsGreen-expressing vector, two lineages of wild-type ZIKV, and other medically important flaviviruses. Benzavir-2 inhibited ZIKV derived reporter gene expression with an EC50 value of 0.8 ± 0.1 µM. Furthermore, ZIKV plaque formation, progeny virus production, and viral RNA expression were strongly inhibited. In addition, 2.5 µM of benzavir-2 reduced infection in vitro in three to five orders of magnitude for five other flaviviruses: WNV, YFV, the tick-borne encephalitis virus, Japanese encephalitis virus, and dengue virus. In conclusion, benzavir-2 was a potent inhibitor of flavivirus infection, which supported the broad-spectrum antiviral activity of benzavir-2.


Subject(s)
Antiviral Agents/pharmacology , Flavivirus/classification , Flavivirus/drug effects , Animals , Cell Line , Cell Survival , Cells, Cultured , Chlorocebus aethiops , Dose-Response Relationship, Drug , Flavivirus/genetics , Flavivirus/isolation & purification , Vero Cells , Viral Plaque Assay , Virus Replication/drug effects
10.
Sci Rep ; 10(1): 2103, 2020 02 07.
Article in English | MEDLINE | ID: mdl-32034212

ABSTRACT

Bacterial infections continue to threaten humankind and the rapid spread of antibiotic resistant bacteria is alarming. Current antibiotics target essential bacterial processes and thereby apply a strong selective pressure on pathogenic and non-pathogenic bacteria alike. One alternative strategy is to block bacterial virulence systems that are essential for the ability to cause disease but not for general bacterial viability. We have previously show that the plant natural product (-)-hopeaphenol blocks the type III secretion system (T3SS) in the Gram-negative pathogens Yersinia pseudotuberculosis and Pseudomonas aeruginosa. (-)-Hopeaphenol is a resveratrol tetramer and in the present study we explore various resveratrol dimers, including partial structures of (-)-hopeaphenol, as T3SS inhibitors. To allow rapid and efficient assessment of T3SS inhibition in P. aeruginosa, we developed a new screening method by using a green fluorescent protein reporter under the control of the ExoS promoter. Using a panel of assays we showed that compounds with a benzofuran core structure i.e. viniferifuran, dehydroampelopsin B, anigopreissin A, dehydro-δ-viniferin and resveratrol-piceatannol hybrid displayed significant to moderate activities towards the T3SS in Y. pseudotuberculosis and P. aeruginosa.


Subject(s)
Anti-Bacterial Agents/pharmacology , Pseudomonas aeruginosa/drug effects , Resveratrol/pharmacology , Type III Secretion Systems/antagonists & inhibitors , Yersinia pseudotuberculosis/drug effects , Benzofurans/pharmacology , Drug Discovery , Flavonoids/pharmacology , Genes, Reporter/genetics , Green Fluorescent Proteins/genetics , Phenols , Pseudomonas aeruginosa/pathogenicity , Resveratrol/analogs & derivatives , Stilbenes/pharmacology , Virulence/drug effects , Yersinia pseudotuberculosis/pathogenicity
11.
Article in English | MEDLINE | ID: mdl-31355152

ABSTRACT

Antibiotics are becoming less effective in treatment of infections caused by multidrug-resistant Pseudomonas aeruginosa. Antimicrobial therapies based on the inhibition of specific virulence-related traits, as opposed to growth inhibitors, constitute an innovative and appealing approach to tackle the threat of P. aeruginosa infections. The twin-arginine translocation (Tat) pathway plays an important role in the pathogenesis of P. aeruginosa, and constitutes a promising target for the development of anti-pseudomonal drugs. In this study we developed and optimized a whole-cell, one-well assay, based on native phospholipase C activity, to identify compounds active against the Tat system. Statistical robustness, sensitivity and consequently suitability for high-throughput screening (HTS) were confirmed by a dry run/pre-screening test scoring a Z' of 0.82 and a signal-to-noise ratio of 49. Using this assay, we evaluated ca. 40,000 molecules and identified 59 initial hits as possible Tat inhibitors. Since phospholipase C is exported into the periplasm by Tat, and subsequently translocated across the outer membrane by the type II secretion system (T2SS), our assay could also identify T2SS inhibitors. To validate our hits and discriminate between compounds that inhibited either Tat or T2SS, two separate counter assays were developed and optimized. Finally, three Tat inhibitors and one T2SS inhibitor were confirmed by means of dose-response analysis and additional counter and confirming assays. Although none of the identified inhibitors was suitable as a lead compound for drug development, this study validates our assay as a simple, efficient, and HTS compatible method for the identification of Tat and T2SS inhibitors.


Subject(s)
Anti-Bacterial Agents/pharmacology , High-Throughput Screening Assays , Pseudomonas aeruginosa/drug effects , Small Molecule Libraries/pharmacology , Twin-Arginine-Translocation System/drug effects , Type II Secretion Systems/drug effects , Anti-Bacterial Agents/chemistry , Dose-Response Relationship, Drug , Drug Discovery , Drug Resistance, Multiple, Bacterial/drug effects , Protein Transport/drug effects , Pseudomonas aeruginosa/genetics , Pseudomonas aeruginosa/growth & development , Pseudomonas aeruginosa/metabolism , Small Molecule Libraries/chemistry , Twin-Arginine-Translocation System/genetics , Twin-Arginine-Translocation System/metabolism , Type C Phospholipases/antagonists & inhibitors , Type C Phospholipases/genetics , Type C Phospholipases/metabolism , Type II Secretion Systems/genetics , Type II Secretion Systems/metabolism
12.
Viruses ; 11(5)2019 04 27.
Article in English | MEDLINE | ID: mdl-31035532

ABSTRACT

Human adenoviruses (HAdV) are the most common cause of ocular infections. Species B human adenovirus type 3 (HAdV-B3) causes pharyngoconjunctival fever (PCF), whereas HAdV-D8, -D37, and -D64 cause epidemic keratoconjunctivitis (EKC). Recently, HAdV-D53, -D54, and -D56 emerged as new EKC-causing agents. HAdV-E4 is associated with both PCF and EKC. We have previously demonstrated that HAdV-D37 uses sialic acid (SA)-containing glycans as cellular receptors on human corneal epithelial (HCE) cells, and the virus interaction with SA is mediated by the knob domain of the viral fiber protein. Here, by means of cell-based assays and using neuraminidase (a SA-cleaving enzyme), we investigated whether ocular HAdVs other than HAdV-D37 also use SA-containing glycans as receptors on HCE cells. We found that HAdV-E4 and -D56 infect HCE cells independent of SAs, whereas HAdV-D53 and -D64 use SAs as cellular receptors. HAdV-D8 and -D54 fiber knobs also bound to cell-surface SAs. Surprisingly, HCE cells were found resistant to HAdV-B3 infection. We also demonstrated that the SA-based molecule i.e., ME0462, designed to bind to SA-binding sites on the HAdV-D37 fiber knob, efficiently prevents binding and infection of several EKC-causing HAdVs. Surface plasmon resonance analysis confirmed a direct interaction between ME0462 and fiber knobs. Altogether, we demonstrate that SA-containing glycans serve as receptors for multiple EKC-causing HAdVs, and, that SA-based compound function as a broad-spectrum antiviral against known and emerging EKC-causing HAdVs.


Subject(s)
Adenovirus Infections, Human/metabolism , Adenovirus Infections, Human/virology , Adenoviruses, Human/physiology , N-Acetylneuraminic Acid/metabolism , Polysaccharides/metabolism , Receptors, Virus/metabolism , Viral Tropism , A549 Cells , Amino Acid Sequence , Capsid Proteins/chemistry , Capsid Proteins/metabolism , Humans , Keratoconjunctivitis/metabolism , Keratoconjunctivitis/virology , Sequence Analysis, DNA
13.
Biochem Biophys Res Commun ; 511(1): 117-121, 2019 03 26.
Article in English | MEDLINE | ID: mdl-30773257

ABSTRACT

The effect of corticosteroids on human physiology is complex and their use in tuberculosis patients remains controversial. In a high-throughput screening approach designed to discover virulence inhibitors, several corticosteroids were found to prevent cytolysis of fibroblasts infected with mycobacteria. Further experiments with Mycobacterium tuberculosis showed anti-cytolytic activity in the 10 nM range, but no effect on bacterial growth or survival in the absence of host cells at 20 µM. The results from a panel of corticosteroids with various affinities to the glucocorticoid- and mineralocorticoid receptors indicate that the inhibition of cytolysis most likely is mediated through the glucocorticoid receptor. Using live-imaging of M. tuberculosis-infected human monocyte-derived macrophages, we also show that corticosteroids to some extent control intracellular bacteria. In vitro systems with reduced complexity are to further study and understand the interactions between bacterial infection, immune defense and cell signaling.


Subject(s)
Adrenal Cortex Hormones/pharmacology , Fibroblasts/drug effects , Macrophages/drug effects , Protective Agents/pharmacology , Tuberculosis/drug therapy , Antitubercular Agents/pharmacology , Cell Line , Cell Survival/drug effects , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/microbiology , Humans , Macrophages/cytology , Macrophages/metabolism , Macrophages/microbiology , Mycobacterium tuberculosis/drug effects , Receptors, Glucocorticoid/metabolism , Tuberculosis/metabolism , Tuberculosis/microbiology
14.
Sci Rep ; 9(1): 26, 2019 01 10.
Article in English | MEDLINE | ID: mdl-30631100

ABSTRACT

High-throughput screening facilities do not generally support biosafety level 3 organisms such as Mycobacterium tuberculosis. To discover not only antibacterials, but also virulence inhibitors with either bacterial or host cell targets, an assay monitoring lung fibroblast survival upon infection was developed and optimized for 384-plate format and robotic liquid handling. By using Mycobacterium marinum as surrogate organism, 28,000 compounds were screened at biosafety level 2 classification, resulting in 49 primary hits. Exclusion of substances with unfavourable properties and known antimicrobials resulted in 11 validated hits of which 7 had virulence inhibiting properties and one had bactericidal effect also in wild type Mycobacterium tuberculosis. This strategy to discover virulence inhibitors using a model organism in high-throughput screening can be a valuable tool for other researchers working on drug discovery against tuberculosis and other biosafety level 3 infectious agents.


Subject(s)
Anti-Bacterial Agents/isolation & purification , Mycobacterium marinum/drug effects , Mycobacterium marinum/pathogenicity , Virulence Factors/antagonists & inhibitors , Cell Survival , Fibroblasts/physiology , High-Throughput Screening Assays/methods , Virulence
15.
Mol Plant Pathol ; 20(1): 20-32, 2019 01.
Article in English | MEDLINE | ID: mdl-30062690

ABSTRACT

The identification of chemical compounds that prevent and combat bacterial diseases is fundamental for crop production. Bacterial virulence inhibitors are a promising alternative to classical control treatments, because they have a low environmental impact and are less likely to generate bacterial resistance. The major virulence determinant of most animal and plant bacterial pathogens is the type III secretion system (T3SS). In this work, we screened nine plant extracts and 12 isolated compounds-including molecules effective against human pathogens-for their capacity to inhibit the T3SS of plant pathogens and for their applicability as virulence inhibitors for crop protection. The screen was performed using a luminescent reporter system developed in the model pathogenic bacterium Ralstonia solanacearum. Five synthetic molecules, one natural product and two plant extracts were found to down-regulate T3SS transcription, most through the inhibition of the regulator hrpB. In addition, for three of the molecules, corresponding to salicylidene acylhydrazide derivatives, the inhibitory effect caused a dramatic decrease in the secretion capacity, which was translated into impaired plant responses. These candidate virulence inhibitors were then tested for their ability to protect plants. We demonstrated that salicylidene acylhydrazides can limit R. solanacearum multiplication in planta and protect tomato plants from bacterial speck caused by Pseudomonas syringae pv. tomato. Our work validates the efficiency of transcription reporters to discover compounds or natural product extracts that can be potentially applied to prevent bacterial plant diseases.


Subject(s)
Plant Diseases/microbiology , Ralstonia solanacearum/physiology , Type III Secretion Systems , Anhydrides/pharmacology , Gene Expression Regulation, Bacterial/drug effects , Solanum lycopersicum/drug effects , Solanum lycopersicum/microbiology , Ralstonia solanacearum/drug effects , Ralstonia solanacearum/genetics , Ralstonia solanacearum/growth & development , Transcription, Genetic/drug effects , Type III Secretion Systems/drug effects , Type III Secretion Systems/genetics
16.
Cell Death Differ ; 26(9): 1615-1630, 2019 09.
Article in English | MEDLINE | ID: mdl-30442946

ABSTRACT

PARP3 has been shown to be a key driver of TGFß-induced epithelial-to-mesenchymal transition (EMT) and stemness in breast cancer cells, emerging as an attractive therapeutic target. Nevertheless, the therapeutic value of PARP3 inhibition has not yet been assessed. Here we investigated the impact of the absence of PARP3 or its inhibition on the tumorigenicity of BRCA1-proficient versus BRCA1-deficient breast cancer cell lines, focusing on the triple-negative breast cancer subtype (TNBC). We show that PARP3 knockdown exacerbates centrosome amplification and genome instability and reduces survival of BRCA1-deficient TNBC cells. Furthermore, we engineered PARP3-/- BRCA1-deficient or BRCA1-proficient TNBC cell lines using the CRISPR/nCas9D10A gene editing technology and demonstrate that the absence of PARP3 selectively suppresses the growth, survival and in vivo tumorigenicity of BRCA1-deficient TNBC cells, mechanistically via effects associated with an altered Rictor/mTORC2 signaling complex resulting from enhanced ubiquitination of Rictor. Accordingly, PARP3 interacts with and ADP-ribosylates GSK3ß, a positive regulator of Rictor ubiquitination and degradation. Importantly, these phenotypes were rescued by re-expression of a wild-type PARP3 but not by a catalytic mutant, demonstrating the importance of PARP3's catalytic activity. Accordingly, reduced survival and compromised Rictor/mTORC2 signaling were also observed using a cell-permeable PARP3-specific inhibitor. We conclude that PARP3 and BRCA1 are synthetic lethal and that targeting PARP3's catalytic activity is a promising therapeutic strategy for BRCA1-associated cancers via the Rictor/mTORC2 signaling pathway.


Subject(s)
BRCA1 Protein/genetics , Cell Cycle Proteins/genetics , Poly(ADP-ribose) Polymerases/genetics , Rapamycin-Insensitive Companion of mTOR Protein/genetics , Triple Negative Breast Neoplasms/genetics , Animals , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Heterografts , Humans , Mechanistic Target of Rapamycin Complex 2/genetics , Mice , Signal Transduction , Transforming Growth Factor beta/genetics , Triple Negative Breast Neoplasms/pathology
17.
ChemistryOpen ; 7(12): 953-956, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30524921

ABSTRACT

We demonstrate the versatile use of the cyclopropylmethyl group to protect phenols through the total synthesis of two benzofuran-based natural products, that is, anigopreissin A and the resveratrol-piceatannol hybrid. This protecting group is a good alternative to the conventional methyl group, owing to the feasibility of introduction, stability under a variety of conditions, and its relative ease of removal under different acidic conditions.

18.
Org Lett ; 20(21): 6650-6654, 2018 11 02.
Article in English | MEDLINE | ID: mdl-30350667

ABSTRACT

A convenient synthetic strategy to obtain viniferifuran and (±)-dehydroampelopsin B analogues based on the heterocyclic cores of indole, benzo[ b]thiophene, and benzo[ b]selenophene is presented. The key transformations utilized in the described syntheses include Sonogashira couplings, Cacchi and alkyne electrophilic cyclizations, Horner-Wadsworth-Emmons (HWE) reaction, chemoselective Suzuki-Miyaura couplings, and acid-promoted intramolecular cyclization to form the seven-membered ring of (±)-dehydroampelopsin B.

19.
Nat Commun ; 9(1): 3785, 2018 09 17.
Article in English | MEDLINE | ID: mdl-30224724

ABSTRACT

Pseudomonas are a common cause of hospital-acquired infections that may be lethal. ADP-ribosyltransferase activities of Pseudomonas exotoxin-S and -T depend on 14-3-3 proteins inside the host cell. By binding in the 14-3-3 phosphopeptide binding groove, an amphipathic C-terminal helix of ExoS and ExoT has been thought to be crucial for their activation. However, crystal structures of the 14-3-3ß:ExoS and -ExoT complexes presented here reveal an extensive hydrophobic interface that is sufficient for complex formation and toxin activation. We show that C-terminally truncated ExoS ADP-ribosyltransferase domain lacking the amphipathic binding motif is active when co-expressed with 14-3-3. Moreover, swapping the amphipathic C-terminus with a fragment from Vibrio Vis toxin creates a 14-3-3 independent toxin that ADP-ribosylates known ExoS targets. Finally, we show that 14-3-3 stabilizes ExoS against thermal aggregation. Together, this indicates that 14-3-3 proteins activate exotoxin ADP-ribosyltransferase domains by chaperoning their hydrophobic surfaces independently of the amphipathic C-terminal segment.


Subject(s)
14-3-3 Proteins/chemistry , ADP Ribose Transferases/chemistry , ADP Ribose Transferases/metabolism , Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , GTPase-Activating Proteins/chemistry , GTPase-Activating Proteins/metabolism , 14-3-3 Proteins/metabolism , ADP Ribose Transferases/genetics , Bacterial Toxins/genetics , Binding Sites , Crystallography, X-Ray , Escherichia coli/genetics , GTPase-Activating Proteins/genetics , Host-Pathogen Interactions , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Chaperones/chemistry , Molecular Chaperones/metabolism , Protein Conformation , Protein Domains , Pseudomonas aeruginosa/pathogenicity , Saccharomyces cerevisiae/genetics
20.
SLAS Discov ; 23(8): 815-822, 2018 09.
Article in English | MEDLINE | ID: mdl-29630847

ABSTRACT

Acetaldehyde dehydrogenase (AdhE) is a bifunctional acetaldehyde-coenzyme A (CoA) dehydrogenase and alcohol dehydrogenase involved in anaerobic metabolism in gram-negative bacteria. This enzyme was recently found to be a key regulator of the type three secretion (T3S) system in Escherichia coli. AdhE inhibitors can be used as tools to study bacterial virulence and a starting point for discovery of novel antibacterial agents. We developed a robust enzymatic assay, based on the acetaldehyde-CoA dehydrogenase activity of AdhE using both absorption and fluorescence detection models (Z' > 0.7). This assay was used to screen ~11,000 small molecules in 384-well format that resulted in three hits that were confirmed by resynthesis and validation. All three compounds are noncompetitive with respect to acetaldehyde and display a clear dose-response effect with hill slopes of 1-2. These new inhibitors will be used as chemical tools to study the interplay between metabolism and virulence and the role of AdhE in T3S regulation in gram-negative bacteria, and as starting points for the development of novel antibacterial agents.


Subject(s)
Alcohol Dehydrogenase/antagonists & inhibitors , Aldehyde Oxidoreductases/antagonists & inhibitors , Anti-Bacterial Agents/pharmacology , Drug Evaluation, Preclinical , Enterohemorrhagic Escherichia coli/drug effects , Enterohemorrhagic Escherichia coli/enzymology , Enzyme Inhibitors/pharmacology , Escherichia coli Proteins/antagonists & inhibitors , Alcohol Dehydrogenase/genetics , Alcohol Dehydrogenase/metabolism , Aldehyde Oxidoreductases/genetics , Aldehyde Oxidoreductases/metabolism , Animals , Anti-Bacterial Agents/chemistry , Cell Line , Dose-Response Relationship, Drug , Drug Discovery/methods , Drug Evaluation, Preclinical/methods , Enterohemorrhagic Escherichia coli/genetics , Enzyme Activation/drug effects , Enzyme Inhibitors/chemistry , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Mice , Workflow
SELECTION OF CITATIONS
SEARCH DETAIL