Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 30
Filter
2.
mBio ; 13(2): e0024022, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35258332

ABSTRACT

Bacterial type IV secretion systems (T4SSs) are macromolecular machines that translocate effector proteins across multiple membranes into infected host cells. Loss of function mutations in genes encoding protein components of the T4SS render bacteria avirulent, highlighting the attractiveness of T4SSs as drug targets. Here, we designed an automated high-throughput screening approach for the identification of compounds that interfere with the delivery of a reporter-effector fusion protein from Legionella pneumophila into RAW264.7 mouse macrophages. Using a fluorescence resonance energy transfer (FRET)-based detection assay in a bacteria/macrophage coculture format, we screened a library of over 18,000 compounds and, upon vetting compound candidates in a variety of in vitro and cell-based secondary screens, isolated several hits that efficiently interfered with biological processes that depend on a functional T4SS, such as intracellular bacterial proliferation or lysosomal avoidance, but had no detectable effect on L. pneumophila growth in culture medium, conditions under which the T4SS is dispensable. Notably, the same hit compounds also attenuated, to varying degrees, effector delivery by the closely related T4SS from Coxiella burnetii, notably without impacting growth of this organism within synthetic media. Together, these results support the idea that interference with T4SS function is a possible therapeutic intervention strategy, and the emerging compounds provide tools to interrogate at a molecular level the regulation and dynamics of these virulence-critical translocation machines. IMPORTANCE Multi-drug-resistant pathogens are an emerging threat to human health. Because conventional antibiotics target not only the pathogen but also eradicate the beneficial microbiota, they often cause additional clinical complications. Thus, there is an urgent need for the development of "smarter" therapeutics that selectively target pathogens without affecting beneficial commensals. The bacterial type IV secretion system (T4SS) is essential for the virulence of a variety of pathogens but dispensable for bacterial viability in general and can, thus, be considered a pathogen's Achilles heel. By identifying small molecules that interfere with cargo delivery by the T4SS from two important human pathogens, Legionella pneumophila and Coxiella burnetii, our study represents the first step in our pursuit toward precision medicine by developing pathogen-selective therapeutics capable of treating the infections without causing harm to commensal bacteria.


Subject(s)
Coxiella burnetii , Legionella pneumophila , Animals , Bacterial Secretion Systems/metabolism , Legionella pneumophila/metabolism , Mice , Type IV Secretion Systems/genetics , Type IV Secretion Systems/metabolism , Virulence Factors/genetics
3.
PLoS One ; 17(1): e0261821, 2022.
Article in English | MEDLINE | ID: mdl-35041689

ABSTRACT

The global health emergency posed by the outbreak of Zika virus (ZIKV), an arthropod-borne flavivirus causing severe neonatal neurological conditions, has subsided, but there continues to be transmission of ZIKV in endemic regions. As such, there is still a medical need for discovering and developing therapeutical interventions against ZIKV. To identify small-molecule compounds that inhibit ZIKV disease and transmission, we screened multiple small-molecule collections, mostly derived from natural products, for their ability to inhibit wild-type ZIKV. As a primary high-throughput screen, we used a viral cytopathic effect (CPE) inhibition assay conducted in Vero cells that was optimized and miniaturized to a 1536-well format. Suitably active compounds identified from the primary screen were tested in a panel of orthogonal assays using recombinant Zika viruses, including a ZIKV Renilla luciferase reporter assay and a ZIKV mCherry reporter system. Compounds that were active in the wild-type ZIKV inhibition and ZIKV reporter assays were further evaluated for their inhibitory effects against other flaviviruses. Lastly, we demonstrated that wild-type ZIKV is able to infect a 3D-bioprinted outer-blood-retina barrier tissue model and disrupt its barrier function, as measured by electrical resistance. One of the identified compounds (3-Acetyl-13-deoxyphomenone, NCGC00380955) was able to prevent the pathological effects of the viral infection on this clinically relevant ZIKV infection model.


Subject(s)
Antiviral Agents/pharmacology , Models, Biological , Printing, Three-Dimensional , Retina , Virus Replication/drug effects , Zika Virus Infection , Zika Virus/physiology , Animals , Antiviral Agents/chemistry , Chlorocebus aethiops , Drug Evaluation, Preclinical , Hep G2 Cells , Humans , Retina/metabolism , Retina/virology , Vero Cells , Virus Replication/genetics , Zika Virus Infection/drug therapy , Zika Virus Infection/genetics , Zika Virus Infection/metabolism
4.
Stem Cell Reports ; 16(12): 3076-3092, 2021 12 14.
Article in English | MEDLINE | ID: mdl-34861164

ABSTRACT

Efficient translation of human induced pluripotent stem cells (hiPSCs) requires scalable cell manufacturing strategies for optimal self-renewal and functional differentiation. Traditional manual cell culture is variable and labor intensive, posing challenges for high-throughput applications. Here, we established a robotic platform and automated all essential steps of hiPSC culture and differentiation under chemically defined conditions. This approach allowed rapid and standardized manufacturing of billions of hiPSCs that can be produced in parallel from up to 90 different patient- and disease-specific cell lines. Moreover, we established automated multi-lineage differentiation and generated functional neurons, cardiomyocytes, and hepatocytes. To validate our approach, we compared robotic and manual cell culture operations and performed comprehensive molecular and cellular characterizations (e.g., single-cell transcriptomics, mass cytometry, metabolism, electrophysiology) to benchmark industrial-scale cell culture operations toward building an integrated platform for efficient cell manufacturing for disease modeling, drug screening, and cell therapy.


Subject(s)
Cell Culture Techniques/methods , Cell Differentiation , Induced Pluripotent Stem Cells/cytology , Robotics , Automation , Cell Lineage , Cells, Cultured , Embryoid Bodies/cytology , Hepatocytes/cytology , Hepatocytes/virology , Human Embryonic Stem Cells/cytology , Humans , Myocytes, Cardiac/cytology , Myocytes, Cardiac/virology , Neurons/cytology , RNA-Seq , Reference Standards , Single-Cell Analysis , Zika Virus Infection/pathology
5.
ACS Pharmacol Transl Sci ; 4(3): 1214-1226, 2021 Jun 11.
Article in English | MEDLINE | ID: mdl-34151211

ABSTRACT

Activation-induced deaminase (AID) not only mutates DNA within the immunoglobulin loci to generate antibody diversity, but it also promotes development of B cell lymphomas. To tame this mutagen, we performed a quantitative high-throughput screen of over 90 000 compounds to see if AID activity could be mitigated. The enzymatic activity was assessed in biochemical assays to detect cytosine deamination and in cellular assays to measure class switch recombination. Three compounds showed promise via inhibition of switching in a transformed B cell line and in murine splenic B cells. These compounds have similar chemical structures, which suggests a shared mechanism of action. Importantly, the inhibitors blocked AID, but not a related cytosine DNA deaminase, APOBEC3B. We further determined that AID was continually expressed for several days after B cell activation to induce switching. This first report of small molecules that inhibit AID can be used to gain regulatory control over base editors.

6.
Cancers (Basel) ; 13(8)2021 Apr 14.
Article in English | MEDLINE | ID: mdl-33919707

ABSTRACT

FEN1 plays critical roles in long patch base excision repair (LP-BER), Okazaki fragment maturation, and rescue of stalled replication forks. In a clinical cohort, FEN1 overexpression is associated with aggressive phenotype and poor progression-free survival after platinum chemotherapy. Pre-clinically, FEN1 is induced upon cisplatin treatment, and nuclear translocation of FEN1 is dependent on physical interaction with importin ß. FEN1 depletion, gene inactivation, or inhibition re-sensitizes platinum-resistant ovarian cancer cells to cisplatin. BRCA2 deficient cells exhibited synthetic lethality upon treatment with a FEN1 inhibitor. FEN1 inhibitor-resistant PEO1R cells were generated, and these reactivated BRCA2 and overexpressed the key repair proteins, POLß and XRCC1. FEN1i treatment was selectively toxic to POLß deficient but not XRCC1 deficient ovarian cancer cells. High throughput screening of 391,275 compounds identified several FEN1 inhibitor hits that are suitable for further drug development. We conclude that FEN1 is a valid target for ovarian cancer therapy.

7.
J Med Chem ; 64(4): 2291-2309, 2021 02 25.
Article in English | MEDLINE | ID: mdl-33573376

ABSTRACT

A novel diazaspiro[3.4]octane series was identified from a Plasmodium falciparum whole-cell high-throughput screening campaign. Hits displayed activity against multiple stages of the parasite lifecycle, which together with a novel sp3-rich scaffold provided an attractive starting point for a hit-to-lead medicinal chemistry optimization and biological profiling program. Structure-activity-relationship studies led to the identification of compounds that showed low nanomolar asexual blood-stage activity (<50 nM) together with strong gametocyte sterilizing properties that translated to transmission-blocking activity in the standard membrane feeding assay. Mechanistic studies through resistance selection with one of the analogues followed by whole-genome sequencing implicated the P. falciparum cyclic amine resistance locus in the mode of resistance.


Subject(s)
Antimalarials/pharmacology , Plasmodium falciparum/drug effects , Spiro Compounds/pharmacology , Animals , Anopheles/drug effects , Antimalarials/chemical synthesis , Antimalarials/metabolism , Female , Germ Cells/drug effects , High-Throughput Screening Assays , Humans , Male , Mice , Microsomes, Liver/metabolism , Molecular Structure , Parasitic Sensitivity Tests , Rats , Spiro Compounds/chemical synthesis , Spiro Compounds/metabolism , Structure-Activity Relationship
8.
Sci Rep ; 11(1): 2121, 2021 01 22.
Article in English | MEDLINE | ID: mdl-33483532

ABSTRACT

The spread of Plasmodium falciparum parasites resistant to most first-line antimalarials creates an imperative to enrich the drug discovery pipeline, preferably with curative compounds that can also act prophylactically. We report a phenotypic quantitative high-throughput screen (qHTS), based on concentration-response curves, which was designed to identify compounds active against Plasmodium liver and asexual blood stage parasites. Our qHTS screened over 450,000 compounds, tested across a range of 5 to 11 concentrations, for activity against Plasmodium falciparum asexual blood stages. Active compounds were then filtered for unique structures and drug-like properties and subsequently screened in a P. berghei liver stage assay to identify novel dual-active antiplasmodial chemotypes. Hits from thiadiazine and pyrimidine azepine chemotypes were subsequently prioritized for resistance selection studies, yielding distinct mutations in P. falciparum cytochrome b, a validated antimalarial drug target. The thiadiazine chemotype was subjected to an initial medicinal chemistry campaign, yielding a metabolically stable analog with sub-micromolar potency. Our qHTS methodology and resulting dataset provides a large-scale resource to investigate Plasmodium liver and asexual blood stage parasite biology and inform further research to develop novel chemotypes as causal prophylactic antimalarials.


Subject(s)
Antimalarials/pharmacology , High-Throughput Screening Assays/methods , Liver/drug effects , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Antimalarials/chemistry , Drug Evaluation, Preclinical/methods , Hep G2 Cells , Humans , Liver/parasitology , Malaria, Falciparum/blood , Malaria, Falciparum/parasitology , Molecular Structure , Parasitic Sensitivity Tests , Plasmodium berghei/drug effects , Plasmodium berghei/physiology , Plasmodium falciparum/genetics , Plasmodium falciparum/physiology , Protective Agents/chemistry , Protective Agents/pharmacology , Reproducibility of Results , Structure-Activity Relationship , Thiadiazines/chemistry , Thiadiazines/pharmacology
9.
Proc Natl Acad Sci U S A ; 117(49): 31365-31375, 2020 12 08.
Article in English | MEDLINE | ID: mdl-33229545

ABSTRACT

When Zika virus emerged as a public health emergency there were no drugs or vaccines approved for its prevention or treatment. We used a high-throughput screen for Zika virus protease inhibitors to identify several inhibitors of Zika virus infection. We expressed the NS2B-NS3 Zika virus protease and conducted a biochemical screen for small-molecule inhibitors. A quantitative structure-activity relationship model was employed to virtually screen ∼138,000 compounds, which increased the identification of active compounds, while decreasing screening time and resources. Candidate inhibitors were validated in several viral infection assays. Small molecules with favorable clinical profiles, especially the five-lipoxygenase-activating protein inhibitor, MK-591, inhibited the Zika virus protease and infection in neural stem cells. Members of the tetracycline family of antibiotics were more potent inhibitors of Zika virus infection than the protease, suggesting they may have multiple mechanisms of action. The most potent tetracycline, methacycline, reduced the amount of Zika virus present in the brain and the severity of Zika virus-induced motor deficits in an immunocompetent mouse model. As Food and Drug Administration-approved drugs, the tetracyclines could be quickly translated to the clinic. The compounds identified through our screening paradigm have the potential to be used as prophylactics for patients traveling to endemic regions or for the treatment of the neurological complications of Zika virus infection.


Subject(s)
Antiviral Agents/analysis , Antiviral Agents/pharmacology , Drug Evaluation, Preclinical , High-Throughput Screening Assays , Protease Inhibitors/analysis , Protease Inhibitors/pharmacology , Zika Virus/drug effects , Animals , Antiviral Agents/therapeutic use , Artificial Intelligence , Chlorocebus aethiops , Disease Models, Animal , Immunocompetence , Inhibitory Concentration 50 , Methacycline/pharmacology , Mice, Inbred C57BL , Protease Inhibitors/therapeutic use , Quantitative Structure-Activity Relationship , Small Molecule Libraries , Vero Cells , Zika Virus Infection/drug therapy , Zika Virus Infection/virology
10.
bioRxiv ; 2020 Aug 03.
Article in English | MEDLINE | ID: mdl-32793899

ABSTRACT

Efficient translation of human induced pluripotent stem cells (hiPSCs) depends on implementing scalable cell manufacturing strategies that ensure optimal self-renewal and functional differentiation. Currently, manual culture of hiPSCs is highly variable and labor-intensive posing significant challenges for high-throughput applications. Here, we established a robotic platform and automated all essential steps of hiPSC culture and differentiation under chemically defined conditions. This streamlined approach allowed rapid and standardized manufacturing of billions of hiPSCs that can be produced in parallel from up to 90 different patient-and disease-specific cell lines. Moreover, we established automated multi-lineage differentiation to generate primary embryonic germ layers and more mature phenotypes such as neurons, cardiomyocytes, and hepatocytes. To validate our approach, we carefully compared robotic and manual cell culture and performed molecular and functional cell characterizations (e.g. bulk culture and single-cell transcriptomics, mass cytometry, metabolism, electrophysiology, Zika virus experiments) in order to benchmark industrial-scale cell culture operations towards building an integrated platform for efficient cell manufacturing for disease modeling, drug screening, and cell therapy. Combining stem cell-based models and non-stop robotic cell culture may become a powerful strategy to increase scientific rigor and productivity, which are particularly important during public health emergencies (e.g. opioid crisis, COVID-19 pandemic).

11.
Cancer Res ; 80(10): 1914-1926, 2020 05 15.
Article in English | MEDLINE | ID: mdl-32193286

ABSTRACT

Estrogen receptor α (ERα) is a key transcriptional regulator in the majority of breast cancers. ERα-positive patients are frequently treated with tamoxifen, but resistance is common. In this study, we refined a previously identified 111-gene outcome prediction-classifier, revealing FEN1 as the strongest determining factor in ERα-positive patient prognostication. FEN1 levels were predictive of outcome in tamoxifen-treated patients, and FEN1 played a causal role in ERα-driven cell growth. FEN1 impacted the transcriptional activity of ERα by facilitating coactivator recruitment to the ERα transcriptional complex. FEN1 blockade induced proteasome-mediated degradation of activated ERα, resulting in loss of ERα-driven gene expression and eradicated tumor cell proliferation. Finally, a high-throughput 465,195 compound screen identified a novel FEN1 inhibitor, which effectively blocked ERα function and inhibited proliferation of tamoxifen-resistant cell lines as well as ex vivo-cultured ERα-positive breast tumors. Collectively, these results provide therapeutic proof of principle for FEN1 blockade in tamoxifen-resistant breast cancer. SIGNIFICANCE: These findings show that pharmacologic inhibition of FEN1, which is predictive of outcome in tamoxifen-treated patients, effectively blocks ERα function and inhibits proliferation of tamoxifen-resistant tumor cells.


Subject(s)
Breast Neoplasms/pathology , Drug Resistance, Neoplasm/genetics , Estrogen Receptor alpha/metabolism , Flap Endonucleases/metabolism , Antineoplastic Agents, Hormonal/therapeutic use , Breast Neoplasms/genetics , Cell Line, Tumor , Estrogen Receptor alpha/genetics , Female , Flap Endonucleases/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Tamoxifen/therapeutic use
12.
Oncogene ; 38(19): 3710-3728, 2019 05.
Article in English | MEDLINE | ID: mdl-30674989

ABSTRACT

Melanoma is an aggressive neoplasm with increasing incidence that is classified by the NCI as a recalcitrant cancer, i.e., a cancer with poor prognosis, lacking progress in diagnosis and treatment. In addition to conventional therapy, melanoma treatment is currently based on targeting the BRAF/MEK/ERK signaling pathway and immune checkpoints. As drug resistance remains a major obstacle to treatment success, advanced therapeutic approaches based on novel targets are still urgently needed. We reasoned that the base excision repair enzyme thymine DNA glycosylase (TDG) could be such a target for its dual role in safeguarding the genome and the epigenome, by performing the last of the multiple steps in DNA demethylation. Here we show that TDG knockdown in melanoma cell lines causes cell cycle arrest, senescence, and death by mitotic alterations; alters the transcriptome and methylome; and impairs xenograft tumor formation. Importantly, untransformed melanocytes are minimally affected by TDG knockdown, and adult mice with conditional knockout of Tdg are viable. Candidate TDG inhibitors, identified through a high-throughput fluorescence-based screen, reduced viability and clonogenic capacity of melanoma cell lines and increased cellular levels of 5-carboxylcytosine, the last intermediate in DNA demethylation, indicating successful on-target activity. These findings suggest that TDG may provide critical functions specific to cancer cells that make it a highly suitable anti-melanoma drug target. By potentially disrupting both DNA repair and the epigenetic state, targeting TDG may represent a completely new approach to melanoma therapy.


Subject(s)
Enzyme Inhibitors/pharmacology , Melanoma/pathology , Thymine DNA Glycosylase/genetics , Animals , Cell Cycle/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cytosine/analogs & derivatives , Cytosine/metabolism , DNA Methylation , Female , Gene Expression Regulation, Neoplastic , Humans , Melanoma/drug therapy , Melanoma/genetics , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice, Knockout , Mice, SCID , Mice, Transgenic , Molecular Targeted Therapy/methods , Thymine DNA Glycosylase/antagonists & inhibitors , Thymine DNA Glycosylase/metabolism , Xenograft Model Antitumor Assays
13.
PLoS One ; 14(1): e0210525, 2019.
Article in English | MEDLINE | ID: mdl-30625228

ABSTRACT

Werner syndrome (WS), an autosomal recessive genetic disorder, displays accelerated clinical symptoms of aging leading to a mean lifespan less than 50 years. The WS helicase-nuclease (WRN) is involved in many important pathways including DNA replication, recombination and repair. Replicating cells are dependent on helicase activity, leading to the pursuit of human helicases as potential therapeutic targets for cancer treatment. Small molecule inhibitors of DNA helicases can be used to induce synthetic lethality, which attempts to target helicase-dependent compensatory DNA repair pathways in tumor cells that are already genetically deficient in a specific pathway of DNA repair. Alternatively, helicase inhibitors may be useful as tools to study the specialized roles of helicases in replication and DNA repair. In this study, approximately 350,000 small molecules were screened based on their ability to inhibit duplex DNA unwinding by a catalytically active WRN helicase domain fragment in a high-throughput fluorometric assay to discover new non-covalent small molecule inhibitors of the WRN helicase. Select compounds were screened to exclude ones that inhibited DNA unwinding by other helicases in the screen, bound non-specifically to DNA, acted as irreversible inhibitors, or possessed unfavorable chemical properties. Several compounds were tested for their ability to impair proliferation of cultured tumor cells. We observed that two of the newly identified WRN helicase inhibitors inhibited proliferation of cancer cells in a lineage-dependent manner. These studies represent the first high-throughput screen for WRN helicase inhibitors and the results have implications for anti-cancer strategies targeting WRN in different cancer cells and genetic backgrounds.


Subject(s)
Enzyme Inhibitors/pharmacology , High-Throughput Screening Assays/methods , Small Molecule Libraries/analysis , Small Molecule Libraries/pharmacology , Werner Syndrome Helicase/antagonists & inhibitors , Biocatalysis , Cell Line, Tumor , Cell Proliferation/drug effects , DNA/metabolism , Enzyme Assays , Enzyme Inhibitors/chemistry , Fluorometry , Humans , Inhibitory Concentration 50 , Reproducibility of Results , Small Molecule Libraries/chemistry , Werner Syndrome Helicase/metabolism
14.
ACS Cent Sci ; 4(12): 1727-1741, 2018 Dec 26.
Article in English | MEDLINE | ID: mdl-30648156

ABSTRACT

Natural products and their derivatives continue to be wellsprings of nascent therapeutic potential. However, many laboratories have limited resources for biological evaluation, leaving their previously isolated or synthesized compounds largely or completely untested. To address this issue, the Canvass library of natural products was assembled, in collaboration with academic and industry researchers, for quantitative high-throughput screening (qHTS) across a diverse set of cell-based and biochemical assays. Characterization of the library in terms of physicochemical properties, structural diversity, and similarity to compounds in publicly available libraries indicates that the Canvass library contains many structural elements in common with approved drugs. The assay data generated were analyzed using a variety of quality control metrics, and the resultant assay profiles were explored using statistical methods, such as clustering and compound promiscuity analyses. Individual compounds were then sorted by structural class and activity profiles. Differential behavior based on these classifications, as well as noteworthy activities, are outlined herein. One such highlight is the activity of (-)-2(S)-cathafoline, which was found to stabilize calcium levels in the endoplasmic reticulum. The workflow described here illustrates a pilot effort to broadly survey the biological potential of natural products by utilizing the power of automation and high-throughput screening.

15.
J Biomol Screen ; 21(10): 1125-1131, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27269812

ABSTRACT

We describe a polyacrylamide gel casting cassette that overcomes limitations of commercially available gel electrophoresis equipment. This apparatus molds a single polyacrylamide gel that can evaluate more than 200 samples in parallel, is loaded with a multichannel pipettor, and is flexible with respect to composition of the separating matrix. We demonstrate its use to characterize inhibitors of enzymes that modify protein and nucleic acid substrates. Throughputs of greater than 1000 samples per day were achieved when this system was paired with a quantitative laser-based imaging system, yielding data of remarkable quality.


Subject(s)
Electrophoretic Mobility Shift Assay/methods , Enzyme Inhibitors/isolation & purification , High-Throughput Screening Assays/methods , Small Molecule Libraries/chemistry , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/genetics , Enzyme Inhibitors/chemistry , Humans , Protein Binding , Small Molecule Libraries/analysis
16.
Mol Carcinog ; 55(5): 688-704, 2016 May.
Article in English | MEDLINE | ID: mdl-25865359

ABSTRACT

The apurinic/apyrimidinic endonuclease 1 (APE1) is a protein central to the base excision DNA repair pathway and operates in the modulation of gene expression through redox-dependent and independent mechanisms. Aberrant expression and localization of APE1 in tumors are recurrent hallmarks of aggressiveness and resistance to therapy. We identified and characterized the molecular association between APE1 and nucleophosmin (NPM1), a multifunctional protein involved in the preservation of genome stability and rRNA maturation. This protein-protein interaction modulates subcellular localization and endonuclease activity of APE1. Moreover, we reported a correlation between APE1 and NPM1 expression levels in ovarian cancer, with NPM1 overexpression being a marker of poor prognosis. These observations suggest that tumors that display an augmented APE1/NPM1 association may exhibit increased aggressiveness and resistance. Therefore, targeting the APE1/NPM1 interaction might represent an innovative strategy for the development of anticancer drugs, as tumor cells relying on higher levels of APE1 and NPM1 for proliferation and survival may be more sensitive than untransformed cells. We set up a chemiluminescence-based high-throughput screening assay in order to find small molecules able to interfere with the APE1/NPM1 interaction. This screening led to the identification of a set of bioactive compounds that impair the APE1/NPM1 association in living cells. Interestingly, some of these molecules display anti-proliferative activity and sensitize cells to therapeutically relevant genotoxins. Given the prognostic significance of APE1 and NPM1, these compounds might prove effective in the treatment of tumors that show abundant levels of both proteins, such as ovarian or hepatic carcinomas.


Subject(s)
Antineoplastic Agents/pharmacology , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Neoplasms/metabolism , Nuclear Proteins/metabolism , Small Molecule Libraries/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Female , HeLa Cells , High-Throughput Screening Assays , Humans , MCF-7 Cells , Neoplasms/pathology , Nuclear Proteins/antagonists & inhibitors , Nucleophosmin , Protein Binding/drug effects
17.
Mol Oncol ; 8(7): 1326-38, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24880630

ABSTRACT

FEN1 has key roles in Okazaki fragment maturation during replication, long patch base excision repair, rescue of stalled replication forks, maintenance of telomere stability and apoptosis. FEN1 may be dysregulated in breast and ovarian cancers and have clinicopathological significance in patients. We comprehensively investigated FEN1 mRNA expression in multiple cohorts of breast cancer [training set (128), test set (249), external validation (1952)]. FEN1 protein expression was evaluated in 568 oestrogen receptor (ER) negative breast cancers, 894 ER positive breast cancers and 156 ovarian epithelial cancers. FEN1 mRNA overexpression was highly significantly associated with high grade (p = 4.89 × 10(-57)), high mitotic index (p = 5.25 × 10(-28)), pleomorphism (p = 6.31 × 10(-19)), ER negative (p = 9.02 × 10(-35)), PR negative (p = 9.24 × 10(-24)), triple negative phenotype (p = 6.67 × 10(-21)), PAM50.Her2 (p = 5.19 × 10(-13)), PAM50. Basal (p = 2.7 × 10(-41)), PAM50.LumB (p = 1.56 × 10(-26)), integrative molecular cluster 1 (intClust.1) (p = 7.47 × 10(-12)), intClust.5 (p = 4.05 × 10(-12)) and intClust. 10 (p = 7.59 × 10(-38)) breast cancers. FEN1 mRNA overexpression is associated with poor breast cancer specific survival in univariate (p = 4.4 × 10(-16)) and multivariate analysis (p = 9.19 × 10(-7)). At the protein level, in ER positive tumours, FEN1 overexpression remains significantly linked to high grade, high mitotic index and pleomorphism (ps < 0.01). In ER negative tumours, high FEN1 is significantly associated with pleomorphism, tumour type, lymphovascular invasion, triple negative phenotype, EGFR and HER2 expression (ps < 0.05). In ER positive as well as in ER negative tumours, FEN1 protein overexpression is associated with poor survival in univariate and multivariate analysis (ps < 0.01). In ovarian epithelial cancers, similarly, FEN1 overexpression is associated with high grade, high stage and poor survival (ps < 0.05). We conclude that FEN1 is a promising biomarker in breast and ovarian epithelial cancer.


Subject(s)
Breast Neoplasms/genetics , Breast/pathology , Flap Endonucleases/genetics , Neoplasms, Glandular and Epithelial/genetics , Ovarian Neoplasms/genetics , Ovary/pathology , Aged , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Breast/metabolism , Breast Neoplasms/diagnosis , Breast Neoplasms/pathology , Carcinoma, Ovarian Epithelial , Female , Flap Endonucleases/analysis , Gene Expression Regulation, Neoplastic , Humans , Neoplasms, Glandular and Epithelial/diagnosis , Neoplasms, Glandular and Epithelial/pathology , Ovarian Neoplasms/diagnosis , Ovarian Neoplasms/pathology , Ovary/metabolism , Prognosis
18.
PLoS One ; 8(12): e81667, 2013.
Article in English | MEDLINE | ID: mdl-24349107

ABSTRACT

Following the formation of oxidatively-induced DNA damage, several DNA glycosylases are required to initiate repair of the base lesions that are formed. Recently, NEIL1 and other DNA glycosylases, including OGG1 and NTH1 were identified as potential targets in combination chemotherapeutic strategies. The potential therapeutic benefit for the inhibition of DNA glycosylases was validated by demonstrating synthetic lethality with drugs that are commonly used to limit DNA replication through dNTP pool depletion via inhibition of thymidylate synthetase and dihydrofolate reductase. Additionally, NEIL1-associated synthetic lethality has been achieved in combination with Fanconi anemia, group G. As a prelude to the development of strategies to exploit the potential benefits of DNA glycosylase inhibition, it was necessary to develop a reliable high-throughput screening protocol for this class of enzymes. Using NEIL1 as the proof-of-principle glycosylase, a fluorescence-based assay was developed that utilizes incision of site-specifically modified oligodeoxynucleotides to detect enzymatic activity. This assay was miniaturized to a 1536-well format and used to screen small molecule libraries for inhibitors of the combined glycosylase/AP lyase activities. Among the top hits of these screens were several purine analogs, whose postulated presence in the active site of NEIL1 was consistent with the paradigm of NEIL1 recognition and excision of damaged purines. Although a subset of these small molecules could inhibit other DNA glycosylases that excise oxidatively-induced DNA adducts, they could not inhibit a pyrimidine dimer-specific glycosylase.


Subject(s)
DNA Glycosylases/antagonists & inhibitors , DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Deoxyribonuclease (Pyrimidine Dimer)/antagonists & inhibitors , Enzyme Inhibitors/chemistry , Purines/chemistry , Small Molecule Libraries/chemistry , Animals , DNA Glycosylases/chemistry , DNA Glycosylases/metabolism , DNA-(Apurinic or Apyrimidinic Site) Lyase/chemistry , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Deoxyribonuclease (Pyrimidine Dimer)/chemistry , Deoxyribonuclease (Pyrimidine Dimer)/metabolism , Enzyme Assays , Enzyme Inhibitors/pharmacology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/enzymology , Humans , Kinetics , Mice , Oxidation-Reduction , Protein Binding , Purines/pharmacology , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism , Small Molecule Libraries/pharmacology , Substrate Specificity
19.
PLoS One ; 7(10): e47974, 2012.
Article in English | MEDLINE | ID: mdl-23110144

ABSTRACT

The major human apurinic/apyrimidinic endonuclease APE1 plays a pivotal role in the repair of base damage via participation in the DNA base excision repair (BER) pathway. Increased activity of APE1, often observed in tumor cells, is thought to contribute to resistance to various anticancer drugs, whereas down-regulation of APE1 sensitizes cells to DNA damaging agents. Thus, inhibiting APE1 repair endonuclease function in cancer cells is considered a promising strategy to overcome therapeutic agent resistance. Despite ongoing efforts, inhibitors of APE1 with adequate drug-like properties have yet to be discovered. Using a kinetic fluorescence assay, we conducted a fully-automated high-throughput screen (HTS) of the NIH Molecular Libraries Small Molecule Repository (MLSMR), as well as additional public collections, with each compound tested as a 7-concentration series in a 4 µL reaction volume. Actives identified from the screen were subjected to a panel of confirmatory and counterscreen tests. Several active molecules were identified that inhibited APE1 in two independent assay formats and exhibited potentiation of the genotoxic effect of methyl methanesulfonate with a concomitant increase in AP sites, a hallmark of intracellular APE1 inhibition; a number of these chemotypes could be good starting points for further medicinal chemistry optimization. To our knowledge, this represents the largest-scale HTS to identify inhibitors of APE1, and provides a key first step in the development of novel agents targeting BER for cancer treatment.


Subject(s)
DNA-(Apurinic or Apyrimidinic Site) Lyase/antagonists & inhibitors , Drug Evaluation, Preclinical/methods , Enzyme Inhibitors/pharmacology , Small Molecule Libraries , Cell Survival/drug effects , Cell Survival/genetics , DNA Damage , DNA Repair/drug effects , DNA-(Apurinic or Apyrimidinic Site) Lyase/metabolism , Dose-Response Relationship, Drug , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Methyl Methanesulfonate/antagonists & inhibitors , Methyl Methanesulfonate/pharmacology , Molecular Structure , Structure-Activity Relationship
20.
PLoS One ; 7(10): e45032, 2012.
Article in English | MEDLINE | ID: mdl-23056190

ABSTRACT

Human DNA polymerase kappa (pol κ) is a translesion synthesis (TLS) polymerase that catalyzes TLS past various minor groove lesions including N(2)-dG linked acrolein- and polycyclic aromatic hydrocarbon-derived adducts, as well as N(2)-dG DNA-DNA interstrand cross-links introduced by the chemotherapeutic agent mitomycin C. It also processes ultraviolet light-induced DNA lesions. Since pol κ TLS activity can reduce the cellular toxicity of chemotherapeutic agents and since gliomas overexpress pol κ, small molecule library screens targeting pol κ were conducted to initiate the first step in the development of new adjunct cancer therapeutics. A high-throughput, fluorescence-based DNA strand displacement assay was utilized to screen ∼16,000 bioactive compounds, and the 60 top hits were validated by primer extension assays using non-damaged DNAs. Candesartan cilexetil, manoalide, and MK-886 were selected as proof-of-principle compounds and further characterized for their specificity toward pol κ by primer extension assays using DNAs containing a site-specific acrolein-derived, ring-opened reduced form of γ-HOPdG. Furthermore, candesartan cilexetil could enhance ultraviolet light-induced cytotoxicity in xeroderma pigmentosum variant cells, suggesting its inhibitory effect against intracellular pol κ. In summary, this investigation represents the first high-throughput screening designed to identify inhibitors of pol κ, with the characterization of biochemical and biologically relevant endpoints as a consequence of pol κ inhibition. These approaches lay the foundation for the future discovery of compounds that can be applied to combination chemotherapy.


Subject(s)
DNA Damage , DNA Repair/drug effects , Enzyme Inhibitors/pharmacology , Nucleic Acid Synthesis Inhibitors , Acrolein/metabolism , Benzimidazoles/pharmacology , Biphenyl Compounds/pharmacology , Cell Line, Transformed , Cell Survival/drug effects , Cell Survival/radiation effects , DNA/genetics , DNA/metabolism , DNA Adducts/genetics , DNA Adducts/metabolism , DNA Repair/genetics , DNA-Directed DNA Polymerase/metabolism , Deoxyguanosine/analogs & derivatives , Deoxyguanosine/metabolism , Dose-Response Relationship, Drug , Drug Evaluation, Preclinical/methods , Humans , Indoles/pharmacology , Small Molecule Libraries , Terpenes/pharmacology , Tetrazoles/pharmacology , Ultraviolet Rays
SELECTION OF CITATIONS
SEARCH DETAIL
...