Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Bioorg Med Chem Lett ; 29(4): 560-562, 2019 02 15.
Article in English | MEDLINE | ID: mdl-30616904

ABSTRACT

Fluorination of metabolic hotspots in a molecule is a common medicinal chemistry strategy to improve in vivo half-life and exposure and, generally, this strategy offers significant benefits. Here, we report the application of this strategy to a series of poly-ADP ribose glycohydrolase (PARG) inhibitors, resulting in unexpected in vivo toxicity which was attributed to this single-atom modification.


Subject(s)
Cyclopropanes/pharmacology , Glycoside Hydrolases/toxicity , Microsomes, Liver/drug effects , Administration, Oral , Animals , Cyclopropanes/administration & dosage , Cyclopropanes/chemistry , Cyclopropanes/pharmacokinetics , Glycoside Hydrolases/administration & dosage , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/pharmacokinetics , Half-Life , Humans , Mice , Microsomes, Liver/metabolism
2.
Anal Biochem ; 503: 58-64, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27036617

ABSTRACT

Poly(ADP-ribose) (PAR) polymers are transient post-translational modifications, and their formation is catalyzed by poly(ADP-ribose) polymerase (PARP) enzymes. A number of PARP inhibitors are in advanced clinical development for BRCA-mutated breast cancer, and olaparib has recently been approved for BRCA-mutant ovarian cancer; however, there has already been evidence of developed resistance mechanisms. Poly(ADP-ribose) glycohydrolase (PARG) catalyzes the hydrolysis of the endo- and exo-glycosidic bonds within the PAR polymers. As an alternative strategy, PARG is a potentially attractive therapeutic target. There is only one PARG gene, compared with 17 known PARP family members, and therefore a PARG inhibitor may have wider application with fewer compensatory mechanisms. Prior to the initiation of this project, there were no known existing cell-permeable small molecule PARG inhibitors for use as tool compounds to assess these hypotheses and no suitable high-throughput screening (HTS)-compatible biochemical assays available to identify start points for a drug discovery project. The development of this newly described high-throughput homogeneous time-resolved fluorescence (HTRF) assay has allowed HTS to proceed and, from this, the identification and advancement of multiple validated series of tool compounds for PARG inhibition.


Subject(s)
Fluorescence , Glycoside Hydrolases/metabolism , High-Throughput Screening Assays/methods , Luminescent Measurements/methods , Cell Line , Enzyme Inhibitors/pharmacology , Glycoside Hydrolases/analysis , Glycoside Hydrolases/antagonists & inhibitors , Humans , Structure-Activity Relationship , Time Factors
3.
Sci Rep ; 10(1): 3836, 2020 03 02.
Article in English | MEDLINE | ID: mdl-32123273

ABSTRACT

There is mounting evidence of androgen receptor signaling inducing genome instability and changing DNA repair capacity in prostate cancer cells. Expression of genes associated with base excision repair (BER) is increased with prostate cancer progression and correlates with poor prognosis. Poly(ADP-ribose) polymerase (PARP) and poly(ADP-ribose) glycohydrolase (PARG) are key enzymes in BER that elongate and degrade PAR polymers on target proteins. While PARP inhibitors have been tested in clinical trials and are a promising therapy for prostate cancer patients with TMPRSS2-ERG fusions and mutations in DNA repair genes, PARG inhibitors have not been evaluated. We show that PARG is a direct androgen receptor (AR) target gene. AR is recruited to the PARG locus and induces PARG expression. Androgen ablation combined with PARG inhibition synergistically reduces BER capacity in independently derived LNCaP and LAPC4 prostate cancer cell lines. A combination of PARG inhibition with androgen ablation or with the DNA damaging drug, temozolomide, significantly reduces cellular proliferation and increases DNA damage. PARG inhibition alters AR transcriptional output without changing AR protein levels. Thus, AR and PARG are engaged in reciprocal regulation suggesting that the success of androgen ablation therapy can be enhanced by PARG inhibition in prostate cancer patients.


Subject(s)
Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Repair/drug effects , Drug Synergism , Gene Expression Regulation, Neoplastic/drug effects , Glycoside Hydrolases/metabolism , Humans , Male , Molecular Targeted Therapy
4.
Nat Struct Mol Biol ; 27(12): 1152-1164, 2020 12.
Article in English | MEDLINE | ID: mdl-33046907

ABSTRACT

The synthesis of poly(ADP-ribose) (PAR) reconfigures the local chromatin environment and recruits DNA-repair complexes to damaged chromatin. PAR degradation by poly(ADP-ribose) glycohydrolase (PARG) is essential for progression and completion of DNA repair. Here, we show that inhibition of PARG disrupts homology-directed repair (HDR) mechanisms that underpin alternative lengthening of telomeres (ALT). Proteomic analyses uncover a new role for poly(ADP-ribosyl)ation (PARylation) in regulating the chromatin-assembly factor HIRA in ALT cancer cells. We show that HIRA is enriched at telomeres during the G2 phase and is required for histone H3.3 deposition and telomere DNA synthesis. Depletion of HIRA elicits systemic death of ALT cancer cells that is mitigated by re-expression of ATRX, a protein that is frequently inactivated in ALT tumors. We propose that PARylation enables HIRA to fulfill its essential role in the adaptive response to ATRX deficiency that pervades ALT cancers.


Subject(s)
DNA, Neoplasm/genetics , Gene Expression Regulation, Neoplastic , Glycoside Hydrolases/genetics , Poly(ADP-ribose) Polymerases/genetics , Protein Processing, Post-Translational , Recombinational DNA Repair , Telomere/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chromatin/metabolism , Chromatin/ultrastructure , DNA Damage , DNA, Neoplasm/metabolism , Epithelial Cells/metabolism , Epithelial Cells/pathology , G2 Phase , Glycoside Hydrolases/metabolism , HeLa Cells , Histone Chaperones/antagonists & inhibitors , Histone Chaperones/genetics , Histone Chaperones/metabolism , Histones/genetics , Histones/metabolism , Humans , Poly ADP Ribosylation , Poly Adenosine Diphosphate Ribose/metabolism , Poly(ADP-ribose) Polymerases/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Signal Transduction , Telomere/ultrastructure , Telomere Homeostasis , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism
5.
Mol Cell Biol ; 26(20): 7397-408, 2006 Oct.
Article in English | MEDLINE | ID: mdl-17015472

ABSTRACT

Apoptosis, induced by a number of death stimuli, is associated with a fragmentation of the mitochondrial network. These morphological changes in mitochondria have been shown to require proteins, such as Drp1 or hFis1, which are involved in regulating the fission of mitochondria. However, the precise role of mitochondrial fission during apoptosis remains elusive. Here we report that inhibiting the fission machinery in Bax/Bak-mediated apoptosis, by down-regulating of Drp1 or hFis1, prevents the fragmentation of the mitochondrial network and partially inhibits the release of cytochrome c from the mitochondria but fails to block the efflux of Smac/DIABLO. In addition, preventing mitochondrial fragmentation does not inhibit cell death induced by Bax/Bak-dependent death stimuli, in contrast to the effects of Bcl-xL or caspase inhibition. Therefore, the fission of mitochondria is a dispensable event in Bax/Bak-dependent apoptosis.


Subject(s)
Apoptosis , Mitochondria/metabolism , bcl-2 Homologous Antagonist-Killer Protein/metabolism , bcl-2-Associated X Protein/metabolism , Animals , Apoptosis Regulatory Proteins , Cell Line , Chlorocebus aethiops , Cytochromes c/metabolism , Dynamins , GTP Phosphohydrolases/metabolism , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Microscopy, Electron, Transmission , Microtubule-Associated Proteins/metabolism , Mitochondria/ultrastructure , Mitochondrial Membranes/metabolism , Mitochondrial Membranes/ultrastructure , Mitochondrial Proteins/metabolism , bcl-2 Homologous Antagonist-Killer Protein/genetics , bcl-2-Associated X Protein/genetics
6.
Cancer Cell ; 35(3): 519-533.e8, 2019 03 18.
Article in English | MEDLINE | ID: mdl-30889383

ABSTRACT

Inhibitors of poly(ADP-ribose) polymerase (PARP) have demonstrated efficacy in women with BRCA-mutant ovarian cancer. However, only 15%-20% of ovarian cancers harbor BRCA mutations, therefore additional therapies are required. Here, we show that a subset of ovarian cancer cell lines and ex vivo models derived from patient biopsies are sensitive to a poly(ADP-ribose) glycohydrolase (PARG) inhibitor. Sensitivity is due to underlying DNA replication vulnerabilities that cause persistent fork stalling and replication catastrophe. PARG inhibition is synthetic lethal with inhibition of DNA replication factors, allowing additional models to be sensitized by CHK1 inhibitors. Because PARG and PARP inhibitor sensitivity are mutually exclusive, our observations demonstrate that PARG inhibitors have therapeutic potential to complement PARP inhibitor strategies in the treatment of ovarian cancer.


Subject(s)
DNA Replication/drug effects , Enzyme Inhibitors/pharmacology , Ovarian Neoplasms/genetics , Cell Line, Tumor , Checkpoint Kinase 1 , Female , Glycoside Hydrolases/antagonists & inhibitors , Humans , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/enzymology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Quinazolinones/pharmacology
7.
Cancer Res ; 79(17): 4491-4502, 2019 09 01.
Article in English | MEDLINE | ID: mdl-31273064

ABSTRACT

Patients with metastatic pancreatic ductal adenocarcinoma (PDAC) have an average survival of less than 1 year, underscoring the importance of evaluating novel targets with matched targeted agents. We recently identified that poly (ADP) ribose glycohydrolase (PARG) is a strong candidate target due to its dependence on the pro-oncogenic mRNA stability factor HuR (ELAVL1). Here, we evaluated PARG as a target in PDAC models using both genetic silencing of PARG and established small-molecule PARG inhibitors (PARGi), PDDX-01/04. Homologous repair-deficient cells compared with homologous repair-proficient cells were more sensitive to PARGi in vitro. In vivo, silencing of PARG significantly decreased tumor growth. PARGi synergized with DNA-damaging agents (i.e., oxaliplatin and 5-fluorouracil), but not with PARPi therapy. Mechanistically, combined PARGi and oxaliplatin treatment led to persistence of detrimental PARylation, increased expression of cleaved caspase-3, and increased γH2AX foci. In summary, these data validate PARG as a relevant target in PDAC and establish current therapies that synergize with PARGi. SIGNIFICANCE: PARG is a potential target in pancreatic cancer as a single-agent anticancer therapy or in combination with current standard of care.


Subject(s)
Carcinoma, Pancreatic Ductal/drug therapy , Glycoside Hydrolases/antagonists & inhibitors , Pancreatic Neoplasms/drug therapy , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , DNA Damage , Enzyme Inhibitors/pharmacology , Female , Gene Silencing , Glycoside Hydrolases/genetics , Humans , Mice, Nude , Molecular Targeted Therapy , Oxaliplatin/pharmacology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Recombinational DNA Repair , Small Molecule Libraries/pharmacology , Xenograft Model Antitumor Assays
8.
DNA Repair (Amst) ; 61: 25-36, 2018 01.
Article in English | MEDLINE | ID: mdl-29179156

ABSTRACT

Upon DNA binding the poly(ADP-ribose) polymerase family of enzymes (PARPs) add multiple ADP-ribose subunits to themselves and other acceptor proteins. Inhibitors of PARPs have become an exciting and real prospect for monotherapy and as sensitizers to ionising radiation (IR). The action of PARPs are reversed by poly(ADP-ribose) glycohydrolase (PARG). Until recently studies of PARG have been limited by the lack of an inhibitor. Here, a first in class, specific, and cell permeable PARG inhibitor, PDD00017273, is shown to radiosensitize. Further, PDD00017273 is compared with the PARP1/2/3 inhibitor olaparib. Both olaparib and PDD00017273 altered the repair of IR-induced DNA damage, resulting in delayed resolution of RAD51 foci compared with control cells. However, only PARG inhibition induced a rapid increase in IR-induced activation of PRKDC (DNA-PK) and perturbed mitotic progression. This suggests that PARG has additional functions in the cell compared with inhibition of PARP1/2/3, likely via reversal of tankyrase activity and/or that inhibiting the removal of poly(ADP-ribose) (PAR) has a different consequence to inhibiting PAR addition. Overall, our data are consistent with previous genetic findings, reveal new insights into the function of PAR metabolism following IR and demonstrate for the first time the therapeutic potential of PARG inhibitors as radiosensitizing agents.


Subject(s)
Antineoplastic Agents/pharmacology , Phthalazines/pharmacology , Piperazines/pharmacology , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Radiation Tolerance/drug effects , Cell Cycle/drug effects , Cell Cycle/genetics , Cell Cycle/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , DNA Breaks, Double-Stranded , Histones/metabolism , Humans , Micronuclei, Chromosome-Defective , Mitosis/genetics , Phenotype , Poly(ADP-ribose) Polymerases/metabolism , Rad51 Recombinase/metabolism , Radiation Tolerance/genetics , Radiation, Ionizing
9.
Cancer Cell ; 33(6): 1078-1093.e12, 2018 06 11.
Article in English | MEDLINE | ID: mdl-29894693

ABSTRACT

Inhibitors of poly(ADP-ribose) (PAR) polymerase (PARPi) have recently entered the clinic for the treatment of homologous recombination (HR)-deficient cancers. Despite the success of this approach, drug resistance is a clinical hurdle, and we poorly understand how cancer cells escape the deadly effects of PARPi without restoring the HR pathway. By combining genetic screens with multi-omics analysis of matched PARPi-sensitive and -resistant Brca2-mutated mouse mammary tumors, we identified loss of PAR glycohydrolase (PARG) as a major resistance mechanism. We also found the presence of PARG-negative clones in a subset of human serous ovarian and triple-negative breast cancers. PARG depletion restores PAR formation and partially rescues PARP1 signaling. Importantly, PARG inactivation exposes vulnerabilities that can be exploited therapeutically.


Subject(s)
Glycoside Hydrolases/genetics , Poly (ADP-Ribose) Polymerase-1/genetics , Poly(ADP-ribose) Polymerase Inhibitors/pharmacology , Synthetic Lethal Mutations , Animals , BRCA1 Protein/genetics , BRCA1 Protein/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Female , Glycoside Hydrolases/antagonists & inhibitors , Glycoside Hydrolases/metabolism , Homologous Recombination/drug effects , Homologous Recombination/genetics , Humans , Mice, 129 Strain , Mice, Knockout , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/metabolism , Poly ADP Ribosylation/drug effects
10.
J Med Chem ; 61(23): 10767-10792, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30403352

ABSTRACT

DNA damage repair enzymes are promising targets in the development of new therapeutic agents for a wide range of cancers and potentially other diseases. The enzyme poly(ADP-ribose) glycohydrolase (PARG) plays a pivotal role in the regulation of DNA repair mechanisms; however, the lack of potent drug-like inhibitors for use in cellular and in vivo models has limited the investigation of its potential as a novel therapeutic target. Using the crystal structure of human PARG in complex with the weakly active and cytotoxic anthraquinone 8a, novel quinazolinedione sulfonamides PARG inhibitors have been identified by means of structure-based virtual screening and library design. 1-Oxetan-3-ylmethyl derivatives 33d and 35d were selected for preliminary investigations in vivo. X-ray crystal structures help rationalize the observed structure-activity relationships of these novel inhibitors.


Subject(s)
DNA Repair , Drug Design , Glycoside Hydrolase Inhibitors/chemistry , Glycoside Hydrolase Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , Quinazolinones/chemistry , Quinazolinones/pharmacology , Administration, Oral , Animals , Biological Availability , Catalytic Domain , Glycoside Hydrolase Inhibitors/administration & dosage , Glycoside Hydrolase Inhibitors/pharmacokinetics , Glycoside Hydrolases/chemistry , Glycoside Hydrolases/metabolism , HeLa Cells , Humans , Male , Mice , Models, Molecular , Quinazolinones/administration & dosage , Quinazolinones/pharmacokinetics , Structure-Activity Relationship
11.
Mol Cell Biol ; 24(2): 537-49, 2004 Jan.
Article in English | MEDLINE | ID: mdl-14701728

ABSTRACT

The Wilms' tumor suppressor protein WT1 is a transcriptional regulator that plays a key role in the development of the kidneys. The transcriptional activation domain of WT1 is subject to regulation by a suppression region within the N terminus of WT1. Using a functional assay, we provide direct evidence that this requires a transcriptional cosuppressor, which we identify as brain acid soluble protein 1 (BASP1). WT1 and BASP1 associate within the nuclei of cells that naturally express both proteins. BASP1 can confer WT1 cosuppressor activity in transfection assays, and elimination of endogenous BASP1 expression augments transcriptional activation by WT1. BASP1 is present in the developing nephron structures of the embryonic kidney and, coincident with that of WT1, its expression is restricted to the highly specialized podocyte cells of the adult kidney. Taken together, our results show that BASP1 is a WT1-associated factor that can regulate WT1 transcriptional activity.


Subject(s)
Nerve Tissue Proteins/metabolism , Repressor Proteins/metabolism , WT1 Proteins/metabolism , Adult , Amino Acid Sequence , Animals , Base Sequence , COS Cells , Cell Line , DNA, Complementary/genetics , Female , Gene Expression Regulation, Developmental , Genes, Wilms Tumor , HeLa Cells , Humans , In Vitro Techniques , Kidney/growth & development , Kidney/metabolism , Male , Membrane Proteins , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , WT1 Proteins/chemistry , WT1 Proteins/genetics
12.
Methods Mol Biol ; 1608: 415-430, 2017.
Article in English | MEDLINE | ID: mdl-28695524

ABSTRACT

Protein ADP-ribosylation is a conserved posttranslational modification that regulates many major cellular functions, such as DNA repair, transcription, translation, signal transduction, stress response, cell division, aging, and cell death. Protein ADP-ribosyl transferases catalyze the transfer of an ADP-ribose (ADPr) group from the ß-nicotinamide adenine dinucleotide (ß-NAD+) cofactor onto a specific target protein with the subsequent release of nicotinamide. ADP-ribosylation leads to changes in protein structure, function, stability, and localization, thus defining the appropriate cellular response. Signaling processes that are mediated by modifications need to be finely tuned and eventually silenced and one of the ways to achieve this is through the action of enzymes that remove (reverse) protein ADP-ribosylation in a timely fashion such as PARG, TARG1, MACROD1, and MACROD2. Here, we describe several basic methods used to study the enzymatic activity of de-ADP-ribosylating enzymes.


Subject(s)
ADP-Ribosylation/physiology , Poly(ADP-ribose) Polymerases/metabolism , ADP-Ribosylation/genetics , Adenosine Diphosphate Ribose/metabolism , Animals , DNA Repair/genetics , DNA Repair/physiology , Glycosylation , Humans , Poly(ADP-ribose) Polymerases/genetics , Protein Processing, Post-Translational/genetics , Protein Processing, Post-Translational/physiology
13.
DNA Repair (Amst) ; 52: 81-91, 2017 04.
Article in English | MEDLINE | ID: mdl-28254358

ABSTRACT

Poly(ADP-ribosylation) of proteins following DNA damage is well studied and the use of poly(ADP-ribose) polymerase (PARP) inhibitors as therapeutic agents is an exciting prospect for the treatment of many cancers. Poly(ADP-ribose) glycohydrolase (PARG) has endo- and exoglycosidase activities which can cleave glycosidic bonds, rapidly reversing the action of PARP enzymes. Like addition of poly(ADP-ribose) (PAR) by PARP, removal of PAR by PARG is also thought to be required for repair of DNA strand breaks and for continued replication at perturbed forks. Here we use siRNA to show a synthetic lethal relationship between PARG and BRCA1, BRCA2, PALB2, FAM175A (ABRAXAS) and BARD1. In addition, we demonstrate that MCF7 cells depleted of these proteins are sensitive to Gallotannin and a novel and specific PARG inhibitor PDD00017273. We confirm that PARG inhibition increases endogenous DNA damage, stalls replication forks and increases homologous recombination, and propose that it is the lack of homologous recombination (HR) proteins at PARG inhibitor-induced stalled replication forks that induces cell death. Interestingly not all genes that are synthetically lethal with PARP result in sensitivity to PARG inhibitors, suggesting that although there is overlap, the functions of PARP and PARG may not be completely identical. These data together add further evidence to the possibility that single treatment therapy with PARG inhibitors could be used for treatment of certain HR deficient tumours and provide insight into the relationship between PARP, PARG and the processes of DNA repair.


Subject(s)
DNA Damage , DNA Repair , DNA Replication/drug effects , Glycoside Hydrolases/antagonists & inhibitors , Homologous Recombination , BRCA1 Protein/metabolism , BRCA2 Protein/metabolism , Carrier Proteins/metabolism , DNA/metabolism , Fanconi Anemia Complementation Group N Protein , Female , Glycoside Hydrolases/metabolism , Glycoside Hydrolases/physiology , Humans , Hydrolyzable Tannins/pharmacology , Nuclear Proteins/metabolism , Tumor Suppressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism
14.
F1000Res ; 5: 962, 2016.
Article in English | MEDLINE | ID: mdl-27703665

ABSTRACT

We present IncucyteDRC, an R package for the analysis of data from live cell imaging cell proliferation experiments carried out on the Essen Biosciences IncuCyte ZOOM instrument. The package provides a simple workflow for summarising data into a form that can be used to calculate dose response curves and EC50 values for small molecule inhibitors. Data from different cell lines, or cell lines grown under different conditions, can be normalised as to their doubling time. A simple graphical web interface, implemented using shiny, is provided for the benefit of non-R users. The software is potentially useful to any research group studying the impact of small molecule inhibitors on cell proliferation using the IncuCyte ZOOM.

15.
F1000Res ; 5: 736, 2016.
Article in English | MEDLINE | ID: mdl-27610220

ABSTRACT

After a DNA damage signal multiple polymers of ADP ribose attached to poly(ADP) ribose (PAR) polymerases (PARPs) are broken down by the enzyme poly(ADP) ribose glycohydrolase (PARG). Inhibition of PARG leads to a failure of DNA repair and small molecule inhibition of PARG has been a goal for many years. To determine whether biochemical inhibitors of PARG are active in cells we have designed an immunofluorescence assay to detect nuclear PAR after DNA damage. This 384-well assay is suitable for medium throughput high-content screening and can detect cell-permeable inhibitors of PARG from nM to µM potency. In addition, the assay has been shown to work in murine cells and in a variety of human cancer cells. Furthermore, the assay is suitable for detecting the DNA damage response induced by treatment with temozolomide and methylmethane sulfonate (MMS). Lastly, the assay has been shown to be robust over a period of several years.

16.
ACS Chem Biol ; 11(11): 3179-3190, 2016 11 18.
Article in English | MEDLINE | ID: mdl-27689388

ABSTRACT

The enzyme poly(ADP-ribose) glycohydrolase (PARG) performs a critical role in the repair of DNA single strand breaks (SSBs). However, a detailed understanding of its mechanism of action has been hampered by a lack of credible, cell-active chemical probes. Herein, we demonstrate inhibition of PARG with a small molecule, leading to poly(ADP-ribose) (PAR) chain persistence in intact cells. Moreover, we describe two advanced, and chemically distinct, cell-active tool compounds with convincing on-target pharmacology and selectivity. Using one of these tool compounds, we demonstrate pharmacology consistent with PARG inhibition. Further, while the roles of PARG and poly(ADP-ribose) polymerase (PARP) are closely intertwined, we demonstrate that the pharmacology of a PARG inhibitor differs from that observed with the more thoroughly studied PARP inhibitor olaparib. We believe that these tools will facilitate a wider understanding of this important component of DNA repair and may enable the development of novel therapeutic agents exploiting the critical dependence of tumors on the DNA damage response (DDR).


Subject(s)
DNA Repair , Glycoside Hydrolases/chemistry , Molecular Probes/chemistry , Phthalazines/pharmacology , Piperazines/pharmacology , Enzyme Inhibitors/pharmacology , Glycoside Hydrolases/antagonists & inhibitors , HeLa Cells , Humans , Surface Plasmon Resonance
17.
FEBS Lett ; 538(1-3): 53-9, 2003 Mar 13.
Article in English | MEDLINE | ID: mdl-12633852

ABSTRACT

Mitochondrial fusion is a poorly characterized process which has mainly been studied in yeast and Drosophila but is thought to occur in all eukaryotes. Until now, there was only indirect evidence to support such a process in mammalian cells. In this study, using a cell fusion system, we found that mitochondrial fusion occurs rapidly in mammalian cells and is completed in less than 24 h. We report that the fusion of mitochondria requires an intact mitochondrial inner membrane potential but is independent of a functional cytoskeleton.


Subject(s)
Intracellular Membranes/physiology , Membrane Fusion , Membrane Potentials , Mitochondria/physiology , Actins/physiology , Animals , Cells, Cultured , Dogs , HeLa Cells , Humans , Tubulin/physiology
19.
J Med Chem ; 55(9): 4431-45, 2012 May 10.
Article in English | MEDLINE | ID: mdl-22506561

ABSTRACT

Novel derivatives of the steroid DHEA 1, a known uncompetitive inhibitor of G6PD, were designed, synthesized, and tested for their ability to inhibit this dehydrogenase enzyme. Several compounds with approximately 10-fold improved potency in an enzyme assay were identified, and this improved activity translated to efficacy in a cellular assay. The SAR for steroid inhibition of G6PD has been substantially developed; the 3ß-alcohol can be replaced with 3ß-H-bond donors such as sulfamide, sulfonamide, urea, and carbamate. Improved potency was achieved by replacing the androstane nucleus with a pregnane nucleus, provided a ketone at C-20 is present. For pregnan-20-ones incorporation of a 21-hydroxyl group is often beneficial. The novel compounds generally have good physicochemical properties and satisfactory in vitro DMPK parameters. These derivatives may be useful for examining the role of G6PD inhibition in cells and will assist the future design of more potent steroid inhibitors with potential therapeutic utility.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Glucosephosphate Dehydrogenase/antagonists & inhibitors , Pregnanes/chemistry , Pregnanes/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Cell Survival/drug effects , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/pharmacokinetics , Glucosephosphate Dehydrogenase/metabolism , HEK293 Cells , Humans , Inhibitory Concentration 50 , Magnetic Resonance Spectroscopy , Mass Spectrometry , Pregnanes/chemical synthesis , Pregnanes/pharmacokinetics , Structure-Activity Relationship
20.
Dev Cell ; 15(3): 341-343, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18804432

ABSTRACT

Reporting in Molecular Cell, Sheridan et al. (2008) and Breckenridge et al. (2008) show that mitochondrial fragmentation is not required to induce cell death. Meanwhile, Yamaguchi et al. show that proapoptotic Bcl-2 family members promote cytochrome c mobilization through Opa1-mediated cristae remodeling. Therefore, the connection between mitochondrial structure and apoptosis is more complex than previously imagined.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , Cytochromes c/metabolism , Cytoskeletal Proteins/metabolism , Mitochondria/ultrastructure , Proto-Oncogene Proteins c-bcl-2/genetics
SELECTION OF CITATIONS
SEARCH DETAIL