Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 106
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Arterioscler Thromb Vasc Biol ; 43(10): 1808-1817, 2023 10.
Article in English | MEDLINE | ID: mdl-37345522

ABSTRACT

BACKGROUND: Heparin-induced thrombocytopenia (HIT) is a major concern for all individuals that undergo cardiac bypass surgeries or require prolonged heparin exposure. HIT is a life- and limb-threatening adverse drug reaction with an immune response following the formation of ultra-large immune complexes that drive platelet activation through the receptor FcγRIIA. Thrombotic events remain high following the standard of care treatment with anticoagulants, while increasing risk of bleeding complications. This study sought to investigate a novel approach to treatment of HIT. Recent reports demonstrate increased procoagulant activity in HIT; however, these reports required analysis ex vivo, and relevance in vivo remains unclear. METHODS: Using human and mouse model systems, we investigated the cooperativity of PARs (protease-activated receptors) and FcγRIIA in HIT. We challenged humanized FcγRIIA transgenic mice with or without endogenous mouse Par4 (denoted as IIA-Par4+/+ or IIA-Par4-/-, respectively) with a well-established model IgG immune complex (anti [α]-CD9). Furthermore, we assessed the procoagulant phenotype and efficacy to treat HIT utilizing inhibitor of 12-LOX (12[S]-lipoxygenase), VLX-1005, previously reported to decrease platelet activation downstream of FcγRIIA and PAR4, using the triple allele HIT mouse model. RESULTS: IIA-Par4+/+ mice given αCD9 were severely thrombocytopenic, with extensive platelet-fibrin deposition in the lung. In contrast, IIA-Par4-/- mice had negligible thrombocytopenia or pulmonary platelet-fibrin thrombi. We observed that pharmacological inhibition of 12-LOX resulted in a significant reduction in both platelet procoagulant phenotype ex vivo, and thrombocytopenia and thrombosis in our humanized mouse model of HIT in vivo. CONCLUSIONS: These data demonstrate for the first time the need for dual platelet receptor (PAR and FcγRIIA) stimulation for fibrin formation in HIT in vivo. These results extend our understanding of HIT pathophysiology and provide a scientific rationale for targeting the procoagulant phenotype as a possible therapeutic strategy in HIT.


Subject(s)
Thrombocytopenia , Humans , Mice , Animals , Thrombocytopenia/chemically induced , Heparin/adverse effects , Blood Platelets , Anticoagulants/adverse effects , Mice, Transgenic , Phenotype , Fibrin/genetics , Platelet Factor 4/genetics
2.
Biochem Biophys Res Commun ; 590: 49-54, 2022 01 29.
Article in English | MEDLINE | ID: mdl-34971957

ABSTRACT

Acute lymphoblastic leukemia with chromosomal rearrangements involving the mixed-lineage leukemia (MLL) gene (MLL-r ALL) remains an incurable disease. Thus, development of a safe and effective therapeutic agent to treat this disease is crucial to address this unmet medical need. BRD4, a member of the bromodomain and extra-terminal domain (BET) protein family, and cyclic AMP response element binding protein binding protein (CBP) and p300, two paralogous histone acetyltransferases, are all considered cancer drug targets and simultaneous targeting of these proteins may have therapeutic advantages. Here, we demonstrate that a BET/CBP/p300 multi-bromodomain inhibitor, CN470, has anti-tumor activity against MLL-r ALL in vitro and in vivo. CN470, potently inhibited ligand binding to the bromodomains of BRD4, CBP, and p300 and suppressed the growth of MLL-r ALL cell lines and patient-derived cells with MLL rearrangements. CN470 suppressed mRNA and protein expression of MYC and induced apoptosis in MLL-r ALL cells, following a cell cycle arrest in the G1 phase. Moreover, CN470 reduced BRD4 binding to acetylated histone H3. The in vivo effects of CN470 were investigated using SEMLuc/GFP cells expressing luminescent markers in an orthotopic mouse model. Mice administered CN470 daily had prolonged survival compared to the vehicle group. Further, CN470 also showed anti-tumor effects against an MLL-r ALL patient-derived xenograft model. These findings suggest that inhibition of BET/CBP/p300 by the multi-bromodomain inhibitor, CN470, represents a promising therapeutic approach against MLL-r ALL.


Subject(s)
Antineoplastic Agents/pharmacology , E1A-Associated p300 Protein/antagonists & inhibitors , Gene Rearrangement , Histone-Lysine N-Methyltransferase/genetics , Myeloid-Lymphoid Leukemia Protein/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Animals , Apoptosis/drug effects , Apoptosis/genetics , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/genetics , E1A-Associated p300 Protein/metabolism , Gene Expression Regulation, Leukemic/drug effects , Gene Rearrangement/drug effects , Humans , Mice , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , Xenograft Model Antitumor Assays
3.
PLoS Biol ; 16(8): e2006134, 2018 08.
Article in English | MEDLINE | ID: mdl-30080846

ABSTRACT

Cyclic GMP-AMP (cGAMP) synthase (cGAS) stimulator of interferon genes (STING) senses pathogen-derived or abnormal self-DNA in the cytosol and triggers an innate immune defense against microbial infection and cancer. STING agonists induce both innate and adaptive immune responses and are a new class of cancer immunotherapy agents tested in multiple clinical trials. However, STING is commonly silenced in cancer cells via unclear mechanisms, limiting the application of these agonists. Here, we report that the expression of STING is epigenetically suppressed by the histone H3K4 lysine demethylases KDM5B and KDM5C and is activated by the opposing H3K4 methyltransferases. The induction of STING expression by KDM5 blockade triggered a robust interferon response in a cytosolic DNA-dependent manner in breast cancer cells. This response resulted in resistance to infection by DNA and RNA viruses. In human tumors, KDM5B expression is inversely associated with STING expression in multiple cancer types, with the level of intratumoral CD8+ T cells, and with patient survival in cancers with a high level of cytosolic DNA, such as human papilloma virus (HPV)-positive head and neck cancer. These results demonstrate a novel epigenetic regulatory pathway of immune response and suggest that KDM5 demethylases are potential targets for antipathogen treatment and anticancer immunotherapy.


Subject(s)
Histone Demethylases/physiology , Jumonji Domain-Containing Histone Demethylases/physiology , Membrane Proteins/physiology , Nuclear Proteins/physiology , Repressor Proteins/physiology , Cell Line , Cytosol/metabolism , DNA/metabolism , Histone Methyltransferases/physiology , Histones/physiology , Humans , Immunity, Innate/physiology , Immunotherapy , Interferons/metabolism , Interferons/physiology , MCF-7 Cells , Membrane Proteins/metabolism , Signal Transduction
4.
Bioorg Med Chem Lett ; 41: 127974, 2021 06 01.
Article in English | MEDLINE | ID: mdl-33771585

ABSTRACT

Lactate dehydrogenase (LDH) is a critical enzyme in the glycolytic metabolism pathway that is used by many tumor cells. Inhibitors of LDH may be expected to inhibit the metabolic processes in cancer cells and thus selectively delay or inhibit growth in transformed versus normal cells. We have previously disclosed a pyrazole-based series of potent LDH inhibitors with long residence times on the enzyme. Here, we report the elaboration of a new subseries of LDH inhibitors based on those leads. These new compounds potently inhibit both LDHA and LDHB enzymes, and inhibit lactate production in cancer cell lines.


Subject(s)
Aniline Compounds/pharmacology , Antineoplastic Agents/pharmacology , Drug Design , Ethers/pharmacology , L-Lactate Dehydrogenase/antagonists & inhibitors , L-Lactate Dehydrogenase/metabolism , Aniline Compounds/chemistry , Antineoplastic Agents/chemistry , Cell Line, Tumor , Ethers/chemistry , Humans , L-Lactate Dehydrogenase/chemistry
5.
Drug Dev Res ; 82(8): 1235-1246, 2021 12.
Article in English | MEDLINE | ID: mdl-34075610

ABSTRACT

Inhibitors of bromodomain and extra-terminal motif (BET) proteins are emerging epigenetic therapeutics that suppress gene expressions that drive cancer and inflammation. The present study examined anti-inflammatory effects of a quinazoline-based BET inhibitor, CN210, in a murine ileitis model. CN210 was given orally 30 min before and 24 h after a subcutaneous administration of indomethacin. Macroscopic and histological evidences of ileitis, mucosal myeloperoxidase (MPO) activity and cytokine expressions were evaluated 48 h after the indomethacin administration. To further characterize the anti-inflammatory pathways modulated by CN210, its effects on RAW264 cells treated with lipopolysaccharide (LPS) were investigated. Competitive ligand binding and docking studies of CN210 to CREB-binding protein (CBP) and p300 were also performed. Oral administration of CN210 significantly reduced the severity of ileitis, normalized both proinflammatory MPO activity and concomitant cytokine expressions induced by indomethacin administration. Furthermore, CN210 attenuated the expression of cytokines and reversed the activation of nuclear factor κB (NF-κB) and mitogen-activated protein kinases (MAPK) induced by LPS. Competitive ligand binding assays showed that CN210 bound to the bromodomains of two paralogous histone acetyltransferases, CBP and p300, in addition to the bromodomains of BET proteins. Docking studies of CN210 to the bromodomains of CBP and p300 showed a similarity to the binding mode of SGC-CBP30, a specific CBP/p300 inhibitor. CN210 ameliorates indomethacin-induced ileitis by inhibiting the expression of inflammatory cytokines through the attenuation of NF-κB and MAPK pathways. CN210 thus represents a new mode of therapy for non-steroidal anti-inflammatory drug-induced ileitis and inflammatory bowel disease.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Cytokines/antagonists & inhibitors , Ileitis/drug therapy , Indomethacin/adverse effects , Proteins/antagonists & inhibitors , Animals , Cytokines/biosynthesis , E1A-Associated p300 Protein/metabolism , Ileitis/chemically induced , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , NF-kappa B/physiology , Peroxidase/metabolism , Phosphoproteins/metabolism , Quinazolines/pharmacology , RAW 264.7 Cells
6.
Bioorg Med Chem Lett ; 29(10): 1220-1226, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30905542

ABSTRACT

Extensive optimization of quinazoline-based lead 8 is described. The structure-activity relationship studies indicate the S-configuration is preferred for the phenylmorpholine substitution. Together with incorporation of a (2-hydroxyl-2-methylpropyl)pyrazole moiety at the 2-position leads to analogs with comparable potency and marked improvement in the pharmacokinetic profile over our previously reported lead compounds. Further in vivo efficacy studies in Kasumi-1 xenograft mouse model demonstrates that the selected inhibitors are well tolerated and highly efficacious in the inhibition of tumor growth. Additionally, the representative analog 19 also demonstrated significant improvement of arthritis severity in a collagen-induced arthritis (CIA) mouse model. These results indicate potential use of these quinazoline-based BET inhibitors for treatment of cancer and inflammatory diseases. A brief discussion of the co-crystallized structure of 19 with BRD4 (BD1) is also highlighted.


Subject(s)
Anti-Inflammatory Agents/chemistry , Antineoplastic Agents/chemistry , Cell Cycle Proteins/antagonists & inhibitors , Quinazolines/chemistry , Transcription Factors/antagonists & inhibitors , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/therapeutic use , Antineoplastic Agents/pharmacokinetics , Antineoplastic Agents/therapeutic use , Arthritis/drug therapy , Arthritis/pathology , Cell Cycle Proteins/metabolism , Disease Models, Animal , Half-Life , Humans , Kinetics , Mice , Neoplasms/drug therapy , Quinazolines/pharmacokinetics , Quinazolines/therapeutic use , Structure-Activity Relationship , Transcription Factors/metabolism
7.
Proc Natl Acad Sci U S A ; 113(50): 14366-14371, 2016 12 13.
Article in English | MEDLINE | ID: mdl-28182563

ABSTRACT

X-chromosome inactivation is a mechanism of dosage compensation in which one of the two X chromosomes in female mammals is transcriptionally silenced. Once established, silencing of the inactive X (Xi) is robust and difficult to reverse pharmacologically. However, the Xi is a reservoir of >1,000 functional genes that could be potentially tapped to treat X-linked disease. To identify compounds that could reactivate the Xi, here we screened ∼367,000 small molecules in an automated high-content screen using an Xi-linked GFP reporter in mouse fibroblasts. Given the robust nature of silencing, we sensitized the screen by "priming" cells with the DNA methyltransferase inhibitor, 5-aza-2'-deoxycytidine (5azadC). Compounds that elicited GFP activity include VX680, MLN8237, and 5azadC, which are known to target the Aurora kinase and DNA methylation pathways. We demonstrate that the combinations of VX680 and 5azadC, as well as MLN8237 and 5azadC, synergistically up-regulate genes on the Xi. Thus, our work identifies a synergism between the DNA methylation and Aurora kinase pathways as being one of interest for possible pharmacological reactivation of the Xi.


Subject(s)
Aurora Kinases/antagonists & inhibitors , DNA Methylation/drug effects , X Chromosome Inactivation/drug effects , Animals , Aurora Kinase A/antagonists & inhibitors , Aurora Kinase A/genetics , Aurora Kinase B/antagonists & inhibitors , Aurora Kinase B/genetics , Aurora Kinases/genetics , Azacitidine/administration & dosage , Azacitidine/analogs & derivatives , Azepines/administration & dosage , Cell Line , Decitabine , Drug Evaluation, Preclinical , Drug Synergism , Female , Gene Knockdown Techniques , Genes, X-Linked , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , High-Throughput Screening Assays , Mice , Mice, Transgenic , Piperazines/administration & dosage , Pyrimidines/administration & dosage , X Chromosome/drug effects , X Chromosome/genetics
8.
Arterioscler Thromb Vasc Biol ; 37(10): 1828-1839, 2017 10.
Article in English | MEDLINE | ID: mdl-28775075

ABSTRACT

OBJECTIVE: Adequate platelet reactivity is required for maintaining hemostasis. However, excessive platelet reactivity can also lead to the formation of occlusive thrombi. Platelet 12(S)-lipoxygenase (12-LOX), an oxygenase highly expressed in the platelet, has been demonstrated to regulate platelet function and thrombosis ex vivo, supporting a key role for 12-LOX in the regulation of in vivo thrombosis. However, the ability to pharmacologically target 12-LOX in vivo has not been established to date. Here, we studied the effect of the first highly selective 12-LOX inhibitor, ML355, on in vivo thrombosis and hemostasis. APPROACH AND RESULTS: ML355 dose-dependently inhibited human platelet aggregation and 12-LOX oxylipin production, as confirmed by mass spectrometry. Interestingly, the antiplatelet effects of ML355 were reversed after exposure to high concentrations of thrombin in vitro. Ex vivo flow chamber assays confirmed that human platelet adhesion and thrombus formation at arterial shear over collagen were attenuated in whole blood treated with ML355 comparable to aspirin. Oral administration of ML355 in mice showed reasonable plasma drug levels by pharmacokinetic assessment. ML355 treatment impaired thrombus growth and vessel occlusion in FeCl3-induced mesenteric and laser-induced cremaster arteriole thrombosis models in mice. Importantly, hemostatic plug formation and bleeding after treatment with ML355 was minimal in mice in response to laser ablation on the saphenous vein or in a cremaster microvasculature laser-induced rupture model. CONCLUSIONS: Our data strongly support 12-LOX as a key determinant of platelet reactivity in vivo, and inhibition of platelet 12-LOX with ML355 may represent a new class of antiplatelet therapy.


Subject(s)
Hemostasis/drug effects , Lipoxygenase Inhibitors/pharmacology , Platelet Aggregation/drug effects , Sulfonamides/pharmacology , Thrombosis/prevention & control , Animals , Dose-Response Relationship, Drug , Humans , Lipoxygenase Inhibitors/administration & dosage , Lipoxygenase Inhibitors/blood , Mice , Platelet Adhesiveness/drug effects , Sulfonamides/administration & dosage , Sulfonamides/blood , Thrombin/physiology
9.
Bioorg Med Chem Lett ; 28(21): 3483-3488, 2018 11 15.
Article in English | MEDLINE | ID: mdl-30268702

ABSTRACT

A new series of quinazoline-based analogs as potent bromodomain-containing protein 4 (BRD4) inhibitors is described. The structure-activity relationships on 2- and 4-position of quinazoline ring, and the substitution at 6-position that mimic the acetylated lysine are discussed. A co-crystallized structure of 48 (CN750) with BRD4 (BD1) including key inhibitor-protein interactions is also highlighted. Together with preliminary rodent pharmacokinetic results, a new lead (65, CN427) is identified which is suitable for further lead optimization.


Subject(s)
Nuclear Proteins/antagonists & inhibitors , Quinazolines/pharmacology , Transcription Factors/antagonists & inhibitors , Animals , Binding Sites , Cell Cycle Proteins , Cell Line, Tumor , Drug Discovery , Humans , Mice , Microsomes, Liver/metabolism , Molecular Structure , Nuclear Proteins/chemistry , Quinazolines/chemical synthesis , Quinazolines/chemistry , Quinazolines/pharmacokinetics , Structure-Activity Relationship , Transcription Factors/chemistry
10.
Proc Natl Acad Sci U S A ; 111(6): 2349-54, 2014 Feb 11.
Article in English | MEDLINE | ID: mdl-24469833

ABSTRACT

The clinical development of drug combinations is typically achieved through trial-and-error or via insight gained through a detailed molecular understanding of dysregulated signaling pathways in a specific cancer type. Unbiased small-molecule combination (matrix) screening represents a high-throughput means to explore hundreds and even thousands of drug-drug pairs for potential investigation and translation. Here, we describe a high-throughput screening platform capable of testing compounds in pairwise matrix blocks for the rapid and systematic identification of synergistic, additive, and antagonistic drug combinations. We use this platform to define potential therapeutic combinations for the activated B-cell-like subtype (ABC) of diffuse large B-cell lymphoma (DLBCL). We identify drugs with synergy, additivity, and antagonism with the Bruton's tyrosine kinase inhibitor ibrutinib, which targets the chronic active B-cell receptor signaling that characterizes ABC DLBCL. Ibrutinib interacted favorably with a wide range of compounds, including inhibitors of the PI3K-AKT-mammalian target of rapamycin signaling cascade, other B-cell receptor pathway inhibitors, Bcl-2 family inhibitors, and several components of chemotherapy that is the standard of care for DLBCL.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , B-Lymphocytes/immunology , Lymphoma, Large B-Cell, Diffuse/pathology , Pyrazoles/pharmacology , Pyrimidines/pharmacology , Adenine/analogs & derivatives , Cell Line, Tumor , High-Throughput Screening Assays , Humans , Lymphoma, Large B-Cell, Diffuse/immunology , Phosphatidylinositol 3-Kinases/metabolism , Piperidines
11.
Biochemistry ; 55(23): 3329-40, 2016 06 14.
Article in English | MEDLINE | ID: mdl-27226387

ABSTRACT

Pseudomonas aeruginosa is an opportunistic pathogen that can cause nosocomial and chronic infections in immunocompromised patients. P. aeruginosa secretes a lipoxygenase, LoxA, but the biological role of this enzyme is currently unknown. LoxA is poorly similar in sequence to both soybean LOX-1 (s15-LOX-1) and human 15-LOX-1 (37 and 39%, respectively) yet has kinetics comparably fast versus those of s15-LOX-1 (at pH 6.5, Kcat = 181 ± 6 s(-1) and Kcat/KM = 16 ± 2 µM(-1) s(-1)). LoxA is capable of efficiently catalyzing the peroxidation of a broad range of free fatty acid (FA) substrates (e.g., AA and LA) with high positional specificity, indicating a 15-LOX. Its mechanism includes hydrogen atom abstraction [a kinetic isotope effect (KIE) of >30], yet LoxA is a poor catalyst against phosphoester FAs, suggesting that LoxA is not involved in membrane decomposition. LoxA also does not react with 5- or 15-HETEs, indicating poor involvement in lipoxin production. A LOX high-throughput screen of the LOPAC library yielded a variety of low-micromolar inhibitors; however, none selectively targeted LoxA over the human LOX isozymes. With respect to cellular activity, the level of LoxA expression is increased when P. aeruginosa undergoes the transition to a biofilm mode of growth, but LoxA is not required for biofilm growth on abiotic surfaces. However, LoxA does appear to be required for biofilm growth in association with the host airway epithelium, suggesting a role for LoxA in mediating bacterium-host interactions during colonization.


Subject(s)
Arachidonate 15-Lipoxygenase/chemistry , Arachidonate 15-Lipoxygenase/metabolism , Hydroxyeicosatetraenoic Acids/metabolism , Lipoxygenase Inhibitors/metabolism , Pseudomonas aeruginosa/enzymology , Amino Acid Sequence , Animals , Antibody Formation , Arachidonate 15-Lipoxygenase/immunology , Humans , Kinetics , Rabbits , Substrate Specificity
12.
Retrovirology ; 13: 11, 2016 Feb 16.
Article in English | MEDLINE | ID: mdl-26880034

ABSTRACT

BACKGROUND: Nonnucleoside reverse transcriptase inhibitors (NNRTIs) are a class of antiretroviral compounds that bind in an allosteric binding pocket in HIV-1 RT, located about 10 Å from the polymerase active site. Binding of an NNRTI causes structural changes that perturb the alignment of the primer terminus and polymerase active site, preventing viral DNA synthesis. Rilpivirine (RPV) is the most recent NNRTI approved by the FDA, but like all other HIV-1 drugs, suboptimal treatment can lead to the development of resistance. To generate better compounds that could be added to the current HIV-1 drug armamentarium, we have developed several RPV analogs to combat viral variants that are resistant to the available NNRTIs. RESULTS: Using a single-round infection assay, we identified several RPV analogs that potently inhibited a broad panel of NNRTI resistant mutants. Additionally, we determined that several resistant mutants selected by either RPV or Doravirine (DOR) caused only a small increase in susceptibility to the most promising RPV analogs. CONCLUSIONS: The antiviral data suggested that there are RPV analogs that could be candidates for further development as NNRTIs, and one of the most promising compounds was modeled in the NNRTI binding pocket. This model can be used to explain why this compound is broadly effective against the panel of NNRTI resistance mutants.


Subject(s)
Anti-HIV Agents/pharmacology , Drug Resistance, Viral , HIV-1/drug effects , Mutation , Rilpivirine/analogs & derivatives , Rilpivirine/pharmacology , Humans , Microbial Sensitivity Tests
13.
Antimicrob Agents Chemother ; 60(8): 4471-81, 2016 08.
Article in English | MEDLINE | ID: mdl-27161622

ABSTRACT

Filoviruses are highly infectious, and no FDA-approved drug therapy for filovirus infection is available. Most work to find a treatment has involved only a few strains of Ebola virus and testing of relatively small drug libraries or compounds that have shown efficacy against other virus types. Here we report the findings of a high-throughput screening of 319,855 small molecules from the Molecular Libraries Small Molecule Repository library for their activities against Marburg virus and Ebola virus. Nine of the most potent, novel compounds that blocked infection by both viruses were analyzed in detail for their mechanisms of action. The compounds inhibited known key steps in the Ebola virus infection mechanism by blocking either cell surface attachment, macropinocytosis-mediated uptake, or endosomal trafficking. To date, very few specific inhibitors of macropinocytosis have been reported. The 2 novel macropinocytosis inhibitors are more potent inhibitors of Ebola virus infection and less toxic than ethylisopropylamiloride, one commonly accepted macropinocytosis inhibitor. Each compound blocked infection of primary human macrophages, indicating their potential to be developed as new antifiloviral therapies.


Subject(s)
Antiviral Agents/pharmacology , Ebolavirus/drug effects , Marburgvirus/drug effects , Animals , Cell Line , Chlorocebus aethiops , Ebolavirus/genetics , Ebolavirus/metabolism , HeLa Cells , Humans , Marburgvirus/genetics , Marburgvirus/metabolism , Vero Cells
14.
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
15.
Blood ; 124(14): 2271-9, 2014 Oct 02.
Article in English | MEDLINE | ID: mdl-25100742

ABSTRACT

Platelets are essential in maintaining hemostasis following inflammation or injury to the vasculature. Dysregulated platelet activity often results in thrombotic complications leading to myocardial infarction and stroke. Activation of the FcγRIIa receptor leads to immune-mediated thrombosis, which is often life threatening in patients undergoing heparin-induced thrombocytopenia or sepsis. Inhibiting FcγRIIa-mediated activation in platelets has been shown to limit thrombosis and is the principal target for prevention of immune-mediated platelet activation. In this study, we show for the first time that platelet 12(S)-lipoxygenase (12-LOX), a highly expressed oxylipin-producing enzyme in the human platelet, is an essential component of FcγRIIa-mediated thrombosis. Pharmacologic inhibition of 12-LOX in human platelets resulted in significant attenuation of FcγRIIa-mediated aggregation. Platelet 12-LOX was shown to be essential for FcγRIIa-induced phospholipase Cγ2 activity leading to activation of calcium mobilization, Rap1 and protein kinase C activation, and subsequent activation of the integrin αIIbß3. Additionally, platelets from transgenic mice expressing human FcγRIIa but deficient in platelet 12-LOX, failed to form normal platelet aggregates and exhibited deficiencies in Rap1 and αIIbß3 activation. These results support an essential role for 12-LOX in regulating FcγRIIa-mediated platelet function and identifies 12-LOX as a potential therapeutic target to limit immune-mediated thrombosis.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Blood Platelets/metabolism , Receptors, IgG/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/chemistry , Animals , Calcium/metabolism , Enzyme Activation , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Phospholipase C gamma/metabolism , Phosphorylation , Platelet Activation , Platelet Aggregation , Protein Kinase C/metabolism , Signal Transduction , Thrombosis/immunology
16.
Nat Chem Biol ; 10(4): 298-304, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24531842

ABSTRACT

Protein ubiquitination and deubiquitination are central to the control of a large number of cellular pathways and signaling networks in eukaryotes. Although the essential roles of ubiquitination have been established in the eukaryotic DNA damage response, the deubiquitination process remains poorly defined. Chemical probes that perturb the activity of deubiquitinases (DUBs) are needed to characterize the cellular function of deubiquitination. Here we report ML323 (2), a highly potent inhibitor of the USP1-UAF1 deubiquitinase complex with excellent selectivity against human DUBs, deSUMOylase, deneddylase and unrelated proteases. Using ML323, we interrogated deubiquitination in the cellular response to UV- and cisplatin-induced DNA damage and revealed new insights into the requirement of deubiquitination in the DNA translesion synthesis and Fanconi anemia pathways. Moreover, ML323 potentiates cisplatin cytotoxicity in non-small cell lung cancer and osteosarcoma cells. Our findings point to USP1-UAF1 as a key regulator of the DNA damage response and a target for overcoming resistance to the platinum-based anticancer drugs.


Subject(s)
Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Arabidopsis Proteins/antagonists & inhibitors , DNA Damage/physiology , Nuclear Proteins/antagonists & inhibitors , Ubiquitin-Specific Proteases/antagonists & inhibitors , Ubiquitination/drug effects , Algorithms , Butyrates/pharmacology , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Colony-Forming Units Assay , DNA Damage/genetics , DNA, Neoplasm/antagonists & inhibitors , DNA, Neoplasm/biosynthesis , Drug Resistance, Neoplasm , Electrophoresis, Polyacrylamide Gel , Fanconi Anemia/genetics , Fanconi Anemia Complementation Group D2 Protein/antagonists & inhibitors , High-Throughput Screening Assays , Humans , Indicators and Reagents , Phenylurea Compounds/pharmacology , Pimozide/pharmacology , Proliferating Cell Nuclear Antigen/drug effects , Proliferating Cell Nuclear Antigen/metabolism , RNA, Small Interfering/genetics , Recombinant Proteins/chemistry , Recombination, Genetic/drug effects , Sister Chromatid Exchange/drug effects
17.
Bioorg Med Chem ; 24(6): 1183-90, 2016 Mar 15.
Article in English | MEDLINE | ID: mdl-26899595

ABSTRACT

Human reticulocyte 12/15-lipoxygenase (h12/15-LOX) is a lipid-oxidizing enzyme that can directly oxidize lipid membranes in the absence of a phospholipase, leading to a direct attack on organelles, such as the mitochondria. This cytotoxic activity of h12/15-LOX is up-regulated in neurons and endothelial cells after a stroke and thought to contribute to both neuronal cell death and blood-brain barrier leakage. The discovery of inhibitors that selectively target recombinant h12/15-LOX in vitro, as well as possessing activity against the murine ortholog ex vivo, could potentially support a novel therapeutic strategy for the treatment of stroke. Herein, we report a new family of inhibitors discovered in a High Throughput Screen (HTS) that are selective and potent against recombinant h12/15-LOX and cellular mouse 12/15-LOX (m12/15-LOX). MLS000099089 (compound 99089), the parent molecule, exhibits an IC50 potency of 3.4±0.5 µM against h12/15-LOX in vitro and an ex vivo IC50 potency of approximately 10 µM in a mouse neuronal cell line, HT-22. Compound 99089 displays greater than 30-fold selectivity versus h5-LOX and COX-2, 15-fold versus h15-LOX-2 and 10-fold versus h12-LOX, when tested at 20 µM inhibitor concentration. Steady-state inhibition kinetics reveals that the mode of inhibition of 99089 against h12/15-LOX is that of a mixed inhibitor with a Kic of 1.0±0.08 µM and a Kiu of 6.0±3.3 µM. These data indicate that 99089 and related derivatives may serve as a starting point for the development of anti-stroke therapeutics due to their ability to selectively target h12/15-LOX in vitro and m12/15-LOX ex vivo.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Arachidonate 15-Lipoxygenase/metabolism , Lipoxygenase Inhibitors/pharmacology , Animals , Cell Line , Dose-Response Relationship, Drug , High-Throughput Screening Assays , Humans , Lipoxygenase Inhibitors/chemistry , Mice , Models, Molecular , Molecular Structure , Recombinant Proteins/metabolism , Structure-Activity Relationship , Substrate Specificity
18.
Biochem J ; 466(2): 359-67, 2015 Mar 01.
Article in English | MEDLINE | ID: mdl-25495341

ABSTRACT

NIH-12848 (NCGC00012848-02), a putative phosphatidylinositol 5-phosphate 4-kinase γ (PI5P4Kγ) inhibitor, was explored as a tool for investigating this enigmatic, low activity, lipid kinase. PI5P4K assays in vitro showed that NIH-12848 inhibited PI5P4Kγ with an IC50 of approximately 1 µM but did not inhibit the α and ß PI5P4K isoforms at concentrations up to 100 µM. A lack of inhibition of PI5P4Kγ ATPase activity suggested that NIH-12848 does not interact with the enzyme's ATP-binding site and direct exploration of binding using hydrogen-deuterium exchange (HDX)-MS (HDX-MS) revealed the putative PI5P-binding site of PI5P4Kγ to be the likely region of interaction. This was confirmed by a series of mutation experiments which led to the identification of a single PI5P4Kγ amino acid residue that can be mutated to its PI5P4Ks α and ß homologue to render PI5P4Kγ resistant NIH-12848 inhibition. NIH-12848 (10 µM) was applied to cultured mouse principal kidney cortical collecting duct (mpkCCD) cells which, we show, express PI5P4Kγ that increases when the cells grow to confluence and polarize. NIH-12848 inhibited the translocation of Na⁺/K⁺-ATPase to the plasma membrane that occurs when mpkCCD cells grow to confluence and also prevented reversibly their forming of 'domes' on the culture dish. Both these NIH-12848-induced effects were mimicked by specific RNAi knockdown of PI5P4Kγ, but not that of PI5P4Ks α or ß. Overall, the data reveal a probable contribution of PI5P4Kγ to the development and maintenance of epithelial cell functional polarity and show that NIH-12848 is a potentially powerful tool for exploring the cell physiology of PI5P4Ks.


Subject(s)
Cell Differentiation/drug effects , Cell Polarity/drug effects , Enzyme Inhibitors/pharmacology , Kidney Cortex/enzymology , Models, Molecular , Phosphatidylinositol Phosphates/metabolism , Phosphotransferases (Alcohol Group Acceptor)/metabolism , Quinazolines/pharmacology , Thiophenes/pharmacology , Amino Acid Substitution , Animals , Binding Sites , Cell Line , Cell Membrane/drug effects , Cell Membrane/enzymology , Cell Membrane/metabolism , Deuterium Exchange Measurement , Isoenzymes/antagonists & inhibitors , Isoenzymes/genetics , Isoenzymes/metabolism , Kidney Cortex/cytology , Kidney Cortex/drug effects , Kidney Cortex/metabolism , Mice , Mutant Proteins/antagonists & inhibitors , Mutant Proteins/chemistry , Mutant Proteins/metabolism , Phosphatidylinositol Phosphates/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/antagonists & inhibitors , Phosphotransferases (Alcohol Group Acceptor)/chemistry , Phosphotransferases (Alcohol Group Acceptor)/genetics , Protein Conformation , Protein Transport/drug effects , RNA Interference , Recombinant Proteins/chemistry , Recombinant Proteins/metabolism
19.
Diabetologia ; 58(3): 549-57, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25417214

ABSTRACT

AIMS/HYPOTHESIS: Islet inflammation leads to loss of functional pancreatic beta cell mass. Increasing evidence suggests that activation of 12-lipoxygenase leads to inflammatory beta cell loss. This study evaluates new specific small-molecule inhibitors of 12-lipoxygenase for protecting rodent and human beta cells from inflammatory damage. METHODS: Mouse beta cell lines and mouse and human islets were treated with inflammatory cytokines IL-1ß, TNFα and IFNγ in the absence or presence of novel selective 12-lipoxygenase inhibitors. Glucose-stimulated insulin secretion (GSIS), gene expression, cell survival and 12-S-hydroxyeicosatetraenoic acid (12-S-HETE) levels were evaluated using established methods. Pharmacokinetic analysis was performed with the lead inhibitor in CD1 mice. RESULTS: Inflammatory cytokines led to the loss of human beta cell function, elevated cell death, increased inflammatory gene expression and upregulation of 12-lipoxygenase expression and activity (measured by 12-S-HETE generation). Two 12-lipoxygenase inhibitors, Compounds 5 and 9, produced a concentration-dependent reduction of stimulated 12-S-HETE levels. GSIS was preserved in the presence of the 12-lipoxygenase inhibitors. 12-Lipoxygenase inhibition preserved survival of primary mouse and human islets. When administered orally, Compound 5 reduced plasma 12-S-HETE in CD1 mice. Compounds 5 and 9 preserved the function and survival of human donor islets exposed to inflammatory cytokines. CONCLUSIONS/INTERPRETATION: Selective inhibition of 12-lipoxygenase activity confers protection to beta cells during exposure to inflammatory cytokines. These concept validation studies identify 12-lipoxygenase as a promising target in the prevention of loss of functional beta cells in diabetes.


Subject(s)
Arachidonate 12-Lipoxygenase/metabolism , Enzyme Inhibitors/pharmacology , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , 12-Hydroxy-5,8,10,14-eicosatetraenoic Acid/metabolism , Animals , Apoptosis/drug effects , Cell Line , Cell Survival/drug effects , Enzyme-Linked Immunosorbent Assay , Humans , In Vitro Techniques , Interleukin-1beta/metabolism , Male , Mice , Mice, Inbred C57BL , Real-Time Polymerase Chain Reaction , Tumor Necrosis Factor-alpha/metabolism
20.
Anesthesiology ; 122(2): 325-33, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25603205

ABSTRACT

BACKGROUND: The development of novel anesthetics has historically been a process of combined serendipity and empiricism, with most recent new anesthetics developed via modification of existing anesthetic structures. METHODS: Using a novel high-throughput screen employing the fluorescent anesthetic 1-aminoanthracene and apoferritin as a surrogate for on-pathway anesthetic protein target(s), we screened a 350,000 compound library for competition with 1-aminoanthracene-apoferritin binding. Hit compounds meeting structural criteria had their binding affinities for apoferritin quantified with isothermal titration calorimetry and were tested for γ-aminobutyric acid type A receptor binding using a flunitrazepam binding assay. Chemotypes with a strong presence in the top 700 and exhibiting activity via isothermal titration calorimetry were selected for medicinal chemistry optimization including testing for anesthetic potency and toxicity in an in vivo Xenopus laevis tadpole assay. Compounds with low toxicity and high potency were tested for anesthetic potency in mice. RESULTS: From an initial chemical library of more than 350,000 compounds, we identified 2,600 compounds that potently inhibited 1-aminoanthracene binding to apoferritin. A subset of compounds chosen by structural criteria (700) was successfully reconfirmed using the initial assay. Based on a strong presence in both the initial and secondary screens the 6-phenylpyridazin-3(2H)-one chemotype was assessed for anesthetic activity in tadpoles. Medicinal chemistry efforts identified four compounds with high potency and low toxicity in tadpoles, two were found to be effective novel anesthetics in mice. CONCLUSION: The authors demonstrate the first use of a high-throughput screen to successfully identify a novel anesthetic chemotype and show mammalian anesthetic activity for members of that chemotype.


Subject(s)
Anesthetics/chemistry , Anesthetics/pharmacology , High-Throughput Screening Assays/methods , Animals , Calorimetry , Female , Flunitrazepam/metabolism , Larva , Mice , Mice, Inbred C57BL , Phenols/chemistry , Phenols/pharmacology , Receptors, GABA-A/drug effects , Reflex/drug effects , Xenopus
SELECTION OF CITATIONS
SEARCH DETAIL