Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 107
Filtrar
1.
bioRxiv ; 2024 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-39091839

RESUMEN

Type 1 diabetes (T1D) is characterized by the autoimmune destruction of insulin-producing ß cells and involves an interplay between ß cells and cells of the innate and adaptive immune systems. We investigated the therapeutic potential of targeting 12-lipoxygenase (12-LOX), an enzyme implicated in inflammatory pathways in ß cells and macrophages, using a mouse model in which the endogenous mouse Alox15 gene is replaced by the human ALOX12 gene. Our findings demonstrate that VLX-1005, a potent 12-LOX inhibitor, effectively delays the onset of autoimmune diabetes in human gene replacement non-obese diabetic (NOD) mice. By spatial proteomics analysis, VLX-1005 treatment resulted in marked reductions in infiltrating T and B cells and macrophages with accompanying increases in immune checkpoint molecules PD-L1 and PD-1, suggesting a shift towards an immune-suppressive microenvironment. RNA sequencing analysis of isolated islets from inhibitor-treated mice revealed significant alteration of cytokine-responsive pathways. RNA sequencing of polarized proinflammatory macrophages showed that VLX-1005 significantly reduced the interferon response. Our studies demonstrate that the ALOX12 human replacement gene mouse provides a platform for the preclinical evaluation of LOX inhibitors and supports VLX-1005 as an inhibitor of human 12-LOX that engages the enzymatic target and alters the inflammatory phenotypes of islets and macrophages to promote the delay of autoimmune diabetes.

2.
Arterioscler Thromb Vasc Biol ; 43(10): 1808-1817, 2023 10.
Artículo en Inglés | MEDLINE | ID: mdl-37345522

RESUMEN

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.


Asunto(s)
Trombocitopenia , Humanos , Ratones , Animales , Trombocitopenia/inducido químicamente , Heparina/efectos adversos , Plaquetas , Anticoagulantes/efectos adversos , Ratones Transgénicos , Fenotipo , Fibrina/genética , Factor Plaquetario 4/genética
3.
Biochem Biophys Res Commun ; 590: 49-54, 2022 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-34971957

RESUMEN

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.


Asunto(s)
Antineoplásicos/farmacología , Proteína p300 Asociada a E1A/antagonistas & inhibidores , Reordenamiento Génico , N-Metiltransferasa de Histona-Lisina/genética , Proteína de la Leucemia Mieloide-Linfoide/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Animales , Apoptosis/efectos de los fármacos , Apoptosis/genética , Puntos de Control del Ciclo Celular/efectos de los fármacos , Puntos de Control del Ciclo Celular/genética , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Proteína p300 Asociada a E1A/metabolismo , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Reordenamiento Génico/efectos de los fármacos , Humanos , Ratones , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia , Ensayos Antitumor por Modelo de Xenoinjerto
4.
Drug Dev Res ; 82(8): 1235-1246, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34075610

RESUMEN

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.


Asunto(s)
Antiinflamatorios/farmacología , Citocinas/antagonistas & inhibidores , Ileítis/tratamiento farmacológico , Indometacina/efectos adversos , Proteínas/antagonistas & inhibidores , Animales , Citocinas/biosíntesis , Proteína p300 Asociada a E1A/metabolismo , Ileítis/inducido químicamente , Masculino , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/fisiología , Peroxidasa/metabolismo , Fosfoproteínas/metabolismo , Quinazolinas/farmacología , Células RAW 264.7
5.
Bioorg Med Chem Lett ; 41: 127974, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33771585

RESUMEN

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.


Asunto(s)
Compuestos de Anilina/farmacología , Antineoplásicos/farmacología , Diseño de Fármacos , Éteres/farmacología , L-Lactato Deshidrogenasa/antagonistas & inhibidores , L-Lactato Deshidrogenasa/metabolismo , Compuestos de Anilina/química , Antineoplásicos/química , Línea Celular Tumoral , Éteres/química , Humanos , L-Lactato Deshidrogenasa/química
6.
Sci Rep ; 11(1): 2121, 2021 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-33483532

RESUMEN

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.


Asunto(s)
Antimaláricos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Hígado/efectos de los fármacos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Antimaláricos/química , Evaluación Preclínica de Medicamentos/métodos , Células Hep G2 , Humanos , Hígado/parasitología , Malaria Falciparum/sangre , Malaria Falciparum/parasitología , Estructura Molecular , Pruebas de Sensibilidad Parasitaria , Plasmodium berghei/efectos de los fármacos , Plasmodium berghei/fisiología , Plasmodium falciparum/genética , Plasmodium falciparum/fisiología , Sustancias Protectoras/química , Sustancias Protectoras/farmacología , Reproducibilidad de los Resultados , Relación Estructura-Actividad , Tiadiazinas/química , Tiadiazinas/farmacología
7.
J Med Chem ; 63(19): 10984-11011, 2020 10 08.
Artículo en Inglés | MEDLINE | ID: mdl-32902275

RESUMEN

Lactate dehydrogenase (LDH) catalyzes the conversion of pyruvate to lactate, with concomitant oxidation of reduced nicotinamide adenine dinucleotide as the final step in the glycolytic pathway. Glycolysis plays an important role in the metabolic plasticity of cancer cells and has long been recognized as a potential therapeutic target. Thus, potent, selective inhibitors of LDH represent an attractive therapeutic approach. However, to date, pharmacological agents have failed to achieve significant target engagement in vivo, possibly because the protein is present in cells at very high concentrations. We report herein a lead optimization campaign focused on a pyrazole-based series of compounds, using structure-based design concepts, coupled with optimization of cellular potency, in vitro drug-target residence times, and in vivo PK properties, to identify first-in-class inhibitors that demonstrate LDH inhibition in vivo. The lead compounds, named NCATS-SM1440 (43) and NCATS-SM1441 (52), possess desirable attributes for further studying the effect of in vivo LDH inhibition.


Asunto(s)
Inhibidores Enzimáticos/farmacología , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Pirazoles/farmacología , Animales , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacocinética , Semivida , Humanos , Ratones , Relación Estructura-Actividad , Ensayos Antitumor por Modelo de Xenoinjerto
8.
Cell Rep ; 30(6): 1798-1810.e4, 2020 02 11.
Artículo en Inglés | MEDLINE | ID: mdl-32049011

RESUMEN

The reliance of many cancers on aerobic glycolysis has stimulated efforts to develop lactate dehydrogenase (LDH) inhibitors. However, despite significant efforts, LDH inhibitors (LDHi) with sufficient specificity and in vivo activity to determine whether LDH is a feasible drug target are lacking. We describe an LDHi with potent, on-target, in vivo activity. Using hyperpolarized magnetic resonance spectroscopic imaging (HP-MRSI), we demonstrate in vivo LDH inhibition in two glycolytic cancer models, MIA PaCa-2 and HT29, and we correlate depth and duration of LDH inhibition with direct anti-tumor activity. HP-MRSI also reveals a metabolic rewiring that occurs in vivo within 30 min of LDH inhibition, wherein pyruvate in a tumor is redirected toward mitochondrial metabolism. Using HP-MRSI, we show that inhibition of mitochondrial complex 1 rapidly redirects tumor pyruvate toward lactate. Inhibition of both mitochondrial complex 1 and LDH suppresses metabolic plasticity, causing metabolic quiescence in vitro and tumor growth inhibition in vivo.


Asunto(s)
Quimioterapia Combinada/métodos , L-Lactato Deshidrogenasa/antagonistas & inhibidores , Neoplasias/inmunología , Animales , Humanos , Ratones , Neoplasias/tratamiento farmacológico
9.
Bioorg Med Chem Lett ; 29(10): 1220-1226, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30905542

RESUMEN

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.


Asunto(s)
Antiinflamatorios/química , Antineoplásicos/química , Proteínas de Ciclo Celular/antagonistas & inhibidores , Quinazolinas/química , Factores de Transcripción/antagonistas & inhibidores , Animales , Antiinflamatorios/farmacocinética , Antiinflamatorios/uso terapéutico , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Artritis/tratamiento farmacológico , Artritis/patología , Proteínas de Ciclo Celular/metabolismo , Modelos Animales de Enfermedad , Semivida , Humanos , Cinética , Ratones , Neoplasias/tratamiento farmacológico , Quinazolinas/farmacocinética , Quinazolinas/uso terapéutico , Relación Estructura-Actividad , Factores de Transcripción/metabolismo
11.
PLoS One ; 14(1): e0210525, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30625228

RESUMEN

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.


Asunto(s)
Inhibidores Enzimáticos/farmacología , Ensayos Analíticos de Alto Rendimiento/métodos , Bibliotecas de Moléculas Pequeñas/análisis , Bibliotecas de Moléculas Pequeñas/farmacología , Helicasa del Síndrome de Werner/antagonistas & inhibidores , Biocatálisis , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , ADN/metabolismo , Pruebas de Enzimas , Inhibidores Enzimáticos/química , Fluorometría , Humanos , Concentración 50 Inhibidora , Reproducibilidad de los Resultados , Bibliotecas de Moléculas Pequeñas/química , Helicasa del Síndrome de Werner/metabolismo
12.
Mol Cancer Ther ; 18(2): 421-436, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30420565

RESUMEN

Systematic approaches for accurate repurposing of targeted therapies are needed. We developed and aimed to biologically validate our therapy predicting tool (TPT) for the repurposing of targeted therapies for specific tumor types by testing the role of Bromodomain and Extra-Terminal motif inhibitors (BETi) in inhibiting BRD4 function and downregulating Notch3 signaling in ovarian cancer.Utilizing established ovarian cancer preclinical models, we carried out in vitro and in vivo studies with clinically relevant BETis to determine their therapeutic effect and impact on Notch3 signaling.Treatment with BETis or siRNA-mediated BRD4 knockdown resulted in decreased cell viability, reduced cell proliferation, and increased cell apoptosis in vitro. In vivo studies with orthotopic mouse models demonstrated that treatment with BETi decreased tumor growth. In addition, knockdown of BRD4 with doxycycline-inducible shRNA increased survival up to 50% (P < 0.001). Treatment with either BETis or BRD4 siRNA decreased Notch3 expression both in vitro and in vivo BRD4 inhibition also decreased the expression of NOTCH3 targets, including HES1 Chromatin immunoprecipitation revealed that BRD4 was present at the NOTCH3 promoter.Our findings provide biological validation for the TPT by demonstrating that BETis can be an effective therapeutic agent for ovarian cancer by downregulating Notch3 expression.The TPT could rapidly identify candidate drugs for ovarian or other cancers along with novel companion biomarkers.


Asunto(s)
Acetamidas/administración & dosificación , Azepinas/administración & dosificación , Proteínas Nucleares/metabolismo , Neoplasias Ováricas/tratamiento farmacológico , Receptor Notch3/metabolismo , Factores de Transcripción/metabolismo , Acetamidas/farmacología , Animales , Azepinas/farmacología , Proteínas de Ciclo Celular , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Proteínas Nucleares/genética , Neoplasias Ováricas/genética , Neoplasias Ováricas/metabolismo , Factores de Transcripción/genética , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cancer Cell ; 34(6): 939-953.e9, 2018 12 10.
Artículo en Inglés | MEDLINE | ID: mdl-30472020

RESUMEN

Members of the KDM5 histone H3 lysine 4 demethylase family are associated with therapeutic resistance, including endocrine resistance in breast cancer, but the underlying mechanism is poorly defined. Here we show that genetic deletion of KDM5A/B or inhibition of KDM5 activity increases sensitivity to anti-estrogens by modulating estrogen receptor (ER) signaling and by decreasing cellular transcriptomic heterogeneity. Higher KDM5B expression levels are associated with higher transcriptomic heterogeneity and poor prognosis in ER+ breast tumors. Single-cell RNA sequencing, cellular barcoding, and mathematical modeling demonstrate that endocrine resistance is due to selection for pre-existing genetically distinct cells, while KDM5 inhibitor resistance is acquired. Our findings highlight the importance of cellular phenotypic heterogeneity in therapeutic resistance and identify KDM5A/B as key regulators of this process.


Asunto(s)
Neoplasias de la Mama/genética , Resistencia a Antineoplásicos/genética , Histona Demetilasas con Dominio de Jumonji/genética , Proteínas Nucleares/genética , Proteínas Represoras/genética , Proteína 2 de Unión a Retinoblastoma/genética , Transcriptoma/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Estradiol/farmacología , Moduladores de los Receptores de Estrógeno/farmacología , Femenino , Fulvestrant/farmacología , Heterogeneidad Genética , Humanos , Histona Demetilasas con Dominio de Jumonji/metabolismo , Células MCF-7 , Proteínas Nucleares/metabolismo , Proteínas Represoras/metabolismo , Proteína 2 de Unión a Retinoblastoma/metabolismo , Transcriptoma/efectos de los fármacos , Secuenciación del Exoma/métodos
14.
Elife ; 72018 11 27.
Artículo en Inglés | MEDLINE | ID: mdl-30479275

RESUMEN

Lecithin:cholesterol acyltransferase (LCAT) and LCAT-activating compounds are being investigated as treatments for coronary heart disease (CHD) and familial LCAT deficiency (FLD). Herein we report the crystal structure of human LCAT in complex with a potent piperidinylpyrazolopyridine activator and an acyl intermediate-like inhibitor, revealing LCAT in an active conformation. Unlike other LCAT activators, the piperidinylpyrazolopyridine activator binds exclusively to the membrane-binding domain (MBD). Functional studies indicate that the compound does not modulate the affinity of LCAT for HDL, but instead stabilizes residues in the MBD and facilitates channeling of substrates into the active site. By demonstrating that these activators increase the activity of an FLD variant, we show that compounds targeting the MBD have therapeutic potential. Our data better define the substrate binding site of LCAT and pave the way for rational design of LCAT agonists and improved biotherapeutics for augmenting or restoring reverse cholesterol transport in CHD and FLD patients.


Asunto(s)
HDL-Colesterol/metabolismo , Fosfatidilcolina-Esterol O-Aciltransferasa/metabolismo , Dominio Catalítico , Activadores de Enzimas/química , Activadores de Enzimas/farmacología , Estabilidad de Enzimas/efectos de los fármacos , Células HEK293 , Humanos , Lípidos de la Membrana/metabolismo , Mutación/genética , Fosfatidilcolina-Esterol O-Aciltransferasa/química , Conformación Proteica , Electricidad Estática , Relación Estructura-Actividad
15.
J Med Chem ; 61(23): 10588-10601, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30392349

RESUMEN

The active sites of hundreds of human α-ketoglutarate (αKG) and Fe(II)-dependent dioxygenases are exceedingly well preserved, which challenges the design of selective inhibitors. We identified a noncatalytic cysteine (Cys481 in KDM5A) near the active sites of KDM5 histone H3 lysine 4 demethylases, which is absent in other histone demethylase families, that could be explored for interaction with the cysteine-reactive electrophile acrylamide. We synthesized analogs of a thienopyridine-based inhibitor chemotype, namely, 2-((3-aminophenyl)(2-(piperidin-1-yl)ethoxy)methyl)thieno[3,2- b]pyridine-7-carboxylic acid (N70) and a derivative containing a (dimethylamino)but-2-enamido)phenyl moiety (N71) designed to form a covalent interaction with Cys481. We characterized the inhibitory and binding activities against KDM5A and determined the cocrystal structures of the catalytic domain of KDM5A in complex with N70 and N71. Whereas the noncovalent inhibitor N70 displayed αKG-competitive inhibition that could be reversed after dialysis, inhibition by N71 was dependent on enzyme concentration and persisted even after dialysis, consistent with covalent modification.


Asunto(s)
Diseño de Fármacos , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Proteína 2 de Unión a Retinoblastoma/antagonistas & inhibidores , Acrilamida/química , Línea Celular , Humanos , Modelos Moleculares , Conformación Proteica , Proteína 2 de Unión a Retinoblastoma/química
16.
Bioorg Med Chem Lett ; 28(21): 3483-3488, 2018 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-30268702

RESUMEN

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.


Asunto(s)
Proteínas Nucleares/antagonistas & inhibidores , Quinazolinas/farmacología , Factores de Transcripción/antagonistas & inhibidores , Animales , Sitios de Unión , Proteínas de Ciclo Celular , Línea Celular Tumoral , Descubrimiento de Drogas , Humanos , Ratones , Microsomas Hepáticos/metabolismo , Estructura Molecular , Proteínas Nucleares/química , Quinazolinas/síntesis química , Quinazolinas/química , Quinazolinas/farmacocinética , Relación Estructura-Actividad , Factores de Transcripción/química
17.
Pain ; 159(12): 2620-2629, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-30130298

RESUMEN

Although nonsteroidal anti-inflammatory drugs are the first line of therapeutics for the treatment of mild to moderate somatic pain, they are not generally considered to be effective for neuropathic pain. In the current study, direct activation of spinal Toll-like 4 receptors (TLR4) by the intrathecal (IT) administration of KDO2 lipid A (KLA), the active component of lipopolysaccharide, elicits a robust tactile allodynia that is unresponsive to cyclooxygenase inhibition, despite elevated expression of cyclooxygenase metabolites in the spinal cord. Intrathecal KLA increases 12-lipoxygenase-mediated hepoxilin production in the lumbar spinal cord, concurrent with expression of the tactile allodynia. The TLR4-induced hepoxilin production was also observed in primary spinal microglia, but not in astrocytes, and was accompanied by increased microglial expression of the 12/15-lipoxygenase enzyme 15-LOX-1. Intrathecal KLA-induced tactile allodynia was completely prevented by spinal pretreatment with the 12/15-lipoxygenase inhibitor CDC or a selective antibody targeting rat 15-LOX-1. Similarly, pretreatment with the selective inhibitors ML127 or ML351 both reduced activity of the rat homolog of 15-LOX-1 heterologously expressed in HEK-293T cells and completely abrogated nonsteroidal anti-inflammatory drug-unresponsive allodynia in vivo after IT KLA. Finally, spinal 12/15-lipoxygenase inhibition by nordihydroguaiaretic acid (NDGA) both prevents phase II formalin flinching and reverses formalin-induced persistent tactile allodynia. Taken together, these findings suggest that spinal TLR4-mediated hyperpathic states are mediated at least in part through activation of microglial 15-LOX-1.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Hiperalgesia/tratamiento farmacológico , Hiperalgesia/metabolismo , Lipooxigenasas/uso terapéutico , Neuroglía/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Ácido 8,11,14-Eicosatrienoico/análogos & derivados , Ácido 8,11,14-Eicosatrienoico/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Cromatografía Liquida , Inhibidores Enzimáticos/uso terapéutico , Lipopolisacáridos/toxicidad , Masculino , Espectrometría de Masas , Estimulación Física/efectos adversos , ARN Mensajero , Ratas , Ratas Sprague-Dawley , Médula Espinal/citología , Receptor Toll-Like 4/antagonistas & inhibidores , Transfección
18.
PLoS Biol ; 16(8): e2006134, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30080846

RESUMEN

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.


Asunto(s)
Histona Demetilasas/fisiología , Histona Demetilasas con Dominio de Jumonji/fisiología , Proteínas de la Membrana/fisiología , Proteínas Nucleares/fisiología , Proteínas Represoras/fisiología , Línea Celular , Citosol/metabolismo , ADN/metabolismo , Histona Metiltransferasas/fisiología , Histonas/fisiología , Humanos , Inmunidad Innata/fisiología , Inmunoterapia , Interferones/metabolismo , Interferones/fisiología , Células MCF-7 , Proteínas de la Membrana/metabolismo , Transducción de Señal
20.
J Med Chem ; 61(11): 4883-4903, 2018 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-29767973

RESUMEN

Aldehyde dehydrogenases (ALDHs) are responsible for the metabolism of aldehydes (exogenous and endogenous) and possess vital physiological and toxicological functions in areas such as CNS, inflammation, metabolic disorders, and cancers. Overexpression of certain ALDHs (e.g., ALDH1A1) is an important biomarker in cancers and cancer stem cells (CSCs) indicating the potential need for the identification and development of small molecule ALDH inhibitors. Herein, a newly designed series of quinoline-based analogs of ALDH1A1 inhibitors is described. Extensive medicinal chemistry optimization and biological characterization led to the identification of analogs with significantly improved enzymatic and cellular ALDH inhibition. Selected analogs, e.g., 86 (NCT-505) and 91 (NCT-506), demonstrated target engagement in a cellular thermal shift assay (CETSA), inhibited the formation of 3D spheroid cultures of OV-90 cancer cells, and potentiated the cytotoxicity of paclitaxel in SKOV-3-TR, a paclitaxel resistant ovarian cancer cell line. Lead compounds also exhibit high specificity over other ALDH isozymes and unrelated dehydrogenases. The in vitro ADME profiles and pharmacokinetic evaluation of selected analogs are also highlighted.


Asunto(s)
Aldehído Deshidrogenasa/antagonistas & inhibidores , Descubrimiento de Drogas , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/farmacología , Quinolinas/química , Quinolinas/farmacología , Administración Oral , Familia de Aldehído Deshidrogenasa 1 , Animales , Disponibilidad Biológica , Línea Celular Tumoral , Resistencia a Antineoplásicos/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Inhibidores Enzimáticos/farmacocinética , Humanos , Masculino , Ratones , Paclitaxel/farmacología , Quinolinas/administración & dosificación , Quinolinas/farmacocinética , Retinal-Deshidrogenasa
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...