Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 167(1): 171-186.e15, 2016 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-27641501

RESUMEN

While acute myeloid leukemia (AML) comprises many disparate genetic subtypes, one shared hallmark is the arrest of leukemic myeloblasts at an immature and self-renewing stage of development. Therapies that overcome differentiation arrest represent a powerful treatment strategy. We leveraged the observation that the majority of AML, despite their genetically heterogeneity, share in the expression of HoxA9, a gene normally downregulated during myeloid differentiation. Using a conditional HoxA9 model system, we performed a high-throughput phenotypic screen and defined compounds that overcame differentiation blockade. Target identification led to the unanticipated discovery that inhibition of the enzyme dihydroorotate dehydrogenase (DHODH) enables myeloid differentiation in human and mouse AML models. In vivo, DHODH inhibitors reduced leukemic cell burden, decreased levels of leukemia-initiating cells, and improved survival. These data demonstrate the role of DHODH as a metabolic regulator of differentiation and point to its inhibition as a strategy for overcoming differentiation blockade in AML.


Asunto(s)
Antineoplásicos/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/patología , Terapia Molecular Dirigida , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/antagonistas & inhibidores , Animales , Antineoplásicos/química , Antineoplásicos/aislamiento & purificación , Diferenciación Celular , Dihidroorotato Deshidrogenasa , Inhibidores Enzimáticos/química , Inhibidores Enzimáticos/aislamiento & purificación , Ensayos Analíticos de Alto Rendimiento , Proteínas de Homeodominio/genética , Humanos , Leucemia Mieloide Aguda/genética , Ratones , Células Mieloides/patología , Oxidorreductasas actuantes sobre Donantes de Grupo CH-CH/metabolismo , Pirimidinas/metabolismo , Bibliotecas de Moléculas Pequeñas/química , Bibliotecas de Moléculas Pequeñas/aislamiento & purificación , Bibliotecas de Moléculas Pequeñas/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
3.
Cell ; 150(3): 575-89, 2012 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-22863010

RESUMEN

The mechanism by which cells decide to skip mitosis to become polyploid is largely undefined. Here we used a high-content image-based screen to identify small-molecule probes that induce polyploidization of megakaryocytic leukemia cells and serve as perturbagens to help understand this process. Our study implicates five networks of kinases that regulate the switch to polyploidy. Moreover, we find that dimethylfasudil (diMF, H-1152P) selectively increased polyploidization, mature cell-surface marker expression, and apoptosis of malignant megakaryocytes. An integrated target identification approach employing proteomic and shRNA screening revealed that a major target of diMF is Aurora kinase A (AURKA). We further find that MLN8237 (Alisertib), a selective inhibitor of AURKA, induced polyploidization and expression of mature megakaryocyte markers in acute megakaryocytic leukemia (AMKL) blasts and displayed potent anti-AMKL activity in vivo. Our findings provide a rationale to support clinical trials of MLN8237 and other inducers of polyploidization and differentiation in AMKL.


Asunto(s)
Azepinas/farmacología , Descubrimiento de Drogas , Leucemia Megacarioblástica Aguda/tratamiento farmacológico , Megacariocitos/metabolismo , Poliploidía , Pirimidinas/farmacología , Bibliotecas de Moléculas Pequeñas , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/análogos & derivados , 1-(5-Isoquinolinesulfonil)-2-Metilpiperazina/farmacología , Animales , Aurora Quinasa A , Aurora Quinasas , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Humanos , Leucemia Megacarioblástica Aguda/genética , Megacariocitos/citología , Megacariocitos/patología , Ratones , Ratones Endogámicos C57BL , Mapas de Interacción de Proteínas , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/metabolismo , Quinasas Asociadas a rho/metabolismo
4.
Nat Chem Biol ; 18(6): 615-624, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35332332

RESUMEN

The ability to understand and predict variable responses to therapeutic agents may improve outcomes in patients with cancer. We hypothesized that the basal gene-transcription state of cancer cell lines, coupled with cell viability profiles of small molecules, might be leveraged to nominate specific mechanisms of intrinsic resistance and to predict drug combinations that overcome resistance. We analyzed 564,424 sensitivity profiles to identify candidate gene-compound pairs, and validated nine such relationships. We determined the mechanism of a novel relationship, in which expression of the serine hydrolase enzymes monoacylglycerol lipase (MGLL) or carboxylesterase 1 (CES1) confers resistance to the histone lysine demethylase inhibitor GSK-J4 by direct enzymatic modification. Insensitive cell lines could be sensitized to GSK-J4 by inhibition or gene knockout. These analytical and mechanistic studies highlight the potential of integrating gene-expression features with small-molecule response to identify patient populations that are likely to benefit from treatment, to nominate rational candidates for combinations and to provide insights into mechanisms of action.


Asunto(s)
Histona Demetilasas , Monoacilglicerol Lipasas , Biomarcadores , Supervivencia Celular , Combinación de Medicamentos , Histona Demetilasas/metabolismo , Humanos
5.
J Org Chem ; 89(5): 2825-2839, 2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38334085

RESUMEN

Fluorescence-based biological imaging in the shortwave infrared (SWIR, 1000-1700 nm) is an attractive replacement for modern in vivo imaging techniques currently employed in both medical and research settings. Xanthene-based fluorophores containing heterocycle donors have recently emerged as a way to access deep SWIR emitting fluorophores. A concern for xanthene-based SWIR fluorophores though is chemical stability toward ambient nucleophiles due to the high electrophilicity of the cationic fluorophore core. Herein, a series of SWIR emitting silicon-rosindolizine (SiRos) fluorophores with emission maxima >1300 nm (up to 1550 nm) are synthesized. The SiRos fluorophore photophysical properties and chemical stability toward nucleophiles are examined through systematic derivatization of the silicon-core alkyl groups, indolizine donor substitution, and the use of o-tolyl or o-xylyl groups appended to the fluorophore core. The dyes are studied via absorption spectroscopy, steady-state emission spectroscopy, solution-based cyclic voltammetry, time-dependent density functional theory (TD-DFT) computational analysis, X-ray diffraction crystallography, and relative chemical stability over time. Optimal chemical stability is observed via the incorporation of the 2-ethylhexyl silicon substituent and the o-xylyl group to protect the core of the fluorophore.

6.
Nature ; 538(7625): 344-349, 2016 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-27602946

RESUMEN

Antimalarial drugs have thus far been chiefly derived from two sources-natural products and synthetic drug-like compounds. Here we investigate whether antimalarial agents with novel mechanisms of action could be discovered using a diverse collection of synthetic compounds that have three-dimensional features reminiscent of natural products and are underrepresented in typical screening collections. We report the identification of such compounds with both previously reported and undescribed mechanisms of action, including a series of bicyclic azetidines that inhibit a new antimalarial target, phenylalanyl-tRNA synthetase. These molecules are curative in mice at a single, low dose and show activity against all parasite life stages in multiple in vivo efficacy models. Our findings identify bicyclic azetidines with the potential to both cure and prevent transmission of the disease as well as protect at-risk populations with a single oral dose, highlighting the strength of diversity-oriented synthesis in revealing promising therapeutic targets.


Asunto(s)
Antimaláricos/síntesis química , Antimaláricos/farmacología , Azetidinas/uso terapéutico , Descubrimiento de Drogas , Estadios del Ciclo de Vida/efectos de los fármacos , Malaria Falciparum/tratamiento farmacológico , Plasmodium falciparum/efectos de los fármacos , Plasmodium falciparum/crecimiento & desarrollo , Animales , Antimaláricos/administración & dosificación , Antimaláricos/uso terapéutico , Compuestos de Azabiciclo/administración & dosificación , Compuestos de Azabiciclo/síntesis química , Compuestos de Azabiciclo/farmacología , Compuestos de Azabiciclo/uso terapéutico , Azetidinas/administración & dosificación , Azetidinas/efectos adversos , Azetidinas/farmacología , Citosol/enzimología , Modelos Animales de Enfermedad , Femenino , Hígado/efectos de los fármacos , Hígado/parasitología , Macaca mulatta/parasitología , Malaria Falciparum/prevención & control , Malaria Falciparum/transmisión , Masculino , Ratones , Fenilalanina-ARNt Ligasa/antagonistas & inhibidores , Compuestos de Fenilurea/administración & dosificación , Compuestos de Fenilurea/síntesis química , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Plasmodium falciparum/citología , Plasmodium falciparum/enzimología , Seguridad
7.
J Biol Chem ; 295(11): 3431-3446, 2020 03 13.
Artículo en Inglés | MEDLINE | ID: mdl-32005668

RESUMEN

Cytotoxic molecules can kill cancer cells by disrupting critical cellular processes or by inducing novel activities. 6-(4-(Diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one (DNMDP) is a small molecule that kills cancer cells by generation of novel activity. DNMDP induces complex formation between phosphodiesterase 3A (PDE3A) and schlafen family member 12 (SLFN12) and specifically kills cancer cells expressing elevated levels of these two proteins. Here, we examined the characteristics and covariates of the cancer cell response to DNMDP. On average, the sensitivity of human cancer cell lines to DNMDP is correlated with PDE3A expression levels. However, DNMDP could also bind the related protein, PDE3B, and PDE3B supported DNMDP sensitivity in the absence of PDE3A expression. Although inhibition of PDE3A catalytic activity did not account for DNMDP sensitivity, we found that expression of the catalytic domain of PDE3A in cancer cells lacking PDE3A is sufficient to confer sensitivity to DNMDP, and substitutions in the PDE3A active site abolish compound binding. Moreover, a genome-wide CRISPR screen identified the aryl hydrocarbon receptor-interacting protein (AIP), a co-chaperone protein, as required for response to DNMDP. We determined that AIP is also required for PDE3A-SLFN12 complex formation. Our results provide mechanistic insights into how DNMDP induces PDE3A-SLFN12 complex formation, thereby killing cancer cells with high levels of PDE3A and SLFN12 expression.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Neoplasias/patología , Secuencia de Bases , Biomarcadores de Tumor/metabolismo , Sistemas CRISPR-Cas/genética , Dominio Catalítico , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/química , Mutación del Sistema de Lectura/genética , Genoma , Heterocigoto , Humanos , Unión Proteica/efectos de los fármacos , Piridazinas/farmacología
8.
Nat Chem Biol ; 12(12): 1023-1030, 2016 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-27694802

RESUMEN

Asparagine (N)-linked glycosylation is a protein modification critical for glycoprotein folding, stability, and cellular localization. To identify small molecules that inhibit new targets in this biosynthetic pathway, we initiated a cell-based high-throughput screen and lead-compound-optimization campaign that delivered a cell-permeable inhibitor, NGI-1. NGI-1 targets oligosaccharyltransferase (OST), a hetero-oligomeric enzyme that exists in multiple isoforms and transfers oligosaccharides to recipient proteins. In non-small-cell lung cancer cells, NGI-1 blocks cell-surface localization and signaling of the epidermal growth factor receptor (EGFR) glycoprotein, but selectively arrests proliferation in only those cell lines that are dependent on EGFR (or fibroblast growth factor, FGFR) for survival. In these cell lines, OST inhibition causes cell-cycle arrest accompanied by induction of p21, autofluorescence, and cell morphology changes, all hallmarks of senescence. These results identify OST inhibition as a potential therapeutic approach for treating receptor-tyrosine-kinase-dependent tumors and provides a chemical probe for reversibly regulating N-linked glycosylation in mammalian cells.


Asunto(s)
Benzamidas/farmacología , Senescencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Hexosiltransferasas/antagonistas & inhibidores , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas Tirosina Quinasas Receptoras/antagonistas & inhibidores , Sulfonamidas/farmacología , Benzamidas/química , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/química , Hexosiltransferasas/metabolismo , Ensayos Analíticos de Alto Rendimiento , Humanos , Proteínas de la Membrana/metabolismo , Estructura Molecular , Proteínas Tirosina Quinasas Receptoras/metabolismo , Relación Estructura-Actividad , Sulfonamidas/química
9.
Nat Chem Biol ; 12(2): 102-8, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26656089

RESUMEN

High cancer death rates indicate the need for new anticancer therapeutic agents. Approaches to discovering new cancer drugs include target-based drug discovery and phenotypic screening. Here, we identified phosphodiesterase 3A modulators as cell-selective cancer cytotoxic compounds through phenotypic compound library screening and target deconvolution by predictive chemogenomics. We found that sensitivity to 6-(4-(diethylamino)-3-nitrophenyl)-5-methyl-4,5-dihydropyridazin-3(2H)-one, or DNMDP, across 766 cancer cell lines correlates with expression of the gene PDE3A, encoding phosphodiesterase 3A. Like DNMDP, a subset of known PDE3A inhibitors kill selected cancer cells, whereas others do not. Furthermore, PDE3A depletion leads to DNMDP resistance. We demonstrated that DNMDP binding to PDE3A promotes an interaction between PDE3A and Schlafen 12 (SLFN12), suggestive of a neomorphic activity. Coexpression of SLFN12 with PDE3A correlates with DNMDP sensitivity, whereas depletion of SLFN12 results in decreased DNMDP sensitivity. Our results implicate PDE3A modulators as candidate cancer therapeutic agents and demonstrate the power of predictive chemogenomics in small-molecule discovery.


Asunto(s)
Fosfodiesterasas de Nucleótidos Cíclicos Tipo 3/metabolismo , Citotoxinas/farmacología , Neoplasias/terapia , Piridazinas/química , Piridazinas/farmacología , Línea Celular , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Citotoxinas/química , Citotoxinas/aislamiento & purificación , Sistemas de Liberación de Medicamentos , Activación Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Genómica , Humanos , Immunoblotting
11.
Proc Natl Acad Sci U S A ; 109(14): 5364-9, 2012 Apr 03.
Artículo en Inglés | MEDLINE | ID: mdl-22434908

RESUMEN

Under the instruction of cell-fate-determining, DNA-binding transcription factors, chromatin-modifying enzymes mediate and maintain cell states throughout development in multicellular organisms. Currently, small molecules modulating the activity of several classes of chromatin-modifying enzymes are available, including clinically approved histone deacetylase (HDAC) and DNA methyltransferase (DNMT) inhibitors. We describe the genome-wide expression changes induced by 29 compounds targeting HDACs, DNMTs, histone lysine methyltransferases (HKMTs), and protein arginine methyltransferases (PRMTs) in pancreatic α- and ß-cell lines. HDAC inhibitors regulate several hundred transcripts irrespective of the cell type, with distinct clusters of dissimilar activity for hydroxamic acids and orthoamino anilides. In contrast, compounds targeting histone methyltransferases modulate the expression of restricted gene sets in distinct cell types. For example, we find that G9a/GLP methyltransferase inhibitors selectively up-regulate the cholesterol biosynthetic pathway in pancreatic but not liver cells. These data suggest that, despite their conservation across the entire genome and in different cell types, chromatin pathways can be targeted to modulate the expression of selected transcripts.


Asunto(s)
Cromatina/metabolismo , Páncreas/efectos de los fármacos , Transcripción Genética , Línea Celular , Regulación hacia Abajo , Expresión Génica , Inhibidores de Histona Desacetilasas/farmacología , Humanos , Páncreas/citología , Páncreas/metabolismo , Regulación hacia Arriba
12.
Bioorg Med Chem Lett ; 24(24): 5801-5804, 2014 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-25454272

RESUMEN

As ENT inhibitors including dilazep have shown efficacy improving oHSV1 targeted oncolytic cancer therapy, a series of dilazep analogues was synthesized and biologically evaluated to examine both ENT1 and ENT2 inhibition. The central diamine core, alkyl chains, ester linkage and substituents on the phenyl ring were all varied. Compounds were screened against ENT1 and ENT2 using a radio-ligand cell-based assay. Dilazep and analogues with minor structural changes are potent and selective ENT1 inhibitors. No selective ENT2 inhibitors were found, although some analogues were more potent against ENT2 than the parent dilazep.


Asunto(s)
Dilazep/análogos & derivados , Tranportador Equilibrativo 1 de Nucleósido/antagonistas & inhibidores , Transportador Equilibrativo 2 de Nucleósido/antagonistas & inhibidores , Animales , Transporte Biológico/efectos de los fármacos , Línea Celular , Dilazep/síntesis química , Dilazep/farmacología , Tranportador Equilibrativo 1 de Nucleósido/genética , Tranportador Equilibrativo 1 de Nucleósido/metabolismo , Transportador Equilibrativo 2 de Nucleósido/genética , Transportador Equilibrativo 2 de Nucleósido/metabolismo , Humanos , Unión Proteica , Ratas , Proteínas Recombinantes/biosíntesis , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Porcinos
13.
Proc Natl Acad Sci U S A ; 108(21): 8773-8, 2011 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-21555567

RESUMEN

Activating K-RAS mutations are the most frequent oncogenic mutations in human cancer. Numerous downstream signaling pathways have been shown to be deregulated by oncogenic K-ras. However, to date there are still no effective targeted therapies for this genetically defined subset of patients. Here we report the results of a small molecule, synthetic lethal screen using mouse embryonic fibroblasts derived from a mouse model harboring a conditional oncogenic K-ras(G12D) allele. Among the >50,000 compounds screened, we identified a class of drugs with selective activity against oncogenic K-ras-expressing cells. The most potent member of this class, lanperisone, acts by inducing nonapoptotic cell death in a cell cycle- and translation-independent manner. The mechanism of cell killing involves the induction of reactive oxygen species that are inefficiently scavenged in K-ras mutant cells, leading to oxidative stress and cell death. In mice, treatment with lanperisone suppresses the growth of K-ras-driven tumors without overt toxicity. Our findings establish the specific antitumor activity of lanperisone and reveal oxidative stress pathways as potential targets in Ras-mediated malignancies.


Asunto(s)
Neoplasias/tratamiento farmacológico , Estrés Oxidativo/efectos de los fármacos , Proteínas ras , Animales , Antineoplásicos/farmacología , Muerte Celular/efectos de los fármacos , Células Cultivadas , Evaluación Preclínica de Medicamentos , Fibroblastos/citología , Ratones , Neoplasias/metabolismo , Neoplasias/patología , Especies Reactivas de Oxígeno/agonistas
14.
RSC Adv ; 14(13): 9254-9261, 2024 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-38505385

RESUMEN

Latent bloodstain detection remains imperative for crime scene investigators. Widely used luminol offers high sensitivity to human blood, but can produce untrustworthy results from a bleach-cleaned crime scene or in a room not dark enough. Furthermore, dark pigments impede imaging bloodstains covered by dark materials with previously reported bloodstain detection agents. A novel on/off human albumin-sensing dye (SO3C7) is reported herein with a longer emission wavelength (942 nm) than previous materials that allows imaging behind ∼5 mm of black fabric. The switch-on emission of SO3C7 is selective and sensitive to human albumin and lasts longer than luminol (24-48 hours). Emission studies, transient absorption spectra (TAS), and near-infrared (NIR) photographs herein describe the albumin sensing properties of the dye.

15.
PLoS Biol ; 8(1): e1000287, 2010 Jan 26.
Artículo en Inglés | MEDLINE | ID: mdl-20126263

RESUMEN

DNA damage checkpoints arrest cell cycle progression to facilitate DNA repair. The ability to survive genotoxic insults depends not only on the initiation of cell cycle checkpoints but also on checkpoint maintenance. While activation of DNA damage checkpoints has been studied extensively, molecular mechanisms involved in sustaining and ultimately inactivating cell cycle checkpoints are largely unknown. Here, we explored feedback mechanisms that control the maintenance and termination of checkpoint function by computationally identifying an evolutionary conserved mitotic phosphorylation network within the DNA damage response. We demonstrate that the non-enzymatic checkpoint adaptor protein 53BP1 is an in vivo target of the cell cycle kinases Cyclin-dependent kinase-1 and Polo-like kinase-1 (Plk1). We show that Plk1 binds 53BP1 during mitosis and that this interaction is required for proper inactivation of the DNA damage checkpoint. 53BP1 mutants that are unable to bind Plk1 fail to restart the cell cycle after ionizing radiation-mediated cell cycle arrest. Importantly, we show that Plk1 also phosphorylates the 53BP1-binding checkpoint kinase Chk2 to inactivate its FHA domain and inhibit its kinase activity in mammalian cells. Thus, a mitotic kinase-mediated negative feedback loop regulates the ATM-Chk2 branch of the DNA damage signaling network by phosphorylating conserved sites in 53BP1 and Chk2 to inactivate checkpoint signaling and control checkpoint duration.


Asunto(s)
Proteína Quinasa CDC2/metabolismo , Proteínas de Ciclo Celular/metabolismo , División Celular/fisiología , Daño del ADN , Fase G2/fisiología , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Línea Celular , Quinasa de Punto de Control 2 , Retroalimentación Fisiológica , Humanos , Fosforilación , Transducción de Señal , Proteína 1 de Unión al Supresor Tumoral P53 , Quinasa Tipo Polo 1
16.
Bioorg Med Chem Lett ; 23(10): 3039-43, 2013 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-23562243

RESUMEN

A high-throughput screen (HTS) of the MLPCN library using a homogenous fluorescence polarization assay identified a small molecule as a first-in-class direct inhibitor of Keap1-Nrf2 protein-protein interaction. The HTS hit has three chiral centers; a combination of flash and chiral chromatographic separation demonstrated that Keap1-binding activity resides predominantly in one stereoisomer (SRS)-5 designated as ML334 (LH601A), which is at least 100× more potent than the other stereoisomers. The stereochemistry of the four cis isomers was assigned using X-ray crystallography and confirmed using stereospecific synthesis. (SRS)-5 is functionally active in both an ARE gene reporter assay and an Nrf2 nuclear translocation assay. The stereospecific nature of binding between (SRS)-5 and Keap1 as well as the preliminary but tractable structure-activity relationships support its use as a lead for our ongoing optimization.


Asunto(s)
Descubrimiento de Drogas , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Isoquinolinas/farmacología , Imagen Molecular , Sondas Moleculares/farmacología , Factor 2 Relacionado con NF-E2/metabolismo , Ftalimidas/farmacología , Bibliotecas de Moléculas Pequeñas/farmacología , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Polarización de Fluorescencia , Ensayos Analíticos de Alto Rendimiento , Humanos , Isoquinolinas/química , Proteína 1 Asociada A ECH Tipo Kelch , Modelos Moleculares , Sondas Moleculares/química , Estructura Molecular , Ftalimidas/química , Unión Proteica/efectos de los fármacos , Bibliotecas de Moléculas Pequeñas/química , Relación Estructura-Actividad
17.
J Racial Ethn Health Disparities ; 10(4): 2039-2053, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-36068482

RESUMEN

The heroin epidemic has existed for decades, but a sharp rise in opioid overdose deaths (OODs) jolted the nation in the mid-twenty-teens and continues as a major health crisis to this day. Although the new wave of OODs was initially approached as a rural problem impacting a White/Caucasian demographic, surveillance records suggest severe impacts on African Americans and urban-dwelling individuals, which have been largely underreported. The focus of this report is on specific trends in OOD rates in Black and White residents in states with a significant Black urban population and declared as hotspots for OOD: (Maryland (MD), Illinois (IL), Michigan (MI), and Pennsylvania (PA)), and Washington District of Columbia (DC). We compare OODs by type of opioid, across ethnicities, across city/rural demographics, and to homicide rates using 2013-2020 data acquired from official Chief Medical Examiners' or Departments of Health (DOH) reports. With 2013 or 2014 as baseline, the OOD rate in major cities (Baltimore, Chicago, Detroit, Philadelphia) were elevated two-fold over all other regions of their respective state. In DC, Wards 7 and 8 OODs were consistently greater than other jurisdictions, until 2020 when the rate of change of OODs increased for the entire city. Ethnicity-wise, Black OOD rates exceeded White rates by four- to six-fold, with fentanyl and heroin having a disproportionate impact on Black opioid deaths. This disparity was aggravated by its intersection with the COVID-19 pandemic in 2020. African Americans and America's urban dwellers are vulnerable populations in need of social and political resources to address the ongoing opioid epidemic in under-resourced communities.


Asunto(s)
Negro o Afroamericano , Disparidades en Atención de Salud , Sobredosis de Opiáceos , Epidemia de Opioides , Determinantes Sociales de la Salud , Población Urbana , Adolescente , Humanos , Analgésicos Opioides/efectos adversos , Negro o Afroamericano/estadística & datos numéricos , COVID-19/epidemiología , COVID-19/etnología , Heroína/envenenamiento , Epidemia de Opioides/estadística & datos numéricos , Epidemia de Opioides/tendencias , Pandemias , Philadelphia/epidemiología , Estados Unidos/epidemiología , Población Urbana/estadística & datos numéricos , Blanco/estadística & datos numéricos , Sobredosis de Opiáceos/epidemiología , Sobredosis de Opiáceos/etnología , Sobredosis de Opiáceos/mortalidad , Determinantes Sociales de la Salud/etnología , Determinantes Sociales de la Salud/estadística & datos numéricos , Determinantes Sociales de la Salud/tendencias , Disparidades en Atención de Salud/etnología , Disparidades en Atención de Salud/estadística & datos numéricos , Disparidades en Atención de Salud/tendencias , Disparidades en el Estado de Salud
18.
J Virol ; 85(6): 2859-68, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21209116

RESUMEN

Latent Epstein-Barr virus (EBV) infection causes human lymphomas and carcinomas. EBV usually persists as an episome in malignant cells. EBV episome persistence, replication, and gene expression are dependent on EBNA1 binding to multiple cognate sites in oriP. To search for inhibitors of EBNA1- and oriP-dependent episome maintenance or transcription, a library of 40,550 small molecules was screened for compounds that inhibit EBNA1- and oriP-dependent transcription and do not inhibit EBNA1- and oriP-independent transcription. This screening identified roscovitine, a selective inhibitor of cyclin-dependent kinase 1 (CDK1), CDK2, CDK5, and CDK7. Based on motif predictions of EBNA1 serine 393 as a CDK phosphorylation site and (486)RALL(489) and (580)KDLVM(584) as potential cyclin binding domains, we hypothesized that cyclin binding to EBNA1 may enable CDK1, -2, -5, or -7 to phosphorylate serine 393. We found that Escherichia coli-expressed EBNA1 amino acids 387 to 641 were phosphorylated in vitro by CDK1-, -2-, -5-, and -7/cyclin complexes and serine 393 phosphorylation was roscovitine inhibited. Further, S393A mutation abrogated phosphorylation. S393A mutant EBNA1 was deficient in supporting EBNA1- and oriP-dependent transcription and episome persistence, and roscovitine had little further effect on the diminished S393A mutant EBNA1-mediated transcription or episome persistence. Immunoprecipitated FLAG-EBNA1 was phosphorylated in vitro, and roscovitine inhibited this phosphorylation. Moreover, roscovitine decreased nuclear EBNA1 and often increased cytoplasmic EBNA1, whereas S393A mutant EBNA1 was localized equally in the nucleus and cytoplasm and was unaffected by roscovitine treatment. These data indicate that roscovitine effects are serine 393 specific and that serine 393 is important in EBNA1- and oriPCp-dependent transcription and episome persistence.


Asunto(s)
Antivirales/metabolismo , Antígenos Nucleares del Virus de Epstein-Barr/metabolismo , Herpesvirus Humano 4/efectos de los fármacos , Herpesvirus Humano 4/fisiología , Purinas/metabolismo , Replicación Viral , Evaluación Preclínica de Medicamentos/métodos , Escherichia coli/genética , Humanos , Fosforilación , Plásmidos/efectos de los fármacos , Roscovitina , Transcripción Genética/efectos de los fármacos
19.
Bioorg Med Chem Lett ; 22(4): 1822-6, 2012 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-22297109

RESUMEN

Synthetic lethal screening is a chemical biology approach to identify small molecules that selectively kill oncogene-expressing cell lines with the goal of identifying pathways that provide specific targets against cancer cells. We performed a high-throughput screen of 303,282 compounds from the National Institutes of Health-Molecular Libraries Small Molecule Repository (NIH-MLSMR) against immortalized BJ fibroblasts expressing HRAS(G12V) followed by a counterscreen of lethal compounds in a series of isogenic cells lacking the HRAS(G12V) oncogene. This effort led to the identification of two novel molecular probes (PubChem CID 3689413, ML162 and CID 49766530, ML210) with nanomolar potencies and 4-23-fold selectivities, which can potentially be used for identifying oncogene-specific pathways and targets in cancer cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas Proto-Oncogénicas p21(ras)/antagonistas & inhibidores , Bibliotecas de Moléculas Pequeñas/farmacología , Animales , Línea Celular , Línea Celular Tumoral , Ensayos de Selección de Medicamentos Antitumorales , Fibroblastos , Humanos , Concentración 50 Inhibidora , Estructura Molecular , Mutación , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Ratas , Relación Estructura-Actividad , Tiofenos/química , Tiofenos/farmacología
20.
Bioorg Med Chem Lett ; 22(10): 3571-4, 2012 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-22503247

RESUMEN

A high-throughput screen (HTS) with the National Institute of Health-Molecular Libraries Small Molecule Repository (NIH-MLSMR) compound collection identified a class of acyl hydrazones to be selectively lethal to breast cancer stem cell (CSC) enriched populations. Medicinal chemistry efforts were undertaken to optimize potency and selectivity of this class of compounds. The optimized compound was declared as a probe (ML239) with the NIH Molecular Libraries Program and displayed greater than 20-fold selective inhibition of the breast CSC-like cell line (HMLE_sh_Ecad) over the isogenic control line (HMLE_sh_GFP).


Asunto(s)
Neoplasias de la Mama/tratamiento farmacológico , Hidrazonas/farmacología , Células Madre Neoplásicas/citología , Pirroles/farmacología , Neoplasias de la Mama/patología , Femenino , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA