Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Sci Adv ; 7(43): eabh3794, 2021 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-34669463

RESUMO

Cancer therapies are being considered for treating rare noncancerous diseases like pulmonary hypertension (PH), but effective computational screening is lacking. Via transcriptomic differential dependency analyses leveraging parallels between cancer and PH, we mapped a landscape of cancer drug functions dependent upon rewiring of PH gene clusters. Bromodomain and extra-terminal motif (BET) protein inhibitors were predicted to rely upon several gene clusters inclusive of galectin-8 (LGALS8). Correspondingly, LGALS8 was found to mediate the BET inhibitor­dependent control of endothelial apoptosis, an essential role for PH in vivo. Separately, a piperlongumine analog's actions were predicted to depend upon the iron-sulfur biogenesis gene ISCU. Correspondingly, the analog was found to inhibit ISCU glutathionylation, rescuing oxidative metabolism, decreasing endothelial apoptosis, and improving PH. Thus, we identified crucial drug-gene axes central to endothelial dysfunction and therapeutic priorities for PH. These results establish a wide-ranging, network dependency platform to redefine cancer drugs for use in noncancerous conditions.

2.
Cell ; 184(5): 1142-1155, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33667368

RESUMO

The characterization of cancer genomes has provided insight into somatically altered genes across tumors, transformed our understanding of cancer biology, and enabled tailoring of therapeutic strategies. However, the function of most cancer alleles remains mysterious, and many cancer features transcend their genomes. Consequently, tumor genomic characterization does not influence therapy for most patients. Approaches to understand the function and circuitry of cancer genes provide complementary approaches to elucidate both oncogene and non-oncogene dependencies. Emerging work indicates that the diversity of therapeutic targets engendered by non-oncogene dependencies is much larger than the list of recurrently mutated genes. Here we describe a framework for this expanded list of cancer targets, providing novel opportunities for clinical translation.


Assuntos
Sistemas de Liberação de Medicamentos , Neoplasias/tratamento farmacológico , Animais , Ensaios Clínicos como Assunto , Modelos Animais de Doenças , Genômica , Humanos , Neoplasias/genética , Neoplasias/patologia , Evasão Tumoral/efeitos dos fármacos , Microambiente Tumoral/efeitos dos fármacos
3.
Int J Mol Sci ; 21(21)2020 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-33137873

RESUMO

Selective inhibition of histone deacetylase 3 (HDAC3) prevents glucolipotoxicity-induced ß-cell dysfunction and apoptosis by alleviation of proapoptotic endoplasmic reticulum (ER) stress-signaling, but the precise molecular mechanisms of alleviation are unexplored. By unbiased microarray analysis of the ß-cell gene expression profile of insulin-producing cells exposed to glucolipotoxicity in the presence or absence of a selective HDAC3 inhibitor, we identified Enhancer of zeste homolog 2 (EZH2) as the sole target candidate. ß-Cells were protected against glucolipotoxicity-induced ER stress and apoptosis by EZH2 attenuation. Small molecule inhibitors of EZH2 histone methyltransferase activity rescued human islets from glucolipotoxicity-induced apoptosis. Moreover, EZH2 knockdown cells were protected against glucolipotoxicity-induced downregulation of the protective non-canonical Nuclear factor of kappa light polypeptide gene enhancer in B-cells (NFκB) pathway. We conclude that EZH2 deficiency protects from glucolipotoxicity-induced ER stress, apoptosis and downregulation of the non-canonical NFκB pathway, but not from insulin secretory dysfunction. The mechanism likely involves transcriptional regulation via EZH2 functioning as a methyltransferase and/or as a methylation-dependent transcription factor.


Assuntos
Apoptose , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Glucose/efeitos adversos , Secreção de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Lipídeos/efeitos adversos , Células Cultivadas , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Transdução de Sinais , Edulcorantes/efeitos adversos
4.
Nat Commun ; 11(1): 4043, 2020 08 13.
Artigo em Inglês | MEDLINE | ID: mdl-32792475

RESUMO

Genetically fusing protein domains to Cas9 has yielded several transformative technologies; however, the genetic modifications are limited to natural polypeptide chains at the Cas9 termini, which excludes a diverse array of molecules useful for gene editing. Here, we report chemical modifications that allow site-specific and multiple-site conjugation of a wide assortment of molecules on both the termini and internal sites of Cas9, creating a platform for endowing Cas9 with diverse functions. Using this platform, Cas9 can be modified to more precisely incorporate exogenously supplied single-stranded oligonucleotide donor (ssODN) at the DNA break site. We demonstrate that the multiple-site conjugation of ssODN to Cas9 significantly increases the efficiency of precision genome editing, and such a platform is compatible with ssODNs of diverse lengths. By leveraging the conjugation platform, we successfully engineer INS-1E, a ß-cell line, to repurpose the insulin secretion machinery, which enables the glucose-dependent secretion of protective immunomodulatory factor interleukin-10.


Assuntos
Proteína 9 Associada à CRISPR/química , Proteína 9 Associada à CRISPR/metabolismo , Engenharia Celular/métodos , Biologia Sintética/métodos , Proteína 9 Associada à CRISPR/genética , Linhagem Celular , Edição de Genes , Humanos , Sistemas de Infusão de Insulina
5.
Cell Chem Biol ; 26(9): 1306-1314.e5, 2019 09 19.
Artigo em Inglês | MEDLINE | ID: mdl-31204288

RESUMO

The proteinaceous extracellular matrix (ECM) is vital for the survival, proliferation, migration, and differentiation of many types of cancer. However, little is known regarding metabolic pathways required for ECM secretion. By using an unbiased computational approach, we searched for enzymes whose suppression may lead to disruptions in protein secretion. Here, we show that 6-phosphogluconate dehydrogenase (PGD), a cytosolic enzyme involved in carbohydrate metabolism, is required for ER structural integrity and protein secretion. Chemical inhibition or genetic suppression of PGD activity led to cell stress accompanied by significantly expanded ER volume and was rescued by compensating endogenous glutathione supplies. Our results also suggest that this characteristic ER-dilation phenotype may be a general marker indicating increased ECM protein congestion inside cells and decreased secretion. Thus, PGD serves as a link between cytosolic carbohydrate metabolism and protein secretion.


Assuntos
Metabolismo dos Carboidratos/fisiologia , Glutationa/metabolismo , Fosfogluconato Desidrogenase/metabolismo , Sistemas de Translocação de Proteínas/metabolismo , Linhagem Celular Tumoral , Citosol/metabolismo , Retículo Endoplasmático/metabolismo , Estresse do Retículo Endoplasmático/fisiologia , Matriz Extracelular/metabolismo , Humanos , Fosfogluconato Desidrogenase/fisiologia , Sistemas de Translocação de Proteínas/fisiologia
6.
Nat Commun ; 10(1): 1617, 2019 04 08.
Artigo em Inglês | MEDLINE | ID: mdl-30962421

RESUMO

Clear-cell carcinomas (CCCs) are a histological group of highly aggressive malignancies commonly originating in the kidney and ovary. CCCs are distinguished by aberrant lipid and glycogen accumulation and are refractory to a broad range of anti-cancer therapies. Here we identify an intrinsic vulnerability to ferroptosis associated with the unique metabolic state in CCCs. This vulnerability transcends lineage and genetic landscape, and can be exploited by inhibiting glutathione peroxidase 4 (GPX4) with small-molecules. Using CRISPR screening and lipidomic profiling, we identify the hypoxia-inducible factor (HIF) pathway as a driver of this vulnerability. In renal CCCs, HIF-2α selectively enriches polyunsaturated lipids, the rate-limiting substrates for lipid peroxidation, by activating the expression of hypoxia-inducible, lipid droplet-associated protein (HILPDA). Our study suggests targeting GPX4 as a therapeutic opportunity in CCCs, and highlights that therapeutic approaches can be identified on the basis of cell states manifested by morphological and metabolic features in hard-to-treat cancers.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Carcinoma de Células Renais/patologia , Glutationa Peroxidase/metabolismo , Neoplasias Renais/patologia , Proteínas de Neoplasias/metabolismo , Idoso , Animais , Apoptose/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Sistemas CRISPR-Cas/genética , Carcinoma de Células Renais/genética , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Glutationa Peroxidase/genética , Células HEK293 , Humanos , Ferro/metabolismo , Neoplasias Renais/genética , Peroxidação de Lipídeos/genética , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Fosfolipídeo Hidroperóxido Glutationa Peroxidase , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Proc Natl Acad Sci U S A ; 114(20): E4030-E4039, 2017 05 16.
Artigo em Inglês | MEDLINE | ID: mdl-28461505

RESUMO

Children with Down syndrome (DS) are prone to development of high-risk B-cell precursor ALL (DS-ALL), which differs genetically from most sporadic pediatric ALLs. Increased expression of cytokine receptor-like factor 2 (CRLF2), the receptor to thymic stromal lymphopoietin (TSLP), characterizes about half of DS-ALLs and also a subgroup of sporadic "Philadelphia-like" ALLs. To understand the pathogenesis of relapsed DS-ALL, we performed integrative genomic analysis of 25 matched diagnosis-remission and -relapse DS-ALLs. We found that the CRLF2 rearrangements are early events during DS-ALL evolution and generally stable between diagnoses and relapse. Secondary activating signaling events in the JAK-STAT/RAS pathway were ubiquitous but highly redundant between diagnosis and relapse, suggesting that signaling is essential but that no specific mutations are "relapse driving." We further found that activated JAK2 may be naturally suppressed in 25% of CRLF2pos DS-ALLs by loss-of-function aberrations in USP9X, a deubiquitinase previously shown to stabilize the activated phosphorylated JAK2. Interrogation of large ALL genomic databases extended our findings up to 25% of CRLF2pos, Philadelphia-like ALLs. Pharmacological or genetic inhibition of USP9X, as well as treatment with low-dose ruxolitinib, enhanced the survival of pre-B ALL cells overexpressing mutated JAK2. Thus, somehow counterintuitive, we found that suppression of JAK-STAT "hypersignaling" may be beneficial to leukemic B-cell precursors. This finding and the reduction of JAK mutated clones at relapse suggest that the therapeutic effect of JAK specific inhibitors may be limited. Rather, combined signaling inhibitors or direct targeting of the TSLP receptor may be a useful therapeutic strategy for DS-ALL.


Assuntos
Síndrome de Down/complicações , Janus Quinases/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Fatores de Transcrição STAT/metabolismo , Adolescente , Linhagem Celular Tumoral , Criança , Pré-Escolar , Feminino , Humanos , Masculino , Mutação , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Receptores de Citocinas/genética , Recidiva , Transdução de Sinais , Ubiquitina Tiolesterase/genética , Adulto Jovem
8.
Diabetes ; 65(6): 1660-71, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26953159

RESUMO

Restoring functional ß-cell mass is an important therapeutic goal for both type 1 and type 2 diabetes (1). While proliferation of existing ß-cells is the primary means of ß-cell replacement in rodents (2), it is unclear whether a similar principle applies to humans, as human ß-cells are remarkably resistant to stimulation of division (3,4). Here, we show that 5-iodotubercidin (5-IT), an annotated adenosine kinase inhibitor previously reported to increase proliferation in rodent and porcine islets (5), strongly and selectively increases human ß-cell proliferation in vitro and in vivo. Remarkably, 5-IT also increased glucose-dependent insulin secretion after prolonged treatment. Kinome profiling revealed 5-IT to be a potent and selective inhibitor of the dual-specificity tyrosine phosphorylation-regulated kinase (DYRK) and cell division cycle-like kinase families. Induction of ß-cell proliferation by either 5-IT or harmine, another natural product DYRK1A inhibitor, was suppressed by coincubation with the calcineurin inhibitor FK506, suggesting involvement of DYRK1A and nuclear factor of activated T cells signaling. Gene expression profiling in whole islets treated with 5-IT revealed induction of proliferation- and cell cycle-related genes, suggesting that true proliferation is induced by 5-IT. Furthermore, 5-IT promotes ß-cell proliferation in human islets grafted under the kidney capsule of NOD-scid IL2Rg(null) mice. These results point to inhibition of DYRK1A as a therapeutic strategy to increase human ß-cell proliferation.


Assuntos
Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Tubercidina/análogos & derivados , Animais , Proliferação de Células/genética , Perfilação da Expressão Gênica , Humanos , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos NOD , Fosforilação/efeitos dos fármacos , Tubercidina/farmacologia , Quinases Dyrk
9.
Nat Chem Biol ; 12(2): 102-8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26656089

RESUMO

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.


Assuntos
Nucleotídeo Cíclico Fosfodiesterase do Tipo 3/metabolismo , Citotoxinas/farmacologia , Neoplasias/terapia , Piridazinas/química , Piridazinas/farmacologia , Linhagem Celular , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citotoxinas/química , Citotoxinas/isolamento & purificação , Sistemas de Liberação de Medicamentos , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Genômica , Humanos , Immunoblotting
10.
Nat Chem Biol ; 12(2): 109-16, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26656090

RESUMO

Changes in cellular gene expression in response to small-molecule or genetic perturbations have yielded signatures that can connect unknown mechanisms of action (MoA) to ones previously established. We hypothesized that differential basal gene expression could be correlated with patterns of small-molecule sensitivity across many cell lines to illuminate the actions of compounds whose MoA are unknown. To test this idea, we correlated the sensitivity patterns of 481 compounds with ∼19,000 basal transcript levels across 823 different human cancer cell lines and identified selective outlier transcripts. This process yielded many novel mechanistic insights, including the identification of activation mechanisms, cellular transporters and direct protein targets. We found that ML239, originally identified in a phenotypic screen for selective cytotoxicity in breast cancer stem-like cells, most likely acts through activation of fatty acid desaturase 2 (FADS2). These data and analytical tools are available to the research community through the Cancer Therapeutics Response Portal.


Assuntos
Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Aflatoxinas/química , Aflatoxinas/farmacologia , Western Blotting , Neoplasias da Mama/tratamento farmacológico , Linhagem Celular Tumoral , Simulação por Computador , Sistemas de Liberação de Medicamentos , Feminino , Humanos , Estrutura Molecular , Análise de Componente Principal , Reação em Cadeia da Polimerase em Tempo Real
11.
Trends Genet ; 31(1): 16-23, 2015 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-25498789

RESUMO

Over the past decade, tremendous progress in high-throughput small molecule-screening methods has facilitated the rapid expansion of phenotype-based data. Parallel advances in genomic characterization methods have complemented these efforts by providing a growing list of annotated cell line features. Together, these developments have paved the way for feature-based identification of novel, exploitable cellular dependencies, subsequently expanding our therapeutic toolkit in cancer and other diseases. Here, we provide an overview of the evolution of phenotypic small-molecule profiling and discuss the most significant and recent profiling and analytical efforts, their impact on the field, and their clinical ramifications. We additionally provide a perspective for future developments in phenotypic profiling efforts guided by genomic science.


Assuntos
Descoberta de Drogas , Genética Médica , Bibliotecas de Moléculas Pequenas/farmacologia , Resistência a Medicamentos/genética , Genoma Humano/genética , Humanos , Fenótipo
12.
Curr Protoc Chem Biol ; 6(3): 157-168, 2014 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-25205564

RESUMO

The identification of novel small molecules that promote pancreatic beta-cell proliferation is an important approach to therapeutic discovery for diabetes. Because human islets are not easy to culture, and attach poorly to plates, it had not been feasible to run high-throughput phenotypic assays in these cells. Therefore, most laboratories have turned to rodent islets for ease of culture and accessibility. However, rodent islets are not physiologically similar to human islets, either in terms of islet architecture or endocrine cell interactions within the islet, and data generated in rodent islets do not typically translate to human islet biology. To address this challenge, we developed a human islet culture system for high-throughput screening using a thymidine analog, EdU, to detect beta-cell replication during screening. Simultaneous monitoring of EdU incorporation and beta cell numbers provides a robust assay for beta-cell replication, and is now becoming a standard protocol enabling screening in human islets.


Assuntos
Desoxiuridina/análogos & derivados , Ilhotas Pancreáticas/citologia , Cultura Primária de Células/métodos , Adenoviridae , Biomarcadores/metabolismo , Proliferação de Células , Desoxiuridina/metabolismo , Humanos , Ilhotas Pancreáticas/química , Ilhotas Pancreáticas/metabolismo
13.
J Biomol Screen ; 19(5): 771-81, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24464433

RESUMO

High-throughput screening allows rapid identification of new candidate compounds for biological probe or drug development. Here, we describe a principled method to generate "assay performance profiles" for individual compounds that can serve as a basis for similarity searches and cluster analyses. Our method overcomes three challenges associated with generating robust assay performance profiles: (1) we transform data, allowing us to build profiles from assays having diverse dynamic ranges and variability; (2) we apply appropriate mathematical principles to handle missing data; and (3) we mitigate the fact that loss-of-signal assay measurements may not distinguish between multiple mechanisms that can lead to certain phenotypes (e.g., cell death). Our method connected compounds with similar mechanisms of action, enabling prediction of new targets and mechanisms both for known bioactives and for compounds emerging from new screens. Furthermore, we used Bayesian modeling of promiscuous compounds to distinguish between broadly bioactive and narrowly bioactive compound communities. Several examples illustrate the utility of our method to support mechanism-of-action studies in probe development and target identification projects.


Assuntos
Avaliação Pré-Clínica de Medicamentos/métodos , Ensaios de Triagem em Larga Escala/métodos , Bibliotecas de Moléculas Pequenas/química , Algoritmos , Animais , Teorema de Bayes , Linhagem Celular Tumoral , Análise por Conglomerados , Humanos , Potencial da Membrana Mitocondrial , Camundongos , Modelos Moleculares , Fenótipo , Reprodutibilidade dos Testes
14.
Chem Biol ; 20(12): 1513-22, 2013 Dec 19.
Artigo em Inglês | MEDLINE | ID: mdl-24290880

RESUMO

Pancreatic and duodenal homeobox 1 (PDX1), a member of the homeodomain-containing transcription factor family, is a key transcription factor important for both pancreas development and mature ß cell function. The ectopic overexpression of Pdx1, Neurog3, and MafA in mice reprograms acinar cells to insulin-producing cells. We developed a quantitative PCR-based gene expression assay to screen more than 60,000 compounds for expression of each of these genes in the human PANC-1 ductal carcinoma cell line. We identified BRD7552, which upregulated PDX1 expression in both primary human islets and ductal cells, and induced epigenetic changes in the PDX1 promoter consistent with transcriptional activation. Prolonged compound treatment induced both insulin mRNA and protein and also enhanced insulin expression induced by the three-gene combination. These results provide a proof of principle for identifying small molecules that induce expression of transcription factors to control cellular reprogramming.


Assuntos
Carcinoma Ductal/genética , Carcinoma Ductal Pancreático/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Proteínas de Homeodomínio/genética , Bibliotecas de Moléculas Pequenas/farmacologia , Transativadores/genética , Animais , Linhagem Celular Tumoral , Ensaios de Triagem em Larga Escala/métodos , Humanos , Camundongos , Reação em Cadeia da Polimerase/métodos , Regiões Promotoras Genéticas/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos
15.
ACS Chem Biol ; 8(5): 923-9, 2013 May 17.
Artigo em Inglês | MEDLINE | ID: mdl-23477340

RESUMO

Elevation of reactive oxygen species (ROS) levels has been observed in many cancer cells relative to nontransformed cells, and recent reports have suggested that small-molecule enhancers of ROS may selectively kill cancer cells in various in vitro and in vivo models. We used a high-throughput screening approach to identify several hundred small-molecule enhancers of ROS in a human osteosarcoma cell line. A minority of these compounds diminished the viability of cancer cell lines, indicating that ROS elevation by small molecules is insufficient to induce death of cancer cell lines. Three chemical probes (BRD5459, BRD56491, BRD9092) are highlighted that most strongly elevate markers of oxidative stress without causing cell death and may be of use in a variety of cellular settings. For example, combining nontoxic ROS-enhancing probes with nontoxic doses of L-buthionine sulfoximine, an inhibitor of glutathione synthesis previously studied in cancer patients, led to potent cell death in more than 20 cases, suggesting that even nontoxic ROS-enhancing treatments may warrant exploration in combination strategies. Additionally, a few ROS-enhancing compounds that contain sites of electrophilicity, including piperlongumine, show selective toxicity for transformed cells over nontransformed cells in an engineered cell-line model of tumorigenesis. These studies suggest that cancer cell lines are more resilient to chemically induced increases in ROS levels than previously thought and highlight electrophilicity as a property that may be more closely associated with cancer-selective cell death than ROS elevation.


Assuntos
Antineoplásicos/farmacologia , Ensaios de Triagem em Larga Escala/métodos , Sondas Moleculares/metabolismo , Sondas Moleculares/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Ósseas/tratamento farmacológico , Neoplasias Ósseas/patologia , Butionina Sulfoximina/farmacologia , Morte Celular/efeitos dos fármacos , Linhagem Celular Transformada , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dioxolanos/farmacologia , Ensaios de Seleção de Medicamentos Antitumorais/métodos , Humanos , Sondas Moleculares/química , Osteossarcoma/tratamento farmacológico , Osteossarcoma/patologia , Estresse Oxidativo/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia
16.
Proc Natl Acad Sci U S A ; 109(38): 15115-20, 2012 Sep 18.
Artigo em Inglês | MEDLINE | ID: mdl-22949699

RESUMO

Piperlongumine is a naturally occurring small molecule recently identified to be toxic selectively to cancer cells in vitro and in vivo. This compound was found to elevate cellular levels of reactive oxygen species (ROS) selectively in cancer cell lines. The synthesis of 80 piperlongumine analogs has revealed structural modifications that retain, enhance, and ablate key piperlongumine-associated effects on cells, including elevation of ROS, cancer cell death, and selectivity for cancer cells over nontransformed cell types. Structure/activity relationships suggest that the electrophilicity of the C2-C3 olefin is critical for the observed effects on cells. Furthermore, we show that analogs lacking a reactive C7-C8 olefin can elevate ROS to levels observed with piperlongumine but show markedly reduced cell death, suggesting that ROS-independent mechanisms, including cellular cross-linking events, may also contribute to piperlongumine's induction of apoptosis. In particular, we have identified irreversible protein glutathionylation as a process associated with cellular toxicity. We propose a mechanism of action for piperlongumine that may be relevant to other small molecules having two sites of reactivity, one with greater and the other with lesser electrophilicity.


Assuntos
Dioxolanos/química , Trifosfato de Adenosina/química , Alcenos/química , Alelos , Antineoplásicos/farmacologia , Apoptose , Linhagem Celular Tumoral , Dioxolanos/síntese química , Relação Dose-Resposta a Droga , Células HeLa , Humanos , Modelos Químicos , Estresse Oxidativo , Fenótipo , Espécies Reativas de Oxigênio , Compostos de Sulfidrila/química
17.
Chem Biol ; 19(6): 669-73, 2012 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-22726680

RESUMO

Cytokine-induced beta-cell apoptosis is important to the etiology of type-1 diabetes. Although previous reports have shown that general inhibitors of histone deacetylase (HDAC) activity, such as suberoylanilide hydroxamic acid and trichostatin A, can partially prevent beta-cell death, they do not fully restore beta-cell function. To understand HDAC isoform selectivity in beta cells, we measured the cellular effects of 11 structurally diverse HDAC inhibitors on cytokine-induced apoptosis in the rat INS-1E cell line. All 11 compounds restored ATP levels and reduced nitrite secretion. However, caspase-3 activity was reduced only by MS-275 and CI-994, both of which target HDAC1, 2, and 3. Importantly, both MS-275 and genetic knockdown of Hdac3 alone were sufficient to restore glucose-stimulated insulin secretion in the presence of cytokines. These results suggest that HDAC3-selective inhibitors may be effective in preventing cytokine-induced beta-cell apoptosis.


Assuntos
Apoptose/efeitos dos fármacos , Benzamidas/farmacologia , Citocinas/metabolismo , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/metabolismo , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Fenilenodiaminas/farmacologia , Piridinas/farmacologia , Animais , Relação Dose-Resposta a Droga , Glucose/farmacologia , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/metabolismo , Ratos , Relação Estrutura-Atividade , Células Tumorais Cultivadas
18.
ACS Chem Biol ; 7(7): 1152-7, 2012 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-22536950

RESUMO

Post-translational modifications of histones alter chromatin structure and play key roles in gene expression and specification of cell states. Small molecules that target chromatin-modifying enzymes selectively are useful as probes and have promise as therapeutics, although very few are currently available. G9a (also named euchromatin histone methyltransferase 2 (EHMT2)) catalyzes methylation of lysine 9 on histone H3 (H3K9), a modification linked to aberrant silencing of tumor-suppressor genes, among others. Here, we report the discovery of a novel histone methyltransferase inhibitor, BRD4770. This compound reduced cellular levels of di- and trimethylated H3K9 without inducing apoptosis, induced senescence, and inhibited both anchorage-dependent and -independent proliferation in the pancreatic cancer cell line PANC-1. ATM-pathway activation, caused by either genetic or small-molecule inhibition of G9a, may mediate BRD4770-induced cell senescence. BRD4770 may be a useful tool to study G9a and its role in senescence and cancer cell biology.


Assuntos
Adenocarcinoma/enzimologia , Senescência Celular/efeitos dos fármacos , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Sondas Moleculares/farmacologia , Neoplasias Pancreáticas/enzimologia , Adenocarcinoma/patologia , Benzamidas/química , Benzamidas/metabolismo , Benzamidas/farmacologia , Benzimidazóis/química , Benzimidazóis/metabolismo , Benzimidazóis/farmacologia , Linhagem Celular Tumoral , Senescência Celular/imunologia , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Células HeLa , Antígenos de Histocompatibilidade/metabolismo , Histona-Lisina N-Metiltransferase/metabolismo , Humanos , Sondas Moleculares/química , Sondas Moleculares/metabolismo , Neoplasias Pancreáticas/patologia
19.
PLoS One ; 7(1): e28808, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22242153

RESUMO

BACKGROUND: Expression of insulin in terminally differentiated non-beta cell types in the pancreas could be important to treating type-1 diabetes. Previous findings led us to hypothesize involvement of kinase inhibition in induction of insulin expression in pancreatic alpha cells. METHODOLOGY/PRINCIPAL FINDINGS: Alpha (αTC1.6) cells and human islets were treated with GW8510 and other small-molecule inhibitors for up to 5 days. Alpha cells were assessed for gene- and protein-expression levels, cell-cycle status, promoter occupancy status by chromatin immunoprecipitation (ChIP), and p53-dependent transcriptional activity. GW8510, a putative CDK2 inhibitor, up-regulated insulin expression in mouse alpha cells and enhanced insulin secretion in dissociated human islets. Gene-expression profiling and gene-set enrichment analysis of GW8510-treated alpha cells suggested up-regulation of the p53 pathway. Accordingly, the compound increased p53 transcriptional activity and expression levels of p53 transcriptional targets. A predicted p53 response element in the promoter region of the mouse Ins2 gene was verified by chromatin immunoprecipitation (ChIP). Further, inhibition of Jun N-terminal kinase (JNK) and p38 kinase activities suppressed insulin induction by GW8510. CONCLUSIONS/SIGNIFICANCE: The induction of Ins2 by GW8510 occurred through p53 in a JNK- and p38-dependent manner. These results implicate p53 activity in modulation of Ins2 expression levels in pancreatic alpha cells, and point to a potential approach toward using small molecules to generate insulin in an alternative cell type.


Assuntos
Células Secretoras de Glucagon/efeitos dos fármacos , Células Secretoras de Glucagon/metabolismo , Indóis/farmacologia , Insulina/metabolismo , Transcrição Gênica/efeitos dos fármacos , Ativação Transcricional/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Animais , Ciclo Celular/efeitos dos fármacos , Linhagem Celular , Células Secretoras de Glucagon/citologia , Humanos , Camundongos , Pessoa de Meia-Idade , Modelos Biológicos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Elementos de Resposta/genética , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ativação Transcricional/genética
20.
J Biomol Screen ; 17(4): 509-18, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22156222

RESUMO

A small-molecule inducer of beta-cell proliferation in human islets represents a potential regeneration strategy for treating type 1 diabetes. However, the lack of suitable human beta cell lines makes such a discovery a challenge. Here, we adapted an islet cell culture system to high-throughput screening to identify such small molecules. We prepared microtiter plates containing extracellular matrix from a human bladder carcinoma cell line. Dissociated human islets were seeded onto these plates, cultured for up to 7 days, and assessed for proliferation by simultaneous Ki67 and C-peptide immunofluorescence. Importantly, this environment preserved beta-cell physiological function, as measured by glucose-stimulated insulin secretion. Adenoviral overexpression of cdk-6 and cyclin D(1), known inducers of human beta cell proliferation, was used as a positive control in our assay. This induction was inhibited by cotreatment with rapamycin, an immunosuppressant often used in islet transplantation. We then performed a pilot screen of 1280 compounds, observing some phenotypic effects on cells. This high-throughput human islet cell culture method can be used to assess various aspects of beta-cell biology on a relatively large number of compounds.


Assuntos
Ensaios de Triagem em Larga Escala/métodos , Ilhotas Pancreáticas/citologia , Cultura Primária de Células/métodos , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Avaliação Pré-Clínica de Medicamentos , Glucose/metabolismo , Humanos , Insulina/metabolismo , Secreção de Insulina , Células Secretoras de Insulina/citologia , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/metabolismo , Ilhotas Pancreáticas/efeitos dos fármacos , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA