Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Proc Natl Acad Sci U S A ; 113(7): 1778-83, 2016 Feb 16.
Artigo em Inglês | MEDLINE | ID: mdl-26831078

RESUMO

Cancer cells reprogram their metabolism to promote growth and proliferation. The genetic evidence pointing to the importance of the amino acid serine in tumorigenesis is striking. The gene encoding the enzyme 3-phosphoglycerate dehydrogenase (PHGDH), which catalyzes the first committed step of serine biosynthesis, is overexpressed in tumors and cancer cell lines via focal amplification and nuclear factor erythroid-2-related factor 2 (NRF2)-mediated up-regulation. PHGDH-overexpressing cells are exquisitely sensitive to genetic ablation of the pathway. Here, we report the discovery of a selective small molecule inhibitor of PHGDH, CBR-5884, identified by screening a library of 800,000 drug-like compounds. CBR-5884 inhibited de novo serine synthesis in cancer cells and was selectively toxic to cancer cell lines with high serine biosynthetic activity. Biochemical characterization of the inhibitor revealed that it was a noncompetitive inhibitor that showed a time-dependent onset of inhibition and disrupted the oligomerization state of PHGDH. The identification of a small molecule inhibitor of PHGDH not only enables thorough preclinical evaluation of PHGDH as a target in cancers, but also provides a tool with which to study serine metabolism.


Assuntos
Neoplasias/metabolismo , Fosfoglicerato Desidrogenase/antagonistas & inibidores , Serina/biossíntese , Linhagem Celular Tumoral , Proliferação de Células , Humanos , Neoplasias/patologia
2.
Biochemistry ; 57(47): 6604-6614, 2018 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-30365304

RESUMO

Pancreatic cancer cells are characterized by deregulated metabolic programs that facilitate growth and resistance to oxidative stress. Among these programs, pancreatic cancers preferentially utilize a metabolic pathway through the enzyme aspartate aminotransferase 1 [also known as glutamate oxaloacetate transaminase 1 (GOT1)] to support cellular redox homeostasis. As such, small molecule inhibitors that target GOT1 could serve as starting points for the development of new therapies for pancreatic cancer. We ran a high-throughput screen for inhibitors of GOT1 and identified a small molecule, iGOT1-01, with in vitro GOT1 inhibitor activity. Application in pancreatic cancer cells revealed metabolic and growth inhibitory activity reflecting a promiscuous inhibitory profile. We then performed an in silico docking analysis to study inhibitor-GOT1 interactions with iGOT1-01 analogues that possess improved solubility and potency properties. These results suggested that the GOT1 inhibitor competed for binding to the pyridoxal 5-phosphate (PLP) cofactor site of GOT1. To analyze how the GOT1 inhibitor bound to GOT1, a series of GOT1 mutant enzymes that abolished PLP binding were generated. Application of the mutants in X-ray crystallography and thermal shift assays again suggested but were unable to formally conclude that the GOT1 inhibitor bound to the PLP site. Mutational studies revealed the relationship between PLP binding and the thermal stability of GOT1 while highlighting the essential nature of several residues for GOT1 catalytic activity. Insight into the mode of action of GOT1 inhibitors may provide leads to the development of drugs that target redox balance in pancreatic cancer.


Assuntos
Aspartato Aminotransferase Citoplasmática/antagonistas & inibidores , Proliferação de Células/efeitos dos fármacos , Neoplasias do Colo/patologia , Inibidores Enzimáticos/farmacologia , Mutação , Neoplasias Pancreáticas/patologia , Aspartato Aminotransferase Citoplasmática/genética , Aspartato Aminotransferase Citoplasmática/metabolismo , Sítios de Ligação , Neoplasias do Colo/tratamento farmacológico , Neoplasias do Colo/metabolismo , Cristalografia por Raios X , Inibidores Enzimáticos/química , Ensaios de Triagem em Larga Escala , Humanos , Metabolômica , Modelos Moleculares , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Conformação Proteica , Relação Estrutura-Atividade , Células Tumorais Cultivadas
3.
Blood ; 115(23): 4824-33, 2010 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-20348394

RESUMO

On-patent and off-patent drugs with previously unrecognized anticancer activity could be rapidly repurposed for this new indication given their prior toxicity testing. To identify such compounds, we conducted chemical screens and identified the antihelmintic flubendazole. Flubendazole induced cell death in leukemia and myeloma cell lines and primary patient samples at nanomolar concentrations. Moreover, it delayed tumor growth in leukemia and myeloma xenografts without evidence of toxicity. Mechanistically, flubendazole inhibited tubulin polymerization by binding tubulin at a site distinct from vinblastine. In addition, cells resistant to vinblastine because of overexpression of P-glycoprotein remained fully sensitive to flubendazole, indicating that flubendazole can overcome some forms of vinblastine resistance. Given the different mechanisms of action, we evaluated the combination of flubendazole and vinblastine in vitro and in vivo. Flubendazole synergized with vinblastine to reduce the viability of OCI-AML2 cells. In addition, combinations of flubendazole with vinblastine or vincristine in a leukemia xenograft model delayed tumor growth more than either drug alone. Therefore, flubendazole is a novel microtubule inhibitor that displays preclinical activity in leukemia and myeloma.


Assuntos
Antinematódeos/farmacologia , Leucemia/tratamento farmacológico , Mebendazol/análogos & derivados , Microtúbulos/metabolismo , Mieloma Múltiplo/tratamento farmacológico , Alcaloides de Vinca/farmacologia , Animais , Antinematódeos/agonistas , Antinematódeos/uso terapêutico , Antineoplásicos Fitogênicos/agonistas , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Morte Celular , Sobrevivência Celular , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Células HeLa , Humanos , Leucemia/metabolismo , Masculino , Mebendazol/agonistas , Mebendazol/farmacologia , Mebendazol/uso terapêutico , Camundongos , Mieloma Múltiplo/metabolismo , Células U937 , Vimblastina/agonistas , Vimblastina/farmacologia , Vimblastina/uso terapêutico , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
4.
Blood ; 115(11): 2251-9, 2010 Mar 18.
Artigo em Inglês | MEDLINE | ID: mdl-20075161

RESUMO

The proteasomal pathway of protein degradation involves 2 discrete steps: ubiquitination and degradation. Here, we evaluated the effects of inhibiting the ubiquitination pathway at the level of the ubiquitin-activating enzyme UBA1 (E1). By immunoblotting, leukemia cell lines and primary patient samples had increased protein ubiquitination. Therefore, we examined the effects of genetic and chemical inhibition of the E1 enzyme. Knockdown of E1 decreased the abundance of ubiquitinated proteins in leukemia and myeloma cells and induced cell death. To further investigate effects of E1 inhibition in malignancy, we discovered a novel small molecule inhibitor, 3,5-dioxopyrazolidine compound, 1-(3-chloro-4-fluorophenyl)-4-[(5-nitro-2-furyl)methylene]-3,5-pyrazolidinedione (PYZD-4409). PYZD-4409 induced cell death in malignant cells and preferentially inhibited the clonogenic growth of primary acute myeloid leukemia cells compared with normal hematopoietic cells. Mechanistically, genetic or chemical inhibition of E1 increased expression of E1 stress markers. Moreover, BI-1 overexpression blocked cell death after E1 inhibition, suggesting ER stress is functionally important for cell death after E1 inhibition. Finally, in a mouse model of leukemia, intraperitoneal administration of PYZD-4409 decreased tumor weight and volume compared with control without untoward toxicity. Thus, our work highlights the E1 enzyme as a novel target for the treatment of hematologic malignancies.


Assuntos
Leucemia/enzimologia , Leucemia/terapia , Mieloma Múltiplo/enzimologia , Mieloma Múltiplo/terapia , Enzimas Ativadoras de Ubiquitina/metabolismo , Animais , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ciclina D3/metabolismo , Modelos Animais de Doenças , Ensaios de Seleção de Medicamentos Antitumorais , Retículo Endoplasmático/efeitos dos fármacos , Retículo Endoplasmático/patologia , Inibidores Enzimáticos/farmacologia , Técnicas de Silenciamento de Genes , Sistema Hematopoético/citologia , Sistema Hematopoético/efeitos dos fármacos , Humanos , Camundongos , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Bibliotecas de Moléculas Pequenas/farmacologia , Estresse Fisiológico/efeitos dos fármacos , Fatores de Tempo , Proteína Supressora de Tumor p53/metabolismo , Enzimas Ativadoras de Ubiquitina/antagonistas & inibidores , Ubiquitinação/efeitos dos fármacos
5.
Blood ; 112(3): 760-9, 2008 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-18502826

RESUMO

D-cyclins are regulators of cell division that act in a complex with cyclin-dependent kinases to commit cells to a program of DNA replication. D-cyclins are overexpressed in many tumors, including multiple myeloma and leukemia, and contribute to disease progression and chemoresistance. To better understand the role and impact of D-cyclins in hematologic malignancies, we conducted a high throughput screen for inhibitors of the cyclin D2 promoter and identified the drug cyproheptadine. In myeloma and leukemia cells, cyproheptadine decreased expression of cyclins D1, D2, and D3 and arrested these cells in the G(0)/G(1) phase. After D-cyclin suppression, cyproheptadine induced apoptosis in myeloma and leukemia cell lines and primary patient samples preferentially over normal hematopoietic cells. In mouse models of myeloma and leukemia, cyproheptadine inhibited tumor growth without significant toxicity. Cyproheptadine-induced apoptosis was preceded by activation of the mitochondrial pathway of caspase activation and was independent of the drug's known activity as an H1 histamine and serotonin receptor antagonist. Thus, cyproheptadine represents a lead for a novel therapeutic agent for the treatment of malignancy. Because the drug is well tolerated and already approved in multiple countries for clinical use as an antihistamine and appetite stimulant, it could be moved directly into clinical trials for cancer.


Assuntos
Ciclinas/genética , Ciproeptadina/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Leucemia Mieloide Aguda/tratamento farmacológico , Mieloma Múltiplo/tratamento farmacológico , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular , Linhagem Celular Tumoral , Ciclina D1/genética , Ciclina D2 , Ciclina D3 , Ciproeptadina/uso terapêutico , Ensaios de Seleção de Medicamentos Antitumorais , Humanos , Leucemia Mieloide Aguda/patologia , Camundongos , Mieloma Múltiplo/patologia
6.
Apoptosis ; 13(6): 748-55, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18415680

RESUMO

Quinolines are a class of chemical compounds with emerging anti-cancer properties. Here, we tested the activity of series of quinolines and quinoline-like molecules for anti-cancer activity and identified a novel diquinoline, 1-methyl-2-[3-(1-methyl-1,2-dihydroquinolin-2-yliden)prop-1-enyl]quinolinium iodide (Q(2)). Q(2 )induced cell death in leukemia, myeloma, and solid tumor cell lines with LD50s in the low to submicromolar range. Moreover, Q(2) induced cell death in primary acute myeloid leukemia (AML) cells preferentially over normal hematopoietic cells. In a mouse model of leukemia, Q(2) delayed tumor growth. Mechanistically, Q(2) induced cell death through caspase independent mechanisms. By electron microscopy, Q(2) increased cytoplasmic vacuolization and mitochondrial swelling. Potentially consistent with the induction of autophagic cell death, Q(2) treatment led to a punctate distribution of LC3 and increased MDC staining. Thus, Q(2) is a novel quinolinium with preclinical activity in malignancies such as leukemia and myeloma and warrants further investigation.


Assuntos
Morte Celular/efeitos dos fármacos , Compostos de Quinolínio/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Animais , Autofagia/efeitos dos fármacos , Caspases/fisiologia , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Células HeLa , Humanos , Células Jurkat , Dose Letal Mediana , Leucemia Linfocítica Crônica de Células B/tratamento farmacológico , Leucemia Mieloide Aguda/tratamento farmacológico , Camundongos , Camundongos Endogâmicos DBA , Compostos de Quinolínio/uso terapêutico , Células Tumorais Cultivadas , Células U937
7.
J Alzheimers Dis ; 14(1): 69-84, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18525129

RESUMO

Dysregulation of iron homeostasis is implicated in Alzheimer's disease (AD). In this pilot study, common variants of the apolipoprotein E (APOE) and HFE genes resulting in the iron overload disorder of hereditary hemochromatosis (C282Y, H63D and S65C) were evaluated as factors in sporadic AD in an Ontario sample in which folic acid fortification has been mandatory since 1998. Laboratory studies also were done to search for genetic effects on blood markers of iron status, red cell folates and serum B12. Participants included 58 healthy volunteers (25 males, 33 females) and 54 patients with probable AD (20 males, 34 females). Statistical analyses were interpreted at the 95% confidence level. Contingency table and odds ratio analyses supported the hypothesis that in females of the given age range, E4 significantly predisposed to AD in the presence but not absence of H63D. In males, E4 significantly predisposed to AD in the absence of H63D, and H63D in the absence of E4 appeared protective against AD. Among E4+ AD patients, H63D was associated with significant lowering of red cell folate concentration, possibly as the result of excessive oxidative stress. However, folate levels in the lowest population quartile did not affect the risk of AD. A model is presented to explain the experimental findings.


Assuntos
Alelos , Doença de Alzheimer/genética , Apolipoproteína E4/genética , Análise Mutacional de DNA , Ácido Fólico/administração & dosagem , Variação Genética/genética , Hemocromatose/genética , Antígenos de Histocompatibilidade Classe I/genética , Sobrecarga de Ferro/genética , Proteínas de Membrana/genética , Idoso de 80 Anos ou mais , Doença de Alzheimer/sangue , Doença de Alzheimer/tratamento farmacológico , Biomarcadores/sangue , Eritrócitos/metabolismo , Feminino , Ácido Fólico/sangue , Predisposição Genética para Doença/genética , Genótipo , Hemocromatose/sangue , Proteína da Hemocromatose , Humanos , Sobrecarga de Ferro/sangue , Masculino , Entrevista Psiquiátrica Padronizada , Pessoa de Meia-Idade , Ontário , Fatores Sexuais , Vitamina B 12/sangue
8.
PLoS One ; 7(9): e43721, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22957033

RESUMO

Oncogenic signaling promotes tumor invasion and metastasis, in part, by increasing the expression of tri- and tetra- branched N-glycans. The branched N-glycans bind to galectins forming a multivalent lattice that enhances cell surface residency of growth factor receptors, and focal adhesion turnover. N-acetylglucosaminyltransferase I (MGAT1), the first branching enzyme in the pathway, is required for the addition of all subsequent branches. Here we have introduced MGAT1 shRNA into human HeLa cervical and PC-3-Yellow prostate tumor cells lines, generating cell lines with reduced transcript, enzyme activity and branched N-glycans at the cell surface. MGAT1 knockdown inhibited HeLa cell migration and invasion, but did not alter cell proliferation rates. Swainsonine, an inhibitor of α-mannosidase II immediately downstream of MGAT1, also inhibited cell invasion and was not additive with MGAT1 shRNA, consistent with a common mechanism of action. Focal adhesion and microfilament organization in MGAT1 knockdown cells also indicate a less motile phenotype. In vivo, MGAT1 knockdown in the PC-3-Yellow orthotopic prostate cancer xenograft model significantly decreased primary tumor growth and the incidence of lung metastases. Our results demonstrate that blocking MGAT1 is a potential target for anti-cancer therapy.


Assuntos
N-Acetilglucosaminiltransferases/genética , Neoplasias/genética , Neoplasias/terapia , RNA Interferente Pequeno/metabolismo , Animais , Linhagem Celular Tumoral , Membrana Celular/metabolismo , Progressão da Doença , Inibidores Enzimáticos/farmacologia , Feminino , Células HeLa , Humanos , Masculino , Camundongos , N-Acetilglucosaminiltransferases/metabolismo , Metástase Neoplásica , Transplante de Neoplasias , Fenótipo , Polissacarídeos/química , Neoplasias da Próstata/genética , Neoplasias da Próstata/terapia , Transdução de Sinais , Swainsonina/farmacologia
9.
Cancer Res ; 69(7): 2739-47, 2009 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-19293189

RESUMO

Normal epithelial cells undergo apoptosis upon detachment from the extracellular matrix, a process termed "anoikis." However, malignant epithelial cells with metastatic potential resist anoikis and can survive in an anchorage-independent fashion. Molecules that sensitize resistant cells to anoikis will be useful chemical probes to understand this pathway. To identify novel anoikis sensitizers in anoikis-resistant PPC-1 prostate adenocarcinoma cells, a library of 2,000 off-patent drugs and natural products was screened for their ability to preferentially induce cell death in suspension over adherent culture conditions. This screen identified five members of the family of cardiac glycosides as anoikis sensitizers, including ouabain, peruvoside, digoxin, digitoxin, and strophanthidin. We conducted further studies with ouabain to discern the mechanism of cardiac glycoside-induced anoikis sensitization. Ouabain initiated anoikis through the mitochondrial pathway of caspase activation. In addition, ouabain sensitized cells to anoikis by inhibiting its known target, the Na(+)/K(+) ATPase pump, and inducing hypoosmotic stress. Resistance to anoikis permits cancer cells to survive in the circulation and facilitates their metastasis to distant organs, so we tested the effects of Na(+)/K(+) ATPase inhibition on distant tumor formation in mouse models. In these mouse models, ouabain inhibited tumor metastases but did not alter the growth of subcutaneous tumors. Thus, we have identified a novel mechanism to sensitize resistant cells to anoikis and decrease tumor metastasis. These results suggest a potential mechanism for the observed clinical reduction in metastasis and relapse in breast cancer patients who have undergone treatments with cardiac glycosides.


Assuntos
Anoikis/efeitos dos fármacos , Glicosídeos Cardíacos/farmacologia , Inibidores Enzimáticos/farmacologia , Neoplasias/tratamento farmacológico , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Animais , Anoikis/fisiologia , Inibidores de Caspase , Caspases/metabolismo , Linhagem Celular Tumoral , Ensaios de Seleção de Medicamentos Antitumorais , Ativação Enzimática/efeitos dos fármacos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Metástase Neoplásica , Neoplasias/enzimologia , Neoplasias/patologia , Pressão Osmótica/efeitos dos fármacos , Ouabaína/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Cancer Res ; 68(16): 6688-97, 2008 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-18701493

RESUMO

Aberrant N-linked glycans promote the malignant potential of cells by enhancing the epithelial-to-mesenchymal transition and the invasive phenotype. To identify small molecule inhibitors of N-glycan biosynthesis, we developed a chemical screen based on the ability of the tetravalent plant lectin L-phytohemagglutinin (L-PHA) to bind and crosslink surface glycoproteins with beta1,6GlcNAc-branched complex type N-glycans and thereby induce agglutination and cell death. In this screen, Jurkat cells were treated with a library of off-patent chemicals (n = 1,280) to identify molecules that blocked L-PHA-induced death. The most potent hit from this screen was the cardiac glycoside (CG) dihydroouabain. In secondary assays, a panel of CGs was tested for their effects on L-PHA-induced agglutination and cell death. All of the CGs tested inhibited L-PHA-induced death in Jurkat cells, and the most potent CG tested was digoxin with an EC(50) of 60 +/- 20 nmol/L. Digoxin also increased the fraction of some concanavalin A-binding N-glycans. Using matrix-assisted laser desorption/ionization time-of-flight mass spectrometry, digoxin specifically increased GlcNAc(1)Man(3)GlcNAc(2)Fuc(1) and GlcNAc(2)Man(3)GlcNAc(2)Fuc(1) oligosaccharides demonstrating an impairment of the N-glycan pathway. Consistent with this effect on the N-glycan pathway, digoxin inhibited N-glycosylation-mediated processes of tumor cell migration and invasion. Furthermore, digoxin prevented distant tumor formation in two mouse models of metastatic prostate cancer. Thus, taken together, our high throughput screen identified CGs as modifiers of the N-glycan pathway. These molecules can be used as tools to better understand the role of N-glycans in normal and malignant cells. Moreover, these results may partly explain the anticancer effect of CGs in cardiovascular patients.


Assuntos
Glicosídeos Digitálicos/farmacologia , Inibidores Enzimáticos/farmacologia , Neoplasias/tratamento farmacológico , Polissacarídeos/metabolismo , ATPase Trocadora de Sódio-Potássio/antagonistas & inibidores , Animais , Movimento Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Técnicas de Química Combinatória , Glicopeptídeos/metabolismo , Glicosilação , Humanos , Masculino , Camundongos , Camundongos SCID , Neoplasias/enzimologia , Neoplasias/patologia , Fito-Hemaglutininas/farmacologia , RNA Interferente Pequeno/farmacologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Espectrometria de Massas por Ionização e Dessorção a Laser Assistida por Matriz , Transfecção , Células Tumorais Cultivadas , Cicatrização
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA