Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 16(13)2024 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-39001392

RESUMO

Rapidly proliferative processes in mammalian tissues including tumorigenesis and embryogenesis rely on the glycolytic pathway for energy and biosynthetic precursors. The enzyme 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-3 (PFKFB3) plays an important regulatory role in glycolysis by activating the key rate-limiting glycolytic enzyme, 6-phosphofructo-1-kinase (PFK-1). We have previously determined that decreased PFKFB3 expression reduced glycolysis and growth in transformed cells in vitro and suppressed xenograft growth in vivo. In earlier studies, we created a constitutive knockout mouse to interrogate the function of PFKFB3 in vivo but failed to generate homozygous offspring due to the requirement for PFKFB3 for embryogenesis. We have now developed a novel transgenic mouse model that exhibits inducible homozygous pan-tissue Pfkfb3 gene deletion (Pfkfb3fl/fl). We have induced Pfkfb3 genomic deletion in these mice and found that it effectively decreased PFKFB3 expression and activity. To evaluate the functional consequences of Pfkfb3 deletion in vivo, we crossed Cre-bearing Pfkfb3fl/fl mice with oncogene-driven tumor models and found that Pfkfb3 deletion markedly decreased their glucose uptake and growth. In summary, our studies reveal a critical regulatory function for PFKFB3 in glycolysis and tumorigenesis in vivo and characterize an effective and powerful model for further investigation of its role in multiple biological processes.

2.
Cells ; 10(7)2021 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-34359849

RESUMO

Tyrosine kinase inhibitors (TKIs) targeting the kinase domain of the epidermal growth factor receptor (EGFR), such as erlotinib, have dramatically improved clinical outcomes of patients with EGFR-driven non-small cell lung carcinomas (NSCLCs). However, intrinsic or acquired resistance remains a clinical barrier to the success of FDA-approved EGFR TKIs. Multiple mechanisms of resistance have been identified, including the activation of prosurvival autophagy. We have previously shown that the expression and activity of PFKFB3-a known driver of glycolysis-is associated with resistance to erlotinib and that PFKFB3 inhibition improves the response of NSCLC cells to erlotinib. This study focuses on investigating the role of PFKFB3 in regulating erlotinib-driven autophagy to escape resistance to erlotinib. We evaluated the consequence of pharmacological inhibition of PFKFB3 on erlotinib-driven autophagy in NSCLC cells with different mutation statuses. Here, we identify PFKFB3 as a mediator of erlotinib-induced autophagy in NSCLCs. We demonstrate that PFKFB3 inhibition sensitizes NCSLCs to erlotinib via impairing autophagy flux. In summary, our studies uncovered a novel crosstalk between PFKFB3 and EGFR that regulates erlotinib-induced autophagy, thus contributing to erlotinib sensitivity in NSCLCs.


Assuntos
Autofagia , Carcinoma Pulmonar de Células não Pequenas/patologia , Cloridrato de Erlotinib/farmacologia , Neoplasias Pulmonares/patologia , Fosfofrutoquinase-2/antagonistas & inibidores , Adenilato Quinase/metabolismo , Autofagossomos/efeitos dos fármacos , Autofagossomos/metabolismo , Autofagia/efeitos dos fármacos , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cloroquina/farmacologia , Ativação Enzimática/efeitos dos fármacos , Receptores ErbB/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Neoplasias Pulmonares/metabolismo , Proteínas Associadas aos Microtúbulos/metabolismo , Proteínas Mutantes/metabolismo , Fosfofrutoquinase-2/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Proteína Sequestossoma-1/metabolismo
3.
Breast Cancer Res Treat ; 186(3): 677-686, 2021 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-33599863

RESUMO

PURPOSE: The management of triple-negative breast cancer (TNBC) remains a significant clinical challenge due to the lack of effective targeted therapies. Inhibitors of the cyclin-dependent kinases 4 and 6 (CDK4/6) are emerging as promising therapeutic agents against TNBC; however, cells can rapidly acquire resistance through multiple mechanisms that are yet to be identified. Therefore, determining the mechanisms underlying resistance to CDK4/6 inhibition is crucial to develop combination therapies that can extend the efficacy of the CDK4/6 inhibitors or delay resistance. This study aims to identify differentially expressed genes (DEG) associated with acquired resistance to palbociclib in ER- breast cancer cells. METHODS: We performed next-generation transcriptomic sequencing (RNA-seq) and pathway analysis in ER- MDA-MB-231 palbociclib-sensitive (231/pS) and palbociclib-resistant (231/pR) cells. RESULTS: We identified 2247 up-regulated and 1427 down-regulated transcripts in 231/pR compared to 231/pS cells. DEGs were subjected to functional analysis using Gene Ontology (GO) and the KEGG database which identified many transduction pathways associated with breast cancer, including the PI3K/AKT, PTEN and mTOR pathways. Additionally, Ingenuity Pathway Analysis (IPA) revealed that resistance to palbociclib is closely associated with altered cholesterol and fatty acid biosynthesis suggesting that resistance to palbociclib may be dependent on lipid metabolic reprograming. CONCLUSION: This study provides evidence that lipid metabolism is altered in TNBC with acquired resistance to palbociclib. Further studies are needed to determine if the observed lipid metabolic rewiring can be exploited to overcome therapy resistance in TNBC.


Assuntos
Neoplasias da Mama , Neoplasias de Mama Triplo Negativas , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Humanos , Fosfatidilinositol 3-Quinases , Piperazinas , Piridinas , RNA-Seq , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/genética
4.
Genes (Basel) ; 11(4)2020 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-32344635

RESUMO

Acquired resistance to cyclin-dependent kinases 4 and 6 (CDK4/6) inhibition in estrogen receptor-positive (ER+) breast cancer remains a significant clinical challenge. Efforts to uncover the mechanisms underlying resistance are needed to establish clinically actionable targets effective against resistant tumors. In this study, we sought to identify differentially expressed genes (DEGs) associated with acquired resistance to palbociclib in ER+ breast cancer. We performed next-generation transcriptomic RNA sequencing (RNA-seq) and pathway analysis in ER+ MCF7 palbociclib-sensitive (MCF7/pS) and MCF7 palbociclib-resistant (MCF7/pR) cells. We identified 2183 up-regulated and 1548 down-regulated transcripts in MCF7/pR compared to MCF7/pS cells. Functional analysis of the DEGs using Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) database identified several pathways associated with breast cancer, including 'cell cycle', 'DNA replication', 'DNA repair' and 'autophagy'. Additionally, Ingenuity Pathway Analysis (IPA) revealed that resistance to palbociclib is closely associated with deregulation of several key canonical and metabolic pathways. Further studies are needed to determine the utility of these DEGs and pathways as therapeutics targets against ER+ palbociclib-resistant breast cancer.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias da Mama/genética , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Piperazinas/farmacologia , Piridinas/farmacologia , Receptores de Estrogênio/metabolismo , Transcriptoma/efeitos dos fármacos , Antineoplásicos/farmacologia , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/metabolismo , Neoplasias da Mama/patologia , Biologia Computacional , Feminino , Humanos , Células Tumorais Cultivadas
5.
Cancers (Basel) ; 11(5)2019 May 16.
Artigo em Inglês | MEDLINE | ID: mdl-31100952

RESUMO

While clinical responses to palbociclib have been promising, metastatic breast cancer remains incurable due to the development of resistance. We generated estrogen receptor-positive (ER+) and ER-negative (ER-) cell line models and determined their permissiveness and cellular responses to an oncolytic adenovirus (OAd) known as Ad5/3-delta24. Analysis of ER+ and ER- palbociclib-resistant cells revealed two clearly distinguishable responses to the OAd. While ER+ palbociclib-resistant cells displayed a hypersensitive phenotype to the effects of the OAd, ER- palbociclib-resistant cells showed a resistant phenotype to the OAd. Hypersensitivity to the OAd in ER+ palbociclib-resistant cells correlated with a decrease in type I interferon (IFN) signaling, an increase in viral entry receptor expression, and an increase in cyclin E expression. OAd resistance in ER- palbociclib-resistant cells correlated with an increase in type I IFN signaling and a marked decrease in viral entry receptor. Using the OAd as monotherapy caused significant cytotoxicity to both ER+ and ER- palbociclib-sensitive cell lines. However, the addition of palbociclib increased the oncolytic activity of the OAd only in ER+ palbociclib-sensitive cells. Our studies provide a mechanistic base for a novel anti-cancer regimen composed of an OAd in combination with palbociclib for the treatment of ER+ breast cancer.

6.
PLoS One ; 10(8): e0134144, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26270345

RESUMO

Earlier observations indicate that free heme is selectively toxic to cells lacking heme oxygenase-1 (HO-1) but how this enzyme prevents heme toxicity remains unexplained. Here, using A549 (human lung cancer) and immortalized human bronchial epithelial cells incubated with exogenous heme, we find knock-down of HO-1 using siRNA does promote the accumulation of cell-associated heme and heme-induced cell death. However, it appears that the toxic effects of heme are exerted by "loose" (probably intralysosomal) iron because cytotoxic effects of heme are lessened by pre-incubation of HO-1 deficient cells with desferrioxamine (which localizes preferentially in the lysosomal compartment). Desferrioxamine also decreases lysosomal rupture promoted by intracellularly generated hydrogen peroxide. Supporting the importance of endogenous oxidant production, both chemical and siRNA inhibition of catalase activity predisposes HO-1 deficient cells to heme-mediated killing. Importantly, it appears that HO-1 deficiency somehow blocks the induction of ferritin; control cells exposed to heme show ~10-fold increases in ferritin heavy chain expression whereas in heme-exposed HO-1 deficient cells ferritin expression is unchanged. Finally, overexpression of ferritin H chain in HO-1 deficient cells completely prevents heme-induced cytotoxicity. Although two other products of HO-1 activity--CO and bilirubin--have been invoked to explain HO-1-mediated cytoprotection, we conclude that, at least in this experimental system, HO-1 activity triggers the induction of ferritin and the latter is actually responsible for the cytoprotective effects of HO-1 activity.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Heme/toxicidade , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Desferroxamina/farmacologia , Ferritinas/biossíntese , Técnicas de Silenciamento de Genes , Heme Oxigenase-1/genética , Humanos , Lisossomos/metabolismo , Lisossomos/patologia
7.
Oncotarget ; 6(20): 18001-11, 2015 Jul 20.
Artigo em Inglês | MEDLINE | ID: mdl-26221874

RESUMO

Human tumors exhibit increased glucose uptake and metabolism as a result of high demand for ATP and anabolic substrates and this metabolotype is a negative prognostic indicator for survival. Recent studies have demonstrated that cancer cells from several tissue origins and genetic backgrounds require the expression of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase 4 (PFKFB4), a regulatory enzyme that synthesizes an allosteric activator of glycolysis, fructose-2,6-bisphosphate. We report the discovery of a first-in-class PFKFB4 inhibitor, 5-(n-(8-methoxy-4-quinolyl)amino)pentyl nitrate (5MPN), using structure-based virtual computational screening. We find that 5MPN is a selective inhibitor of PFKFB4 that suppresses the glycolysis and proliferation of multiple human cancer cell lines but not non-transformed epithelial cells in vitro. Importantly, 5MPN has high oral bioavailability and per os administration of a non-toxic dose of 5MPN suppresses the glucose metabolism and growth of tumors in mice.


Assuntos
Aminoquinolinas/farmacologia , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Lewis/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Glicólise/efeitos dos fármacos , Neoplasias Pulmonares/tratamento farmacológico , Nitratos/farmacologia , Fosfofrutoquinase-2/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Administração Oral , Aminoquinolinas/administração & dosagem , Aminoquinolinas/química , Aminoquinolinas/farmacocinética , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacocinética , Disponibilidade Biológica , Carcinoma Pulmonar de Lewis/enzimologia , Carcinoma Pulmonar de Lewis/genética , Carcinoma Pulmonar de Lewis/patologia , Carcinoma Pulmonar de Células não Pequenas/enzimologia , Carcinoma Pulmonar de Células não Pequenas/genética , Carcinoma Pulmonar de Células não Pequenas/patologia , Proliferação de Células/efeitos dos fármacos , Desenho Assistido por Computador , Relação Dose-Resposta a Droga , Feminino , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Células HCT116 , Humanos , Neoplasias Pulmonares/enzimologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Estrutura Molecular , Terapia de Alvo Molecular , Nitratos/administração & dosagem , Nitratos/química , Nitratos/farmacocinética , Fosfofrutoquinase-2/genética , Fosfofrutoquinase-2/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Interferência de RNA , Relação Estrutura-Atividade , Transfecção , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
J Biol Chem ; 289(13): 9440-8, 2014 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-24515104

RESUMO

Estradiol (E2) administered to estrogen receptor-positive (ER(+)) breast cancer patients stimulates glucose uptake by tumors. Importantly, this E2-induced metabolic flare is predictive of the clinical effectiveness of anti-estrogens and, as a result, downstream metabolic regulators of E2 are expected to have utility as targets for the development of anti-breast cancer agents. The family of 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatases (PFKFB1-4) control glycolytic flux via their product, fructose-2,6-bisphosphate (F26BP), which activates 6-phosphofructo-1-kinase (PFK-1). We postulated that E2 might promote PFKFB3 expression, resulting in increased F26BP and glucose uptake. We demonstrate that PFKFB3 expression is highest in stage III lymph node metastases relative to normal breast tissues and that exposure of human MCF-7 breast cancer cells to E2 causes a rapid increase in [(14)C]glucose uptake and glycolysis that is coincident with an induction of PFKFB3 mRNA (via ER binding to its promoter), protein expression and the intracellular concentration of its product, F26BP. Importantly, selective inhibition of PFKFB3 expression and activity using siRNA or a PFKFB3 inhibitor markedly reduces the E2-mediated increase in F26BP, [(14)C]glucose uptake, and glycolysis. Furthermore, co-treatment of MCF-7 cells with the PFKFB3 inhibitor and the anti-estrogen ICI 182,780 synergistically induces apoptotic cell death. These findings demonstrate for the first time that the estrogen receptor directly promotes PFKFB3 mRNA transcription which, in turn, is required for the glucose metabolism and survival of breast cancer cells. Importantly, these results provide essential preclinical information that may allow for the ultimate design of combinatorial trials of PFKFB3 antagonists with anti-estrogen therapies in ER(+) stage IV breast cancer patients.


Assuntos
Estradiol/farmacologia , Glucose/metabolismo , Fosfofrutoquinase-2/metabolismo , Apoptose/efeitos dos fármacos , Transporte Biológico/efeitos dos fármacos , Neoplasias da Mama/patologia , Sobrevivência Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Estradiol/análogos & derivados , Receptor alfa de Estrogênio/metabolismo , Frutosedifosfatos/metabolismo , Fulvestranto , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Glicólise/efeitos dos fármacos , Humanos , Metástase Linfática , Células MCF-7 , Fosfofrutoquinase-2/antagonistas & inibidores , Fosfofrutoquinase-2/genética , Elementos de Resposta/efeitos dos fármacos , Elementos de Resposta/genética
9.
Biochem J ; 449(1): 189-94, 2013 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-22989377

RESUMO

Induction or ectopic overexpression of HO-1 (haem oxygenase 1) protects against a wide variety of disorders. These protective effects have been variably ascribed to generation of carbon monoxide (released during cleavage of the alpha-methene bridge of haem) and/or to production of the antioxidant bilirubin. We investigated HO-1-overexpressing A549 cells and find that, as expected, HO-1-overexpressing cells are resistant to killing by hydrogen peroxide. Surprisingly, these cells have approximately twice the normal amount of intracellular iron which usually tends to amplify oxidant killing. However, HO-1-overexpressing cells contain only ~25% as much 'loose' (probably redox active) iron. Indeed, inhibition of ferritin synthesis [via siRNA (small interfering RNA) directed at the ferritin heavy chain] sensitizes the HO-1-overexpressing cells to peroxide killing. It appears that HO-1 overexpression leads to enhanced destruction of haem, consequent 2-3-fold induction of ferritin, and compensatory increases in transferrin receptor expression and haem synthesis. However, there is no functional haem deficiency because cellular oxygen consumption and catalase activity are similar in both cell types. We conclude that, at least in many cases, the cytoprotective effects of HO-1 induction or forced overexpression may derive from elevated expression of ferritin and consequent reduction of redox active 'loose' iron.


Assuntos
Ferritinas/metabolismo , Regulação Enzimológica da Expressão Gênica , Heme Oxigenase-1/genética , Líquido Intracelular/metabolismo , Ferro/metabolismo , Linhagem Celular Tumoral , Ferritinas/biossíntese , Heme Oxigenase-1/biossíntese , Humanos
10.
Circulation ; 120(13): 1222-30, 2009 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-19752329

RESUMO

BACKGROUND: Although inducible nitric oxide synthase (iNOS) is known to impart powerful protection against myocardial infarction, the mechanism for this salubrious action remains unclear. METHODS AND RESULTS: Adenovirus-mediated iNOS gene transfer in mice resulted 48 to 72 hours later in increased expression not only of iNOS protein but also of heme oxygenase (HO)-1 mRNA and protein; HO-2 protein expression did not change. iNOS gene transfer markedly reduced infarct size in wild-type mice, but this effect was completely abrogated in HO-1(-/-) mice. At 48 hours after iNOS gene transfer, nuclear factor-kappaB was markedly activated. In transgenic mice with cardiomyocyte-restricted expression of a dominant negative mutant of IkappaBalpha (IkappaBalpha(S32A,S36A)), both basal HO-1 levels and upregulation of HO-1 by iNOS gene transfer were suppressed. Chromatin immunoprecipitation analysis of mouse hearts provided direct evidence that nuclear factor-kappaB subunits p50 and p65 were recruited to the HO-1 gene promoter (-468 to -459 bp) 48 hours after iNOS gene transfer. CONCLUSIONS: This study demonstrates for the first time the existence of a close functional coupling between cardiac iNOS and cardiac HO-1: iNOS upregulates HO-1 by augmenting nuclear factor-kappaB binding to the region of the HO-1 gene promoter from -468 to -459 bp, and HO-1 then mediates the cardioprotective effects of iNOS. These results also reveal an important role of nuclear factor-kappaB in both basal and iNOS-induced expression of cardiac HO-1. Collectively, the present findings significantly expand our understanding of the regulation of cardiac HO-1 and of the mechanism whereby iNOS exerts its cardioprotective actions.


Assuntos
Terapia Genética/métodos , Coração/fisiologia , Heme Oxigenase-1/metabolismo , Infarto do Miocárdio/terapia , NF-kappa B/metabolismo , Óxido Nítrico Sintase Tipo II/genética , Adenoviridae/genética , Animais , Bilirrubina/metabolismo , Temperatura Corporal , Feminino , Técnicas de Transferência de Genes , Frequência Cardíaca , Heme Oxigenase-1/genética , Proteínas I-kappa B/genética , Masculino , Camundongos , Camundongos Transgênicos , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/patologia , Inibidor de NF-kappaB alfa , Óxido Nítrico Sintase Tipo II/metabolismo , Regiões Promotoras Genéticas/fisiologia , RNA Mensageiro/metabolismo , Regulação para Cima/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...