Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 61
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
FEBS Open Bio ; 13(8): 1495-1506, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37151134

RESUMO

We have recently shown that IFNγ, produced during cancer therapy, induces expression of the Bcl3 proto-oncogene in ovarian cancer (OC) cells, resulting in their increased proliferation, migration, and invasion, but the mechanisms are unknown. Here, we demonstrate that the IFNγ-induced Bcl3 expression is dependent on JAK1 and STAT1 signaling, and on p65 NFκB. Furthermore, the IFNγ-induced Bcl3 expression is associated with an increased occupancy of Ser-727 phosphorylated STAT1 and acetylated histone H3 at the Bcl3 promoter. Our data indicate that Bcl3 promotes expression of the pro-inflammatory chemokine interleukin-8 (IL-8) in OC cells. These findings identify Bcl3 as a novel target of IFNγ/JAK1/STAT1 signaling and suggest that targeting the JAK1/STAT1 pathway may suppress IFNγ-induced Bcl3 expression in OC.


Assuntos
Interleucina-8 , Neoplasias Ovarianas , Humanos , Feminino , Interleucina-8/metabolismo , Transdução de Sinais , NF-kappa B/metabolismo , Interferon gama/farmacologia , Interferon gama/metabolismo , Neoplasias Ovarianas/genética , Fator de Transcrição STAT1/metabolismo , Janus Quinase 1/genética , Janus Quinase 1/metabolismo
2.
Cell Signal ; 97: 110400, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-35820543

RESUMO

Expression of the immune checkpoint programmed death ligand-1 (PD-L1) is increased in ovarian cancer (OC) and correlates with poor prognosis. Interferon-γ (IFNγ) induces PD-L1 expression in OC cells, resulting in their increased proliferation and tumor growth, but the mechanisms that regulate the PD-L1 expression in OC remain unclear. Here, we show that the IFNγ-induced PD-L1 expression in OC cells is associated with increased levels of STAT1, Tyr-701 pSTAT1 and Ser-727 pSTAT1. Suppression of JAK1 and STAT1 significantly decreases the IFNγ-induced PD-L1 expression in OC cells, and STAT1 overexpression increases the IFNγ-induced PD-L1 expression. In addition, IFNγ induces expression of the transcription factor interferon regulatory factor 1 (IRF1) and IRF1 suppression attenuates the IFNγ-induced gene and protein levels of PD-L1. Chromatin immunoprecipitation results show that IFNγ induces PD-L1 promoter acetylation and recruitment of STAT1, Ser-727 pSTAT1 and IRF1 in OC cells. Together, these findings demonstrate that the IFNγ-induced PD-L1 expression in OC cells is regulated by JAK1, STAT1, and IRF1 signaling, and suggest that targeting the JAK1/ STAT1/IRF1 pathway may provide a leverage to regulate the PD-L1 levels in ovarian cancer.


Assuntos
Antígeno B7-H1 , Neoplasias Ovarianas , Antígeno B7-H1/genética , Antígeno B7-H1/metabolismo , Carcinoma Epitelial do Ovário , Linhagem Celular Tumoral , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Fator Regulador 1 de Interferon/genética , Fator Regulador 1 de Interferon/metabolismo , Interferon gama/metabolismo , Interferon gama/farmacologia , Janus Quinase 1/metabolismo , Neoplasias Ovarianas/genética , Fator de Transcrição STAT1/metabolismo
3.
PLoS One ; 16(11): e0260400, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34807950

RESUMO

Heme is an essential cofactor for enzymes of the electron transport chain (ETC) and ATP synthesis in mitochondrial oxidative phosphorylation (OXPHOS). Heme also binds to and destabilizes Bach1, a transcription regulator that controls expression of several groups of genes important for glycolysis, ETC, and metastasis of cancer cells. Heme synthesis can thus affect pathways through which cells generate energy and precursors for anabolism. In addition, increased heme synthesis may trigger oxidative stress. Since many cancers are characterized by a high glycolytic rate regardless of oxygen availability, targeting glycolysis, ETC, and OXPHOS have emerged as a potential therapeutic strategy. Here, we report that enhancing heme synthesis through exogenous supplementation of heme precursor 5-aminolevulinic acid (ALA) suppresses oxidative metabolism as well as glycolysis and significantly reduces proliferation of both ovarian and breast cancer cells. ALA supplementation also destabilizes Bach1 and inhibits migration of both cell types. Our data indicate that the underlying mechanisms differ in ovarian and breast cancer cells, but involve destabilization of Bach1, AMPK activation, and induction of oxidative stress. In addition, there appears to be an inverse correlation between the activity of oxidative metabolism and ALA sensitivity. Promoting heme synthesis by ALA supplementation may thus represent a promising new anti-cancer strategy, particularly in cancers that are sensitive to altered redox signaling, or in combination with strategies that target the antioxidant systems or metabolic weaknesses of cancer cells.


Assuntos
Neoplasias da Mama/metabolismo , Heme/metabolismo , Neoplasias Ovarianas/metabolismo , Estresse Oxidativo , Vias Biossintéticas , Linhagem Celular Tumoral , Feminino , Glicólise , Humanos , Efeito Warburg em Oncologia
4.
Int J Biochem Cell Biol ; 141: 106093, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34626802

RESUMO

Interferon-γ (IFNγ) is a pleiotropic cytokine that has a crucial role in immune response and tumor immunity. Because of its anti-tumor effects, IFNγ has been used in cancer treatment. However, IFNγ also has tumor-promoting functions that are less well understood. Here, we show that IFNγ induces expression of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8) in ovarian cancer (OC) cells. The IFNγ-induced IL-8 expression is dependent on JAK1, STAT1, and p65 NFκB, and is associated with an increased occupancy of K314/315 acetylated p65 NFκB and Ser-727 phosphorylated STAT1 at the IL-8 promoter. Neutralization of IL-8 using anti-IL-8 antibody reduces IFNγ-induced migration of OC cells, and their invasion ability in 3D spheroids. Together, these findings identify IL-8 as a novel target induced by IFNγ/JAK1/STAT1/p65 NFκB signaling, and indicate that the IFNγ-induced IL-8 contributes to IFNγ pro-tumorigenic effects in ovarian cancer cells.


Assuntos
Neoplasias Ovarianas , Feminino , Humanos , Interferon gama , Interleucina-8 , Fator de Transcrição STAT1
5.
J Biol Chem ; 297(5): 101246, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34582893

RESUMO

Proliferating cells coordinate histone and DNA synthesis to maintain correct stoichiometry for chromatin assembly. Histone mRNA levels must be repressed when DNA replication is inhibited to prevent toxicity and genome instability due to free non-chromatinized histone proteins. In mammalian cells, replication stress triggers degradation of histone mRNAs, but it is unclear if this mechanism is conserved from other species. The aim of this study was to identify the histone mRNA decay pathway in the yeast Saccharomyces cerevisiae and determine the mechanism by which DNA replication stress represses histone mRNAs. Using reverse transcription-quantitative PCR and chromatin immunoprecipitation-quantitative PCR, we show here that histone mRNAs can be degraded by both 5' → 3' and 3' → 5' pathways; however, replication stress does not trigger decay of histone mRNA in yeast. Rather, replication stress inhibits transcription of histone genes by removing the histone gene-specific transcription factors Spt10p and Spt21p from histone promoters, leading to disassembly of the preinitiation complexes and eviction of RNA Pol II from histone genes by a mechanism facilitated by checkpoint kinase Rad53p and histone chaperone Asf1p. In contrast, replication stress does not remove SCB-binding factor transcription complex, another activator of histone genes, from the histone promoters, suggesting that Spt10p and Spt21p have unique roles in the transcriptional downregulation of histone genes during replication stress. Together, our data show that, unlike in mammalian cells, replication stress in yeast does not trigger decay of histone mRNAs but inhibits histone transcription.


Assuntos
Replicação do DNA , DNA Fúngico , Histona Acetiltransferases , Histonas , Regiões Promotoras Genéticas , RNA Fúngico , RNA Mensageiro , Proteínas de Saccharomyces cerevisiae , Saccharomyces cerevisiae , Fatores de Transcrição , Transcrição Gênica , DNA Fúngico/biossíntese , DNA Fúngico/genética , Histona Acetiltransferases/genética , Histona Acetiltransferases/metabolismo , Histonas/biossíntese , Histonas/genética , RNA Fúngico/biossíntese , RNA Fúngico/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Saccharomyces cerevisiae/genética , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/genética , Proteínas de Saccharomyces cerevisiae/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo
7.
Methods Mol Biol ; 2108: 101-106, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31939173

RESUMO

IFNγ is a pleiotropic cytokine that has both antitumor functions and pro-tumorigenic effects. Recent studies have shown that IFNγ induces expression of the immune checkpoint PD-L1 in ovarian cancer (OC) cells, resulting in their increased proliferation and tumor growth. Here, we tested the hypothesis that IFNγ induces migration of OC cells. Using the scratch wound healing assay, our results demonstrate that IFNγ promotes OC cell migration, thus adding to the complexities of IFNγ pro-tumorigenic mechanisms. This chapter describes analysis of the IFNγ-induced migration of OC cells by the wound healing assay followed by quantification of the obtained images using ImageJ software.


Assuntos
Movimento Celular , Interferon gama/metabolismo , Neoplasias Ovarianas/metabolismo , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Análise de Dados , Feminino , Humanos , Interferon gama/farmacologia , Imagem Molecular/métodos , Neoplasias Ovarianas/patologia , Software , Cicatrização
8.
Methods Mol Biol ; 2108: 107-115, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31939174

RESUMO

The pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8) induces proliferation and invasion of solid tumor cells. In many types of solid cancer, including triple-negative breast cancer (TNBC), the IL-8 expression is induced by proteasome inhibition. In this chapter, we describe a protocol for the analysis of TNBC cell invasion induced by IL-8 in response to proteasome inhibition by bortezomib (BZ). Using this approach, we show that BZ increases the invasion ability of TNBC cells, and that the BZ-increased TNBC cell invasion is suppressed by IκB kinase (IKK) inhibition, which also decreases the IL-8 expression. The experimental protocol includes the cell invasion assay, microscopic evaluation of the invading cells, and quantitative analysis of the obtained images. This protocol should be applicable also for measurement of chemokine-induced tumor cell invasion in other types of cancer cells.


Assuntos
Movimento Celular , Interleucina-8/metabolismo , Neoplasias de Mama Triplo Negativas/metabolismo , Biomarcadores , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células , Citocinas/metabolismo , Análise de Dados , Feminino , Humanos , Quinase I-kappa B/metabolismo , Interleucina-8/farmacologia , Imagem Molecular , Inibidores de Proteassoma/farmacologia , Software , Neoplasias de Mama Triplo Negativas/patologia
9.
Methods Mol Biol ; 2108: 117-124, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31939175

RESUMO

Ovarian cancer (OC) is the most common cause of cancer deaths among gynecological malignancies. OC ascites contain multicellular spheroid aggregates, which exhibit increased pro-survival signaling, invasive behavior, and chemotherapeutic resistance. OC cells are characterized by an increased expression of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8), which increases their survival and migration, thus contributing to OC metastasis and angiogenesis. While previous studies have shown that IL-8 increases proliferation of OC cells grown in monolayer cultures, the effect of IL-8 on proliferation of OC cells grown in 3D spheroids has not been investigated. The spheroid 3D culture assays have been particularly useful in translational research since they allow cell-to-cell interactions that resemble tumor growth in vivo, while allowing easy cell manipulations and visualization. Here, we used the 3D spheroid culture assay to investigate the effect of IL-8 on OC cell proliferation. Using this assay, our results show that IL-8 significantly increases proliferation of OC cells grown in 3D spheroids.


Assuntos
Interleucina-8/farmacologia , Biomarcadores , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Feminino , Humanos , Neoplasias Ovarianas/metabolismo , Neoplasias Ovarianas/patologia , Esferoides Celulares
10.
Methods Mol Biol ; 2108: 211-220, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31939183

RESUMO

Expression of the immune checkpoint programmed death ligand 1 (PD-L1, CD274) is increased in many types of cancer, including ovarian cancer (OC), but the mechanisms that regulate the PD-L1 expression are not fully understood. In addition to binding to PD-1 on T cells, thus inhibiting T cell-mediated antitumor responses, PD-L1 has also tumor-intrinsic effects that include increased cancer cell survival and proliferation, and that might be in part mediated by the intracellular PD-L1. In this chapter, we describe a protocol for the analysis of the intracellular PD-L1 protein levels in OC cells by immunoblotting. Our results show that interferon-γ (IFNγ) induces the intracellular levels of PD-L1 and the proto-oncogene Bcl3 in OC cells. However, the PD-L1 expression is significantly decreased in OC cells stably transfected with Bcl3 shRNA, demonstrating that the IFNγ-induced PD-L1 expression in OC cells is mediated by Bcl3. These data identify the IFNγ-Bcl3-PD-L1 axis as a novel therapeutic target in OC, and suggest that targeting Bcl3 may provide a novel strategy to regulate the PD-L1 expression, and especially the tumor-intrinsic PD-L1 effects mediated by the intracellular PD-L1 in OC cells.


Assuntos
Proteína 3 do Linfoma de Células B/genética , Antígeno B7-H1/metabolismo , Immunoblotting , Interferon gama/metabolismo , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , RNA Interferente Pequeno/genética , Proteína 3 do Linfoma de Células B/metabolismo , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Feminino , Expressão Gênica , Humanos , Immunoblotting/métodos , Interferon gama/farmacologia , Neoplasias Ovarianas/patologia , Proto-Oncogene Mas , Transfecção
11.
Methods Mol Biol ; 2108: 221-228, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31939184

RESUMO

Expression of programmed death ligand-1 (PD-L1, CD274) on cancer cells is regulated by interferon-γ (IFNγ) signaling as well as by epigenetic mechanisms. By binding to PD-1 on cytotoxic T cells, PD-L1 inhibits T cell-mediated antitumor responses, resulting in immune escape. This chapter describes analysis of the surface PD-L1 expression in ovarian cancer (OC) cells using flow cytometry (FC). Our data demonstrate that the surface PD-L1 expression in OC cells is induced by IFNγ as well as by the class I histone deacetylase (HDAC) inhibition by romidepsin, suggesting that class I HDAC inhibition might provide a useful strategy to modulate the PD-L1 levels on OC cells.


Assuntos
Antígeno B7-H1/metabolismo , Membrana Celular/metabolismo , Depsipeptídeos/farmacologia , Citometria de Fluxo , Interferon gama/metabolismo , Neoplasias Ovarianas/metabolismo , Antígeno B7-H1/genética , Linhagem Celular Tumoral , Separação Celular/métodos , Análise de Dados , Feminino , Citometria de Fluxo/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Interferon gama/farmacologia , Neoplasias Ovarianas/genética
12.
Methods Mol Biol ; 2108: 229-239, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31939185

RESUMO

The immune checkpoint molecule, programmed death ligand 1 (PD-L1; B7-H1, CD274), induces T cell apoptosis and tolerance, thus inhibiting the antitumor immunity. PD-L1 expression is increased in many types of cancer, including ovarian cancer (OC), and correlates with poor prognosis. However, the mechanisms that regulate the PD-L1 expression in cancer cells are incompletely understood. The transcriptional regulation of PD-L1 expression is orchestrated by several transcription factors, including NFκB. The human PD-L1 promoter contains five NFκB-binding sites. Interferon-γ (IFNγ) stimulation of OC cells induces p65, and particularly K314/315 acetylated p65 recruitment to all five NFκB-binding sites in PD-L1 promoter, resulting in increased PD-L1 expression. In this chapter, we describe a protocol that uses chromatin immunoprecipitation (ChIP) to analyze the transcriptional regulation of PD-L1 by measuring recruitment of NFκB p65 and K314/315 acetylated p65 to PD-L1 promoter in human OC cells.


Assuntos
Antígeno B7-H1/genética , Imunoprecipitação da Cromatina , Proteínas de Ligação a DNA/metabolismo , Regulação Neoplásica da Expressão Gênica , Neoplasias Ovarianas/genética , Neoplasias Ovarianas/metabolismo , Transcrição Gênica , Antígeno B7-H1/metabolismo , Sítios de Ligação , Técnicas de Cultura de Células , Linhagem Celular Tumoral , Imunoprecipitação da Cromatina/métodos , Feminino , Humanos , Interferon gama/metabolismo , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , Ligação Proteica , Reação em Cadeia da Polimerase em Tempo Real
13.
J Biol Chem ; 294(25): 9771-9786, 2019 06 21.
Artigo em Inglês | MEDLINE | ID: mdl-31073026

RESUMO

The DNA damage response (DDR) is an evolutionarily conserved process essential for cell survival. Previously, we found that decreased histone expression induces mitochondrial respiration, raising the question whether the DDR also stimulates respiration. Here, using oxygen consumption and ATP assays, RT-qPCR and ChIP-qPCR methods, and dNTP analyses, we show that DDR activation in the budding yeast Saccharomyces cerevisiae, either by genetic manipulation or by growth in the presence of genotoxic chemicals, induces respiration. We observed that this induction is conferred by reduced transcription of histone genes and globally decreased DNA nucleosome occupancy. This globally altered chromatin structure increased the expression of genes encoding enzymes of tricarboxylic acid cycle, electron transport chain, oxidative phosphorylation, elevated oxygen consumption, and ATP synthesis. The elevated ATP levels resulting from DDR-stimulated respiration drove enlargement of dNTP pools; cells with a defect in respiration failed to increase dNTP synthesis and exhibited reduced fitness in the presence of DNA damage. Together, our results reveal an unexpected connection between respiration and the DDR and indicate that the benefit of increased dNTP synthesis in the face of DNA damage outweighs possible cellular damage due to increased oxygen metabolism.


Assuntos
Dano ao DNA , Nucleotídeos/metabolismo , Fosforilação Oxidativa , Consumo de Oxigênio , Proteínas de Saccharomyces cerevisiae/metabolismo , Saccharomyces cerevisiae/crescimento & desenvolvimento , Trifosfato de Adenosina/metabolismo , Sobrevivência Celular , Montagem e Desmontagem da Cromatina , Regulação Fúngica da Expressão Gênica , Histonas/metabolismo , Mitocôndrias/metabolismo , Saccharomyces cerevisiae/metabolismo
14.
Int J Mol Sci ; 19(11)2018 Oct 25.
Artigo em Inglês | MEDLINE | ID: mdl-30366365

RESUMO

Adenosine monophosphate (AMP)-activated protein kinase (AMPK) serves as an energy sensor and master regulator of metabolism. In general, AMPK inhibits anabolism to minimize energy consumption and activates catabolism to increase ATP production. One of the mechanisms employed by AMPK to regulate metabolism is protein acetylation. AMPK regulates protein acetylation by at least five distinct mechanisms. First, AMPK phosphorylates and inhibits acetyl-CoA carboxylase (ACC) and thus regulates acetyl-CoA homeostasis. Since acetyl-CoA is a substrate for all lysine acetyltransferases (KATs), AMPK affects the activity of KATs by regulating the cellular level of acetyl-CoA. Second, AMPK activates histone deacetylases (HDACs) sirtuins by increasing the cellular concentration of NAD⁺, a cofactor of sirtuins. Third, AMPK inhibits class I and II HDACs by upregulating hepatic synthesis of α-hydroxybutyrate, a natural inhibitor of HDACs. Fourth, AMPK induces translocation of HDACs 4 and 5 from the nucleus to the cytoplasm and thus increases histone acetylation in the nucleus. Fifth, AMPK directly phosphorylates and downregulates p300 KAT. On the other hand, protein acetylation regulates AMPK activity. Sirtuin SIRT1-mediated deacetylation of liver kinase B1 (LKB1), an upstream kinase of AMPK, activates LKB1 and AMPK. AMPK phosphorylates and inactivates ACC, thus increasing acetyl-CoA level and promoting LKB1 acetylation and inhibition. In yeast cells, acetylation of Sip2p, one of the regulatory ß-subunits of the SNF1 complex, results in inhibition of SNF1. This results in activation of ACC and reduced cellular level of acetyl-CoA, which promotes deacetylation of Sip2p and activation of SNF1. Thus, in both yeast and mammalian cells, AMPK/SNF1 regulate protein acetylation and are themselves regulated by protein acetylation.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Acetilcoenzima A/metabolismo , Acetilação , Animais , Epigenômica , Histona Desacetilases/genética , Histona Desacetilases/metabolismo , Humanos , Proteínas Serina-Treonina Quinases/genética
15.
J Biol Chem ; 293(40): 15483-15496, 2018 10 05.
Artigo em Inglês | MEDLINE | ID: mdl-30135206

RESUMO

The proto-oncogene Bcl3 induces survival and proliferation in cancer cells; however, its function and regulation in ovarian cancer (OC) remain unknown. Here, we show that Bcl3 expression is increased in human OC tissues. Surprisingly, however, we found that in addition to promoting survival, proliferation, and migration of OC cells, Bcl3 promotes both constitutive and interferon-γ (IFN)-induced expression of the immune checkpoint molecule PD-L1. The Bcl3 expression in OC cells is further increased by IFN, resulting in increased PD-L1 transcription. The mechanism consists of an IFN-induced, Bcl3- and p300-dependent PD-L1 promoter occupancy by Lys-314/315 acetylated p65 NF-κB. Blocking PD-L1 by neutralizing antibody reduces proliferation of OC cells overexpressing Bcl3, suggesting that the pro-proliferative effect of Bcl3 in OC cells is partly mediated by PD-L1. Together, this work identifies PD-L1 as a novel target of Bcl3, and links Bcl3 to IFNγ signaling and PD-L1-mediated immune escape.


Assuntos
Antígeno B7-H1/genética , Pontos de Checagem do Ciclo Celular/imunologia , Células Epiteliais/imunologia , Regulação Neoplásica da Expressão Gênica , Proteínas Proto-Oncogênicas/genética , Fatores de Transcrição/genética , Evasão Tumoral/genética , Anticorpos Neutralizantes/farmacologia , Apoptose/efeitos dos fármacos , Proteína 3 do Linfoma de Células B , Antígeno B7-H1/antagonistas & inibidores , Antígeno B7-H1/imunologia , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proteína p300 Associada a E1A , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/patologia , Feminino , Humanos , Interferon gama/farmacologia , Ovário/imunologia , Ovário/patologia , Regiões Promotoras Genéticas , Proto-Oncogene Mas , Proteínas Proto-Oncogênicas/imunologia , Transdução de Sinais , Fator de Transcrição RelA/genética , Fator de Transcrição RelA/imunologia , Fatores de Transcrição/imunologia , Transcrição Gênica
16.
Trends Pharmacol Sci ; 39(10): 867-878, 2018 10.
Artigo em Inglês | MEDLINE | ID: mdl-30150001

RESUMO

Metformin has been a frontline therapy for type 2 diabetes (T2D) for many years. Its effectiveness in T2D treatment is mostly attributed to its suppression of hepatic gluconeogenesis; however, the mechanistic aspects of metformin action remain elusive. In addition to its glucose-lowering effect, metformin possesses other pleiotropic health-promoting effects that include reduced cancer risk and tumorigenesis. Metformin inhibits the electron transport chain (ETC) and ATP synthesis; however, recent data reveal that metformin regulates AMP-activated protein kinase (AMPK) and the mechanistic target of rapamycin complex 1 (mTORC1) by multiple, mutually nonexclusive mechanisms that do not necessarily depend on the inhibition of ETC and the cellular ATP level. In this review, we discuss recent advances in elucidating the molecular mechanisms that are relevant for metformin use in cancer treatment.


Assuntos
Antineoplásicos/farmacologia , Hipoglicemiantes/farmacologia , Metformina/farmacologia , Proteínas Quinases Ativadas por AMP/metabolismo , Animais , Antineoplásicos/uso terapêutico , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Complexo I de Transporte de Elétrons/metabolismo , Humanos , Hipoglicemiantes/uso terapêutico , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Metformina/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/metabolismo
17.
PLoS One ; 13(8): e0201858, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30089134

RESUMO

Triple negative breast cancer (TNBC) cells express increased levels of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (IL-8, CXCL8), which promotes their proliferation and migration. Because TNBC patients are unresponsive to current targeted therapies, new therapeutic strategies are urgently needed. While proteasome inhibition by bortezomib (BZ) or carfilzomib (CZ) has been effective in treating hematological malignancies, it has been less effective in solid tumors, including TNBC, but the mechanisms are incompletely understood. Here we report that proteasome inhibition significantly increases expression of IL-8, and its receptors CXCR1 and CXCR2, in TNBC cells. Suppression or neutralization of the BZ-induced IL-8 potentiates the BZ cytotoxic and anti-proliferative effect in TNBC cells. The IL-8 expression induced by proteasome inhibition in TNBC cells is mediated by IκB kinase (IKK), increased nuclear accumulation of p65 NFκB, and by IKK-dependent p65 recruitment to IL-8 promoter. Importantly, inhibition of IKK activity significantly decreases proliferation, migration, and invasion of BZ-treated TNBC cells. These data provide the first evidence demonstrating that proteasome inhibition increases the IL-8 signaling in TNBC cells, and suggesting that IKK inhibitors may increase effectiveness of proteasome inhibitors in treating TNBC.


Assuntos
Antineoplásicos/farmacologia , Quinase I-kappa B/metabolismo , Interleucina-8/metabolismo , Inibidores de Proteassoma/farmacologia , Neoplasias de Mama Triplo Negativas/tratamento farmacológico , Neoplasias de Mama Triplo Negativas/metabolismo , Bortezomib/farmacologia , Morte Celular/efeitos dos fármacos , Morte Celular/fisiologia , Linhagem Celular Tumoral , Forma do Núcleo Celular/efeitos dos fármacos , Forma do Núcleo Celular/fisiologia , Quimioterapia Combinada , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Oligopeptídeos/farmacologia , Complexo de Endopeptidases do Proteassoma/metabolismo , Receptores de Interleucina-8A/metabolismo , Receptores de Interleucina-8B/metabolismo , Fator de Transcrição RelA/metabolismo
18.
Trends Pharmacol Sci ; 39(3): 295-306, 2018 03.
Artigo em Inglês | MEDLINE | ID: mdl-29233541

RESUMO

The rationale for developing histone deacetylase (HDAC) inhibitors (HDACi) as anticancer agents was based on their ability to induce apoptosis and cell cycle arrest in cancer cells. However, while HDACi have been remarkably effective in the treatment of hematological malignancies, clinical studies with HDACi as single agents in solid cancers have been disappointing. Recent studies have shown that, in addition to inducing apoptosis in cancer cells, class I HDACi induce IκB kinase (IKK)-dependent expression of proinflammatory chemokines, such as interleukin-8 (IL8; CXCL8), resulting in the increased proliferation of tumor cells, and limiting the effectiveness of HDACi in solid tumors. Here, we discuss the mechanisms responsible for HDACi-induced CXCL8 expression, and opportunities for combination therapies targeting HDACs and IKK in solid tumors.


Assuntos
Antineoplásicos/uso terapêutico , Inibidores de Histona Desacetilases/uso terapêutico , Quinase I-kappa B/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica , Apoptose/efeitos dos fármacos , Inibidores de Histona Desacetilases/administração & dosagem , Inibidores de Histona Desacetilases/farmacologia , Humanos , Quinase I-kappa B/metabolismo , Interleucina-8/metabolismo , Neoplasias/metabolismo , Inibidores de Proteínas Quinases/administração & dosagem , Inibidores de Proteínas Quinases/farmacologia
19.
Oncotarget ; 8(41): 70798-70810, 2017 Sep 19.
Artigo em Inglês | MEDLINE | ID: mdl-29050320

RESUMO

Although inhibitors of epigenetic regulators have been effective in the treatment of cutaneous T cell lymphoma (CTCL) and other hematopoietic malignancies, they have been less effective in solid tumors, including ovarian cancer (OC). We have previously shown that inhibition of histone deacetylase (HDAC) activity induces expression of the pro-inflammatory and pro-angiogenic chemokine interleukin-8 (CXCL8, IL-8) in OC cells, resulting in their increased survival and proliferation. Here, we show that in addition to ovarian cancer SKOV3, OVCAR3, and CAOV3 cells, HDAC inhibition induces the CXCL8 expression in HeLa cells, but not in CTCL Hut-78 cells. In OC cells, the CXCL8 expression is induced by pharmacological inhibition of class I HDACs. Interestingly, while an individual suppression of HDAC1, HDAC2, or HDAC3 by corresponding siRNAs inhibits the CXCL8 expression, their simultaneous suppression induces the CXCL8 expression. The induced CXCL8 expression in OC cells is dependent on histone acetyltransferase (HAT) activity of CREB-binding protein (CBP), but not p300, and is associated with HAT-dependent p65 recruitment to CXCL8 promoter. Together, our results show that the CXCL8 expression in OC cells is induced by combined inhibition of HDAC1, -2, and -3, and silenced by suppression of HAT activity of CBP. In addition, our data indicate that the induced CXCL8 expression may be responsible for the limited effectiveness of HDAC inhibitors in OC and perhaps other solid cancers characterized by CXCL8 overexpression, and suggest that targeting class I HDACs and CBP may provide novel combination strategies by limiting the induced CXCL8 expression.

20.
Oncotarget ; 8(21): 34030-34031, 2017 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-28504965
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...