Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
1.
Mol Pharmacol ; 91(3): 189-196, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-28003426

RESUMO

Hepatocellular carcinoma (HCC) is a common form of cancer with prevalence worldwide. There are many factors that lead to the development and progression of HCC. This study aimed to identify potential new tumor suppressors, examine their function as cell cycle modulators, and investigate their impact on the cyclin family of proteins and cyclin-dependent kinases (CDKs). In this study, the pyruvate dehydrogenase kinase (PDK)4 gene was shown to have potential tumor suppressor characteristics. PDK4 expression was significantly downregulated in human HCC. Pdk4-/- mouse liver exhibited a consistent increase in cell cycle regulator proteins, including cyclin D1, cyclin E1, cyclin A2, some associated CDKs, and transcription factor E2F1. PDK4-knockdown HCC cells also progressed faster through the cell cycle, which concurrently expressed high levels of cyclins and E2F1 as seen in the Pdk4-/- mice. Interestingly, the induced cyclin E1 and cyclin A2 caused by Pdk4 deficiency was repressed by arsenic treatment in mouse liver and in HCC cells. E2f1 deficiency in E2f1-/- mouse liver or knockdown E2F1 using small hairpin RNAs in HCC cells significantly decreased cyclin E1, cyclin A2, and E2F1 proteins. In contrast, inhibition of PDK4 activity in HCC cells increased cyclin E1, cyclin A2, and E2F1 proteins. These findings demonstrate that PDK4 is a critical regulator of hepatocyte proliferation via E2F1-mediated regulation of cyclins.


Assuntos
Ciclinas/metabolismo , Fator de Transcrição E2F1/metabolismo , Proteínas Quinases/deficiência , Animais , Arsênio/farmacologia , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Proliferação de Células , Progressão da Doença , Citometria de Fluxo , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/patologia , Fígado/efeitos dos fármacos , Fígado/patologia , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Masculino , Camundongos Endogâmicos C57BL , Proteínas Quinases/metabolismo , Análise de Sequência de RNA
2.
Toxicol Appl Pharmacol ; 304: 42-7, 2016 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-27217333

RESUMO

It is well established that increased liver cancer incidence is strongly associated with epigenetic silencing of tumor suppressor genes; the latter is contributed by the environmental exposure to arsenic. Pyruvate dehydrogenase kinase 4 (PDK4) is a mitochondrial protein that regulates the TCA cycle. However, the epigenetic mechanisms mediated by arsenic to control PDK4 expression remain elusive. In the present study, we showed that histone methyltransferase G9a- and Suv39H-mediated histone H3 lysine 9 (H3K9) methylations contributed to PDK4 silencing in hepatic cells. The PDK4 expression was induced by G9a inhibitor BRD4770 (BRD) and Suv39H inhibitor Chaetocin (CHA). In contrast, arsenic exposure decreased PDK4 expression by inducing G9a and increasing H3K9 di- and tri-methylations levels (H3K9me2/3). In addition, arsenic exposure antagonizes the effect of BRD by enhancing the enrichment of H3K9me2/3 in the PKD4 promoter. Moreover, knockdown of G9a using siRNA induced PDK4 expression in HCC cells. Furthermore, arsenic decreased hepatic PDK4 expression as well as diminished the induction of PDK4 by BRD in mouse liver and hepatocytes. Overall, the results suggest that arsenic causes aberrant repressive histone modification to silence PDK4 in both HCC cells and in mouse liver.


Assuntos
Arsênio/toxicidade , Histona-Lisina N-Metiltransferase/metabolismo , Histonas/metabolismo , Metiltransferases/metabolismo , Proteínas Quinases/biossíntese , Proteínas Repressoras/metabolismo , Animais , Benzamidas/farmacologia , Benzimidazóis/farmacologia , Sobrevivência Celular/efeitos dos fármacos , Humanos , Masculino , Metilação/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Piperazinas/farmacologia , RNA Interferente Pequeno
3.
Sci Adv ; 8(45): eabn6579, 2022 11 11.
Artigo em Inglês | MEDLINE | ID: mdl-36351019

RESUMO

Although major organ toxicities frequently arise in patients treated with cytotoxic or targeted cancer therapies, the mechanisms that drive them are poorly understood. Here, we report that vascular endothelial cells (ECs) are more highly primed for apoptosis than parenchymal cells across many adult tissues. Consequently, ECs readily undergo apoptosis in response to many commonly used anticancer agents including cytotoxic and targeted drugs and are more sensitive to ionizing radiation and BH3 mimetics than parenchymal cells in vivo. Further, using differentiated isogenic human induced pluripotent stem cell models of ECs and vascular smooth muscle cells (VSMCs), we find that these vascular cells exhibit distinct drug toxicity patterns, which are linked to divergent therapy-induced vascular toxicities in patients. Collectively, our results demonstrate that vascular cells are highly sensitive to apoptosis-inducing stress across life span and may represent a "weakest link" vulnerability in multiple tissues for development of toxicities.


Assuntos
Células-Tronco Pluripotentes Induzidas , Neoplasias , Adulto , Humanos , Músculo Liso Vascular/fisiologia , Células Endoteliais , Longevidade , Células-Tronco Pluripotentes Induzidas/fisiologia , Células Cultivadas , Neoplasias/etiologia
4.
Nat Commun ; 13(1): 5789, 2022 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-36184661

RESUMO

Immunoglobulin light chain (AL) amyloidosis is an incurable hematologic disorder typically characterized by the production of amyloidogenic light chains by clonal plasma cells. These light chains misfold and aggregate in healthy tissues as amyloid fibrils, leading to life-threatening multi-organ dysfunction. Here we show that the clonal plasma cells in AL amyloidosis are highly primed to undergo apoptosis and dependent on pro-survival proteins MCL-1 and BCL-2. Notably, this MCL-1 dependency is indirectly targeted by the proteasome inhibitor bortezomib, currently the standard of care for this disease and the related plasma cell disorder multiple myeloma, due to upregulation of pro-apoptotic Noxa and its inhibitory binding to MCL-1. BCL-2 inhibitors sensitize clonal plasma cells to multiple front-line therapies including bortezomib, dexamethasone and lenalidomide. Strikingly, in mice bearing AL amyloidosis cell line xenografts, single agent treatment with the BCL-2 inhibitor ABT-199 (venetoclax) produces deeper remissions than bortezomib and triples median survival. Mass spectrometry-based proteomic analysis reveals rewiring of signaling pathways regulating apoptosis, proliferation and mitochondrial metabolism between isogenic AL amyloidosis and multiple myeloma cells that divergently alter their sensitivity to therapies. These findings provide a roadmap for the use of BH3 mimetics to exploit endogenous and induced apoptotic vulnerabilities in AL amyloidosis.


Assuntos
Antineoplásicos , Amiloidose de Cadeia Leve de Imunoglobulina , Mieloma Múltiplo , Amiloide/uso terapêutico , Animais , Antineoplásicos/farmacologia , Bortezomib/farmacologia , Bortezomib/uso terapêutico , Compostos Bicíclicos Heterocíclicos com Pontes , Dexametasona/farmacologia , Dexametasona/uso terapêutico , Humanos , Cadeias Leves de Imunoglobulina , Amiloidose de Cadeia Leve de Imunoglobulina/tratamento farmacológico , Lenalidomida/farmacologia , Lenalidomida/uso terapêutico , Camundongos , Mieloma Múltiplo/tratamento farmacológico , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Inibidores de Proteassoma , Proteômica , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Sulfonamidas
5.
Liver Res ; 2(2): 100-107, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31815032

RESUMO

BACKGROUND AND AIM: Pyruvate dehydrogenase kinase 4 (PDK4) is a metabolism switch that regulates glucose oxidation and the tricarboxylic acid cycle (TCA cycle) in the mitochondria. Liver detoxifies xenobiotics and is constantly challenged by various injuries. This study aims at understanding how the loss of the metabolism regulator PDK4 contributes to liver injuries. METHODS: Wild-type (WT) and Pdk4 knockout (Pdk4 -/-) mice of different ages were examined for spontaneous hepatic apoptosis. Juvenile or adult mice of two genotypes were insulted by diethylnitrosamine (DEN), arsenic, galactosamine (GalN)/lipopolysaccharide (LPS), anti-CD95 (Jo2) antibody or carbon tetrachloride (CCl4). Liver injury was monitored by blood biochemistry test. Apoptosis was determined by terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) staining, poly (ADP-ribose) polymerase (PARP) cleavage, and caspase activity assay. Inflammatory response was determined by nuclear factor (NF)-κB activation and the activation of NF-κB target genes. Primary hepatocytes were isolated and cell viability was evaluated by MTS assay. RESULTS: We showed that systematic Pdk4 -/- in mice resulted in age-dependent spontaneous hepatic apoptosis. PDK4-deficiency increased the toxicity of DEN in juvenile mice, which correlated with a lethal consequence and massive hepatic apoptosis. Similarly, chronic arsenic administration induced more severe hepatic apoptosis in Pdk4 -/- mice compared to WT control mice. An aggravated hepatic NF-κB mediated-inflammatory response was observed in Pdk4 -/- mice livers. In vitro, Pdk4-deficient primary hepatocytes were more vulnerable to DEN and arsenic challenges and displayed higher caspase activity than wild type cells. Notably, hepatic PDK4 mRNA level was remarkably reduced during acute liver failure induced by GalN/LPS or Jo2 antibody. The diminished PDK4 expression was also observed in CCl4-induced acute liver injury. CONCLUSIONS: PDK4 may contribute to the protection from apoptotic injury in mouse liver.

6.
Acta Pharm Sin B ; 6(5): 426-429, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27709011

RESUMO

Arsenic is a carcinogenic environmental factor found in food and drinking water around the world. The mechanisms in which arsenic alters homeostasis are not fully understood. Over the past few decades, light has been shed on varying mechanisms in which arsenic induces cancer. Such mechanisms include gut microbe perturbations, genotoxic effects, and epigenetic modification. Gut microbe perturbations have been shown to increase the level of pathogen-associated molecular patterns such as lipopolysaccharide (LPS) leading to uncontained inflammation. Increase in inflammation is the major factor in cirrhosis leading to hepatocellular carcinoma. Alterations in gut permeability and metabolites have also been observed as a fallout of arsenic induced gut microbe modification. The guts proximity and interaction through portal flow make the liver susceptible to gut perturbations and ensuing inflammatory responses. Genotoxic and epigenetic dysregulation induced by arsenic and its toxic metabolites present a more direct mechanism that works synergistically with gut microbe perturbations to induce the incidence of cancers. These pathways combined could be some of the main causes of arsenic-induced carcinogenesis.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA