Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
bioRxiv ; 2024 Jan 19.
Artigo em Inglês | MEDLINE | ID: mdl-38293110

RESUMO

Copper (Cu) is an essential trace element required for mitochondrial respiration. Late-stage clear cell renal cell carcinoma (ccRCC) accumulates Cu and allocates it to mitochondrial cytochrome c oxidase. We show that Cu drives coordinated metabolic remodeling of bioenergy, biosynthesis and redox homeostasis, promoting tumor growth and progression of ccRCC. Specifically, Cu induces TCA cycle-dependent oxidation of glucose and its utilization for glutathione biosynthesis to protect against H 2 O 2 generated during mitochondrial respiration, therefore coordinating bioenergy production with redox protection. scRNA-seq determined that ccRCC progression involves increased expression of subunits of respiratory complexes, genes in glutathione and Cu metabolism, and NRF2 targets, alongside a decrease in HIF activity, a hallmark of ccRCC. Spatial transcriptomics identified that proliferating cancer cells are embedded in clusters of cells with oxidative metabolism supporting effects of metabolic states on ccRCC progression. Our work establishes novel vulnerabilities with potential for therapeutic interventions in ccRCC. Accumulation of copper is associated with progression and relapse of ccRCC and drives tumor growth.Cu accumulation and allocation to cytochrome c oxidase (CuCOX) remodels metabolism coupling energy production and nucleotide biosynthesis with maintenance of redox homeostasis.Cu induces oxidative phosphorylation via alterations in the mitochondrial proteome and lipidome necessary for the formation of the respiratory supercomplexes. Cu stimulates glutathione biosynthesis and glutathione derived specifically from glucose is necessary for survival of Cu Hi cells. Biosynthesis of glucose-derived glutathione requires activity of glutamyl pyruvate transaminase 2, entry of glucose-derived pyruvate to mitochondria via alanine, and the glutamate exporter, SLC25A22. Glutathione derived from glucose maintains redox homeostasis in Cu-treated cells, reducing Cu-H 2 O 2 Fenton-like reaction mediated cell death. Progression of human ccRCC is associated with gene expression signature characterized by induction of ETC/OxPhos/GSH/Cu-related genes and decrease in HIF/glycolytic genes in subpopulations of cancer cells. Enhanced, concordant expression of genes related to ETC/OxPhos, GSH, and Cu characterizes metabolically active subpopulations of ccRCC cells in regions adjacent to proliferative subpopulations of ccRCC cells, implicating oxidative metabolism in supporting tumor growth.

2.
Elife ; 122023 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-36692000

RESUMO

Genetic alterations that activate protein kinase A (PKA) are found in many tumor types. Yet, their downstream oncogenic signaling mechanisms are poorly understood. We used global phosphoproteomics and kinase activity profiling to map conserved signaling outputs driven by a range of genetic changes that activate PKA in human cancer. Two signaling networks were identified downstream of PKA: RAS/MAPK components and an Aurora Kinase A (AURKA)/glycogen synthase kinase (GSK3) sub-network with activity toward MYC oncoproteins. Findings were validated in two PKA-dependent cancer models: a novel, patient-derived fibrolamellar carcinoma (FLC) line that expresses a DNAJ-PKAc fusion and a PKA-addicted melanoma model with a mutant type I PKA regulatory subunit. We identify PKA signals that can influence both de novo translation and stability of the proto-oncogene c-MYC. However, the primary mechanism of PKA effects on MYC in our cell models was translation and could be blocked with the eIF4A inhibitor zotatifin. This compound dramatically reduced c-MYC expression and inhibited FLC cell line growth in vitro. Thus, targeting PKA effects on translation is a potential treatment strategy for FLC and other PKA-driven cancers.


Assuntos
Carcinoma Hepatocelular , Proteínas Quinases Dependentes de AMP Cíclico , Humanos , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Quinase 3 da Glicogênio Sintase/metabolismo , Carcinoma Hepatocelular/genética , Transdução de Sinais , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , Linhagem Celular Tumoral
3.
Nat Commun ; 13(1): 899, 2022 02 16.
Artigo em Inglês | MEDLINE | ID: mdl-35173161

RESUMO

Hexokinase 2 (HK2), which catalyzes the first committed step in glucose metabolism, is induced in cancer cells. HK2's role in tumorigenesis has been attributed to its glucose kinase activity. Here, we describe a kinase independent HK2 activity, which contributes to metastasis. HK2 binds and sequesters glycogen synthase kinase 3 (GSK3) and acts as a scaffold forming a ternary complex with the regulatory subunit of protein kinase A (PRKAR1a) and GSK3ß to facilitate GSK3ß phosphorylation and inhibition by PKA. Thus, HK2 functions as an A-kinase anchoring protein (AKAP). Phosphorylation by GSK3ß targets proteins for degradation. Consistently, HK2 increases the level and stability of GSK3 targets, MCL1, NRF2, and particularly SNAIL. In addition to GSK3 inhibition, HK2 kinase activity mediates SNAIL glycosylation, which prohibits its phosphorylation by GSK3. Finally, in mouse models of breast cancer metastasis, HK2 deficiency decreases SNAIL protein levels and inhibits SNAIL-mediated epithelial mesenchymal transition and metastasis.


Assuntos
Glucose/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Hexoquinase/metabolismo , Neoplasias/patologia , Proteínas de Ancoragem à Quinase A/metabolismo , Células A549 , Animais , Células CHO , Carcinogênese/patologia , Linhagem Celular Tumoral , Cricetulus , Subunidade RIalfa da Proteína Quinase Dependente de AMP Cíclico/metabolismo , Desoxiglucose/farmacologia , Transição Epitelial-Mesenquimal/fisiologia , Feminino , Glicogênio Sintase Quinase 3 beta/antagonistas & inibidores , Glicosilação , Células HCT116 , Células HEK293 , Hexoquinase/genética , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Metástase Neoplásica/patologia , Fosforilação/efeitos dos fármacos , Ratos , Fatores de Transcrição da Família Snail/metabolismo
4.
World J Hepatol ; 13(3): 343-361, 2021 Mar 27.
Artigo em Inglês | MEDLINE | ID: mdl-33815677

RESUMO

BACKGROUND: BIR repeat-containing ubiquitin conjugating enzyme (BRUCE) is a liver tumor suppressor, which is downregulated in a large number of patients with liver diseases. BRUCE facilitates DNA damage repair to protect the mouse liver against the hepatocarcinogen diethylnitrosamine (DEN)-dependent acute liver injury and carcinogenesis. While there exists an established pathologic connection between fibrosis and hepatocellular carcinoma (HCC), DEN exposure alone does not induce robust hepatic fibrosis. Further studies are warranted to identify new suppressive mechanisms contributing to DEN-induced fibrosis and HCC. AIM: To investigate the suppressive mechanisms of BRUCE in hepatic fibrosis and HCC development. METHODS: Male C57/BL6/J control mice [loxp/Loxp; albumin-cre (Alb-cre)-] and BRUCE Alb-Cre KO mice (loxp/Loxp; Alb-Cre+) were injected with a single dose of DEN at postnatal day 15 and sacrificed at different time points to examine liver disease progression. RESULTS: By using a liver-specific BRUCE knockout (LKO) mouse model, we found that BRUCE deficiency, in conjunction with DEN exposure, induced hepatic fibrosis in both premalignant as well as malignant stages, thus recapitulating the chronic fibrosis background often observed in HCC patients. Activated in fibrosis and HCC, ß-catenin activity depends on its stabilization and subsequent translocation to the nucleus. Interestingly, we observed that livers from BRUCE KO mice demonstrated an increased nuclear accumulation and elevated activity of ß-catenin in the three stages of carcinogenesis: Pre-malignancy, tumor initiation, and HCC. This suggests that BRUCE negatively regulates ß-catenin activity during liver disease progression. ß-catenin can be activated by phosphorylation by protein kinases, such as protein kinase A (PKA), which phosphorylates it at Ser-675 (pSer-675-ß-catenin). Mechanistically, BRUCE and PKA were colocalized in the cytoplasm of hepatocytes where PKA activity is maintained at the basal level. However, in BRUCE deficient mouse livers or a human liver cancer cell line, both PKA activity and pSer-675-ß-catenin levels were observed to be elevated. CONCLUSION: Our data support a "BRUCE-PKA-ß-catenin" signaling axis in the mouse liver. The BRUCE interaction with PKA in hepatocytes suppresses PKA-dependent phosphorylation and activation of ß-catenin. This study implicates BRUCE as a novel negative regulator of both PKA and ß-catenin in chronic liver disease progression. Furthermore, BRUCE-liver specific KO mice serve as a promising model for understanding hepatic fibrosis and HCC in patients with aberrant activation of PKA and ß-catenin.

5.
Dev Cell ; 50(3): 261-263, 2019 08 05.
Artigo em Inglês | MEDLINE | ID: mdl-31386860

RESUMO

Macropinocytosis coordinates non-specific uptake of macromolecules and fluid. Cancers employ macropinocytosis to obtain nutrients to support their metabolic homeostasis. In this issue of Developmental Cell, Lee et al. (2019) report that glutamine deprivation boosts macropinocytosis via EGFR signaling induction, providing a fine-tuned mechanism by which cancers adapt to nutrient supply.


Assuntos
Glutamina , Pinocitose , Aminoácidos , Receptores ErbB , Transdução de Sinais
6.
Cancer Discov ; 9(8): 1064-1079, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31109923

RESUMO

ATP-competitive fibroblast growth factor receptor (FGFR) kinase inhibitors, including BGJ398 and Debio 1347, show antitumor activity in patients with intrahepatic cholangiocarcinoma (ICC) harboring activating FGFR2 gene fusions. Unfortunately, acquired resistance develops and is often associated with the emergence of secondary FGFR2 kinase domain mutations. Here, we report that the irreversible pan-FGFR inhibitor TAS-120 demonstrated efficacy in 4 patients with FGFR2 fusion-positive ICC who developed resistance to BGJ398 or Debio 1347. Examination of serial biopsies, circulating tumor DNA (ctDNA), and patient-derived ICC cells revealed that TAS-120 was active against multiple FGFR2 mutations conferring resistance to BGJ398 or Debio 1347. Functional assessment and modeling the clonal outgrowth of individual resistance mutations from polyclonal cell pools mirrored the resistance profiles observed clinically for each inhibitor. Our findings suggest that strategic sequencing of FGFR inhibitors, guided by serial biopsy and ctDNA analysis, may prolong the duration of benefit from FGFR inhibition in patients with FGFR2 fusion-positive ICC. SIGNIFICANCE: ATP-competitive FGFR inhibitors (BGJ398, Debio 1347) show efficacy in FGFR2-altered ICC; however, acquired FGFR2 kinase domain mutations cause drug resistance and tumor progression. We demonstrate that the irreversible FGFR inhibitor TAS-120 provides clinical benefit in patients with resistance to BGJ398 or Debio 1347 and overcomes several FGFR2 mutations in ICC models.This article is highlighted in the In This Issue feature, p. 983.


Assuntos
Trifosfato de Adenosina/metabolismo , Colangiocarcinoma/genética , Colangiocarcinoma/metabolismo , Resistencia a Medicamentos Antineoplásicos/genética , Inibidores de Proteínas Quinases/farmacologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/antagonistas & inibidores , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Adulto , Idoso , Linhagem Celular Tumoral , Colangiocarcinoma/diagnóstico , DNA Tumoral Circulante , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Proteínas de Fusão Oncogênica/antagonistas & inibidores , Proteínas de Fusão Oncogênica/genética , Compostos de Fenilureia/farmacologia , Inibidores de Proteínas Quinases/química , Pirimidinas/farmacologia , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/química , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/metabolismo , Transdução de Sinais/efeitos dos fármacos , Relação Estrutura-Atividade , Tomografia Computadorizada por Raios X
7.
Cell Metab ; 29(2): 238-240, 2019 02 05.
Artigo em Inglês | MEDLINE | ID: mdl-30612899

RESUMO

Cancer cells must adapt to metabolic stress during tumor progression. In this issue of Cell Metabolism, Eichner et al. (2019) report that lung cancer development in genetically engineered mice requires the energy sensor AMP-activated protein kinase (AMPK). Their findings suggest that AMPK-mediated induction of lysosomal function supports cancer cell fitness, particularly during the early stages of tumorigenesis.


Assuntos
Proteínas Quinases Ativadas por AMP , Neoplasias Pulmonares , Animais , Carcinogênese , Proliferação de Células , Lisossomos , Camundongos
8.
Nat Cell Biol ; 20(7): 811-822, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29941929

RESUMO

G protein αs (GNAS) mediates receptor-stimulated cAMP signalling, which integrates diverse environmental cues with intracellular responses. GNAS is mutationally activated in multiple tumour types, although its oncogenic mechanisms remain elusive. We explored this question in pancreatic tumourigenesis where concurrent GNAS and KRAS mutations characterize pancreatic ductal adenocarcinomas (PDAs) arising from intraductal papillary mucinous neoplasms (IPMNs). By developing genetically engineered mouse models, we show that GnasR201C cooperates with KrasG12D to promote initiation of IPMN, which progress to invasive PDA following Tp53 loss. Mutant Gnas remains critical for tumour maintenance in vivo. This is driven by protein-kinase-A-mediated suppression of salt-inducible kinases (Sik1-3), associated with induction of lipid remodelling and fatty acid oxidation. Comparison of Kras-mutant pancreatic cancer cells with and without Gnas mutations reveals striking differences in the functions of this network. Thus, we uncover Gnas-driven oncogenic mechanisms, identify Siks as potent tumour suppressors, and demonstrate unanticipated metabolic heterogeneity among Kras-mutant pancreatic neoplasms.


Assuntos
Carcinoma Ductal Pancreático/enzimologia , Carcinoma Ductal Pancreático/genética , Transformação Celular Neoplásica/genética , Transformação Celular Neoplásica/metabolismo , Reprogramação Celular/genética , Cromograninas/genética , Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Subunidades alfa Gs de Proteínas de Ligação ao GTP/genética , Metabolismo dos Lipídeos/genética , Mutação , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Proteínas Serina-Treonina Quinases/metabolismo , Animais , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Transformação Celular Neoplásica/patologia , Cromograninas/metabolismo , Proteínas Quinases Dependentes de AMP Cíclico/genética , Repressão Enzimática , Ácidos Graxos/metabolismo , Feminino , Subunidades alfa Gs de Proteínas de Ligação ao GTP/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes ras , Predisposição Genética para Doença , Humanos , Masculino , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Endogâmicos NOD , Camundongos Mutantes , Camundongos Transgênicos , Oxirredução , Neoplasias Pancreáticas/patologia , Fenótipo , Proteínas Serina-Treonina Quinases/genética , Transdução de Sinais , Fatores de Tempo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
9.
Dev Cell ; 45(6): 665-666, 2018 06 18.
Artigo em Inglês | MEDLINE | ID: mdl-29920272

RESUMO

The factors determining longevity of different animals are incompletely defined. In this issue of Developmental Cell, Anzi et al. (2018) show that distinct strategies for postnatal pancreatic growth operate in different mammals and correlate with lifespan, with short-lived species exhibiting increasing pancreatic cell size and long-lived animals increasing cell number.


Assuntos
Longevidade , Mamíferos , Envelhecimento , Animais , Tamanho Celular
10.
Nat Commun ; 9(1): 2539, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29946147

RESUMO

In the originally published version of this Article, the colours of the bars in Fig. 4b were inadvertently switched during the production process, such that 'HK2-Dox' and 'HK2+Dox' were depicted in red and 'Nt-Dox' and 'Nt+Dox' were depicted in blue. These errors have now been corrected in both the PDF and HTML versions of the Article.

11.
Elife ; 72018 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-29687779

RESUMO

Akt activation in human cancers exerts chemoresistance, but pan-Akt inhibition elicits adverse consequences. We exploited the consequences of Akt-mediated mitochondrial and glucose metabolism to selectively eradicate and evade chemoresistance of prostate cancer displaying hyperactive Akt. PTEN-deficient prostate cancer cells that display hyperactivated Akt have high intracellular reactive oxygen species (ROS) levels, in part, because of Akt-dependent increase of oxidative phosphorylation. High intracellular ROS levels selectively sensitize cells displaying hyperactive Akt to ROS-induced cell death enabling a therapeutic strategy combining a ROS inducer and rapamycin in PTEN-deficient prostate tumors in mouse models. This strategy elicited tumor regression, and markedly increased survival even after the treatment was stopped. By contrast, exposure to antioxidant increased prostate tumor progression. To increase glucose metabolism, Akt activation phosphorylated HK2 and induced its expression. Indeed, HK2 deficiency in mouse models of Pten-deficient prostate cancer elicited a marked inhibition of tumor development and extended lifespan.


Assuntos
Antibióticos Antineoplásicos/administração & dosagem , Proteína Oncogênica v-akt/metabolismo , Neoplasias da Próstata/tratamento farmacológico , Sirolimo/administração & dosagem , Animais , Antibióticos Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Modelos Biológicos , Transplante de Neoplasias , Fosforilação Oxidativa , Espécies Reativas de Oxigênio/metabolismo , Sirolimo/farmacologia , Resultado do Tratamento
12.
Nat Commun ; 9(1): 446, 2018 01 31.
Artigo em Inglês | MEDLINE | ID: mdl-29386513

RESUMO

Hepatocellular carcinoma (HCC) cells are metabolically distinct from normal hepatocytes by expressing the high-affinity hexokinase (HK2) and suppressing glucokinase (GCK). This is exploited to selectively target HCC. Hepatic HK2 deletion inhibits tumor incidence in a mouse model of hepatocarcinogenesis. Silencing HK2 in human HCC cells inhibits tumorigenesis and increases cell death, which cannot be restored by GCK or mitochondrial binding deficient HK2. Upon HK2 silencing, glucose flux to pyruvate and lactate is inhibited, but TCA fluxes are maintained. Serine uptake and glycine secretion are elevated suggesting increased requirement for one-carbon contribution. Consistently, vulnerability to serine depletion increases. The decrease in glycolysis is coupled to elevated oxidative phosphorylation, which is diminished by metformin, further increasing cell death and inhibiting tumor growth. Neither HK2 silencing nor metformin alone inhibits mTORC1, but their combination inhibits mTORC1 in an AMPK-independent and REDD1-dependent mechanism. Finally, HK2 silencing synergizes with sorafenib to inhibit tumor growth.


Assuntos
Carcinoma Hepatocelular/enzimologia , Hexoquinase/metabolismo , Neoplasias Hepáticas/enzimologia , Terapia de Alvo Molecular , Animais , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carcinogênese , Carcinoma Hepatocelular/tratamento farmacológico , Glicólise , Células Hep G2 , Hexoquinase/antagonistas & inibidores , Humanos , Hipoglicemiantes/farmacologia , Hipoglicemiantes/uso terapêutico , Neoplasias Hepáticas/tratamento farmacológico , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Análise do Fluxo Metabólico , Metformina/farmacologia , Metformina/uso terapêutico , Camundongos Nus , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Niacinamida/uso terapêutico , Fosforilação Oxidativa , Compostos de Fenilureia/farmacologia , Compostos de Fenilureia/uso terapêutico , Sorafenibe , Ensaios Antitumorais Modelo de Xenoenxerto
13.
Clin Transl Gastroenterol ; 8(4): e86, 2017 Apr 06.
Artigo em Inglês | MEDLINE | ID: mdl-28383565

RESUMO

Pancreatic ductal adenocarcinoma (PDA), one of the most lethal cancers worldwide, is associated with two main types of morphologically distinct precursors-pancreatic intraepithelial neoplasia (PanIN) and intraductal papillary mucinous neoplasm (IPMN). Although the progression of PanIN into invasive cancer has been well characterized, there remains an urgent need to understand the biology of IPMNs, which are larger radiographically detectable cystic tumors. IPMNs comprise a number of subtypes with heterogeneous histopathologic and clinical features. Although frequently remaining benign, a significant proportion exhibits malignant progression. Unfortunately, there are presently no accurate prognosticators for assessing cancer risk in individuals with IPMN. Moreover, the fundamental mechanisms differentiating PanIN and IPMN remain largely obscure, as do those that distinguish IPMN subtypes. Recent studies, however, have identified distinct genetic profiles between PanIN and IPMN, providing a framework to better understand the diversity of the precursors for PDA. Here, we review the clinical, biological, and genetic properties of IPMN and discuss various models for progression of these tumors to invasive PDA.

14.
Cell Rep ; 12(4): 610-21, 2015 Jul 28.
Artigo em Inglês | MEDLINE | ID: mdl-26190111

RESUMO

Akt is frequently activated in human cancers. However, it is unknown whether systemic inhibition of a single Akt isoform could regress cancer progression in cancers that are not driven by Akt activation. We systemically deleted Akt1 after tumor onset in p53(-/-) mice, which develop tumors independently of Akt activation. Systemic Akt1 deletion regresses thymic lymphoma in p53(-/-) mice emulating p53 restoration. Furthermore, pharmacological inhibition of Akt selectively kills thymic lymphoma cells and not primary thymocytes. Mechanistically, Akt1 inhibition in p53(-/-) thymic lymphoma inhibits Skp2 expression and induces FasL, which is the primary cause of cell death. Skp2 exerts resistance to cell death by antagonizing the induction of FasL and reducing FAS expression, which is linked to cyclin D1 expression. The results established a paradigm whereby systemic Akt1 inhibition is sufficient to regress tumors that are not driven by Akt activation and a mechanism of cell survival by Skp2.


Assuntos
Longevidade , Linfoma/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Timócitos/metabolismo , Proteína Supressora de Tumor p53/genética , Animais , Apoptose , Linhagem Celular Tumoral , Proteína Ligante Fas/metabolismo , Humanos , Linfoma/patologia , Camundongos , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Timócitos/patologia , Proteína Supressora de Tumor p53/metabolismo
15.
Trends Biochem Sci ; 39(8): 347-54, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25037503

RESUMO

The pentose phosphate pathway (PPP), which branches from glycolysis at the first committed step of glucose metabolism, is required for the synthesis of ribonucleotides and is a major source of NADPH. NADPH is required for and consumed during fatty acid synthesis and the scavenging of reactive oxygen species (ROS). Therefore, the PPP plays a pivotal role in helping glycolytic cancer cells to meet their anabolic demands and combat oxidative stress. Recently, several neoplastic lesions were shown to have evolved to facilitate the flux of glucose into the PPP. This review summarizes the fundamental functions of the PPP, its regulation in cancer cells, and its importance in cancer cell metabolism and survival.


Assuntos
Neoplasias/metabolismo , Neoplasias/patologia , Estresse Oxidativo , Via de Pentose Fosfato , Animais , Glicólise , Humanos
17.
Cancer Cell ; 24(2): 213-228, 2013 Aug 12.
Artigo em Inglês | MEDLINE | ID: mdl-23911236

RESUMO

Accelerated glucose metabolism is a common feature of cancer cells. Hexokinases catalyze the first committed step of glucose metabolism. Hexokinase 2 (HK2) is expressed at high level in cancer cells, but only in a limited number of normal adult tissues. Using Hk2 conditional knockout mice, we showed that HK2 is required for tumor initiation and maintenance in mouse models of KRas-driven lung cancer, and ErbB2-driven breast cancer, despite continued HK1 expression. Similarly, HK2 ablation inhibits the neoplastic phenotype of human lung and breast cancer cells in vitro and in vivo. Systemic Hk2 deletion is therapeutic in mice bearing lung tumors without adverse physiological consequences. Hk2 deletion in lung cancer cells suppressed glucose-derived ribonucleotides and impaired glutamine-derived carbon utilization in anaplerosis.


Assuntos
Neoplasias da Mama/enzimologia , Hexoquinase/metabolismo , Neoplasias Pulmonares/enzimologia , Animais , Neoplasias da Mama/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Feminino , Glicólise , Hexoquinase/biossíntese , Hexoquinase/genética , Humanos , Neoplasias Pulmonares/genética , Masculino , Camundongos , Camundongos Knockout , Transplante Heterólogo
18.
Cancer Res ; 73(12): 3771-82, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23610444

RESUMO

Colon cancer is one of the deadliest cancers worldwide because of its metastasis to other essential organs. Metastasis of colon cancer involves a complex set of events, including epithelial-to-mesenchymal transition (EMT) that increases invasiveness of the tumor cells. Here, we show that the xeroderma pigmentosum group E (XPE) gene product, damaged DNA-binding protein (DDB)-2, is downregulated in high-grade colon cancers, and it plays a dominant role in the suppression of EMT of the colon cancer cells. Depletion of DDB2 promotes mesenchymal phenotype, whereas expression of DDB2 promotes epithelial phenotype. DDB2 constitutively represses genes that are the key activators of EMT, indicating that DDB2 is a master regulator of EMT of the colon cancer cells. Moreover, we observed evidence that DDB2 functions as a barrier for EMT induced by hypoxia and TGF-ß. Also, we provide evidence that DDB2 inhibits metastasis of colon cancer. The results presented here identify a transcriptional regulatory pathway of DDB2 that is directly linked to the mechanisms that suppress metastasis of colon cancer.


Assuntos
Neoplasias do Colo/genética , Proteínas de Ligação a DNA/genética , Transição Epitelial-Mesenquimal/genética , Regulação Neoplásica da Expressão Gênica , Animais , Western Blotting , Caderinas/metabolismo , Hipóxia Celular , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Proteínas de Ligação a DNA/metabolismo , Células HCT116 , Humanos , Imuno-Histoquímica , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/metabolismo , Neoplasias Pulmonares/secundário , Masculino , Neoplasias Mamárias Experimentais/genética , Neoplasias Mamárias Experimentais/metabolismo , Neoplasias Mamárias Experimentais/patologia , Camundongos , Camundongos Nus , Camundongos SCID , Invasividade Neoplásica , Interferência de RNA , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transplante Heterólogo
19.
Mol Cell Biol ; 29(18): 5136-47, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19620286

RESUMO

The current concept is that Tsc-deficient cells are sensitized to apoptosis due to the inhibition of Akt activity by the negative feedback mechanism induced by the hyperactive mTORC1. Unexpectedly, however, we found that Tsc1/2-deficient cells exhibit increased resistance to serum deprivation-induced apoptosis. mTORC1 hyperactivity contributes to the apoptotic resistance of serum-deprived Tsc1/2-deficient cells in part by increasing the growth factor-independent expression of hexokinase II (HKII) and GLUT1. mTORC1-mediated increase in hypoxia-inducible factor 1alpha (HIF1alpha) abundance, which occurs in the absence of serum in normoxic Tsc2-deficient cells, contributes to these changes. Increased HIF1alpha abundance in these cells is attributed to both an increased level and the sustained translation of HIF1alpha mRNA. Sustained glycogen synthase kinase 3beta inhibition and Mcl-1 expression also contribute to the apoptotic resistance of Tsc2-deficient cells to serum deprivation. The inhibition of mTORC1 activity by either rapamycin or Raptor knockdown cannot resensitize these cells to serum deprivation-induced apoptosis because of elevated Akt activity that is an indirect consequence of mTORC1 inhibition. However, the increased HIF1alpha abundance and the maintenance of Mcl-1 protein expression in serum-deprived Tsc2(-/)(-) cells are dependent largely on the hyperactive eIF4E in these cells. Consistently, the reduction of eIF4E levels abrogates the resistance of Tsc2(-/)(-) cells to serum deprivation-induced apoptosis.


Assuntos
Apoptose , Transportador de Glucose Tipo 1/metabolismo , Quinase 3 da Glicogênio Sintase/antagonistas & inibidores , Hexoquinase/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Soro/metabolismo , Fatores de Transcrição/metabolismo , Animais , Apoptose/efeitos dos fármacos , Embrião de Mamíferos/citologia , Ativação Enzimática/efeitos dos fármacos , Fator de Iniciação 4E em Eucariotos/metabolismo , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/enzimologia , Glicogênio Sintase Quinase 3 beta , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Peptídeos e Proteínas de Sinalização Intercelular/deficiência , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Complexos Multiproteicos , Proteína de Sequência 1 de Leucemia de Células Mieloides , Fosforilação/efeitos dos fármacos , Proteínas , Proteínas Proto-Oncogênicas c-akt/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR , Fatores de Transcrição/antagonistas & inibidores , Proteína 1 do Complexo Esclerose Tuberosa , Proteína 2 do Complexo Esclerose Tuberosa , Proteínas Supressoras de Tumor/deficiência , Proteínas Supressoras de Tumor/metabolismo , Proteína de Morte Celular Associada a bcl/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...