Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 5.641
Filtrar
Mais filtros

Eixos temáticos
Intervalo de ano de publicação
1.
Immunity ; 54(5): 976-987.e7, 2021 05 11.
Artigo em Inglês | MEDLINE | ID: mdl-33979589

RESUMO

Aerobic glycolysis-the Warburg effect-converts glucose to lactate via the enzyme lactate dehydrogenase A (LDHA) and is a metabolic feature of effector T cells. Cells generate ATP through various mechanisms and Warburg metabolism is comparatively an energy-inefficient glucose catabolism pathway. Here, we examined the effect of ATP generated via aerobic glycolysis in antigen-driven T cell responses. Cd4CreLdhafl/fl mice were resistant to Th17-cell-mediated experimental autoimmune encephalomyelitis and exhibited defective T cell activation, migration, proliferation, and differentiation. LDHA deficiency crippled cellular redox balance and inhibited ATP production, diminishing PI3K-dependent activation of Akt kinase and thereby phosphorylation-mediated inhibition of Foxo1, a transcriptional repressor of T cell activation programs. Th17-cell-specific expression of an Akt-insensitive Foxo1 recapitulated the defects seen in Cd4CreLdhafl/fl mice. Induction of LDHA required PI3K signaling and LDHA deficiency impaired PI3K-catalyzed PIP3 generation. Thus, Warburg metabolism augments glycolytic ATP production, fueling a PI3K-centered positive feedback regulatory circuit that drives effector T cell responses.


Assuntos
Trifosfato de Adenosina/metabolismo , Fosfatidilinositol 3-Quinase/metabolismo , Transdução de Sinais/fisiologia , Células Th17/metabolismo , Animais , Diferenciação Celular/fisiologia , Linhagem Celular , Proliferação de Células/fisiologia , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Glucose/metabolismo , Doença de Depósito de Glicogênio/metabolismo , Glicólise/fisiologia , L-Lactato Desidrogenase/deficiência , L-Lactato Desidrogenase/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos
2.
Mol Cell ; 76(5): 838-851.e5, 2019 12 05.
Artigo em Inglês | MEDLINE | ID: mdl-31564558

RESUMO

Intermediary metabolism in cancer cells is regulated by diverse cell-autonomous processes, including signal transduction and gene expression patterns, arising from specific oncogenotypes and cell lineages. Although it is well established that metabolic reprogramming is a hallmark of cancer, we lack a full view of the diversity of metabolic programs in cancer cells and an unbiased assessment of the associations between metabolic pathway preferences and other cell-autonomous processes. Here, we quantified metabolic features, mostly from the 13C enrichment of molecules from central carbon metabolism, in over 80 non-small cell lung cancer (NSCLC) cell lines cultured under identical conditions. Because these cell lines were extensively annotated for oncogenotype, gene expression, protein expression, and therapeutic sensitivity, the resulting database enables the user to uncover new relationships between metabolism and these orthogonal processes.


Assuntos
Carcinoma Pulmonar de Células não Pequenas/metabolismo , Carcinoma Pulmonar de Células não Pequenas/patologia , Linhagem Celular Tumoral/metabolismo , Metaboloma/fisiologia , Biomarcadores Tumorais/metabolismo , Cromatografia Gasosa-Espectrometria de Massas/métodos , Regulação Neoplásica da Expressão Gênica/fisiologia , Glucose/metabolismo , Glutamina/metabolismo , Humanos , Redes e Vias Metabólicas/genética , Metabolômica/métodos , Neoplasias/metabolismo
3.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35135884

RESUMO

Mitochondrial inner NEET (MiNT) and the outer mitochondrial membrane (OMM) mitoNEET (mNT) proteins belong to the NEET protein family. This family plays a key role in mitochondrial labile iron and reactive oxygen species (ROS) homeostasis. NEET proteins contain labile [2Fe-2S] clusters which can be transferred to apo-acceptor proteins. In eukaryotes, the biogenesis of [2Fe-2S] clusters occurs within the mitochondria by the iron-sulfur cluster (ISC) system; the clusters are then transferred to [2Fe-2S] proteins within the mitochondria or exported to cytosolic proteins and the cytosolic iron-sulfur cluster assembly (CIA) system. The last step of export of the [2Fe-2S] is not yet fully characterized. Here we show that MiNT interacts with voltage-dependent anion channel 1 (VDAC1), a major OMM protein that connects the intermembrane space with the cytosol and participates in regulating the levels of different ions including mitochondrial labile iron (mLI). We further show that VDAC1 is mediating the interaction between MiNT and mNT, in which MiNT transfers its [2Fe-2S] clusters from inside the mitochondria to mNT that is facing the cytosol. This MiNT-VDAC1-mNT interaction is shown both experimentally and by computational calculations. Additionally, we show that modifying MiNT expression in breast cancer cells affects the dynamics of mitochondrial structure and morphology, mitochondrial function, and breast cancer tumor growth. Our findings reveal a pathway for the transfer of [2Fe-2S] clusters, which are assembled inside the mitochondria, to the cytosol.


Assuntos
Citosol/metabolismo , Compostos Ferrosos/metabolismo , Mitocôndrias/metabolismo , Canal de Ânion 1 Dependente de Voltagem/metabolismo , Animais , Neoplasias da Mama , Linhagem Celular Tumoral , Simulação por Computador , Matriz Extracelular , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Glicólise , Humanos , Concentração de Íons de Hidrogênio , Camundongos , Camundongos Nus , Neoplasias Experimentais , Consumo de Oxigênio , Canal de Ânion 1 Dependente de Voltagem/genética
4.
Proc Natl Acad Sci U S A ; 119(7)2022 02 15.
Artigo em Inglês | MEDLINE | ID: mdl-35149558

RESUMO

Immune evasion is a significant contributor to tumor evolution, and the immunoinhibitory axis PD-1/PD-L1 is a frequent mechanism employed to escape tumor immune surveillance. To identify cancer drivers involved in immune evasion, we performed a CRISPR-Cas9 screen of tumor suppressor genes regulating the basal and interferon (IFN)-inducible cell surface levels of PD-L1. Multiple regulators of PD-L1 were identified, including IRF2, ARID2, KMT2D, and AAMP. We also identified CTCF and the cohesin complex proteins, known regulators of chromatin architecture and transcription, among the most potent negative regulators of PD-L1 cell surface expression. Additionally, loss of the cohesin subunit RAD21 was shown to up-regulate PD-L2 and MHC-I surface expression. PD-L1 and MHC-I suppression by cohesin were shown to be conserved in mammary epithelial and myeloid cells. Comprehensive examination of the transcriptional effect of STAG2 deficiency in epithelial and myeloid cells revealed an activation of strong IFN and NF-κB expression signatures. Inhibition of JAK-STAT or NF-κB pathways did not result in rescue of PD-L1 up-regulation in RAD21-deficient cells, suggesting more complex or combinatorial mechanisms at play. Discovery of the PD-L1 and IFN up-regulation in cohesin-mutant cells expands our understanding of the biology of cohesin-deficient cells as well as molecular regulation of the PD-L1 molecule.


Assuntos
Antígeno B7-H1/metabolismo , Fator de Ligação a CCCTC/metabolismo , Proteínas de Ciclo Celular/metabolismo , Proteínas Cromossômicas não Histona/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias/metabolismo , Antígeno B7-H1/genética , Fator de Ligação a CCCTC/genética , Proteínas de Ciclo Celular/genética , Linhagem Celular , Proteínas Cromossômicas não Histona/genética , Humanos , Janus Quinases/genética , Janus Quinases/metabolismo , NF-kappa B/genética , NF-kappa B/metabolismo , Fatores de Transcrição STAT/genética , Fatores de Transcrição STAT/metabolismo , Regulação para Cima , Coesinas
5.
Proc Natl Acad Sci U S A ; 118(51)2021 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-34916293

RESUMO

G protein-coupled receptors (GPCRs) are the largest family of human proteins. They have a common structure and, signaling through a much smaller set of G proteins, arrestins, and effectors, activate downstream pathways that often modulate hallmark mechanisms of cancer. Because there are many more GPCRs than effectors, mutations in different receptors could perturb signaling similarly so as to favor a tumor. We hypothesized that somatic mutations in tumor samples may not be enriched within a single gene but rather that cognate mutations with similar effects on GPCR function are distributed across many receptors. To test this possibility, we systematically aggregated somatic cancer mutations across class A GPCRs and found a nonrandom distribution of positions with variant amino acid residues. Individual cancer types were enriched for highly impactful, recurrent mutations at selected cognate positions of known functional motifs. We also discovered that no single receptor drives this pattern, but rather multiple receptors contain amino acid substitutions at a few cognate positions. Phenotypic characterization suggests these mutations induce perturbation of G protein activation and/or ß-arrestin recruitment. These data suggest that recurrent impactful oncogenic mutations perturb different GPCRs to subvert signaling and promote tumor growth or survival. The possibility that multiple different GPCRs could moonlight as drivers or enablers of a given cancer through mutations located at cognate positions across GPCR paralogs opens a window into cancer mechanisms and potential approaches to therapeutics.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , beta-Arrestinas/metabolismo , Cálcio , Linhagem Celular Tumoral , Simulação por Computador , Ensaio de Imunoadsorção Enzimática , Humanos , Mutação , Neoplasias/genética , Conformação Proteica , Receptores Acoplados a Proteínas G/genética , beta-Arrestinas/genética
6.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34330832

RESUMO

UDP-glucose pyrophosphorylase 2 (UGP2), the enzyme that synthesizes uridine diphosphate (UDP)-glucose, rests at the convergence of multiple metabolic pathways, however, the role of UGP2 in tumor maintenance and cancer metabolism remains unclear. Here, we identify an important role for UGP2 in the maintenance of pancreatic ductal adenocarcinoma (PDAC) growth in both in vitro and in vivo tumor models. We found that transcription of UGP2 is directly regulated by the Yes-associated protein 1 (YAP)-TEA domain transcription factor (TEAD) complex, identifying UGP2 as a bona fide YAP target gene. Loss of UGP2 leads to decreased intracellular glycogen levels and defects in N-glycosylation targets that are important for the survival of PDACs, including the epidermal growth factor receptor (EGFR). These critical roles of UGP2 in cancer maintenance, metabolism, and protein glycosylation may offer insights into therapeutic options for otherwise intractable PDACs.


Assuntos
Carcinoma Ductal Pancreático/enzimologia , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glicogênio/biossíntese , Neoplasias Pancreáticas/enzimologia , UTP-Glucose-1-Fosfato Uridililtransferase/metabolismo , Animais , Carcinoma Ductal Pancreático/patologia , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Glicosilação , Humanos , Camundongos , Camundongos Nus , Neoplasias Experimentais , Neoplasias Pancreáticas/patologia , Fatores de Transcrição de Domínio TEA/genética , Fatores de Transcrição de Domínio TEA/metabolismo , UTP-Glucose-1-Fosfato Uridililtransferase/genética , Proteínas de Sinalização YAP/genética , Proteínas de Sinalização YAP/metabolismo
7.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Artigo em Inglês | MEDLINE | ID: mdl-34326251

RESUMO

The p53 pathway is a universal tumor suppressor mechanism that limits tumor progression by triggering apoptosis or permanent cell cycle arrest, called senescence. In recent years, efforts to reactivate p53 function in cancer have proven to be a successful therapeutic strategy in murine models and have gained traction with the development of a range of small molecules targeting mutant p53. However, knowledge of the downstream mediators of p53 reactivation in different oncogenic contexts has been limited. Here, we utilized a panel of murine cancer cell lines from three distinct tumor types susceptible to alternative outcomes following p53 restoration to define unique and shared p53 transcriptional signatures. While we found that the majority of p53-bound sites and p53-responsive transcripts are tumor-type specific, analysis of shared targets identified a core signature of genes activated by p53 across all contexts. Furthermore, we identified repression of E2F and Myc target genes as a key feature of senescence. Characterization of p53-induced transcripts revealed core and senescence-specific long noncoding RNAs (lncRNAs) that are predominantly chromatin associated and whose production is coupled to cis-regulatory activities. Functional investigation of the contributions of p53-induced lncRNAs to p53-dependent outcomes highlighted Pvt1b, the p53-dependent isoform of Pvt1, as a mediator of p53-dependent senescence via Myc repression. Inhibition of Pvt1b led to decreased activation of senescence markers and increased levels of markers of proliferation. These findings shed light on the core and outcome-specific p53 restoration signatures across different oncogenic contexts and underscore the key role of the p53-Pvt1b-Myc regulatory axis in mediating proliferative arrest.


Assuntos
Senescência Celular/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias/metabolismo , RNA Longo não Codificante/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Carcinogênese , Linhagem Celular Tumoral , Proliferação de Células , Dano ao DNA , Fatores de Transcrição E2F/genética , Fatores de Transcrição E2F/metabolismo , Estudo de Associação Genômica Ampla , Camundongos , Proteínas Proto-Oncogênicas c-myc/genética , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Longo não Codificante/genética , Transdução de Sinais , Estresse Fisiológico , Proteína Supressora de Tumor p53/genética
8.
Proc Natl Acad Sci U S A ; 118(33)2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34389675

RESUMO

To identify regulators of triple-negative breast cancer (TNBC), gene expression profiles of malignant parts of TNBC (mTNBC) and normal adjacent (nadj) parts of the same breasts have been compared. We are interested in the roles of estrogen receptor ß (ERß) and the cytochrome P450 family (CYPs) as drivers of TNBC. We examined by RNA sequencing the mTNBC and nadj parts of five women. We found more than a fivefold elevation in mTNBC of genes already known to be expressed in TNBC: BIRC5/survivin, Wnt-10A and -7B, matrix metalloproteinases (MMPs), chemokines, anterior gradient proteins, and lysophosphatidic acid receptor and the known basal characteristics of TNBC, sox10, ROPN1B, and Col9a3. There were two unexpected findings: 1) a strong induction of CYPs involved in activation of fatty acids (CYP4), and in inactivation of calcitriol (CYP24A1) and retinoic acid (CYP26A1); and 2) a marked down-regulation of FOS, FRA1, and JUN, known tethering partners of ERß. ERß is expressed in 20 to 30% of TNBCs and is being evaluated as a target for treating TNBC. We used ERß+ TNBC patient-derived xenografts in mice and found that the ERß agonist LY500703 had no effect on growth or proliferation. Expression of CYPs was confirmed by immunohistochemistry in formalin-fixed and paraffin-embedded (FFPE) TNBC. In TNBC cell lines, the CYP4Z1-catalyzed fatty acid metabolite 20-hydroxyeicosatetraenoic acid (20-HETE) increased proliferation, while calcitriol decreased proliferation but only after inhibition of CYP24A1. We conclude that CYP-mediated pathways can be drivers of TNBC but that ERß is unlikely to be a tumor suppressor because the absence of its main tethering partners renders ERß functionless on genes involved in proliferation and inflammation.


Assuntos
Sistema Enzimático do Citocromo P-450/metabolismo , Receptor beta de Estrogênio/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias de Mama Triplo Negativas/metabolismo , Proteínas de Anfíbios/genética , Proteínas de Anfíbios/metabolismo , Animais , Benzopiranos/farmacologia , Calcitriol/farmacologia , Sistema Enzimático do Citocromo P-450/genética , Regulação para Baixo , Receptor alfa de Estrogênio/genética , Receptor alfa de Estrogênio/metabolismo , Receptor beta de Estrogênio/genética , Ácidos Graxos/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Camundongos , Neoplasias Experimentais , Distribuição Aleatória , Survivina/genética , Survivina/metabolismo , Transcriptoma , Tretinoína/farmacologia , Neoplasias de Mama Triplo Negativas/genética , Proteínas Wnt/genética , Proteínas Wnt/metabolismo
9.
PLoS Genet ; 17(7): e1009708, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34314419

RESUMO

The TMPRSS2/ERG gene rearrangement occurs in 50% of prostate tumors and results in expression of the transcription factor ERG, which is normally silent in prostate cells. ERG expression promotes prostate tumor formation and luminal epithelial cell fates when combined with PI3K/AKT pathway activation, however the mechanism of synergy is not known. In contrast to luminal fates, expression of ERG alone in immortalized normal prostate epithelial cells promotes cell migration and epithelial to mesenchymal transition (EMT). Migration requires ERG serine 96 phosphorylation via endogenous Ras/ERK signaling. We found that a phosphomimetic mutant, S96E ERG, drove tumor formation and clonogenic survival without activated AKT. S96 was only phosphorylated on nuclear ERG, and differential recruitment of ERK to a subset of ERG-bound chromatin associated with ERG-activated, but not ERG-repressed genes. S96E did not alter ERG genomic binding, but caused a loss of ERG-mediated repression, EZH2 binding and H3K27 methylation. In contrast, AKT activation altered the ERG cistrome and promoted expression of luminal cell fate genes. These data suggest that, depending on AKT status, ERG can promote either luminal or EMT transcription programs, but ERG can promote tumorigenesis independent of these cell fates and tumorigenesis requires only the transcriptional activation function.


Assuntos
Regulação Neoplásica da Expressão Gênica/genética , Neoplasias da Próstata/metabolismo , Animais , Carcinogênese/genética , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Transformação Celular Neoplásica/genética , Transição Epitelial-Mesenquimal , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Genes Reguladores , Xenoenxertos , Humanos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Nus , Fosfatidilinositol 3-Quinases/genética , Próstata/patologia , Neoplasias da Próstata/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/genética , Fatores de Transcrição/genética , Ativação Transcricional , Regulador Transcricional ERG/genética , Regulador Transcricional ERG/metabolismo , Proteínas ras/metabolismo
10.
PLoS Pathog ; 17(3): e1009419, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-33720992

RESUMO

Epstein-Barr virus (EBV) infection is associated with the development of specific types of lymphoma and some epithelial cancers. EBV infection of resting B-lymphocytes in vitro drives them to proliferate as lymphoblastoid cell lines (LCLs) and serves as a model for studying EBV lymphomagenesis. EBV nuclear antigen 3C (EBNA3C) is one of the genes required for LCL growth and previous work has suggested that suppression of the CDKN2A encoded tumor suppressor p16INK4A and possibly p14ARF is central to EBNA3C's role in this growth transformation. To directly assess whether loss of p16 and/or p14 was sufficient to explain EBNA3C growth effects, we used CRISPR/Cas9 to disrupt specific CDKN2A exons in EBV transformed LCLs. Disruption of p16 specific exon 1α and the p16/p14 shared exon 2 were each sufficient to restore growth in the absence of EBNA3C. Using EBNA3C conditional LCLs knocked out for either exon 1α or 2, we identified EBNA3C induced and repressed genes. By trans-complementing with EBNA3C mutants, we determined specific genes that require EBNA3C interaction with RBPJ or CtBP for their regulation. Unexpectedly, interaction with the CtBP repressor was required not only for repression, but also for EBNA3C induction of many host genes. Contrary to previously proposed models, we found that EBNA3C does not recruit CtBP to the promoters of these genes. Instead, our results suggest that CtBP is bound to these promoters in the absence of EBNA3C and that EBNA3C interaction with CtBP interferes with the repressive function of CtBP, leading to EBNA3C mediated upregulation.


Assuntos
Oxirredutases do Álcool/metabolismo , Transformação Celular Neoplásica/genética , Proteínas de Ligação a DNA/metabolismo , Infecções por Vírus Epstein-Barr/complicações , Antígenos Nucleares do Vírus Epstein-Barr/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Linfoma/virologia , Linfócitos B/patologia , Linfócitos B/virologia , Linhagem Celular , Humanos , Regulação para Cima
11.
Exp Cell Res ; 412(2): 113033, 2022 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-35041823

RESUMO

Exosomes mediate cellular communications in cancer by transmitting active molecules. However, the CAFs-derived molecular determinants that regulate esophageal squamous cell carcinoma (ESCC) metastasis have not been fully characterized. The purpose of this study was to investigate the potential roles of exosomal LINC01410 of ESCC cells. The characteristics of exosomes were identified using transmission electron microscope (TEM), Nanoparticle Tracking Analysis (NTA). The expression of LINC01410 and miR-122-5p was analyzed by quantitative real-time polymerase chain reaction (qRT-PCR) assay. The biological roles of LINC01410 in ESCC cells were investigated using transwell assay. Western blot assay was employed to detect protein levels. The potential downstream molecular mechanism of LINC01410 was demonstrated with dual-luciferase reporter assay, RNA immunoprecipitation (RIP) assay and RNA pull down. CAFs promote the metastasis and epithelial-mesenchymal transition (EMT) of ESCC cells. CAFs exert their roles by transferring exosomes to ESCC cells, leading to a significant increase of LINC01410 level in ESCC cells. Mechanically, LINC01410 secreted by CAFs-Exo could contribute to metastasis and EMT by sponging miR-122-5p and increasing PKM2 level in TE-1 and Eca-109 cells. Additionally, LINC01410/miR-122-5p/PKM2 axis affecting ESCC metastasis and EMT in vitro and in vivo.


Assuntos
Fibroblastos Associados a Câncer/metabolismo , Transição Epitelial-Mesenquimal/fisiologia , Neoplasias Esofágicas/metabolismo , Carcinoma de Células Escamosas do Esôfago/metabolismo , Exossomos/metabolismo , Animais , Linhagem Celular Tumoral , Movimento Celular/fisiologia , Proliferação de Células/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/metabolismo
12.
Semin Cancer Biol ; 76: 110-119, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34020028

RESUMO

The complex role of NRF2 in the context of cancer continues to evolve. As a transcription factor, NRF2 regulates various genes involved in redox homeostasis, protein degradation, DNA repair, and xenobiotic metabolism. As such, NRF2 is critical in preserving cell function and viability, particularly during stress. Importantly, NRF2 itself is regulated via a variety of mechanisms, and the mode of NRF2 activation often dictates the duration of NRF2 signaling and its role in either preventing cancer initiation or promoting cancer progression. Herein, different modes of NRF2 regulation, including oxidative stress, autophagy dysfunction, protein-protein interactions, and epigenetics, as well as pharmacological modulators targeting this cascade in cancer, are explored. Specifically, how the timing and duration of these different mechanisms of NRF2 induction affect tumor initiation, progression, and metastasis are discussed. Additionally, progress in the discovery and development of NRF2 inhibitors for the treatment of NRF2-addicted cancers is highlighted, including modulators that inhibit specific NRF2 downstream targets. Overall, a better understanding of the intricate nature of NRF2 regulation in specific cancer contexts should facilitate the generation of novel therapeutics designed to not only prevent tumor initiation, but also halt progression and ultimately improve patient wellbeing and survival.


Assuntos
Fator 2 Relacionado a NF-E2/metabolismo , Neoplasias/metabolismo , Neoplasias/patologia , Animais , Transformação Celular Neoplásica/metabolismo , Transformação Celular Neoplásica/patologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos
13.
Mol Pharmacol ; 101(2): 106-119, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34862309

RESUMO

Elevated expression of lysine demethylase 6A (KDM6A) and lysine demethylase 6B (KDM6B) has been reported in prostate cancer (PCa). However, the mechanism underlying the specific role of KDM6A/B in PCa is still fragmentary. Here, we report novel KDM6A/B downstream targets involved in controlling PCa cell proliferation. KDM6A and KDM6B mRNAs were higher in prostate adenocarcinoma, lymph node metastatic site (LNCaP) but not in prostate adenocarcinoma, bone metastatic site (PC3) and prostate adenocarcinoma, brain metastatic site (DU145) cells. Higher KDM6A mRNA was confirmed at the protein level. A metastasis associated gene focused oligonucleotide array was performed to identify KDM6A/B dependent genes in LNCaP cells treated with a KDM6 family selective inhibitor, ethyl-3-(6-(4,5-dihydro-1H-benzo[d]azepin-3(2H)-yl)-2-(pyridin-2-yl)pyrimidin-4-ylamino)propanoate (GSK-J4). This identified five genes [V-myc myelocytomatosis viral oncogene homolog (avian) (c-MYC), neurofibromin 2 (merlin) (NF2), C-terminal binding protein 1 (CTBP1), EPH receptor B2 (EPHB2), and plasminogen activator urokinase receptor (PLAUR)] that were decreased more than 50% by GSK-J4, and c-MYC was the most downregulated gene. Array data were validated by quantitative reverse transcription polymerase chain reaction (qRT-PCR), which detected a reduction in c-MYC steady state mRNA and prespliced mRNA, indicative of transcriptional repression of c-MYC gene expression. Furthermore, c-MYC protein was also decreased by GSK-J4. Importantly, GSK-J4 reduced mRNA and protein levels of c-MYC target gene, cyclinD1 (CCND1). Silencing of KDM6A/B with small interfering RNA (siRNA) confirmed that expression of both c-MYC and CCND1 are dependent on KDM6B. Phosphorylated retinoblastoma (pRb), a marker of G1 to S-phase transition, was decreased by GSK-J4 and KDM6B silencing. GSK-J4 treatment resulted in a decrease in cell proliferation and cell number, detected by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium, inner salt (MTS) assay and conventional cell counting, respectively. Consequently, we conclude that KDM6B controlling c-MYC, CCND1, and pRb contribute regulation of PCa cell proliferation, which represents KDM6B as a promising epigenetic target for the treatment of advanced PCa. SIGNIFICANCE STATEMENT: Lysine demethylase 6A (KDM6A) and 6B (KDM6B) were upregulated in prostate cancer (PCa). We reported novel KDM6A/B downstream targets controlling proliferation. Amongst 84 metastasis associated genes, V-myc myelocytomatosis viral oncogene homolog (avian) (c-MYC) was the most inhibited gene by KDM6 inhibitor, ethyl-3-(6-(4,5-dihydro-1H-benzo[d]azepin-3(2H)-yl)-2-(pyridin-2-yl)pyrimidin-4-ylamino)propanoate (GSK-J4). This was accompanied by decreased c-MYC targets, cyclinD1 (CCND1) and phosphorylated retinoblastoma (pRb), which were KDM6B dependent. GSK-J4 decreased proliferation and cell counting. We conclude that KDM6B controlling c-MYC, CCND1, and pRb contribute regulation of PCa proliferation.


Assuntos
Proliferação de Células/fisiologia , Proteínas de Ligação a DNA/biossíntese , Regulação Neoplásica da Expressão Gênica/fisiologia , Histona Desmetilases com o Domínio Jumonji/biossíntese , Neoplasias da Próstata/metabolismo , Fatores de Transcrição/biossíntese , Linhagem Celular Tumoral , Proteínas de Ligação a DNA/genética , Humanos , Histona Desmetilases com o Domínio Jumonji/genética , Masculino , Neoplasias da Próstata/genética , Fatores de Transcrição/genética
14.
Mol Pharmacol ; 101(2): 78-86, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34862308

RESUMO

Prohibitin-2 (PHB2) is a scaffold protein that has pleiotropic functions, which include interacting with γ-glutamylcyclotransferase (GGCT) in the cytoplasm and repressing the transcriptional activities of the p21Waf1/Cip (p21) gene in the nucleus. The cytotoxic drug fluorizoline binds to PHB1/2 and exerts antiproliferative actions on cancer cells. However, the precise mechanism underlying the antiproliferative effects of fluorizoline is not fully elucidated. In the present study, we first show that fluorizoline induces p21 expression in several human cancer cell lines, including MCF7 breast cancer cells. Treatment of MCF7 cells with fluorizoline suppressed proliferation and prevented cells from entering into the DNA synthesis phase. Knockdown of p21 rescued the suppressed proliferation, indicating that fluorizoline inhibited MCF7 cell growth via the induction of p21. Overexpression of PHB2 in MCF7 cells prevented the induction of p21 expression by fluorizoline and restored the antiproliferative effects and blockade of cell cycle progression. Moreover, treatment of MCF7 cells with fluorizoline inhibited the interaction between endogenous PHB2 and GGCT proteins and reduced the level of nuclear localization of PHB2 proteins. These results indicate that targeting PHB2 with fluorizoline induces the expression of p21 and consequently blocks proliferation of cancer cells. SIGNIFICANCE STATEMENT: This study shows that fluorizoline may be a promising novel anticancer drug candidate that induces p21 expression and blocks cell-cycle progression in human cancer cell lines. In addition, we show that fluorizoline inhibits the interaction between PHB2 and GGCT and reduces the nuclear localization of PHB2 proteins.


Assuntos
Antineoplásicos/farmacologia , Neoplasias da Mama/metabolismo , Inibidor de Quinase Dependente de Ciclina p21/biossíntese , Regulação Neoplásica da Expressão Gênica/fisiologia , Proibitinas/metabolismo , gama-Glutamilciclotransferase/metabolismo , Antineoplásicos/síntese química , Neoplasias da Mama/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/fisiologia , Inibidor de Quinase Dependente de Ciclina p21/genética , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Proibitinas/antagonistas & inibidores , gama-Glutamilciclotransferase/antagonistas & inibidores
15.
Am J Pathol ; 191(2): 368-384, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33181138

RESUMO

Canonical Wnt signaling is critical for melanocyte lineage commitment and melanoma development. RAD6B, a ubiquitin-conjugating enzyme critical for translesion DNA synthesis, potentiates ß-catenin stability/activity by inducing proteasome-insensitive polyubiquitination. RAD6B expression is induced by ß-catenin, triggering a positive feedback loop between the two proteins. RAD6B function in melanoma development/progression was investigated by targeting RAD6B using CrispR/Cas9 or an RAD6-selective small-molecule inhibitor #9 (SMI#9). SMI#9 treatment inhibited melanoma cell proliferation but not normal melanocytes. RAD6B knockout or inhibition in metastatic melanoma cells downregulated ß-catenin, ß-catenin-regulated microphthalmia-associated transcription factor (MITF), sex-determining region Y-box 10, vimentin proteins, and MITF-regulated melan A. RAD6B knockout or inhibition decreased migration/invasion, tumor growth, and lung metastasis. RNA-sequencing and stem cell pathway real-time RT-PCR analysis revealed profound reductions in WNT1 expressions in RAD6B knockout M14 cells compared with control. Expression levels of ß-catenin-regulated genes VIM, MITF-M, melan A, and TYRP1 (a tyrosinase family member critical for melanin biosynthesis) were reduced in RAD6B knockout cells. Pathway analysis identified gene networks regulating stem cell pluripotency, Wnt signaling, melanocyte development, pigmentation signaling, and protein ubiquitination, besides DNA damage response signaling, as being impacted by RAD6B gene disruption. These data reveal an important and early role for RAD6B in melanoma development besides its bonafide translesion DNA synthesis function, and suggest that targeting RAD6B may provide a novel strategy to treat melanomas with dysregulated canonical Wnt signaling.


Assuntos
Regulação Neoplásica da Expressão Gênica/fisiologia , Melanoma/metabolismo , Melanoma/patologia , Enzimas de Conjugação de Ubiquitina/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Linhagem Celular , Xenoenxertos , Humanos , Camundongos , Camundongos Nus , Fenótipo
16.
Am J Pathol ; 191(6): 1094-1107, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33705753

RESUMO

Patients with advanced prostate cancer are frequently treated with the antiandrogen enzalutamide. However, resistance eventually develops in virtually all patients, and various mechanisms have been associated with this process. The histone acetyltransferases EP300 and CREBBP are involved in regulation of cellular events in advanced prostate cancer. This study investigated the role of EP300/CREBBP inhibitors in enzalutamide-resistant prostate cancer. EP300/CREBBP inhibitors led to the same inhibition of androgen receptor activity in enzalutamide-resistant and -sensitive cells. However, enzalutamide-resistant cells were more sensitive to these inhibitors in viability assays. As indicated by the RNA-sequencing-based pathway analysis, genes related to the ribosome and MYC activity were significantly altered upon EP300/CREBBP inhibitor treatment. EP300/CREBBP inhibitors led to the down-regulation of ribosomal proteins RPL36 and RPL29. High-level ribosomal proteins amplifications and MYC amplifications were observed in castration-resistant prostate cancer samples of the publicly available Stand Up to Cancer data set. An inhibitor of RNA polymerase I-mediated transcription was used to evaluate the functional implications of these findings. The enzalutamide-resistant cell lines were more sensitive to this treatment. In addition, the migration rate of enzalutamide-resistant cells was strongly inhibited by this treatment. Taken together, the current data show that EP300/CREBBP inhibitors affect the MYC/ribosomal protein axis in enzalutamide-resistant cells and may have promising therapeutic implications.


Assuntos
Proteína de Ligação a CREB/metabolismo , Resistencia a Medicamentos Antineoplásicos/fisiologia , Proteína p300 Associada a E1A/metabolismo , Neoplasias de Próstata Resistentes à Castração/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Proteínas Ribossômicas/metabolismo , Antagonistas de Androgênios , Benzamidas , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Masculino , Nitrilas , Feniltioidantoína
17.
Am J Pathol ; 191(11): 1999-2008, 2021 11.
Artigo em Inglês | MEDLINE | ID: mdl-34358517

RESUMO

The efficacy of programmed death ligand (PD-L)-1/PD-1 checkpoint blockade in renal cell carcinoma (RCC) remains unknown. The effects of mTOR inhibitors are uncertain, and patients may develop resistance to them. The limited understanding of cancer cell-intrinsic mTOR-mediated pathways remains a challenge in developing effective treatments. Whether transcription factor (TF)-E3 regulates PD-L1 expression and the tumor microenvironment was investigated, and the effects of an mammalian target of rapamycin (mTOR) inhibitor on translocation RCC were explored. TFE3 was overexpressed in clear cell RCC cell lines, and PD-L1 expression was analyzed by Western blot analysis. PD-L1 activity in translocation RCC was analyzed in relation to TFE3 expression via TFE3 knockdown and treatment with an mTOR inhibitor. The results were correlated with the gene expression profile, evaluated using digital multiplex analysis. TFE3 and PD-L1 expression were positively correlated in RCC cells. TFE3 overexpression was associated with the expression of PD-L1 in RCC. Furthermore, mTOR inhibition was associated with enhanced PD-L1 expression via TFE3 activation in translocation RCC. These data support the feasibility of combination therapy based on mTOR inhibition and PD-L1 blockade as a novel strategy for the treatment of patients with translocation RCC.


Assuntos
Antígeno B7-H1/metabolismo , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos/metabolismo , Carcinoma de Células Renais/metabolismo , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias Renais/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Linhagem Celular Tumoral , Humanos
18.
Am J Pathol ; 191(1): 180-193, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33129761

RESUMO

Several studies have confirmed the function of Su(var)3-9, Enhancer of zeste, and Trithorax (SET) domain-containing 5 (SETD5) in post-translational modifications of nonhistone proteins. Mutation of the SETD5 gene has been implicated in the progression of many human cancers, such as breast cancer (BC), but its functional role in BC progression is still unknown. The current article investigates the clinical significance and the functional role of SETD5 in BC. Our studies show that SETD5 expression in BC was related to poor clinical outcomes, including lymph node metastasis and advanced clinical stage. SETD5 expression positively correlated with tumor-associated macrophages. SETD5 was an independent predictor of poor overall survival in BC. Furthermore, these studies show that down-regulation of SETD5 significantly decreased BC cell proliferation, metastasis, and angiogenesis, and increased apoptosis of BC cells. The mechanistic analysis showed that SETD5 contributes BC progression by interacting with AKT1 pathway. Also, in vivo experiments show that blocking of SETD5 expression significantly inhibited tumor growth and pulmonary metastasis of BC cells. These findings indicate that SETD5 is a potential prognosis marker and facilitates tumor progression of BC.


Assuntos
Neoplasias da Mama/patologia , Carcinoma Ductal de Mama/patologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Metiltransferases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Biomarcadores Tumorais/metabolismo , Neoplasias da Mama/metabolismo , Carcinoma Ductal de Mama/metabolismo , Proliferação de Células/fisiologia , Progressão da Doença , Feminino , Xenoenxertos , Humanos , Neoplasias Pulmonares/secundário , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica/patologia , Transdução de Sinais/fisiologia , Regulação para Cima
19.
Hepatology ; 73(6): 2361-2379, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33205519

RESUMO

BACKGROUND AND AIMS: The mechanism by which tumor cells resist metabolic stress remains unclear, but many oncogenes are known to regulate this process. Accordingly, metabolic stress is closely associated with tumor metastasis. In this study, gene chip technology showed that Ras homolog family member F, filopodia associated (RHOF), a member of the Rho guanosine triphosphatase family, is an oncogene that is significantly related to hepatocellular carcinoma (HCC) metastasis; however, it has rarely been reported in tumors. Our aim was to determine the clinicopathological significance and role of RHOF in HCC progression and investigate the associated mechanisms. APPROACH AND RESULTS: The results showed that compared to expression in adjacent noncancerous tissues, RHOF was frequently up-regulated in HCC tumor samples and elevated under conditions of glucose deprivation. RHOF expression was associated with tumor-node-metastasis stage, T grade, metastasis status, recurrence, and survival in HCC. RHOF also affected cell morphology and promoted migration, invasion, and epithelial-mesenchymal transition (EMT) of HCC cell lines. Analysis of the underlying mechanism showed that RHOF promoted the Warburg effect by up-regulating the expression and function of several glycolytic enzymes in HCC cells. This metabolic shift enhanced HCC cell migration and invasion. Specifically, RHOF exerted a tumor-promoting effect by directly interacting with AMP-activated protein kinase (AMPK) and increasing the phosphorylation of AMPK. This subsequently affected RAB3D mRNA stability and led to elevated RAB3D expression, thereby amplifying the Warburg effect and malignant biological behaviors of HCC cells. CONCLUSIONS: RHOF helps tumor cells resist metabolic stress through modulating the Warburg effect and plays a critical role in promoting HCC cell migration, invasion, and EMT, highlighting its important role in remodeling the metastatic microenvironment and regulating tumor metastasis. RHOF shows potential as a therapeutic target and prognostic biomarker for HCC.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Carcinoma Hepatocelular , Regulação Neoplásica da Expressão Gênica/fisiologia , Neoplasias Hepáticas , Estresse Fisiológico/fisiologia , Proteínas rho de Ligação ao GTP/metabolismo , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Linhagem Celular Tumoral , Descoberta de Drogas , Transição Epitelial-Mesenquimal/fisiologia , Humanos , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Invasividade Neoplásica , Metástase Neoplásica , Estadiamento de Neoplasias , Fosforilação , Prognóstico , Regulação para Cima , Proteínas rab3 de Ligação ao GTP/metabolismo
20.
Exp Eye Res ; 216: 108955, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35074341

RESUMO

Krüppel-like factor 2 (KLF2) belongs to the KLF family of zinc-finger transcription factors and mediates the occurrence and progression of various cancers. However, little is known about its expression pattern and biological role in retinoblastoma (RB). In the present study, we showed that KLF2 was markedly downregulated in human RB tissue compared with retina. KLF2 overexpression significantly inhibited RB cell proliferation and decreased proliferating cell nuclear antigen (PCNA) expression. Subsequently, we confirmed that KLF2 arrested cells at the G1-S phase transition, accompanied by the upregulation of p21 and downregulation of CyclinD1, as well as the activation of mitochondria-mediated apoptosis in RB cells. In addition, KLF2 overexpression contributed to suppressing RB cell migration and invasion by downregulating matrix metallopeptidase 9 (MMP9). On the contrary, KLF2 downregulation promoted RB cells proliferation, migration and invasion. Notably, the KLF2 expression pattern was opposite to that of C-X-C chemokine receptor 4 (CXCR4) in the two RB cell lines, KLF2 overexpression significantly decreased CXCR4 expression, silencing KLF2 had the opposite effect. Furthermore, dual-luciferase reporter and chromatin immunoprecipitation (ChIP) assays confirmed that KLF2 directly bound to the CXCR4 promoter and negatively regulated its expression in RB cells. Collectively, our results suggested that KLF2 function as a tumor suppressor in RB and may represent a potential therapeutic target for RB.


Assuntos
Fatores de Transcrição Kruppel-Like/fisiologia , Neoplasias da Retina/metabolismo , Retinoblastoma/metabolismo , Proteínas Supressoras de Tumor/fisiologia , Apoptose/fisiologia , Western Blotting , Ciclo Celular/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Ciclina D1/genética , Regulação Neoplásica da Expressão Gênica/fisiologia , Humanos , Marcação In Situ das Extremidades Cortadas , Plasmídeos , Antígeno Nuclear de Célula em Proliferação/genética , RNA Interferente Pequeno/genética , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias da Retina/patologia , Retinoblastoma/patologia , Transfecção , Quinases Ativadas por p21/genética
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa