Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
1.
Mol Cell ; 53(4): 534-48, 2014 Feb 20.
Artigo em Inglês | MEDLINE | ID: mdl-24486017

RESUMO

Mitochondrial pyruvate dehydrogenase complex (PDC) is crucial for glucose homeostasis in mammalian cells. The current understanding of PDC regulation involves inhibitory serine phosphorylation of pyruvate dehydrogenase (PDH) by PDH kinase (PDK), whereas dephosphorylation of PDH by PDH phosphatase (PDP) activates PDC. Here, we report that lysine acetylation of PDHA1 and PDP1 is common in epidermal growth factor (EGF)-stimulated cells and diverse human cancer cells. K321 acetylation inhibits PDHA1 by recruiting PDK1, and K202 acetylation inhibits PDP1 by dissociating its substrate PDHA1, both of which are important in promoting glycolysis in cancer cells and consequent tumor growth. Moreover, we identified mitochondrial ACAT1 and SIRT3 as the upstream acetyltransferase and deacetylase, respectively, of PDHA1 and PDP1, while knockdown of ACAT1 attenuates tumor growth. Furthermore, Y381 phosphorylation of PDP1 dissociates SIRT3 and recruits ACAT1 to PDC. Together, hierarchical, distinct posttranslational modifications act in concert to control molecular composition of PDC and contribute to the Warburg effect.


Assuntos
Acetil-CoA C-Acetiltransferase/metabolismo , Piruvato Desidrogenase (Lipoamida)-Fosfatase/metabolismo , Piruvato Desidrogenase (Lipoamida)/metabolismo , Sirtuína 3/metabolismo , Tirosina/química , Animais , Linhagem Celular Tumoral , Proliferação de Células , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glicólise , Humanos , Lisina/química , Masculino , Camundongos , Camundongos Nus , Mitocôndrias/metabolismo , Transplante de Neoplasias , Neoplasias/metabolismo , Fosforilação
2.
J Enzyme Inhib Med Chem ; 32(1): 992-1001, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28766956

RESUMO

While progress has been made in treating cancer, cytotoxic chemotherapeutic agents are still the most widely used drugs and are associated with severe side-effects. Drugs that target unique molecular signalling pathways are needed for treating cancer with low or no intrinsic toxicity to normal cells. Our goal is to target hypoxic tumours and specifically the hypoxia inducible factor (HIF) pathway for the development of new cancer therapies. To this end, we have previously developed benzopyran-based HIF-1 inhibitors such as arylsulfonamide KCN1. However, KCN1 and its earlier analogs have poor water solubility, which hamper their applications. Herein, we describe a series of KCN1 analogs that incorporate a morpholine moiety at various positions. We found that replacing the benzopyran group of KCN1 with a phenyl group with a morpholinomethyl moiety at the para positions had minimal effect on potency and improved the water solubility of two new compounds by more than 10-fold compared to KCN1, the lead compound.


Assuntos
Benzopiranos/farmacologia , Desenho de Fármacos , Fator 1 Induzível por Hipóxia/antagonistas & inibidores , Benzopiranos/síntese química , Benzopiranos/química , Solubilidade , Água/química
3.
Bioorg Med Chem Lett ; 21(18): 5528-32, 2011 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-21831638

RESUMO

Solid tumors generally grow under hypoxic conditions, a pathophysiological change, which activates the expression of genes responsible for malignant, aggressive, and treatment-refractory properties. Hypoxia inducible factor (HIF) is the chief transcription factor regulating hypoxia-driven gene expression. Therefore, the HIF pathway has become a critical target for cancer therapeutics development. We screened a privileged library of about 10,000 natural-product-like compounds using a cell-based assay for HIF-dependent transcriptional activity and identified several arylsulfonamide HIF pathway inhibitors. Among these compounds, the most potent ones showed an IC(50) of ∼0.5 µM in the hypoxia-responsive element (HRE)-luciferase reporter system. Further studies are needed to fully elucidate the mechanism of action of this class of compounds and their structure-activity relationship.


Assuntos
Sulfonamidas/farmacologia , Fatores de Transcrição/antagonistas & inibidores , Fosfatase Alcalina/antagonistas & inibidores , Fosfatase Alcalina/metabolismo , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Fator 1 Induzível por Hipóxia , Estrutura Molecular , Bibliotecas de Moléculas Pequenas , Estereoisomerismo , Relação Estrutura-Atividade , Sulfonamidas/química , Fatores de Transcrição/metabolismo
4.
Oncogene ; 40(33): 5182-5191, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34218269

RESUMO

Uveal melanoma (UM) is the most prevalent primary intraocular malignancy in adults, and patients that develop metastases (~50%) survive <1 year, highlighting the urgent need for new therapies. TCGA has recently revealed that a hypoxia gene signature is associated with poor UM patient prognosis. Here we show that expression of hypoxia-regulated collagen prolyl-4-hydroxylase genes P4HA1 and P4HA2 is significantly upregulated in UM patients with metastatic disease and correlates with poor prognosis, suggesting these enzymes might be key tumor drivers. We targeted hypoxia-induced expression of P4HA1/2 in UM with KCN1, a hypoxia inducible factor-1 (HIF-1) pathway inhibitor and found potent inhibition of primary and metastatic disease and extension of animal survival, without overt side effects. At the molecular level, KCN1 antagonized hypoxia-induced expression of P4HA1 and P4HA2, which regulate collagen maturation and deposition in the extracellular matrix. The treatment decreased prolyl hydroxylation, induced proteolytic cleavage and rendered a disordered structure to collagen VI, the main collagen produced by UM, and reduced UM cell invasion. Together, these data demonstrate that extracellular collagen matrix formation can be targeted in UM by inhibiting hypoxia-induced P4HA1 and P4HA2 expression, warranting further development of this strategy in patients with uveal melanoma.


Assuntos
Prolina Dioxigenases do Fator Induzível por Hipóxia , Matriz Extracelular , Humanos , Hidroxilação , Melanoma , Ativação Transcricional , Regulação para Cima , Neoplasias Uveais
5.
Biochim Biophys Acta ; 1795(2): 162-72, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19344680

RESUMO

Transcriptional activation by hypoxia is mediated by the hypoxia-inducible factor (HIF) via binding to the hypoxia-responsive element (HRE). Hypoxia in solid tumors associates with poorer outcome of the disease and reliable cellular markers of tumor hypoxia would represent a valuable diagnostic marker and a potential therapeutic target. In this category, carbonic anhydrase IX (CAIX) is one of the most promising candidates. Here, we summarize the knowledge about transcriptional regulation of CA9. The HRE is the central regulatory element in the CA9 promoter, whereas other elements are limited to lesser roles of amplification of signals received at the HRE. The analysis of known mechanisms of activation of CA9 reveals the prominent role of the HIF-1 pathway. Experimental paradigms with uncoupled HIF-1alpha stability and transcriptional activity (pericellular hypoxia, proteasomal inhibitor) provide evidence that CA9 expression monitors transcriptional activity of HIF-1, rather than the abundance of HIF-1alpha. Furthermore, these paradigms could provide a corollary to some of the apparently discordant cases (CAIX+, HIF-1alpha-) or (CAIX-, HIF-1alpha+) observed in vivo. In conclusion, the existing data support the notion that CA9, due to the unique structure of its promoter, is one of the most sensitive endogenous sensors of HIF-1 activity.


Assuntos
Antígenos de Neoplasias/genética , Anidrases Carbônicas/genética , Hipóxia Celular , Regulação Enzimológica da Expressão Gênica , Fator 1 Induzível por Hipóxia/fisiologia , Neoplasias/enzimologia , Sequência de Bases , Biomarcadores , Biomarcadores Tumorais , Anidrase Carbônica IX , Epigênese Genética , Humanos , Dados de Sequência Molecular , Neoplasias/etiologia , Regiões Promotoras Genéticas , Elementos de Resposta/fisiologia , Transcrição Gênica , Proteína Supressora de Tumor p53/fisiologia
6.
Oncogene ; 39(5): 1041-1048, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31582835

RESUMO

Medulloblastoma (MB) is a malignant pediatric brain tumor for which new therapies are urgently needed. We demonstrate that treatment with EPZ-6438 (Tazemetostat), an enhancer of zeste homolog 2 (EZH2) inhibitor approved for clinical trials, blocks MB cell growth in vitro and in vivo, and prolongs survival in orthotopic xenograft models. We show that the therapeutic effect is dependent on epigenetic reactivation of adhesion G-protein-coupled receptor B1 (BAI1/ADGRB1), a tumor suppressor that controls p53 stability by blocking Mdm2. Histone 3 trimethylated on lysine 27 (H3K27me3), a marker of silent chromatin conformation is present at the ADGRB1 promoter, and inhibition of EZH2, the catalytic component of the Polycomb Repressive complex 2 (PRC2) that methylates H3K27, switches the gene into an active chromatin status and reactivates BAI1 expression. Mechanistically, targeting EZH2 promotes transition from H3K27me3 to H3K27ac at the promoter, recruits the C/EBPß (CREB-binding protein) and CBP transcription factors and activates ADGRB1 gene transcription. Taken together, our results identify key molecular players that regulate ADGRB1 gene expression in MB, demonstrate that reactivation of BAI1 expression underlies EPZ-6438 antitumorigenic action, and provide preclinical proof-of-principle evidence for targeting EZH2 in patients with MB.


Assuntos
Proteínas Angiogênicas/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Epigênese Genética , Meduloblastoma/patologia , Receptores Acoplados a Proteínas G/genética , Proteína Supressora de Tumor p53/genética , Proteínas Angiogênicas/deficiência , Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Transformação Celular Neoplásica , Inativação Gênica , Histonas/metabolismo , Humanos , Metilação , Fragmentos de Peptídeos/metabolismo , Receptores Acoplados a Proteínas G/deficiência , Sialoglicoproteínas/metabolismo
7.
Oncogene ; 39(5): 1165, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31676871

RESUMO

An amendment to this paper has been published and can be accessed via a link at the top of the paper.

8.
Mol Cell Biol ; 26(15): 5895-907, 2006 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-16847340

RESUMO

The ubiquitin-proteasome pathway (UPP) is involved in regulation of multiple cellular processes. Hypoxia-inducible factor 1 alpha (HIF-1 alpha) is a prototypic target of the UPP and, as such, is stabilized under conditions of proteasomal inhibition. Using carbonic anhydrase IX (CAIX) and vascular endothelial growth factor (VEGF) expression as paradigmatic markers of HIF-1 activity, we found that proteasomal inhibitors (PI) abrogated hypoxia-induced CAIX expression in all cell lines tested and VEGF expression in two out of three. Mapping of the inhibitory effect identified the C-terminal activation domain (CAD) of HIF-1 alpha as the primary target of PI. PI specifically inhibited the HIF-1 alpha CAD despite activating the HIF-1 alpha coactivator p300 and another p300 cysteine/histidine-rich domain 1-dependent transcription factor, STAT-2. Coimmunoprecipitation and glutathione S-transferase pull downs indicated that PI does not disrupt interactions between HIF-1 alpha and p300. Mutational analysis failed to confirm involvement of sites of known or putative posttranslational modifications in regulation of HIF-1 alpha CAD function by PI. Our data provide evidence for the counterintuitive hypothesis that inhibition of HIF-1 function could be responsible for at least some of the antitumor effects of proteasomal inhibition. Further studies of the mechanism of the PI-induced attenuation of HIF-1alpha will provide important, potentially novel insight into regulation of HIF-1 activity and possibly identify new targets for HIF-directed therapy.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Inibidores de Proteassoma , Transcrição Gênica , Animais , Biomarcadores/metabolismo , Ácidos Borônicos/metabolismo , Bortezomib , Anidrases Carbônicas/metabolismo , Linhagem Celular , Ativação Enzimática , Regulação da Expressão Gênica , Humanos , Hipóxia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Isoenzimas/metabolismo , Inibidores de Proteases/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Estrutura Terciária de Proteína , Pirazinas/metabolismo , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Fatores de Transcrição de p300-CBP/metabolismo
9.
Clin Cancer Res ; 25(7): 2206-2218, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30563937

RESUMO

PURPOSE: Uveal melanoma (UM) is the most prevalent and lethal intraocular malignancy in adults. Here, we examined the importance of hypoxia in UM growth and tested the antitumor effects of arylsulfonamide 64B, an inhibitor of the hypoxia-induced factor (HIF) pathway in animal models of UM and investigated the related mechanisms. EXPERIMENTAL DESIGN: UM cells were implanted in the uvea of mice eyes and mice systemically treated with 64B. Drug effect on primary eye tumor growth, circulating tumor cells, metastasis formation in liver, and survival were examined. 64B effects on UM cell growth, invasion and hypoxia-induced expression of C-X-C chemokine receptor type 4 (CXCR4) and mesenchymal-epithelial transition factor (c-Met) were measured. Luciferase reporter assays, chromatin immunoprecipitation, co-immunoprecipitation, and cellular thermal shift assays were used to determine how 64B interferes with the HIF transcriptional complex. RESULTS: Systemic administration of 64B had potent antitumor effects against UM in several orthotopic mouse models, suppressing UM growth in the eye (∼70% reduction) and spontaneous liver metastasis (∼50% reduction), and extending mice survival (P < 0.001) while being well tolerated. 64B inhibited hypoxia-induced expression of CXCR4 and c-Met, 2 key drivers of tumor invasion and metastasis. 64B disrupted the HIF-1 complex by interfering with HIF-1α binding to p300/CBP co-factors, thus reducing p300 recruitment to the MET and CXCR4 gene promoters. 64B could thermostabilize p300, supporting direct 64B binding to p300. CONCLUSIONS: Our preclinical efficacy studies support the further optimization of the 64B chemical scaffold toward a clinical candidate for the treatment of UM.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Hipóxia/metabolismo , Melanoma/etiologia , Melanoma/metabolismo , Proteínas Proto-Oncogênicas c-met/genética , Receptores CXCR4/genética , Sulfonamidas/farmacologia , Neoplasias Uveais/etiologia , Neoplasias Uveais/metabolismo , Animais , Biomarcadores Tumorais , Biópsia , Linhagem Celular Tumoral , Modelos Animais de Doenças , Proteína p300 Associada a E1A/metabolismo , Humanos , Neoplasias Hepáticas/secundário , Melanoma/tratamento farmacológico , Melanoma/patologia , Camundongos , Prognóstico , Ligação Proteica , Proteínas Proto-Oncogênicas c-met/metabolismo , Receptores CXCR4/metabolismo , Sulfonamidas/química , Neoplasias Uveais/tratamento farmacológico , Neoplasias Uveais/patologia , Ensaios Antitumorais Modelo de Xenoenxerto
10.
J Cell Biochem ; 104(2): 536-44, 2008 May 15.
Artigo em Inglês | MEDLINE | ID: mdl-18059036

RESUMO

At the cellular level hypoxia induces transcriptional response that is mediated by the transcription factor hypoxia-inducible factor (HIF). HIF is regulated at the level of its alpha subunit by 2-oxoglutarate (2OG)-dependent oxygenases that hydroxylate specific prolyl and asparaginyl residues of HIF-alpha, affecting its stability and activity, respectively. In the presence of O(2), the alpha subunit is degraded in a complex process with several distinct steps. In the first step, the degradation process is initiated by prolyl hydroxylases (PHDs). In the second step, the von Hippel-Lindau (VHL)/E3 ligase complex recognizes the hydroxylated HIF-alpha and mediates its polyubiquitylation by the ubiquitin-conjugating enzyme E2. In the third step, the polyubiquitylated HIF-alpha is translocated to the proteasome where it is degraded. Degradation of HIF-alpha can be inhibited at any of the three levels either by various pharmacological inhibitors or due to inactivation of genes whose products regulate the HIF system. The emerging data about inactivation of HIF under conditions of proteasomal inhibition prompted us to provide an overview contrasting the outcome of inhibition at various stages of the degradative pathway for HIF activity.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores Enzimáticos , Humanos , Inibidores de Proteassoma
11.
Biochem Biophys Res Commun ; 370(4): 613-8, 2008 Jun 13.
Artigo em Inglês | MEDLINE | ID: mdl-18402769

RESUMO

The hypoxia-inducible factor (HIF) activates transcription via binding to the highly variable hypoxia-responsive elements (HREs). All hypoxia-inducible constructs described to date utilize multimers of naturally occurring HREs. Here, we describe the rational design of minimal hypoxia-inducible enhancers, conceptually equivalent to using an optimized HIF-binding site (HBS) as the building block. Optimizations of the HBS, spacing between HBSs, the distance from the minimal promoter, and orientation of HBSs allowed us to design constructs with high hypoxic activity. Activation of the 4xopt HBS (36bp) construct by hypoxia or HIF-1alpha and HIF-2alpha was comparable with that of the 4xEPO HRE (208bp) construct. The strong synergism between the properly arranged optimized HBSs was due to stimulation of high affinity HIF binding. Our data prove, for the first time, that it is possible to assemble artificial hypoxia-inducible enhancers from a single type of regulatory element-optimized HBS.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Elementos Facilitadores Genéticos/genética , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , TATA Box/genética , Anaerobiose/genética , Animais , Sequência de Bases , Sítios de Ligação , Linhagem Celular , Humanos , Isoenzimas/genética , L-Lactato Desidrogenase/genética , Lactato Desidrogenase 5 , Camundongos , Fosfoglicerato Quinase/genética , Inibidor 1 de Ativador de Plasminogênio/genética , Ratos
12.
Clin Chim Acta ; 395(1-2): 6-13, 2008 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-18505681

RESUMO

Cells experiencing lowered O(2) levels (hypoxia) undergo a variety of biological responses in order to adapt to these unfavorable conditions. The master switch, orchestrating the cellular response to low O(2) levels, is the transcription factor, termed hypoxia-inducible factor (HIF). The alpha subunits of HIF are regulated by 2-oxoglutarate-dependent oxygenases that, in the presence of O(2), hydroxylate specific prolyl and asparaginyl residues of HIF-alpha, inducing its proteasome-dependent degradation and repression of transcriptional activity, respectively. Hypoxia inhibits oxygenases, stabilized HIF-alpha translocates to the nucleus, dimerizes with HIF-beta, recruits the coactivators p300/CBP, and induces expression of its transcriptional targets via binding to hypoxia-responsive elements (HREs). HREs are composite regulatory elements, comprising a conserved HIF-binding sequence and a highly variable flanking sequence that modulates the transcriptional response. In summary, the transcriptional response of a cell is the end product of two major functions. The first (trans-acting) is the level of activation of the HIF pathway that depends on regulation of stability and transcriptional activity of the HIF-alpha. The second (cis-acting) comprises the characteristics of endogenous HREs that are determined by the availability of transcription factors cooperating with HIF and/or individual HIF-alpha isoforms.


Assuntos
Hipóxia Celular/genética , Regulação da Expressão Gênica , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Elementos de Resposta/genética , Transdução de Sinais/genética , Transcrição Gênica , Animais , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Elementos de Resposta/fisiologia , Transdução de Sinais/fisiologia
13.
Mol Cell Biol ; 24(13): 5757-66, 2004 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15199132

RESUMO

We investigated the relationship between the tumor suppressor p53 and the hypoxia-inducible factor-1 (HIF-1)-dependent expression of the hypoxia marker, carbonic anhydrase IX (CAIX). MCF-7 (wt p53) and Saos-2 (p53-null) cells displayed similar induction of CAIX expression and CA9 promoter activity under hypoxic conditions. Activation of p53 by the DNA damaging agent mitomycin C (MC) was accompanied by a potent repression of CAIX expression and the CA9 promoter in MCF-7 but not in Saos-2 cells. The activated p53 mediated increased proteasomal degradation of HIF-1alpha protein, resulting in considerably lower steady-state levels of HIF-1alpha protein in hypoxic MCF-7 cells but not in Saos-2 cells. Overexpression of HIF-1alpha relieved the MC-induced repression in MCF-7 cells, confirming regulation at the HIF-1alpha level. Similarly, CA9 promoter activity was downregulated by MC in HCT 116 p53(+/+) but not the isogenic p53(-/-) cells. Activated p53 decreased HIF-1alpha protein levels by accelerated proteasome-dependent degradation without affecting significantly HIF-1alpha transcription. In summary, our results demonstrate that the presence of wtp53 under hypoxic conditions has an insignificant effect on the stabilization of HIF-1alpha protein and HIF-1-dependent expression of CAIX. However, upon activation by DNA damage, wt p53 mediates an accelerated degradation of HIF-1alpha protein, resulting in reduced activation of CA9 transcription and, correspondingly, decreased levels of CAIX protein. A model outlining the quantitative relationship between p53, HIF-1alpha, and CAIX is presented.


Assuntos
Antígenos de Neoplasias/metabolismo , Anidrases Carbônicas/metabolismo , Cisteína Endopeptidases/metabolismo , Dano ao DNA , Hipóxia/metabolismo , Fatores de Transcrição/metabolismo , Proteína Supressora de Tumor p53/fisiologia , Antígenos de Neoplasias/genética , Anidrase Carbônica IX , Anidrases Carbônicas/genética , Linhagem Celular Tumoral , Regulação para Baixo/efeitos dos fármacos , Humanos , Hipóxia/enzimologia , Subunidade alfa do Fator 1 Induzível por Hipóxia , Mitomicina/farmacologia , Regiões Promotoras Genéticas , Fatores de Transcrição/biossíntese , Transcrição Gênica , Proteína Supressora de Tumor p53/deficiência , Proteína Supressora de Tumor p53/genética
14.
Bio Protoc ; 7(17)2017 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-28966947

RESUMO

The transcription factor Hypoxia-Inducible Factor (HIF) complexes with the coactivator p300, activating the hypoxia response pathway and allowing tumors to grow. The CH1 and CAD domains of each respective protein form the interface between p300 and HIF. Small molecule compounds are in development that target and inhibit HIF/p300 complex formation, with the goal of reducing tumor growth. High resolution NMR spectroscopy is necessary to study ligand interaction with p300-CH1, and purifying high quantities of properly folded p300-CH1 is needed for pursuing structural and biophysical studies. p300-CH1 has 3 zinc fingers and 9 cysteine residues, posing challenges associated with reagent compatibility and protein oxidation. A protocol has been developed to overcome such issues by incorporating zinc during expression and streamlining the purification time, resulting in a high yield of optimally folded protein (120 mg per 4 L expression media) that is suitable for structural NMR studies. The structural integrity of the final recombinant p300-CH1 has been verified to be optimal using onedimensional 1H NMR spectroscopy and circular dichroism. This protocol is applicable for the purification of other zinc finger containing proteins.

15.
Oncotarget ; 8(59): 99245-99260, 2017 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-29245898

RESUMO

Neoplastic cells display reprogrammed metabolism due to the heightened energetic demands and the need for biomass synthesis of a growing tumor. Targeting metabolic vulnerabilities is thus an important goal for cancer therapy. Here, we describe a novel small-molecule arylsulfonamide (N-cyclobutyl-N-((2,2-dimethyl-2H-pyrano[3,2-b]pyridin-6-yl)methyl)-3,4-dimethoxybenzenesulfonamide) that exerts potent cytotoxicity and energetic stress on tumor cells while largely sparing non-cancerous human cells. In tumor cells, it stimulates glycolysis and accelerates glucose consumption. Consequently, intracellular ATP levels plummet, triggering activation of AMP-activated protein kinase (AMPK), and diminishing the mammalian target of rapamycin complex 1 (mTORC1) and hypoxia-inducible factor 1 (HIF-1) signaling. In orthotopic triple-negative breast cancer and subcutaneous lung cancer mouse models, this arylsulfonamide robustly suppresses primary tumor growth, inhibits the formation of distant metastases to the lung, and extends mouse survival while being very well tolerated. These therapeutic effects are further potentiated by co-administration of 2-deoxy-D-glucose (2-DG), a glucose analog and glycolysis inhibitor. Collectively, our findings provide preclinical proof of concept for the further development of this arylsulfonamide in combination with 2-DG towards cancer treatment.

16.
Cancer Res ; 63(5): 917-22, 2003 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-12615703

RESUMO

In the present study, we further studied mechanisms of transcriptional regulation of the tumor-associated carbonic anhydrase IX (CAIX). We identified PR5 in the CA9 promoter as another SP1/SP3-binding site. As shown by electromobility shift assays and block-replacement mutagenesis, PR5 is functionally equivalent to the SP1/SP3-binding PR1 identified previously. However, there is a strong requirement for SP1/SP3 activity in the PR1 position, and SP1/SP3 activity from the PR5 position cannot compensate for this. In various cell lines, the expression of endogenous CAIX and activity of CA9 promoter constructs depend on SP1/SP3 activity as demonstrated by the dose-dependent inhibitory effect of the SP1 inhibitor mithramycin A. The two conditions of the induction of CAIX expression described previously differ in their sensitivity to mithramycin A inhibition; the hypoxia-mimic-induced expression is less sensitive than the cell density (mild hypoxia)-induced expression. Our present study highlights the importance of SP1/SP3 activity for CAIX expression and provides additional evidence for distinct mechanisms responsible for true and mild hypoxia-induced CAIX expression. The presence of a SP1/SP3-binding element in the PR1 position is absolutely required for mild hypoxia-induced activity, and it significantly up-regulates the true hypoxic induction. The SP1/SP3 and hypoxia-response element in the CA9 promoter thus may represent a novel type of enhancer capable of mounting responses to a wider range of hypoxic conditions.


Assuntos
Antígenos de Neoplasias/biossíntese , Anidrases Carbônicas/biossíntese , Proteínas de Neoplasias/biossíntese , Fator de Transcrição Sp1/fisiologia , Antígenos de Neoplasias/genética , Sítios de Ligação , Anidrase Carbônica IX , Anidrases Carbônicas/genética , Hipóxia Celular/fisiologia , Fibrossarcoma , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Células HeLa , Humanos , Proteínas de Neoplasias/genética , Fragmentos de Peptídeos/metabolismo , Regiões Promotoras Genéticas , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Ativação Transcricional , Transfecção , Células Tumorais Cultivadas
17.
Cancer Res ; 62(15): 4469-77, 2002 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-12154057

RESUMO

Transcription of the gene coding for the tumor-associated antigen MN/carbonic anhydrase IX (CAIX) is regulated by hypoxia-inducible factor 1 (HIF-1). Previous studies identified CAIX expression in areas adjacent to hypoxic regions in solid tumors and suggested supplementary/alternative modes of regulation. To better understand the mechanisms activating CAIX expression, we characterized the cell density-dependent induction of CAIX in HeLa cells. This process is anchorage and serum independent and is not mediated by a soluble factor, decreased pH, or lowered glucose concentration. Stabilization of HIF-1 alpha was not observed in dense cultures. In contrast to sparse cell culture conditions, phosphatidylinositol 3'-kinase (PI3K) activity was significantly increased in dense HeLa cultures. The PI3K inhibitors LY294002 and wortmannin inhibited CAIX expression in dense cultures in a dose-dependent manner, specifically targeting the CA9 promoter (-173/+31 region) that was transactivated by constitutively active p110 PI3K subunit. The mechanism controlling CAIX expression in dense cultures is, however, dependent on lowered O(2) tension because stirring abrogates induction of CAIX expression. Hypoxia- and cell density-induced CAIX expressions were mediated by two seemingly independent mechanisms, as documented by the additive effect of increased cell density and treatment with the hypoxia-mimic CoCl(2) on levels of CAIX expression. The minimal cell density-dependent region within the CA9 promoter consists of the juxtaposed protected region 1 and hypoxia-response elements. However cell density-dependent CAIX expression was abrogated in the HIF-1 alpha-deficient Kal3.5 cells, suggesting an important role of HIF-1 in the corresponding mechanism. Thus, induction of CAIX in high-density cultures requires separate but interdependent pathways of PI3K activation and a minimal level of HIF-1 alpha. These interdependent pathways function at a lowered O(2) concentration that is, however, above that necessary for HIF-1 alpha stabilization.


Assuntos
Antígenos de Neoplasias , Anidrases Carbônicas/biossíntese , Anidrases Carbônicas/genética , Proteínas de Neoplasias/biossíntese , Proteínas de Neoplasias/genética , Oxigênio/fisiologia , Fosfatidilinositol 3-Quinases/fisiologia , Fatores de Transcrição/fisiologia , Anidrase Carbônica IX , Contagem de Células , Ativação Enzimática , Fibrossarcoma/enzimologia , Fibrossarcoma/genética , Regulação Enzimológica da Expressão Gênica/fisiologia , Regulação Neoplásica da Expressão Gênica/fisiologia , Glucose/deficiência , Células HeLa , Humanos , Concentração de Íons de Hidrogênio , Subunidade alfa do Fator 1 Induzível por Hipóxia , Proteínas de Neoplasias/antagonistas & inibidores , Oxigênio/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Regiões Promotoras Genéticas , Fatores de Transcrição/metabolismo
18.
Oncotarget ; 7(35): 56958-56975, 2016 08 30.
Artigo em Inglês | MEDLINE | ID: mdl-27486972

RESUMO

Cdc42 is a Rho-GTPase which plays a major role in regulating cell polarity and migration by specifying the localization of filopodia. However, the role of Cdc42 in GBM invasion has not been thoroughly investigated. We generated stable doxycycline-inducible clones expressing wild type (WT)-, constitutively active (CA)-, and dominant negative (DN)-Cdc42 in three different human glioma cell lines. Expression of CA-Cdc42 significantly increased the migration and invasive properties of malignant glioma cells compared to WT and DN-Cdc42 cell clones, and this was accompanied by a greater number of filopodia and focal adhesion structures which co-localize with phosphorylated focal adhesion kinase (FAK). By mass spectrometry and immunoprecipitation studies, we demonstrated that activated Cdc42 binds to IQGAP1. When implanted orthotopically in mice, the CA-Cdc42 expressing glioma cells exhibited enhanced local migration and invasion, and led to larger tumors, which significantly reduced survival. Using the Cancer Genome Atlas dataset, we determined that high Cdc42 expression is associated with poorer progression free survival, and that Cdc42 expression is highest in the proneural and neural subgroups of GBM. In summary, our studies demonstrate that activated Cdc42 is a critical determinant of the migratory and invasive phenotype of malignant gliomas, and that its effect may be mediated, at least in part, through its interaction with IQGAP1 and phosphorylated FAK.


Assuntos
Glioblastoma/metabolismo , Invasividade Neoplásica , Proteína cdc42 de Ligação ao GTP/metabolismo , Animais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Adesão Celular , Linhagem Celular Tumoral , Movimento Celular , Sobrevivência Celular , Progressão da Doença , Intervalo Livre de Doença , Doxiciclina/química , Quinase 1 de Adesão Focal/metabolismo , Regulação Neoplásica da Expressão Gênica , Genes Dominantes , Glioblastoma/patologia , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos , Transplante de Neoplasias , Fenótipo , Fosforilação , Pseudópodes/metabolismo , RNA Interferente Pequeno/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA