Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Breast Cancer Res ; 23(1): 29, 2021 03 04.
Artigo em Inglês | MEDLINE | ID: mdl-33663585

RESUMO

BACKGROUND: MDM2/MDMX proteins are frequently elevated in hormone receptor-positive (ER+) breast cancer. We sought to determine the antitumor efficacy of the combination of ALRN-6924, a dual inhibitor of MDM2/MDMX, with chemotherapy in ER+ breast cancer models. METHODS: Three hundred two cell lines representing multiple tumor types were screened to confirm the role of TP53 status in ALRN-6924 efficacy. ER+ breast cancer cell lines (MCF-7 and ZR-75-1) were used to investigate the antitumor efficacy of ALRN-6924 combination. In vitro cell proliferation, cell cycle, and apoptosis assays were performed. Xenograft tumor volumes were measured, and reverse-phase protein array (RPPA), immunohistochemistry (IHC), and TUNEL assay of tumor tissues were performed to evaluate the in vivo pharmacodynamic effects of ALRN-6924 with paclitaxel. RESULTS: ALRN-6924 was active in wild-type TP53 (WT-TP53) cancer cell lines, but not mutant TP53. On ER+ breast cancer cell lines, it was synergistic in vitro and had enhanced in vivo antitumor activity with both paclitaxel and eribulin. Flow cytometry revealed signs of mitotic crisis in all treatment groups; however, S phase was only decreased in MCF-7 single agent and combinatorial ALRN-6924 arms. RPPA and IHC demonstrated an increase in p21 expression in both combinatorial and single agent ALRN-6924 in vivo treatment groups. Apoptotic assays revealed a significantly enhanced in vivo apoptotic rate in ALRN-6924 combined with paclitaxel treatment arm compared to either single agent. CONCLUSION: The significant synergy observed with ALRN-6924 in combination with chemotherapeutic agents supports further evaluation in patients with hormone receptor-positive breast cancer.


Assuntos
Biomarcadores Tumorais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteína Supressora de Tumor p53/genética , Animais , Antineoplásicos/farmacologia , Apoptose , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Sinergismo Farmacológico , Feminino , Humanos , Camundongos , Mitose , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
J Med Chem ; 66(14): 9401-9417, 2023 07 27.
Artigo em Inglês | MEDLINE | ID: mdl-37439511

RESUMO

We report the discovery of sulanemadlin (ALRN-6924), the first cell-permeating, stabilized α-helical peptide to enter clinical trials. ALRN-6924 is a "stapled peptide" that mimics the N-terminal domain of the p53 tumor suppressor protein. It binds with high affinity to both MDM2 and MDMX (also known as MDM4), the endogenous inhibitors of p53, to activate p53 signaling in cells having a non-mutant, or wild-type TP53 genotype (TP53-WT). Iterative structure-activity optimization endowed ALRN-6924 with favorable cell permeability, solubility, and pharmacokinetic and safety profiles. Intracellular proteolysis of ALRN-6924 forms a long-acting active metabolite with potent MDM2 and MDMX binding affinity and slow dissociation kinetics. At high doses, ALRN-6924 exhibits on-mechanism anticancer activity in TP53-WT tumor models. At lower doses, ALRN-6924 transiently arrests the cell cycle in healthy tissues to protect them from chemotherapy without protecting the TP53-mutant cancer cells. These results support the continued clinical evaluation of ALRN-6924 as an anticancer and chemoprotection agent.


Assuntos
Antineoplásicos , Proteína Supressora de Tumor p53 , Proteína Supressora de Tumor p53/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Ligação Proteica , Peptídeos/química , Antineoplásicos/química , Proteínas de Ciclo Celular/metabolismo
3.
Infect Dis (Lond) ; 52(8): 571-574, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32420777

RESUMO

Introduction: With the emergence of novel coronavirus disease 2019 (COVID-19) in many countries, medical resources currently focus on the treatment of confirmed patients and screening of suspected cases. Asymptomatic patients may be contagious, which makes epidemic control difficult. We describe an asymptomatic patient with a positive real-time polymerase chain reaction (RT-PCR) test in urine.Case report: An asymptomatic girl was identified during the epidemiological investigation of a confirmed COVID-19 patient. When admitted to the hospital on 24 February 2020, she had no clinical manifestations. A throat swab was negative for RT-PCR, but urine was positive. She was given antiviral and symptomatic supportive treatment. On 26 February, a throat swab RT-PCR was positive. RT-PCR in throat swabs and urine were negative on 3 and 5 March, and on 9 and 12 March, throat swabs were still negative. At follow-up on 26 March, she felt well, throat swab RT-PCR was negative, and isolation was lifted.Conclusion: The urine of asymptomatic patients may be contagious. RT-PCR in urine might be a useful supplement in screening when the RT-PCR is negative in throat swabs.


Assuntos
Infecções Assintomáticas , Betacoronavirus/isolamento & purificação , Técnicas de Laboratório Clínico/métodos , Infecções por Coronavirus/urina , Pneumonia Viral/urina , Adolescente , Betacoronavirus/genética , COVID-19 , Teste para COVID-19 , Vacinas contra COVID-19 , Infecções por Coronavirus/diagnóstico , Infecções por Coronavirus/virologia , Feminino , Humanos , Pandemias , Pneumonia Viral/virologia , Reação em Cadeia da Polimerase em Tempo Real/métodos , SARS-CoV-2 , Urina/virologia
4.
J Med Chem ; 62(22): 10098-10107, 2019 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-31657556

RESUMO

Stapled peptides recapitulate the binding affinity and specificity of α-helices in proteins, resist proteolytic degradation, and may provide a novel modality against challenging drug targets such as protein-protein interactions. However, most of the stapled peptides have limited cell permeability or are impermeable to the cell membrane. We show herein that stapled peptides can be rendered highly cell-permeable by conjugating a cyclic cell-penetrating peptide to their N-terminus, C-terminus, or stapling unit. Application of this strategy to two previously reported membrane-impermeable peptidyl inhibitors against the MDM2/p53 and ß-catenin/TCF interactions resulted in the generation of potent proof-of-concept antiproliferative agents against key therapeutic targets.


Assuntos
Peptídeos/química , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , beta Catenina/metabolismo , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Peptídeos Penetradores de Células/química , Humanos , Células MCF-7 , Simulação de Dinâmica Molecular , Peptídeos Cíclicos/química , Estudo de Prova de Conceito , Mapas de Interação de Proteínas/efeitos dos fármacos , Fatores de Transcrição TCF/metabolismo
5.
Nat Commun ; 9(1): 2024, 2018 05 22.
Artigo em Inglês | MEDLINE | ID: mdl-29789628

RESUMO

T- and NK-cell lymphomas (TCL) are a heterogenous group of lymphoid malignancies with poor prognosis. In contrast to B-cell and myeloid malignancies, there are few preclinical models of TCLs, which has hampered the development of effective therapeutics. Here we establish and characterize preclinical models of TCL. We identify multiple vulnerabilities that are targetable with currently available agents (e.g., inhibitors of JAK2 or IKZF1) and demonstrate proof-of-principle for biomarker-driven therapies using patient-derived xenografts (PDXs). We show that MDM2 and MDMX are targetable vulnerabilities within TP53-wild-type TCLs. ALRN-6924, a stapled peptide that blocks interactions between p53 and both MDM2 and MDMX has potent in vitro activity and superior in vivo activity across 8 different PDX models compared to the standard-of-care agent romidepsin. ALRN-6924 induced a complete remission in a patient with TP53-wild-type angioimmunoblastic T-cell lymphoma, demonstrating the potential for rapid translation of discoveries from subtype-specific preclinical models.


Assuntos
Antineoplásicos/farmacologia , Regulação Neoplásica da Expressão Gênica , Linfoma Extranodal de Células T-NK/tratamento farmacológico , Linfoma de Células T/tratamento farmacológico , Proteínas Nucleares/genética , Peptídeos/farmacologia , Proteínas Proto-Oncogênicas c-mdm2/genética , Proteínas Proto-Oncogênicas/genética , Proteína Supressora de Tumor p53/genética , Animais , Proteínas de Ciclo Celular , Depsipeptídeos/farmacologia , Avaliação Pré-Clínica de Medicamentos , Humanos , Fator de Transcrição Ikaros/antagonistas & inibidores , Fator de Transcrição Ikaros/genética , Fator de Transcrição Ikaros/metabolismo , Imidazolinas/farmacologia , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/genética , Janus Quinase 2/metabolismo , Linfoma Extranodal de Células T-NK/genética , Linfoma Extranodal de Células T-NK/metabolismo , Linfoma Extranodal de Células T-NK/patologia , Linfoma de Células T/genética , Linfoma de Células T/metabolismo , Linfoma de Células T/patologia , Camundongos , Proteínas Nucleares/antagonistas & inibidores , Proteínas Nucleares/metabolismo , Ligação Proteica/efeitos dos fármacos , Proteínas Proto-Oncogênicas/antagonistas & inibidores , Proteínas Proto-Oncogênicas/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Indução de Remissão , Transdução de Sinais , Proteína Supressora de Tumor p53/antagonistas & inibidores , Proteína Supressora de Tumor p53/metabolismo , Sequenciamento do Exoma , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Sci Rep ; 7(1): 4537, 2017 07 03.
Artigo em Inglês | MEDLINE | ID: mdl-28674429

RESUMO

In this study we have tested the efficacy of citrate therapy in various cancer models. We found that citrate administration inhibited A549 lung cancer growth and additional benefit accrued in combination with cisplatin. Interestingly, citrate regressed Ras-driven lung tumors. Further studies indicated that citrate induced tumor cell differentiation. Additionally, citrate treated tumor samples showed significantly higher infiltrating T-cells and increased blood levels of numerous cytokines. Moreover, we found that citrate inhibited IGF-1R phosphorylation. In vitro studies suggested that citrate treatment inhibited AKT phosphorylation, activated PTEN and increased expression of p-eIF2a. We also found that p-eIF2a was decreased when PTEN was depleted. These data suggest that citrate acts on the IGF-1R-AKT-PTEN-eIF2a pathway. Additionally, metabolic profiling suggested that both glycolysis and the tricarboxylic acid cycle were suppressed in a similar manner in vitro in tumor cells and in vivo but only in tumor tissue. We reproduced many of these observations in an inducible Her2/Neu-driven breast cancer model and in syngeneic pancreatic tumor (Pan02) xenografts. Our data suggests that citrate can inhibit tumor growth in diverse tumor types and via multiple mechanisms. Dietary supplementation with citrate may be beneficial as a cancer therapy.


Assuntos
Ciclo do Ácido Cítrico , Ácido Cítrico/metabolismo , Modelos Biológicos , Receptor IGF Tipo 1/metabolismo , Transdução de Sinais , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Ácido Cítrico/farmacologia , Ciclo do Ácido Cítrico/efeitos dos fármacos , Citocinas/biossíntese , Modelos Animais de Doenças , Feminino , Glicólise/efeitos dos fármacos , Humanos , Mediadores da Inflamação/metabolismo , Leucócitos/metabolismo , Leucócitos/patologia , Camundongos , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto , Proteínas ras/genética , Proteínas ras/metabolismo
7.
Wei Sheng Wu Xue Bao ; 46(2): 292-6, 2006 Apr.
Artigo em Zh | MEDLINE | ID: mdl-16736594

RESUMO

Citrus bacterial canker is an important disease of Citrus species in China. The disease severely occurs especially in the coastal area. Integrated control system has been used for the control of the disease, in which chemotherapy plays an important role at present. The chemotherapy-dominant control system brought many problems to the environment, such as chemical residua in the products and induction of resistance to fungicide(s) by the pathogen. To solve these problems, an intensive study on biocontrol of citrus bacterial canker is needed. Isolations and characterizations of biocontrol agents are the basis for biocontrol of the disease. A bacterial strain Bt8 with strong inhibiting ability against Xanthomonas axonopodis pv. citri (Hasse) Vauterin, was isolated from citrus orchard soil in Nanning, China. The isolated bacterial strain was identified and characterized as Acinetobacter baumannii Bouvet et Grimont on the base of its morphology and 16S rDNA sequence analysis as well as physiological and biochemical characters. The inhibiting activity of the bacterium suspension against the pathogen was significantly influenced by environmental factors, such as temperatures, pHs and media. At temperatures of 18 degrees C to 33 degrees C, both the inhibiting activity of the bacterium suspension and the biomass of the bacterium increased with the increases of temperatures, suggesting that the influence of temperature on inhibiting activity of the bacterium suspension was in dependence on the bacterial biomass. In NA liquid medium of pH 10, the bacterium suspension showed the highest inhibiting activity against Xanthomonas axonopodis pv. citri, which was not in dependence on biomass of the bacterium. The bacterium suspension provided 55.2% inhibition against bacterial canker under greenhouse conditions. The results showed that Acinetobacter baumannii has potential as biocontrol agent against bacterial canker disease. Acinetobacter baumannii was reported as the pathogens infecting human and animals. The present study enriched the understanding on biological diversity in Acinetobacter baumannii to sciences. This is the first report on the isolation of Acinetobacter baumannii with strong inhibiting ability against plant pathogen.


Assuntos
Acinetobacter baumannii/isolamento & purificação , Acinetobacter baumannii/fisiologia , Antibiose , Citrus/microbiologia , Doenças das Plantas/microbiologia , Xanthomonas axonopodis/fisiologia , Acinetobacter baumannii/classificação , Acinetobacter baumannii/genética , Filogenia , Temperatura , Xanthomonas axonopodis/patogenicidade
8.
Med Hypotheses ; 64(1): 74-8, 2005.
Artigo em Inglês | MEDLINE | ID: mdl-15533615

RESUMO

Pigment epithelium-derived factor (PEDF) is a multiple functional protein, coded by the serine proteinase inhibitor, clade F, member 1 (SERPINF1) gene, which has both anti-angiogenic activity and neurotrophic activity at the same time. Its antiangiogenic activity in the mammalian eye is the most potent known at this time. However, the mechanism(s) by which PEDF works in vivo is still uncertain. Some observations suggest that PEDF can simultaneously inhibit the migration and proliferation induced by vascular endothelial growth factor (VEGF), and then further inhibits angiogenesis by interacting with specific cell surface receptors, but no such receptor has been reported to date. Here we propose a hypothesis that PEDF exerts its function by binding with intergrins. Intergrin can therefore serve as the receptor of PEDF.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Integrinas/metabolismo , Modelos Cardiovasculares , Neovascularização Fisiológica/efeitos dos fármacos , Neovascularização Fisiológica/fisiologia , Transdução de Sinais/fisiologia , Fator A de Crescimento do Endotélio Vascular/metabolismo , Animais , Movimento Celular/efeitos dos fármacos , Movimento Celular/fisiologia , Proliferação de Células/efeitos dos fármacos , Humanos , Transdução de Sinais/efeitos dos fármacos
9.
Nanoscale ; 6(6): 3353-60, 2014 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-24522297

RESUMO

Toward the increasing demands of portable energy storage and electric vehicle applications, silicon has been emerging as a promising anode material for lithium-ion batteries (LIBs) owing to its high specific capacity. However, serious pulverization of bulk silicon during cycling limits its cycle life. Herein, we report a novel hierarchical Si nanowire (Si NW)-reduced graphene oxide (rGO) composite fabricated using a solvothermal method followed by a chemical vapor deposition process. In the composite, the uniform-sized [111]-oriented Si NWs are well dispersed on the rGO surface and in between rGO sheets. The flexible rGO enables us to maintain the structural integrity and to provide a continuous conductive network of the electrode, which results in over 100 cycles serving as an anode in half cells at a high lithium storage capacity of 2300 mA h g(-1). Due to its [111] growth direction and the large contact area with rGO, the Si NWs in the composite show substantially enhanced reaction kinetics compared with other Si NWs or Si particles.


Assuntos
Fontes de Energia Elétrica , Grafite/química , Lítio/química , Nanofios/química , Silício/química , Técnicas Eletroquímicas , Eletrodos , Íons/química , Óxidos/química
10.
Sci Rep ; 4: 5414, 2014 Jun 24.
Artigo em Inglês | MEDLINE | ID: mdl-24957098

RESUMO

Mitochondrial malic enzyme 2 (ME2) catalyzes the oxidative decarboxylation of malate to yield CO2 and pyruvate, with concomitant reduction of dinucleotide cofactor NAD(+) or NADP(+). We find that ME2 is highly expressed in many solid tumors. In the A549 non-small cell lung cancer (NSCLC) cell line, ME2 depletion inhibits cell proliferation and induces cell death and differentiation, accompanied by increased reactive oxygen species (ROS) and NADP(+)/NADPH ratio, a drop in ATP, and increased sensitivity to cisplatin. ME2 knockdown impacts phosphoinositide-dependent protein kinase 1 (PDK1) and phosphatase and tensin homolog (PTEN) expression, leading to AKT inhibition. Depletion of ME2 leads to malate accumulation and pyruvate decrease, and exogenous cell permeable dimethyl-malate (DMM) mimics the ME2 knockdown phenotype. Both ME2 knockdown and DMM treatment reduce A549 cell growth in vivo. Collectively, our data suggest that ME2 is a potential target for cancer therapy.


Assuntos
Neoplasias Pulmonares/enzimologia , Malato Desidrogenase/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Western Blotting , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Criança , Pré-Escolar , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Malato Desidrogenase/genética , Malatos/química , Malatos/farmacologia , Masculino , Camundongos Nus , Pessoa de Meia-Idade , Interferência de RNA , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Ensaios Antitumorais Modelo de Xenoenxerto/métodos , Adulto Jovem
11.
Cell Metab ; 19(5): 795-809, 2014 May 06.
Artigo em Inglês | MEDLINE | ID: mdl-24726384

RESUMO

The lactate dehydrogenase-A (LDH-A) enzyme catalyzes the interconversion of pyruvate and lactate, is upregulated in human cancers, and is associated with aggressive tumor outcomes. Here we use an inducible murine model and demonstrate that inactivation of LDH-A in mouse models of NSCLC driven by oncogenic K-RAS or EGFR leads to decreased tumorigenesis and disease regression in established tumors. We also show that abrogation of LDH-A results in reprogramming of pyruvate metabolism, with decreased lactic fermentation in vitro, in vivo, and ex vivo. This was accompanied by reactivation of mitochondrial function in vitro, but not in vivo or ex vivo. Finally, using a specific small molecule LDH-A inhibitor, we demonstrated that LDH-A is essential for cancer-initiating cell survival and proliferation. Thus, LDH-A can be a viable therapeutic target for NSCLC, including cancer stem cell-dependent drug-resistant tumors.


Assuntos
Carcinogênese/metabolismo , Transformação Celular Neoplásica/metabolismo , L-Lactato Desidrogenase/metabolismo , Neoplasias Pulmonares/metabolismo , Animais , Carcinoma Pulmonar de Células não Pequenas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Sobrevivência Celular/fisiologia , Progressão da Doença , Receptores ErbB/metabolismo , Células Hep G2 , Humanos , Isoenzimas/metabolismo , Lactato Desidrogenase 5 , Camundongos , Mitocôndrias/metabolismo , Proteína Oncogênica p21(ras)/metabolismo , Ácido Pirúvico/metabolismo
12.
Nanoscale ; 5(18): 8689-94, 2013 Sep 21.
Artigo em Inglês | MEDLINE | ID: mdl-23900559

RESUMO

Anode materials play a key role in the performance, in particular the capacity and lifetime, of lithium ion batteries (LIBs). Silicon has been demonstrated to be a promising anode material due to its high specific capacity, but pulverization during cycling and formation of an unstable solid-electrolyte interphase limit its cycle life. Herein, we show that anodes consisting of an active silicon nanowire (Si NW), which is surrounded by a uniform graphene shell and comprises silicon carbide nanocrystals, are capable of serving over 500 cycles in half cells at a high lithium storage capacity of 1650 mA h g(-1). In the anodes, the graphene shell provides a highly-conductive path and prevents direct exposure of Si NWs to electrolytes while the SiC nanocrystals may act as a rigid backbone to retain the integrity of the Si NW in its great deformation process caused by repetitive charging-discharging reactions, resulting in a stable cyclability.


Assuntos
Fontes de Energia Elétrica , Grafite/química , Lítio/química , Nanofios/química , Silício/química , Compostos Inorgânicos de Carbono/química , Eletrodos , Eletrólitos/química , Íons/química , Compostos de Silício/química
13.
Asian Pac J Cancer Prev ; 14(11): 6433-8, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24377546

RESUMO

In hepatocellular cancer (HCC), lack of response to chemotherapy and radiation treatment can be caused by a loss of epigenetic modifications of cancer cells. Methionine adenosyltransferase 1A is inactivated in HCC and may be stimulated by an epigenetic change involving promoter hypermethylation. Therefore, drugs releasing epigenetic repression have been proposed to reverse this process. We studied the effect of the demethylating reagent 5-aza-2<-deoxycitidine (5-Aza-CdR) on MAT1A gene expression, DNA methylation and S-adenosylmethionine (SAMe) production in the HCC cell line Huh7. We found that MAT1A mRNA and protein expression were activated in Huh7 cells with the treatment of 5-Aza-CdR; the status of promoter hypermethylation was reversed. At the same time, MAT2A mRNA and protein expression was significantly reduced in Huh7 cells treated with 5-Aza-CdR, while SAMe production was significantly induced. However, 5-Aza-CdR showed no effects on MAT2A methylation. Furthermore, 5-Aza-CdR inhibited the growth of Huh7 cells and induced apoptosis and through down-regulation of Bcl-2, up-regulation of Bax and caspase-3. Our observations suggest that 5-Aza- CdR exerts its anti-tumor effects in Huh7 cells through an epigenetic change involving increased expression of the methionine adenosyltransferase 1A gene and induction of S-adenosylmethionine production.


Assuntos
Apoptose/efeitos dos fármacos , Azacitidina/análogos & derivados , Carcinoma Hepatocelular/tratamento farmacológico , Neoplasias Hepáticas/tratamento farmacológico , Metionina Adenosiltransferase/biossíntese , S-Adenosilmetionina/biossíntese , Azacitidina/farmacologia , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/metabolismo , Carcinoma Hepatocelular/patologia , Caspase 3/genética , Linhagem Celular Tumoral , Metilação de DNA/efeitos dos fármacos , Decitabina , Regulação para Baixo/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Neoplasias Hepáticas/patologia , Metionina Adenosiltransferase/genética , Proteínas Proto-Oncogênicas c-bcl-2/genética , S-Adenosilmetionina/genética , Regulação para Cima/efeitos dos fármacos , Proteína X Associada a bcl-2/genética
14.
PLoS One ; 5(9)2010 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-20824065

RESUMO

Malic enzyme 2 (ME2) is a mitochondrial enzyme that catalyzes the conversion of malate to pyruvate and CO2 and uses NAD as a cofactor. Higher expression of this enzyme correlates with the degree of cell de-differentiation. We found that ME2 is expressed in K562 erythroleukemia cells, in which a number of agents have been found to induce differentiation either along the erythroid or the myeloid lineage. We found that knockdown of ME2 led to diminished proliferation of tumor cells and increased apoptosis in vitro. These findings were accompanied by differentiation of K562 cells along the erythroid lineage, as confirmed by staining for glycophorin A and hemoglobin production. ME2 knockdown also totally abolished growth of K562 cells in nude mice. Increased ROS levels, likely reflecting increased mitochondrial production, and a decreased NADPH/NADP+ ratio were noted but use of a free radical scavenger to decrease inhibition of ROS levels did not reverse the differentiation or apoptotic phenotype, suggesting that ROS production is not causally involved in the resultant phenotype. As might be expected, depletion of ME2 induced an increase in the NAD+/NADH ratio and ATP levels fell significantly. Inhibition of the malate-aspartate shuttle was insufficient to induce K562 differentiation. We also examined several intracellular signaling pathways and expression of transcription factors and intermediate filament proteins whose expression is known to be modulated during erythroid differentiation in K562 cells. We found that silencing of ME2 leads to phospho-ERK1/2 inhibition, phospho-AKT activation, increased GATA-1 expression and diminished vimentin expression. Metabolomic analysis, conducted to gain insight into intermediary metabolic pathways that ME2 knockdown might affect, showed that ME2 depletion resulted in high orotate levels, suggesting potential impairment of pyrimidine metabolism. Collectively our data point to ME2 as a potentially novel metabolic target for leukemia therapy.


Assuntos
Diferenciação Celular , Células Eritroides/citologia , Células Eritroides/enzimologia , Leucemia Eritroblástica Aguda/enzimologia , Leucemia Eritroblástica Aguda/fisiopatologia , Malato Desidrogenase/deficiência , Animais , Apoptose , Proliferação de Células , Técnicas de Inativação de Genes , Humanos , Células K562 , Leucemia Eritroblástica Aguda/genética , Malato Desidrogenase/genética , Camundongos
15.
J Biol Chem ; 283(34): 22972-82, 2008 Aug 22.
Artigo em Inglês | MEDLINE | ID: mdl-18567582

RESUMO

Ca(2+) and calmodulin modulate numerous cellular functions, ranging from muscle contraction to the cell cycle. Accumulating evidence indicates that Ca(2+) and calmodulin regulate the MAPK signaling pathway at multiple positions in the cascade, but the molecular mechanism underlying these observations is poorly defined. We previously documented that IQGAP1 is a scaffold in the MAPK cascade. IQGAP1 binds to and regulates the activities of ERK, MEK, and B-Raf. Here we demonstrate that IQGAP1 integrates Ca(2+) and calmodulin with B-Raf signaling. In vitro analysis reveals that Ca(2+) promotes the direct binding of IQGAP1 to B-Raf. This interaction is inhibited by calmodulin in a Ca(2+)-regulated manner. Epidermal growth factor (EGF) is unable to stimulate B-Raf activity in fibroblasts treated with the Ca(2+) ionophore A23187. In contrast, chelation of intracellular free Ca(2+) concentrations ([Ca(2+)](i)) significantly enhances EGF-stimulated B-Raf activity, an effect that is dependent on IQGAP1. Incubation of cells with EGF augments the association of B-Raf with IQGAP1. Moreover, Ca(2+) regulates the association of B-Raf with IQGAP1 in cells. Increasing [Ca(2+)](i) with Ca(2+) ionophores significantly reduces co-immunoprecipitation of B-Raf and IQGAP1, whereas chelation of Ca(2+) enhances the interaction. Consistent with these findings, increasing and decreasing [Ca(2+)](i) increase and decrease, respectively, co-immunoprecipitation of calmodulin with IQGAP1. Collectively, our data identify a previously unrecognized mechanism in which the scaffold protein IQGAP1 couples Ca(2+) and calmodulin signaling to B-Raf function.


Assuntos
Cálcio/química , Calmodulina/metabolismo , Regulação da Expressão Gênica , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Ativadoras de ras GTPase/fisiologia , Animais , Calcimicina/farmacologia , Cálcio/metabolismo , Fator de Crescimento Epidérmico/metabolismo , Fibroblastos/metabolismo , Humanos , Ionóforos/farmacologia , Camundongos , Modelos Biológicos , Transdução de Sinais , Proteínas Ativadoras de ras GTPase/metabolismo
16.
Proc Natl Acad Sci U S A ; 104(25): 10465-9, 2007 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-17563371

RESUMO

IQGAP1 modulates several cellular functions, including cell-cell adhesion, transcription, cytoskeletal architecture, and selected signaling pathways. We previously documented that IQGAP1 binds ERK and MAPK kinase (MEK) and regulates EGF-stimulated MEK and ERK activity. Here we characterize the interaction between IQGAP1 and B-Raf, the molecule immediately upstream of MEK in the Ras/MAPK signaling cascade. B-Raf binds directly to IQGAP1 in vitro and coimmunoprecipitates with IQGAP1 from cell lysates. Importantly, IQGAP1 modulates B-Raf function. EGF is unable to stimulate B-Raf activity in IQGAP1-null cells and in cells transfected with an IQGAP1 mutant construct that is unable to bind B-Raf. Interestingly, binding to IQGAP1 significantly enhances B-Raf activity in vitro. Our data identify a previously unrecognized interaction between IQGAP1 and B-Raf and suggest that IQGAP1 is a scaffold necessary for activation of B-Raf by EGF.


Assuntos
Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo , Animais , Linhagem Celular Transformada , Células Cultivadas , Meios de Cultura Livres de Soro , Embrião de Mamíferos , Ativação Enzimática , Fator de Crescimento Epidérmico/farmacologia , Fibroblastos/citologia , Fibroblastos/metabolismo , Glutationa Transferase/metabolismo , Humanos , Camundongos , Camundongos Knockout , Mutação , Plasmídeos/metabolismo , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas B-raf/análise , Proteínas Recombinantes de Fusão/metabolismo , Fatores de Tempo , Transfecção , Proteínas Ativadoras de ras GTPase/química , Proteínas Ativadoras de ras GTPase/genética
17.
J Biol Chem ; 280(41): 34548-57, 2005 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-16105843

RESUMO

The scaffolding protein IQGAP1 participates in numerous cellular functions by binding to target proteins such as actin, calmodulin, E-cadherin, beta-catenin, Cdc42, Rac1, and CLIP-170. IQGAP1 regulates the cytoskeleton, promotes cell motility, and modulates E-cadherin-mediated cell-cell adhesion. However, how IQGAP1 exerts its functions in vivo is still unclear. In this study we investigate the self-association of IQGAP1 and its role in IQGAP1 function. Endogenous IQGAP1 co-immunoprecipitated from MCF-7 cells with IQGAP1 tagged with enhanced green fluorescent protein, indicating that IQGAP1 self-associates in cells. In vitro assays confirmed that IQGAP1 can self-associate and that this effect is mediated by the N-terminal half of the protein. Gel filtration analysis suggested that full-length IQGAP1 exists as a combination of monomers, dimers, and larger oligomers. Analysis performed with multiple fragments of IQGAP1 narrowed the self-association region to amino acids 763-863. In support of this observation, a peptide comprising residues 763-863 disrupted self-association of full-length IQGAP1 in a dose-dependent manner. Similarly, deleting this sequence from IQGAP1 abolished binding to full-length IQGAP1. In addition, the ability of IQGAP1 to increase the amount of active Cdc42 in cells is abrogated upon removal of this region. Consistent with these findings, transfection into cells of a peptide containing the self-association domain significantly reduced the amount of active Cdc42 in cell lysates. These observations define a sequence of IQGAP1 that is necessary for its oligomerization and demonstrate that self-association is required for the normal cellular function of IQGAP1.


Assuntos
Proteínas Ativadoras de ras GTPase/química , Sítios de Ligação , Linhagem Celular , Linhagem Celular Tumoral , Cromatografia em Gel , Relação Dose-Resposta a Droga , Deleção de Genes , Glutationa Transferase/metabolismo , Proteínas de Fluorescência Verde/metabolismo , Humanos , Imunoprecipitação , Peptídeos/química , Plasmídeos/metabolismo , Ligação Proteica , Estrutura Terciária de Proteína , Proteínas Recombinantes de Fusão/metabolismo , Transfecção , Proteína cdc42 de Ligação ao GTP/metabolismo , Proteínas Ativadoras de ras GTPase/metabolismo
18.
Proc Natl Acad Sci U S A ; 101(17): 6605-10, 2004 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-15096582

RESUMO

Vascular permeability plays a key role in a wide array of life-threatening and sight-threatening diseases. Vascular endothelial growth factor can increase vascular permeability. Using a model system for nonproliferative diabetic retinopathy, we found that pigment epithelium-derived factor (PEDF) effectively abated vascular endothelial growth factor-induced vascular permeability. A 44-amino acid region of PEDF was sufficient to confer the antivasopermeability activity. Additionally, we identified four amino acids (glutamate-101, isoleucine-103, leucine-112, and serine-115) critical for this activity. PEDF, or a derivative, could potentially abate or restore vision loss from diabetic macular edema. Furthermore, PEDF may represent a superior therapeutic approach to sepsis-associated hypotension, nephrotic syndrome, and other sight-threatening and life-threatening diseases resulting from excessive vascular permeability.


Assuntos
Permeabilidade Capilar/fisiologia , Proteínas do Olho , Fatores de Crescimento Neural , Proteínas/fisiologia , Serpinas/fisiologia , Sequência de Aminoácidos , Animais , Sítios de Ligação , Linhagem Celular , Angiofluoresceinografia , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Dados de Sequência Molecular , Proteínas/química , Proteínas/metabolismo , Proteínas Recombinantes/metabolismo , Vasos Retinianos/fisiologia , Homologia de Sequência de Aminoácidos , Serpinas/química , Serpinas/metabolismo , Fator A de Crescimento do Endotélio Vascular/fisiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA