Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
1.
Biochim Biophys Acta Mol Cell Res ; 1865(7): 959-969, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29655803

RESUMO

In acute myeloid leukemia (AML), high Galectin 3 (LGALS3) expression is associated with poor prognosis. The role of LGALS3 derived from mesenchymal stromal cells (MSC) in the AML microenvironment is unclear; however, we have recently found high LGALS3 expression in MSC derived from AML patients is associated with relapse. In this study, we used reverse phase protein analysis (RPPA) to correlate LGALS3 expression in AML MSC with 119 other proteins including variants of these proteins such as phosphorylated forms or cleaved forms to identify biologically relevant pathways. RPPA revealed that LGALS3 protein was positively correlated with expression of thirteen proteins including MYC, phosphorylated beta-Catenin (p-CTNNB1), and AKT2 and negatively correlated with expression of six proteins including integrin beta 3 (ITGB3). String analysis revealed that proteins positively correlated with LGALS3 showed strong interconnectivity. Consistent with the RPPA results, LGALS3 suppression by shRNA in MSC resulted in decreased MYC and AKT expression while ITGB3 was induced. In co-culture, the ability of AML cell to adhere to MSC LGALS3 shRNA transductants was reduced compared to AML cell adhesion to MSC control shRNA transductants. Finally, use of novel specific LGALS3 inhibitor CBP.001 in co-culture of AML cells with MSC reduced viable leukemia cell populations with induced apoptosis and augmented the chemotherapeutic effect of AraC. In summary, the current study demonstrates that MSC-derived LGALS3 may be critical for important biological pathways for MSC homeostasis and for regulating AML cell localization and survival in the leukemia microenvironmental niche.


Assuntos
Galectina 3/metabolismo , Leucemia Mieloide Aguda/metabolismo , Células-Tronco Mesenquimais/metabolismo , Regulação para Cima , Proteínas Sanguíneas , Técnicas de Cocultura , Galectinas , Regulação Neoplásica da Expressão Gênica , Humanos , Células-Tronco Mesenquimais/citologia , Fosforilação , Mapas de Interação de Proteínas , Proteômica , Células THP-1 , Células Tumorais Cultivadas , Microambiente Tumoral
2.
Blood ; 128(9): 1260-9, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27268264

RESUMO

Autophagy is a cellular adaptive mechanism to stress, including that induced by chemotherapeutic agents. Reverse phase protein array suggested that high expression of the essential autophagy-related protein, Atg7, was associated with shorter remission in newly diagnosed acute myeloid leukemia (AML) patient samples, indicating a role in chemoresistance. Knockdown of Atg7 in AML cells using short hairpin RNA markedly increased apoptosis and DNA damage following treatment with cytarabine and idarubicin. Interestingly, coculture of AML cells with stromal cells increased autophagy and chemoresistance in the AML cells exposed to chemotherapeutic agents, and this was reversed following Atg7 knockdown. This effect was further enhanced by concomitant knockdown of Atg7 in both AML and stromal cells. These findings strongly suggest that Atg7, and likely microenvironment autophagy in general, plays an important role in AML chemoresistance. Mechanistic studies revealed that Atg7 knockdown induced a proapoptotic phenotype in AML cells, which was manifested by an increased NOXA expression at the transcriptional level. Finally, in a mouse model of human leukemia, Atg7 knockdown extended overall survival after chemotherapy. Thus, the inhibition of Atg7 appears to be a valid strategy to enhance chemosensitivity, and it could indeed improve outcomes in AML therapy.


Assuntos
Proteína 7 Relacionada à Autofagia , Autofagia , Resistencia a Medicamentos Antineoplásicos , Leucemia Mieloide Aguda , Microambiente Tumoral , Animais , Autofagia/efeitos dos fármacos , Autofagia/genética , Proteína 7 Relacionada à Autofagia/genética , Proteína 7 Relacionada à Autofagia/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Células HL-60 , Humanos , Células K562 , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Camundongos , Camundongos Endogâmicos NOD , Camundongos SCID , Proteínas Proto-Oncogênicas c-bcl-2/biossíntese , Proteínas Proto-Oncogênicas c-bcl-2/genética , Células Estromais/metabolismo , Células Estromais/patologia , Microambiente Tumoral/efeitos dos fármacos , Microambiente Tumoral/genética , Ensaios Antitumorais Modelo de Xenoenxerto
3.
Haematologica ; 103(5): 810-821, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29545342

RESUMO

Mesenchymal stromal cells (MSC) support acute myeloid leukemia (AML) cell survival in the bone marrow (BM) microenvironment. Protein expression profiles of AML-derived MSC are unknown. Reverse phase protein array analysis was performed to compare expression of 151 proteins from AML-MSC (n=106) with MSC from healthy donors (n=71). Protein expression differed significantly between the two groups with 19 proteins over-expressed in leukemia stromal cells and 9 over-expressed in normal stromal cells. Unbiased hierarchical clustering analysis of the samples using these 28 proteins revealed three protein constellations whose variation in expression defined four MSC protein expression signatures: Class 1, Class 2, Class 3, and Class 4. These cell populations appear to have clinical relevance. Specifically, patients with Class 3 cells have longer survival and remission duration compared to other groups. Comparison of leukemia MSC at first diagnosis with those obtained at salvage (i.e. relapse/refractory) showed differential expression of 9 proteins reflecting a shift toward osteogenic differentiation. Leukemia MSC are more senescent compared to their normal counterparts, possibly due to the overexpressed p53/p21 axis as confirmed by high ß-galactosidase staining. In addition, overexpression of BCL-XL in leukemia MSC might give survival advantage under conditions of senescence or stress and overexpressed galectin-3 exerts profound immunosuppression. Together, our findings suggest that the identification of specific populations of MSC in AML patients may be an important determinant of therapeutic response.


Assuntos
Biomarcadores Tumorais/metabolismo , Leucemia Mieloide Aguda/mortalidade , Células-Tronco Mesenquimais/metabolismo , Recidiva Local de Neoplasia/mortalidade , Análise Serial de Proteínas , Adulto , Estudos de Casos e Controles , Diferenciação Celular , Proliferação de Células , Feminino , Humanos , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Masculino , Células-Tronco Mesenquimais/patologia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/metabolismo , Recidiva Local de Neoplasia/patologia , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas
4.
Biochim Biophys Acta ; 1863(4): 562-71, 2016 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-26704388

RESUMO

Galectin 3 (LGALS3) expression is prognostic for poor survival in acute myeloid leukemia (AML) patients. GCS-100 is a novel galectin inhibitor that may prove useful for AML therapy. In this study, we found that GCS-100 induced apoptosis in AML cells. The agent reduced MCL-1 expression suggesting that GCS-100 could be more effective when combined with a BH3 mimetic. Indeed, potent synergistic cytotoxicity was achieved when GCS-100 was combined with ABT-737 or ABT-199. Furthermore, the GCS-100/ABT-199 combination was effective against primary AML blast cells from patients with FLT3 ITD mutations, which is another prognostic factor for poor outcome in AML. This activity may involve wild-type p53 as shRNA knockdown of LGALS3 or galectin 1 (LGALS1) sensitized wild-type p53 OCI-AML3 cells to GCS-100/ABT-737-induced apoptosis to a much greater extent than p53 null THP-1 cells. Suppression of LGALS3 by shRNA inhibited MCL-1 expression in OCI-AML3 cells, but not THP-1 cells, suggesting the induced sensitivity to ABT-737 may involve a MCL-1 mediated mechanism. OCI-AML3 cells with LGALS1 shRNA were also sensitized to ABT-737. However, these cells exhibited increased MCL-1 expression, so MCL-1 reduction is apparently not required in this process. A role for p53 appears important as GCS-100 induces p53 expression and shRNA knockdown of p53 protected OCI-AML3 cells from the cytotoxic effects of the GCS-100/ABT-737 treatment combination. Our results suggest that galectins regulate a survival axis in AML cells, which may be targeted via combined inhibition with drugs such as GCS-100 and ABT-199.


Assuntos
Compostos de Bifenilo/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Leucemia Mieloide Aguda/patologia , Nitrofenóis/farmacologia , Polissacarídeos/farmacologia , Sulfonamidas/farmacologia , Proteína Supressora de Tumor p53/genética , Idoso , Idoso de 80 Anos ou mais , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Compostos de Bifenilo/administração & dosagem , Compostos Bicíclicos Heterocíclicos com Pontes/administração & dosagem , Linhagem Celular Tumoral , Sinergismo Farmacológico , Feminino , Galectinas/antagonistas & inibidores , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Masculino , Pessoa de Meia-Idade , Nitrofenóis/administração & dosagem , Fragmentos de Peptídeos/química , Piperazinas/administração & dosagem , Piperazinas/farmacologia , Polissacarídeos/administração & dosagem , Proteínas Proto-Oncogênicas/química , Sulfonamidas/administração & dosagem
5.
Biochim Biophys Acta ; 1843(9): 1969-77, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24858343

RESUMO

We recently discovered that the protein phosphatase 2A (PP2A) B55α subunit (PPP2R2A) is under-expressed in primary blast cells and is unfavorable for remission duration in AML patients. In this study, reverse phase protein analysis (RPPA) of 230 proteins in 511 AML patient samples revealed a strong correlation of B55α with a number of proteins including MYC, PKC α, and SRC. B55α suppression in OCI-AML3 cells by shRNA demonstrated that the B subunit is a PKCα phosphatase. B55α does not target SRC, but rather the kinase suppresses protein expression of the B subunit. Finally, the correlation between B55α and MYC levels reflected a complex stoichiometric competition between B subunits. Loss of B55α in OCI-AML3 cells did not change global PP2A activity and the only isoform that is induced is the one containing B56α. In cells containing B55α shRNA, MYC was suppressed with concomitant induction of the competing B subunit B56α (PPP2R5A). A recent study determined that FTY-720, a drug whose action involves the activation of PP2A, resulted in the induction of B55α In AML cells, and a reduction of the B subunit rendered these cells resistant to FTY-720. Finally, reduction of the B subunit resulted in an increase in the expression of miR-191-5p and a suppression of miR-142-3p. B55α regulation of these miRs was intriguing as high levels of miR-191 portend poor survival in AML, and miR-142-3p is mutated in 2% of AML patient samples. In summary, the suppression of B55α activates signaling pathways that could support leukemia cell survival.


Assuntos
Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/genética , MicroRNAs/genética , Proteína Fosfatase 2/metabolismo , Transdução de Sinais/genética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Resistencia a Medicamentos Antineoplásicos/genética , Cloridrato de Fingolimode , Regulação Leucêmica da Expressão Gênica/efeitos dos fármacos , Humanos , MicroRNAs/metabolismo , Modelos Biológicos , Fosforilação/efeitos dos fármacos , Propilenoglicóis/farmacologia , Proteína Quinase C-alfa/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , RNA Interferente Pequeno/metabolismo , Transdução de Sinais/efeitos dos fármacos , Esfingosina/análogos & derivados , Esfingosina/farmacologia , Quinases da Família src/metabolismo
6.
Apoptosis ; 17(3): 258-68, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22012334

RESUMO

We have demonstrated previously that the dihydroorotate dehydrogenase (DHODH) inhibitor teriflunomide (TFN) encourages apoptosis in transformed human keratinocytes. Here we sought to determine if this cytotoxic effect could be restricted to transformed keratinocytes relative to their normal human epidermal keratinocyte (NHEK) counterparts, and ascertain a potential mechanistic basis for the selectivity. The NHEK cells proliferated much slower than the premalignant HaCaT and malignant COLO 16 keratinocytes, and exogenous uridine added to the culture medium did not affect this growth. Similarly, DHODH expression and the bioenergetic characteristics of the normal cells were markedly dissimilar from those observed in the transformed cells indicating that de novo pyrimidine synthesis was involved with keratinocyte proliferation. Moreover, a short-term exposure to TFN caused a wild-type p53 response in the NHEK cells illustrating that pyrimidine metabolic stress could regulate this tumor suppressor protein in the normal cells. TFN-induced apoptosis occurred primarily in S phase HaCaT cells. This cell death was sensitive to uridine, an antioxidant, and a caspase inhibitor, and the suppression of Bcl-X(L) and the induction of Mn superoxide dismutase preceded it. These events suggested that mitochondrial/redox stress was involved with the cytotoxic effect of TFN. Conversely, a long-term exposure to TFN caused G(0)/G(1) arrest in the NHEK cells, which supported a cytoprotective role for p53 against TFN-induced apoptosis. Together, these results propose that TFN could be useful in the prevention or therapy of non-melanoma skin cancers and possibly other hyperproliferative keratinocytic diseases.


Assuntos
Antineoplásicos/farmacologia , Crotonatos/toxicidade , Metabolismo Energético/fisiologia , Queratinócitos/efeitos dos fármacos , Oxirredutases atuantes sobre Doadores de Grupo CH-CH/fisiologia , Toluidinas/toxicidade , Proteína Supressora de Tumor p53/fisiologia , Antioxidantes/farmacologia , Apoptose/efeitos dos fármacos , Apoptose/fisiologia , Linhagem Celular Transformada , Di-Hidro-Orotato Desidrogenase , Humanos , Hidroxibutiratos , Queratinócitos/metabolismo , Queratinócitos/ultraestrutura , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Nitrilas , Pirimidinas/farmacologia
7.
Mol Pharmacol ; 79(6): 941-52, 2011 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-21415306

RESUMO

The production of reactive aldehydes such as 4-hydroxynonenal (4-HNE) is proposed to be an important factor in the etiology of alcoholic liver disease. To understand the effects of 4-HNE on homeostatic signaling pathways in hepatocytes, cellular models consisting of the human hepatocellular carcinoma cell line (HepG2) and primary rat hepatocytes were evaluated. Treatment of both HepG2 cells and primary hepatocytes with subcytotoxic concentrations of 4-HNE resulted in the activation of Akt within 30 min as demonstrated by increased phosphorylation of residues Ser473 and Thr308. Quantification and subsequent immunocytochemistry of phosphatidylinositol-3,4,5-trisphosphate [PtdIns(3,4,5)P(3)[rsqb] resulted in a 6-fold increase in total PtdIns(3,4,5)P(3) and increased immunostaining at the plasma membrane after 4-HNE treatment. Cotreatment of HepG2 cells with 4-HNE and the phosphatidylinositol 3-kinase (PI3K) inhibitor 2-(4-morpholinyl)-8-phenyl-4H-1-benzopyran-4-one (Ly294002) or the protein phosphatase 2A (PP2A) inhibitor okadaic acid revealed that the mechanism of activation of Akt is PI3K-dependent and PP2A-independent. Using biotin hydrazide detection, it was established that the incubation of HepG2 cells with 4-HNE resulted in increased carbonylation of the lipid phosphatase known as "phosphatase and tensin homolog deleted on chromosome 10" (PTEN), a key regulator of Akt activation. Activity assays both in HepG2 cells and recombinant PTEN revealed a decrease in PTEN lipid phosphatase activity after 4-HNE application. Mass spectral analysis of 4-HNE-treated recombinant PTEN detected a single 4-HNE adduct. Subsequent analysis of Akt dependent physiological consequences of 4-HNE in HepG2 cells revealed significant increases in the accumulation of neutral lipids. These results provide a potential mechanism of Akt activation and cellular consequences of 4-HNE in hepatocytes.


Assuntos
Aldeídos/farmacologia , Hepatócitos/efeitos dos fármacos , PTEN Fosfo-Hidrolase/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Sequência de Bases , Linhagem Celular Tumoral , Células Cultivadas , Primers do DNA , Ativação Enzimática , Transferência Ressonante de Energia de Fluorescência , Hepatócitos/enzimologia , Humanos , Imuno-Histoquímica , Masculino , Microscopia Confocal , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes/antagonistas & inibidores
8.
Hepatology ; 51(3): 1007-16, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20099297

RESUMO

UNLABELLED: Acetaminophen-induced liver injury (AILI) is a significant health problem and represents the most frequent cause of drug-induced liver failure in the United States. The development and implementation of successful therapeutic intervention strategies have been demanding, due to significant limitations associated with the current treatment for AILI. Lactoferrin (Lac), a glycoprotein present in milk, has been demonstrated to possess a multitude of biological functions. Our study demonstrated a profound protective effect of Lac in a murine model of AILI, which was not dependent on its iron-binding ability, inhibition of acetaminophen (APAP) metabolism, or a direct cytoprotective effect on hepatocytes. Instead, Lac treatment significantly attenuated APAP-induced liver sinusoidal endothelial cell dysfunction and ameliorated hepatic microcirculation disorder. This protective effect of Lac appeared to be dependent on hepatic resident macrophages (Kupffer cells [KCs]). CONCLUSION: Collectively, our data indicate that Lac, through activation of KCs, inhibited APAP-induced liver sinusoidal endothelial cell damage and improved hepatic congestion, thereby protecting against AILI. These findings reveal the significant therapeutic potential of Lac during AILI and other types of liver diseases.


Assuntos
Acetaminofen/efeitos adversos , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Lactoferrina/uso terapêutico , Animais , Células Endoteliais/efeitos dos fármacos , Células Endoteliais/fisiologia , Fígado/irrigação sanguínea , Fígado/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Microcirculação
9.
Apoptosis ; 15(10): 1234-46, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-20544286

RESUMO

Teriflunomide (TFN) reportedly inhibits de novo pyrimidine synthesis and exhibits anti-inflammatory, disease-modifying activities in vivo. These qualities would suggest that TFN could be useful in skin cancer chemoprevention or therapy. We investigated some mechanistic aspects of this tenet by characterizing the effects of TFN on premalignant and malignant human cutaneous keratinocytes. TFN promoted a dose- and/or time-dependent cytostasis and in these cells, which was followed by apoptosis. These features occurred in the presence of a physiological concentration of uridine in the culture medium. The short-term S phase arrest triggered by TFN was reversible in the malignant keratinocytes, and the indirect apoptosis induction was apparently preceded by mitochondrial disruption and reactive oxygen species production in both the premalignant and malignant keratinocytes. Respiration deficient malignant keratinocytes were resistant to the acute cytostatic and latent apoptotic effects of TFN implicating de novo pyrimidine synthesis and mitochondrial bioenergetics as the primary targets for TFN in the respiring cells. These novel mechanistic findings support a role for TFN in skin cancer chemoprevention and therapy.


Assuntos
Antineoplásicos/farmacologia , Apoptose , Crotonatos/farmacologia , Queratinócitos/efeitos dos fármacos , Lesões Pré-Cancerosas/patologia , Neoplasias Cutâneas/patologia , Toluidinas/farmacologia , Antineoplásicos/uso terapêutico , Antioxidantes/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/prevenção & controle , Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Crotonatos/uso terapêutico , Citostáticos/farmacologia , Metabolismo Energético/efeitos dos fármacos , Células Epiteliais/efeitos dos fármacos , Citometria de Fluxo , Humanos , Hidroxibutiratos , Queratinócitos/citologia , Queratinócitos/metabolismo , Queratinócitos/patologia , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Nitrilas , Consumo de Oxigênio/efeitos dos fármacos , Lesões Pré-Cancerosas/tratamento farmacológico , Lesões Pré-Cancerosas/metabolismo , Pirimidinas/biossíntese , Espécies Reativas de Oxigênio/metabolismo , Fase S , Neoplasias Cutâneas/tratamento farmacológico , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/prevenção & controle , Toluidinas/uso terapêutico , Uridina/metabolismo
10.
Apoptosis ; 14(7): 849-63, 2009 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-19421858

RESUMO

Prostate tumorigenesis is coupled with an early metabolic switch in transformed prostate epithelial cells that effectively increases their mitochondrial bioenergetic capacity. The synthetic retinoid N-(4-hydroxyphenyl)retinamide (4HPR) inhibits prostate cancer development in vivo, and triggers reactive oxygen species (ROS)-dependent prostate cancer cell apoptosis in vitro. The possibility that 4HPR-induced ROS production is associated with mitochondrial bioenergetics and required for apoptosis induction in transformed prostate epithelial cells in vitro would advocate a prospective mechanistic basis for 4HPR-mediated prostate cancer chemoprevention in vivo. We investigated this tenet by comparing and contrasting 4HPR's effects on premalignant PWR-1E and malignant DU-145 human prostate epithelial cells. 4HPR promoted a dose- and/or time-dependent apoptosis induction in PWR-1E and DU-145 cells, which was preceded by and dependent on an increase in mitochondrial ROS production. In this regard, the PWR-1E cells were more sensitive than the DU-145 cells, and they consumed roughly twice as much oxygen as the DU-145 cells suggesting oxidative phosphorylation was higher in the premalignant cells. Interestingly, increasing the [Ca(2+)] in the culture medium of the PWR-1E cells attenuated their proliferation as well as their mitochondrial bioenergetic capacity and 4HPR's cytotoxic effects. Correspondingly, the respiration-deficient derivatives (i.e., rho(0) cells lacking mitochondrial DNA) of DU-145 cells were markedly resistant to 4HPR-induced ROS production and apoptosis. Together, these observations implied that the reduction of mitochondrial bioenergetics protected PWR-1E and DU-145 cells against the cytotoxic effects of 4HPR, and support the concept that oxidative phosphorylation is an essential determinant in 4HPR's apoptogenic signaling in transformed human prostate epithelial cells.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Células Epiteliais , Fenretinida/farmacologia , Mitocôndrias , Próstata , Neoplasias da Próstata , Animais , Ciclo Celular/fisiologia , Linhagem Celular/efeitos dos fármacos , Linhagem Celular/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Humanos , Masculino , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/metabolismo , Consumo de Oxigênio , Próstata/citologia , Próstata/efeitos dos fármacos , Próstata/patologia , Neoplasias da Próstata/metabolismo , Neoplasias da Próstata/patologia , Espécies Reativas de Oxigênio/metabolismo
11.
Free Radic Biol Med ; 44(7): 1382-93, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18206126

RESUMO

Curcumin exhibits anticancer activity in vivo and triggers tumor cell apoptosis in vivo and in vitro. Several in vitro studies suggest that curcumin-induced apoptosis is associated with reactive oxygen species (ROS) production and/or oxidative stress in transformed cells. This study compared and contrasted the effects of curcumin on human skin cancer cells and their respiration-deficient (rho0) clones to characterize the prospective oxidative stress signaling responsible for initiating apoptosis. Curcumin promoted a dose-and time-dependent G2/M cell cycle arrest and/or apoptosis in COLO 16 cells. Apoptosis induction in COLO 16 cells was associated with DNA fragmentation, cell shrinkage, the externalization of cell membrane phosphatidylserine, and mitochondrial disruption, which were preceded by an increase in intracellular ROS production. Pharmacologically lowering the mitochondrial bioenergetic capacity, as well as the constitutive ROS levels, in COLO 16 cells suppressed the cytotoxic effects of curcumin. Correspondingly, the rho0 counterparts of COLO 16 cells were markedly resistant to ROS production, mitochondrial disruption, and DNA fragmentation following curcumin exposure. These observations implied that the diminution of mitochondrial ROS production protected cells against the cytotoxic effects of curcumin, and support the notion that mitochondrial respiration and redox tone are pivotal determinants in apoptosis signaling by curcumin in human skin cancer cells.


Assuntos
Apoptose , Curcumina/farmacologia , Mitocôndrias/metabolismo , Espécies Reativas de Oxigênio , Ciclo Celular , Linhagem Celular Transformada , Linhagem Celular Tumoral , Fragmentação do DNA , Relação Dose-Resposta a Droga , Humanos , Potenciais da Membrana , Modelos Biológicos , Consumo de Oxigênio , Fosfatidilserinas/química , Fatores de Tempo
12.
Free Radic Biol Med ; 45(2): 97-110, 2008 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-18454943

RESUMO

Cancer chemopreventive agents block the transformation of normal cells and/or suppress the promotion of premalignant cells to malignant cells. Certain agents may achieve these objectives by modulating xenobiotic biotransformation, protecting cellular elements from oxidative damage, or promoting a more differentiated phenotype in target cells. Conversely, various cancer chemopreventive agents can encourage apoptosis in premalignant and malignant cells in vivo and/or in vitro, which is conceivably another anticancer mechanism. Furthermore, it is evident that many of these apoptogenic agents function as prooxidants in vitro. The constitutive intracellular redox environment dictates a cell's response to an agent that alters this environment. Thus, it is highly probable that normal cells, through adaption, could acquire resistance to transformation via exposure to a chemopreventive agent that promotes oxidative stress or disrupts the normal redox tone of these cells. In contrast, transformed cells, which typically endure an oxidizing intracellular environment, would ultimately succumb to apoptosis due to an uncontrollable production of reactive oxygen species caused by the same agent. Here, we provide evidence to support the hypothesis that reactive oxygen species and cellular redox tone are exploitable targets in cancer chemoprevention via the stimulation of cytoprotection in normal cells and/or the induction of apoptosis in transformed cells.


Assuntos
Antineoplásicos/uso terapêutico , Antioxidantes/uso terapêutico , Quimioprevenção/métodos , Neoplasias/prevenção & controle , Estresse Oxidativo/fisiologia , Animais , Transformação Celular Neoplásica/efeitos dos fármacos , Humanos , Fator 2 Relacionado a NF-E2/metabolismo , Espécies Reativas de Oxigênio/toxicidade
13.
Cancer Res ; 66(7): 3737-46, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16585200

RESUMO

Phosphoinositol-3-kinase (PI3K)/protein kinase B (AKT) and Fms-like tyrosine kinase 3 (FLT3) signaling are aberrantly activated in acute myelogenous leukemia (AML) cells. Constitutively activated AKT and FLT3 regulate leukemia cell survival and resistance to chemotherapy. In this study, we investigated the effects of the novel multiple kinase inhibitor KP372-1 on the survival of AML cell lines and primary AML samples. KP372-1 directly inhibited the kinase activity of AKT, PDK1, and FLT3 in a concentration-dependent manner. Western blot analysis indicated that KP372-1 decreased the phosphorylation of AKT on both Ser(473) and Thr(308); abrogated the phosphorylation of p70S6 kinase, BAD, and Foxo3a via PI3K/AKT signaling; and down-regulated expression of PIM-1 through direct inhibition of FLT3. Treatment of AML cell lines with KP372-1 resulted in rapid generation of reactive oxygen species and stimulation of oxygen consumption, followed by mitochondrial depolarization, caspase activation, and phosphatidylserine externalization. KP372-1 induced pronounced apoptosis in AML cell lines and primary samples irrespective of their FLT3 status, but not in normal CD34(+) cells. Moreover, KP372-1 markedly decreased the colony-forming ability of primary AML samples (IC(50) < 200 nmol/L) with minimal cytotoxic effects on normal progenitor cells. Taken together, our results show that the simultaneous inhibition of critical prosurvival kinases by KP372-1 leads to mitochondrial dysfunction and apoptosis of AML but not normal hematopoietic progenitor cells.


Assuntos
Apoptose/efeitos dos fármacos , Compostos Heterocíclicos de 4 ou mais Anéis/farmacologia , Leucemia Mieloide Aguda/enzimologia , Mitocôndrias/efeitos dos fármacos , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Tetrazóis/farmacologia , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Apoptose/fisiologia , Processos de Crescimento Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patologia , Camundongos , Mitocôndrias/fisiologia , Mutação , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Transdução de Sinais/efeitos dos fármacos , Células U937 , Tirosina Quinase 3 Semelhante a fms/genética , Tirosina Quinase 3 Semelhante a fms/metabolismo
14.
Oncotarget ; 8(23): 36936-36949, 2017 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-28415808

RESUMO

We have identified that the ganglioside GD2 is a marker for breast cancer stem cells (BCSCs), and that targeting the enzyme GD3 synthase (GD3S, which regulates GD2 biosynthesis) reduces breast tumorigenesis. The pathways regulating GD2 expression, and their anomalous functions in BCSC, are unclear. Proteomic analysis of GD2+ and GD2- cells from breast cancer cell lines revealed the activation of NFκB signaling in GD2+ cells. Dose- and time-dependent suppression of NFκB signaling by the small molecule inhibitor BMS-345541 reduced GD2+ cells by > 90%. Likewise, BMS-345541 inhibited BCSC GD3S expression, mammosphere formation, and cell migration/invasion in vitro. Breast tumor-bearing mice treated with BMS-345541 showed a statistically significant decrease in tumor volume and exhibited prolonged survival compared to control mice, with a median survival of 78 d for the BMS-345541-treated group vs. 58 d for the controls. Moreover, in an experimental metastases model, treatment with BMS-345541 reduced the lung metastases by > 5-fold. These data suggest that GD2 expression and function,and NFκB signaling, are related, and they control BCSCs tumorigenic characteristics. Thus, the suppression of NFκB signaling by BMS-345541 is a potentially important advance in controlling breast cancer growth and metastases.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Gangliosídeos/metabolismo , Imidazóis/farmacologia , Células-Tronco Neoplásicas/efeitos dos fármacos , Quinoxalinas/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Carcinogênese/efeitos dos fármacos , Carcinogênese/genética , Carcinogênese/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Quinase I-kappa B/antagonistas & inibidores , Quinase I-kappa B/genética , Quinase I-kappa B/metabolismo , Camundongos Endogâmicos NOD , Camundongos Knockout , Camundongos SCID , Metástase Neoplásica , Células-Tronco Neoplásicas/metabolismo , Células-Tronco Neoplásicas/patologia , Interferência de RNA , Sialiltransferases/genética , Sialiltransferases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética
15.
Cancer Res ; 77(6): 1453-1464, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28108519

RESUMO

Leukemia cells in the bone marrow must meet the biochemical demands of increased cell proliferation and also survive by continually adapting to fluctuations in nutrient and oxygen availability. Thus, targeting metabolic abnormalities in leukemia cells located in the bone marrow is a novel therapeutic approach. In this study, we investigated the metabolic role of bone marrow adipocytes in supporting the growth of leukemic blasts. Prevention of nutrient starvation-induced apoptosis of leukemic cells by bone marrow adipocytes, as well as the metabolic and molecular mechanisms involved in this process, was investigated using various analytic techniques. In acute monocytic leukemia (AMoL) cells, the prevention of spontaneous apoptosis by bone marrow adipocytes was associated with an increase in fatty acid ß-oxidation (FAO) along with the upregulation of PPARγ, FABP4, CD36, and BCL2 genes. In AMoL cells, bone marrow adipocyte coculture increased adiponectin receptor gene expression and its downstream target stress response kinase AMPK, p38 MAPK with autophagy activation, and upregulated antiapoptotic chaperone HSPs. Inhibition of FAO disrupted metabolic homeostasis, increased reactive oxygen species production, and induced the integrated stress response mediator ATF4 and apoptosis in AMoL cells cocultured with bone marrow adipocytes. Our results suggest that bone marrow adipocytes support AMoL cell survival by regulating their metabolic energy balance and that the disruption of FAO in bone marrow adipocytes may be an alternative, novel therapeutic strategy for AMoL therapy. Cancer Res; 77(6); 1453-64. ©2017 AACR.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Adipócitos/patologia , Apoptose , Medula Óssea/patologia , Ácidos Graxos/química , Redes Reguladoras de Genes , Leucemia Monocítica Aguda/patologia , Adipócitos/metabolismo , Medula Óssea/metabolismo , Ciclo Celular , Proliferação de Células , Técnicas de Cocultura , Humanos , Leucemia Monocítica Aguda/metabolismo , Metabolismo dos Lipídeos , Células-Tronco Mesenquimais/metabolismo , Oxirredução , Fosforilação , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais , Células Tumorais Cultivadas , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
16.
J Natl Cancer Inst ; 94(17): 1281-92, 2002 Sep 04.
Artigo em Inglês | MEDLINE | ID: mdl-12208893

RESUMO

BACKGROUND: The vanilloids capsaicin and resiniferatoxin are natural products that contain a vanillyl moiety (4-hydroxy-3-methoxybenzyl). Both vanilloids can induce apoptosis in certain cell types by a mechanism that has not been fully elucidated but may involve plasma membrane or mitochondrial targets. We investigated the role of mitochondrial respiration in vanilloid-induced apoptosis. METHODS: Cytofluorometric analysis was used to evaluate the effects of vanilloids on apoptosis, Ca(2+) mobilization, hydroperoxide generation, and DNA content in cells from two human cutaneous squamous cell carcinoma (SCC) cell lines (parental cells) and in their respiration-deficient clones. Oxygen consumption by the cells was determined polarographically. RESULTS: The majority of the parental SCC cells underwent apoptosis after a 12-hour exposure to 100 micro M capsaicin or 10 micro M resiniferatoxin. The induction of apoptosis was associated with the mitochondrial permeability transition (i.e., an increase in the permeability of the inner mitochondrial membrane associated with the opening of a nonspecific pore). Exposure of parental cells to either vanilloid was not associated with an increase in intracellular free Ca(2+) levels but was associated with a rapid increase in hydroperoxide generation and a decrease in oxygen consumption. After vanilloid treatment, the respiration-deficient clones generated less hydroperoxide and were resistant to the mitochondrial permeability transition and the induction of apoptosis. Moreover, vanilloid treatment inhibited cell proliferation in the respiration-deficient clones by promoting G(1) arrest. CONCLUSIONS: Vanilloid-induced apoptosis in the parental SCC cells appears to involve the inhibition of mitochondrial respiration. The apoptogenic effects promoted by vanilloid treatment in parental SCC cells, as well as the antiproliferative effects observed in their respiration-deficient clones, suggest that vanilloids may be useful for preventing or treating skin cancers or other hyperproliferative skin disorders.


Assuntos
Apoptose/efeitos dos fármacos , Capsaicina/farmacologia , Carcinoma de Células Escamosas/tratamento farmacológico , Mitocôndrias/efeitos dos fármacos , Consumo de Oxigênio/efeitos dos fármacos , Espécies Reativas de Oxigênio/farmacologia , Neoplasias Cutâneas/tratamento farmacológico , Cálcio/metabolismo , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Ciclo Celular/efeitos dos fármacos , DNA de Neoplasias/metabolismo , Diterpenos/farmacologia , Feminino , Citometria de Fluxo , Humanos , Peróxido de Hidrogênio/metabolismo , Potenciais da Membrana , Mitocôndrias/metabolismo , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Células Tumorais Cultivadas/efeitos dos fármacos
17.
Pharmacol Ther ; 153: 25-35, 2015 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26048327

RESUMO

Nucleocytoplasmic trafficking of proteins/RNAs is essential to normal cellular function. Indeed, accumulating evidence suggests that cancer cells escape anti-neoplastic mechanisms and benefit from pro-survival signals via the dysregulation of this system. The nuclear exporter chromosome region maintenance 1 (CRM1) protein is the only protein in the karyopherin-ß protein family that contributes to the trafficking of numerous proteins and RNAs from the nucleus. It is considered to be an oncogenic, anti-apoptotic protein in transformed cells, since it reportedly functions as a gatekeeper for cell survival, including affecting p53 function, and ribosomal biogenesis. Furthermore, abnormally high expression of CRM1 is correlated with poor patient prognosis in various malignancies. Therapeutic targeting of CRM1 has emerged as a novel cancer treatment strategy, starting with a clinical trial with leptomycin B, the original specific inhibitor of CRM1, followed by development of several next-generation small molecules. KPT-330, a novel member of the CRM1-selective inhibitors of nuclear export (SINE) class of compounds, is currently undergoing clinical evaluation for the therapy of various malignancies. Results from these trials suggest that SINE compounds may be particularly useful against hematological malignancies, which often become refractory to standard chemotherapeutic agents.


Assuntos
Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Carioferinas/antagonistas & inibidores , Carioferinas/metabolismo , Neoplasias/tratamento farmacológico , Receptores Citoplasmáticos e Nucleares/antagonistas & inibidores , Receptores Citoplasmáticos e Nucleares/metabolismo , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/fisiologia , Humanos , Carioferinas/biossíntese , Modelos Biológicos , Terapia de Alvo Molecular/métodos , Neoplasias/metabolismo , Neoplasias/patologia , Receptores Citoplasmáticos e Nucleares/biossíntese , Proteína Exportina 1
18.
BBA Clin ; 4: 59-68, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26674329

RESUMO

BACKGROUND: Acute myeloid leukemia (AML) patients with highly active AKT tend to do poorly. Cell cycle arrest and apoptosis are tightly regulated by AKT via phosphorylation of GSK3α and ß isoforms which inactivates these kinases. In the current study we examine the prognostic role of AKT mediated GSK3 phosphorylation in AML. METHODS: We analyzed GSK3α/ß phosphorylation by reverse phase protein analysis (RPPA) in a cohort of 511 acute myeloid leukemia (AML) patients. Levels of phosphorylated GSK3 were correlated with patient characteristics including survival and with expression of other proteins important in AML cell survival. RESULTS: High levels of p-GSK3α/ß correlated with adverse overall survival and a lower incidence of complete remission duration in patients with intermediate cytogenetics, but not in those with unfavorable cytogenetics. Intermediate cytogenetic patients with FLT3 mutation also fared better respectively when p-GSK3α/ß levels were lower. Phosphorylated GSK3α/ß expression was compared and contrasted with that of 229 related cell cycle arrest and/or apoptosis proteins. Consistent with p-GSK3α/ß as an indicator of AKT activation, RPPA revealed that p-GSK3α/ß positively correlated with phosphorylation of AKT, BAD, and P70S6K, and negatively correlated with ß-catenin and FOXO3A. PKCδ also positively correlated with p-GSK3α/ß expression, suggesting crosstalk between the AKT and PKC signaling pathways in AML cells. CONCLUSIONS: These findings suggest that AKT-mediated phosphorylation of GSK3α/ß may be beneficial to AML cell survival, and hence detrimental to the overall survival of AML patients. Intrinsically, p-GSK3α/ß may serve as an important adverse prognostic factor for a subset of AML patients.

19.
Front Oncol ; 3: 67, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23565503

RESUMO

Nearly 60 years ago Otto Warburg proposed, in a seminal publication, that an irreparable defect in the oxidative capacity of normal cells supported the switch to glycolysis for energy generation and the appearance of the malignant phenotype (Warburg, 1956). Curiously, this phenotype was also observed by Warburg in embryonic tissues, and recent research demonstrated that normal stem cells may indeed rely on aerobic glycolysis - fermenting pyruvate to lactate in the presence of ample oxygen - rather than on the complete oxidation of pyruvate in the Krebs cycle - to generate cellular energy (Folmes et al., 2012). However, it remains to be determined whether this phenotype is causative for neoplastic development, or rather the result of malignant transformation. In addition, in light of mounting evidence demonstrating that cancer cells can carry out electron transport and oxidative phosphorylation, although in some cases predominantly using electrons from non-glucose carbon sources (Bloch-Frankenthal et al., 1965), Warburg's hypothesis needs to be revisited. Lastly, recent evidence suggests that the leukemia bone marrow microenvironment promotes the Warburg phenotype adding another layer of complexity to the study of metabolism in hematological malignancies. In this review we will discuss some of the evidence for alterations in the intermediary metabolism of leukemia cells and present evidence for a concept put forth decades ago by lipid biochemist Feodor Lynen, and acknowledged by Warburg himself, that cancer cell mitochondria uncouple ATP synthesis from electron transport and therefore depend on glycolysis to meet their energy demands (Lynen, 1951; Warburg, 1956).

20.
PLoS One ; 8(3): e58426, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23484027

RESUMO

CXCR4, the receptor for stromal-derived factor-1, is reportedly involved in breast carcinogenesis. However, the mechanisms through which CXCR4 contributes to breast cancer cell growth and metastases are poorly understood. In this study, we examined the putative in vitro and in vivo anti-cancer effects of the specific CXCR4 inhibitor AMD3465. Here, we report that AMD3465 triggers a reduction in breast cancer cell invasiveness in vitro, and promotes marked changes in oncogenic signaling proteins including a reduction in STAT3, JAK2, AKT, and CXCR4 phosphorylation and the reduced expression of GSK3 and cMYC. Using three breast cancer cell lines as murine syngeneic immunocompetent breast cancer models, we found that AMD3465 inhibited breast tumor formation and reduced tumor cell metastases to the lung and liver. Furthermore, treatment with AMD3465 significantly reduced the infiltration of myeloid CD11b positive cells at the aforementioned metastatic sites as well as the spleen implying this agent could regulate the formation of the tumor microenvironment and conceivably the premetastatic niche. In conclusion, our studies suggest that AMD3465 inhibits breast cancer growth and metastases by acting on tumor cells as well as immune cells that constitute the tumor microenvironment. This process appears to be regulated, at least in part, through the modulation of oncogenic signaling that includes the STAT3 pathway. Thus, CXCR4 could be a novel target for breast cancer therapy.


Assuntos
Neoplasias da Mama/tratamento farmacológico , Invasividade Neoplásica/fisiopatologia , Metástase Neoplásica/prevenção & controle , Piridinas/farmacologia , Receptores CXCR4/antagonistas & inibidores , Transdução de Sinais/fisiologia , Análise de Variância , Animais , Western Blotting , Linhagem Celular Tumoral , Feminino , Citometria de Fluxo , Proteínas de Fluorescência Verde , Técnicas Histológicas , Luciferases , Camundongos , Camundongos Endogâmicos BALB C , Piridinas/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA