Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Metallomics ; 13(7)2021 07 12.
Artigo em Inglês | MEDLINE | ID: mdl-34185060

RESUMO

Copper (Cu) is an essential trace element that plays an important role in maintaining neuronal functions such as the biosynthesis of neurotransmitters. In contrast, exposure to excess Cu results in cell injury. Therefore, intracellular Cu levels are strictly regulated by proteins related to Cu-trafficking, including ATP7A. Parkinson's disease (PD) is a neurodegenerative disorder and is characterized by the loss of dopaminergic neurons in the substantia nigra. Recently, the abnormality of Cu homeostasis was demonstrated to be related to the pathogenesis of PD. However, the association between Cu dyshomeostasis and PD remains unclear. In this study, we examined the effects of 6-hydroxydopamine (6-OHDA), a neurotoxin used for the production of PD model animals, on cellular Cu trafficking in human neuroblastoma SH-SY5Y cells. 6-OHDA reduced the protein levels of the Cu exporter ATP7A and the Cu chaperone Atox1, but not CTR1, a Cu importer; however, it did not affect the expression of ATP7A and Atox1 mRNAs. The decreased levels of ATP7A and Atox1 proteins were restored by the antioxidant N-acetylcysteine and the lysosomal inhibitor bafilomycin A1. This suggests that 6-OHDA-induced oxidative stress facilitates the degradation of these proteins. In addition, the amount of intracellular Cu after exposure to CuCl2 was significantly higher in cells pretreated with 6-OHDA than in untreated cells. Moreover, 6-OHDA reduced the protein levels of the cuproenzyme dopamine ß-hydroxylase that converts dopamine to noradrenaline. Thus, this study suggests that 6-OHDA disrupts Cu homeostasis through the dysregulation of cellular Cu trafficking, resulting in the dysfunction of neuronal cells.


Assuntos
Proteínas de Transporte de Cobre/antagonistas & inibidores , ATPases Transportadoras de Cobre/antagonistas & inibidores , Cobre/metabolismo , Homeostase , Chaperonas Moleculares/antagonistas & inibidores , Neuroblastoma/patologia , Estresse Oxidativo , Oxidopamina/farmacologia , Adrenérgicos/farmacologia , Morte Celular , Humanos , Neuroblastoma/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Células Tumorais Cultivadas
2.
Am J Obstet Gynecol ; 222(1): 64.e1-64.e16, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31351063

RESUMO

BACKGROUND: Multidrug resistance is a major concern in uterine leiomyosarcoma treatment. Development of effective chemotherapies and management of drug resistance in patients is necessary. The copper efflux transporter adenosine triphosphatase copper transporting beta is a member of the P-type adenosine triphosphatase family and is also known as a strong platinum efflux transporter. Various reports have shown the association between adenosine triphosphatase copper transporting beta and platinum resistance; however, suitable inhibitors or methods for inhibiting platinum efflux via adenosine triphosphatase copper transporting beta are not developed. OBJECTIVE: Our study focused on platinum resistance in uterine leiomyosarcoma. The role of adenosine triphosphatase copper transporting beta in uterine leiomyosarcoma resistance to platinum drugs was investigated both in vitro and in vivo. STUDY DESIGN: Adenosine triphosphatase copper transporting beta expression was investigated by Western blotting and the efficacy of copper sulfate pretreatment and cisplatin administration in adenosine triphosphatase copper transporting beta-expressing cells was investigated both in vitro and in vivo. RESULTS: Western blot analysis of SK-LMS-1 cells (uterine leiomyosarcoma cell line) revealed strong adenosine triphosphatase copper transporting beta expression. A permanent SK-LMS-ATPase copper transporting beta-suppressed cell line (SK-LMS-7B cells) was generated, and cisplatin exhibited a significant antitumor effect in SK-LMS-7B cells, both in vitro (SK-LMS-1 cells, half-maximal inhibitory concentration, 17.2 µM; SK-LMS-7B cells, half-maximal inhibitory concentration, 4.2 µM, P < .01) and in xenografts compared with that in SK-LMS-1 cells (5.8% vs 62.8%, P < .01). Copper sulfate was identified as a preferential inhibitor of platinum efflux via adenosine triphosphatase copper transporting beta. In SK-LMS-1 cells pretreated with 15 µM copper sulfate for 3 hours, the cisplatin half-maximal inhibitory concentration decreased significantly compared with that in untreated cells and resulted in significantly increased intracellular platinum accumulation (1.9 pg/cell vs 8.6 pg/cell, P < .01). The combination of copper sulfate pretreatment with cisplatin administration was also effective in vivo and caused cisplatin to exhibit significantly increased antitumor effects in mice with SK-LMS-1 xenografts (3.1% vs 62.7%, P < .01). CONCLUSION: Our study demonstrates that adenosine triphosphatase copper transporting beta is overexpressed in uterine leiomyosarcoma cells and that copper sulfate, which acts as an inhibitor of platinum efflux via adenosine triphosphatase copper transporting beta, may be a therapeutic agent in the treatment of uterine leiomyosarcoma.


Assuntos
Antineoplásicos/uso terapêutico , Cisplatino/uso terapêutico , Sulfato de Cobre/farmacologia , ATPases Transportadoras de Cobre/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Leiomiossarcoma/tratamento farmacológico , Neoplasias Uterinas/tratamento farmacológico , Animais , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Cisplatino/farmacologia , ATPases Transportadoras de Cobre/antagonistas & inibidores , ATPases Transportadoras de Cobre/genética , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Leiomiossarcoma/genética , Leiomiossarcoma/metabolismo , Camundongos , Transplante de Neoplasias , Transplante Heterólogo , Neoplasias Uterinas/genética , Neoplasias Uterinas/metabolismo
3.
Oncol Rep ; 42(6): 2611-2621, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31638244

RESUMO

Platinum­based antitumor agents have been widely used to treat head and neck squamous cell carcinoma (HNSCC) and numerous other malignancies. Cisplatin is the most frequently used platinum­based antitumor agent, however drug resistance and numerous undesirable side effects limit its clinical efficacy for cancer patients. Cancer cells discharge cisplatin into the extracellular space via copper transporters such as ATPase copper transporting beta (ATP7B) in order to escape from cisplatin­induced cell death. In the present study, it was demonstrated for the first time that the copper chelator ammonium tetrathiomolybdate (TM) has several promising effects on cisplatin and HNSCC. First, TM suppressed the ATP7B expression in HNSCC cell lines in vitro, thereby enhancing the accumulation and apoptotic effect of cisplatin in the cancer cells. Next, it was revealed that TM enhanced the antitumor effect of cisplatin in HNSCC cell tumor progression in a mouse model of bone invasion, which is important since HNSCC cells frequently invade to facial bone. Finally, it was demonstrated that TM was able to overcome the cisplatin resistance of a human cancer cell line, A431, via ATP7B depression in vitro.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Cisplatino/farmacologia , ATPases Transportadoras de Cobre/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Molibdênio/farmacologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Neoplasias de Cabeça e Pescoço/patologia , Humanos , Masculino , Camundongos , Molibdênio/uso terapêutico , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Análise Serial de Tecidos , Ensaios Antitumorais Modelo de Xenoenxerto
4.
J Am Chem Soc ; 141(30): 12109-12120, 2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31283225

RESUMO

Copper (Cu) is required for maturation of cuproenzymes, cell proliferation, and angiogenesis, and its transport entails highly specific protein-protein interactions. In humans, the Cu chaperone Atox1 mediates Cu(I) delivery to P-type ATPases Atp7a and Atp7b (the Menkes and Wilson disease proteins, respectively), which are responsible for Cu release to the secretory pathway and excess Cu efflux. Cu(I) handover is believed to occur through the formation of three-coordinate intermediates where the metal ion is simultaneously linked to Atox1 and to a soluble domain of Cu-ATPases, both sharing a CxxC dithiol motif. The ultrahigh thermodynamic stability of chelating S-donor ligands secures the redox-active and potentially toxic Cu(I) ion, while their kinetic lability allows facile metal transfer. The same CxxC motifs can interact with and mediate the biological response to antitumor platinum drugs, which are among the most used chemotherapeutics. We show that cisplatin and an oxaliplatin analogue can specifically bind to the heterodimeric complex Atox1-Cu(I)-Mnk1 (Mnk1 is the first soluble domain of Atp7a), thus leading to a kinetically stable adduct that has been structurally characterized by solution NMR and X-ray crystallography. Of the two possible binding configurations of the Cu(I) ion in the cage made by the CxxC motifs of the two proteins, one (bidentate Atox1 and monodentate Mnk1) is less stable and more reactive toward cis-Pt(II) compounds, as shown by using mutated proteins. A Cu(I) ion can be retained at the Pt(II) coordination site but can be released to glutathione (a physiological thiol) or to other complexing agents. The Pt(II)-supported heterodimeric complex does not form if Zn(II) is used in place of Cu(I) and transplatin instead of cisplatin. The results indicate that Pt(II) drugs can specifically affect Cu(I) homeostasis by interfering with the rapid exchange of Cu(I) between Atox1 and Cu-ATPases with consequences on cancer cell viability and migration.


Assuntos
Antineoplásicos/farmacologia , Cisplatino/farmacologia , Proteínas de Transporte de Cobre/antagonistas & inibidores , ATPases Transportadoras de Cobre/antagonistas & inibidores , Cobre/metabolismo , Chaperonas Moleculares/antagonistas & inibidores , Oxaliplatina/farmacologia , Fragmentos de Peptídeos/antagonistas & inibidores , Antineoplásicos/química , Cisplatino/química , Proteínas de Transporte de Cobre/metabolismo , ATPases Transportadoras de Cobre/metabolismo , Cristalografia por Raios X , Humanos , Cinética , Modelos Moleculares , Chaperonas Moleculares/metabolismo , Estrutura Molecular , Oxaliplatina/química , Fragmentos de Peptídeos/metabolismo , Termodinâmica
5.
Am J Med Sci ; 358(3): 227-234, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31327462

RESUMO

BACKGROUND: Targeting ß-catenin has been shown to have great potential therapeutic value in cervical cancer. Because ß-catenin is also essential for normal cells, strategies to specifically target cancer will require identification of druggable factors capable of distinguishing ß-catenin signaling pathways between cancer and normal cells. METHODS: Expression of p-eIF4E and p-ß-catenin was analyzed in malignant and normal cervical tissues and cells. The effects and its underlying mechanisms of targeting MNK and eukaryotic translation initiation factor 4E (eIF4E) were determined in cervical cancer and normal cells. RESULTS: Inhibiting MNK/eIF4E axis selectively targets cervical cancer without affecting normal cervical cells, via suppressing eIF4E-mediated ß-catenin activation. We found that eIF4E phosphorylation was upregulated in cervical cancer cells and tissues but not normal cervical counterparts, and its phosphorylation at Ser 209 activates Wnt/ß-catenin signaling, promotes growth and migration in cervical cancer, in an MNK-dependent manner. MNK inhibition via genetic small interfering RNA (siRNA) knockdown or pharmacologic inhibitor effectively decreased phosphorylation of eIF4E and ß-catenin, leading to reduced ß-catenin activity and transcript levels of Wnt target genes in cervical cancer cells. Consistently, we found that MNK kinase inhibitor is effective in inhibiting proliferation and migration, and inducing apoptosis in cervical cancer but not normal cervical cells. The combination of MNK kinase inhibitor with paclitaxel achieved greater efficacy in cervical cancer cells than paclitaxel alone. CONCLUSIONS: Our work identifies MNK-eIF4E axis as a specific and critical regulator of ß-catenin activity in cervical cancer but not normal cervical cells, and suggests that targeting MNK is a useful therapeutic strategy in cervical cancer.


Assuntos
Antineoplásicos/uso terapêutico , ATPases Transportadoras de Cobre/antagonistas & inibidores , Fator de Iniciação 4E em Eucariotos/antagonistas & inibidores , Inibidores de Proteínas Quinases/uso terapêutico , Neoplasias do Colo do Útero/tratamento farmacológico , beta Catenina/metabolismo , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células , Fator de Iniciação 4E em Eucariotos/metabolismo , Feminino , Humanos , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Neoplasias do Colo do Útero/enzimologia , Neoplasias do Colo do Útero/patologia
6.
Mol Cancer Res ; 17(6): 1305-1315, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30842251

RESUMO

Medulloblastoma is a highly malignant pediatric brain tumor associated with poor outcome. Developing treatments that target the cancer stem cell (CSC) population in medulloblastoma are important to prevent tumor relapse and induce long-lasting clinical responses. We utilized medulloblastoma neurospheres that display CSC characteristics and found activation of the PI3K/AKT pathway in sphere-forming cells. Of all class IA PI3Ks, only the PI3Kα isoform was required for sphere formation by medulloblastoma cells. Knockdown of p110α, but not p110ß or p110δ, significantly disrupted cancer stem cell frequencies as determined by extreme limiting dilution analysis (ELDA), indicating an essential role for the PI3Kα catalytic isoform in medulloblastoma CSCs. Importantly, pharmacologic inhibition of the MAPK-interacting kinase (MNK) enhanced the antineoplastic effects of targeted PI3Kα inhibition in medulloblastoma. This indicates that MNK signaling promotes survival in medulloblastoma, suggesting dual PI3Kα and MNK inhibition may provide a novel approach to target and eliminate medulloblastoma CSCs. We also observed a significant reduction in tumor formation in subcutaneous and intracranial mouse xenograft models, which further suggests that this combinatorial approach may represent an efficient therapeutic strategy for medulloblastoma. IMPLICATIONS: These findings raise the possibility of a unique therapeutic approach for medulloblastoma, involving MNK targeting to sensitize medulloblastoma CSCs to PI3Kα inhibition.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Cerebelares/tratamento farmacológico , Classe I de Fosfatidilinositol 3-Quinases/antagonistas & inibidores , ATPases Transportadoras de Cobre/antagonistas & inibidores , Meduloblastoma/tratamento farmacológico , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Neoplasias Cerebelares/metabolismo , Feminino , Humanos , Meduloblastoma/metabolismo , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
7.
Sci Rep ; 9(1): 1535, 2019 02 07.
Artigo em Inglês | MEDLINE | ID: mdl-30733544

RESUMO

Wilson's disease (WD) is an inherited metabolic disease arising from ATPase copper transporting beta gene (ATP7B) mutation. Orthotoropic liver transplantation is the only radical treatment of fulminant WD, although appropriate donors are lacking at the onset of emergency. Given the hepatogenic capacity and tissue-integration/reconstruction ability in the liver of stem cells from human exfoliated deciduous teeth (SHED), SHED have been proposed as a source for curing liver diseases. We hypothesized the therapeutic potential of SHED and SHED-converted hepatocyte-like- cells (SHED-Heps) for fulminant WD. SHED and SHED-Heps were transplanted into WD model Atp7b-mutated Long-Evans Cinnamon (LEC) rats received copper overloading to induce a lethal fulminant liver failure. Due to the superior copper tolerance via ATP7B, SHED-Hep transplantation gave more prolonged life-span of fulminant LEC rats than SHED transplantation. The integrated ATP7B-expressing SHED-Heps showed more therapeutic effects on to restoring the hepatic dysfunction and tissue damages in the recipient liver than the integrated naïve SHED without ATP7B expression. Moreover, SHED-Heps could reduce copper-induced oxidative stress via ATP7B- independent stanniocalcin 1 secretion in the fulminant LEC rats, suggesting a possible role for paracrine effect of the integrated SHED-Heps. Taken together, SHED-Heps offer a potential of functional restoring, bridging, and preventive approaches for treating fulminant WD.


Assuntos
Hepatócitos/transplante , Degeneração Hepatolenticular/terapia , Células-Tronco/citologia , Dente Decíduo/citologia , Animais , Diferenciação Celular , Cobre/toxicidade , ATPases Transportadoras de Cobre/antagonistas & inibidores , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Modelos Animais de Doenças , Glicoproteínas/metabolismo , Hepatócitos/citologia , Hepatócitos/metabolismo , Degeneração Hepatolenticular/mortalidade , Degeneração Hepatolenticular/patologia , Humanos , Peptídeos e Proteínas de Sinalização Intercelular/farmacologia , Estresse Oxidativo/efeitos dos fármacos , Comunicação Parácrina , Interferência de RNA , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Endogâmicos LEC , Células-Tronco/metabolismo , Taxa de Sobrevida
8.
Nat Commun ; 10(1): 186, 2019 01 14.
Artigo em Inglês | MEDLINE | ID: mdl-30643139

RESUMO

Tetrathiomolybdate (TM) is used in the clinic for the treatment of Wilson's disease by targeting the cellular copper efflux protein ATP7B (WLN). Interestingly, both TM and WLN are associated with the efficacy of cisplatin, a widely used anticancer drug. Herein, we show that TM induces dimerization of the metal-binding domain of ATP7B (WLN4) through a unique sulfur-bridged Mo2S6O2 cluster. TM expels copper ions from Cu-WLN4 and forms a copper-free dimer. The binding of Mo to cysteine residues of WLN4 inhibits platination of the protein. Reaction with multi-domain proteins indicates that TM can also connect two domains in the same molecule, forming Mo-bridged intramolecular crosslinks. These results provide structural and chemical insight into the mechanism of action of TM against ATPase, and reveal the molecular mechanism by which TM attenuates the cisplatin resistance mediated by copper efflux proteins.


Assuntos
Antineoplásicos/farmacologia , Quelantes/farmacologia , Cisplatino/farmacologia , ATPases Transportadoras de Cobre/metabolismo , Molibdênio/farmacologia , Antineoplásicos/uso terapêutico , Quelantes/uso terapêutico , Cisplatino/uso terapêutico , Cobre/metabolismo , ATPases Transportadoras de Cobre/antagonistas & inibidores , ATPases Transportadoras de Cobre/química , Reagentes de Ligações Cruzadas/química , Reagentes de Ligações Cruzadas/farmacologia , Reagentes de Ligações Cruzadas/uso terapêutico , Cristalografia por Raios X , Cisteína/química , Cisteína/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Humanos , Molibdênio/uso terapêutico , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Platina/metabolismo , Domínios e Motivos de Interação entre Proteínas/efeitos dos fármacos , Multimerização Proteica/efeitos dos fármacos , Estrutura Secundária de Proteína
9.
J Korean Med Sci ; 32(8): 1327-1336, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28665070

RESUMO

Microarray analysis was used to investigate the lack of identified mammalian target of rapamycin (mTOR) pathway downstream genes to overcome cross-talk at non-muscle invasive high-grade (HG)-urothelial carcinoma (UC) of the bladder, gene expression patterns, gene ontology, and gene clustering by triple (p70S6K, S6K, and eIF4E) small interfering RNAs (siRNAs) or rapamycin in 5637 and T24 cell lines. We selected mTOR pathway downstream genes that were suppressed by siRNAs more than 2-fold, or were up-regulated or down-regulated by rapamycin more than 2-fold. We validated mTOR downstream genes with immunohistochemistry using a tissue microarray (TMA) of 125 non-muscle invasive HG-UC patients and knockout study to evaluate the synergistic effect with rapamycin. The microarray analysis selected mTOR pathway downstream genes consisting of 4 rapamycin up-regulated genes (FABP4, H19, ANXA10, and UPK3A) and 4 rapamycin down-regulated genes (FOXD3, ATP7A, plexin D1, and ADAMTS5). In the TMA, FABP4, and ATP7A were more expressed at T1 and FOXD3 was at Ta. ANXA10 and ADAMTS5 were more expressed in tumors ≤ 3 cm in diameter. In a multivariate Cox regression model, ANXA10 was a significant predictor of recurrence and ATP7A was a significant predictor of progression in non-muscle invasive HG-UC of the bladder. In an ATP7A knock-out model, rapamycin treatment synergistically inhibited cell viability, wound healing, and invasion ability compared to rapamycin only. Activity of the ANXA10 and ATP7A mTOR pathway downstream genes might predict recurrence and progression in non-muscle invasive HG-UC of the bladder. ATP7A knockout overcomes rapamycin cross-talk.


Assuntos
Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Neoplasias da Bexiga Urinária/patologia , Idoso , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , ATPases Transportadoras de Cobre/antagonistas & inibidores , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Progressão da Doença , Regulação para Baixo/efeitos dos fármacos , Fator de Iniciação 4E em Eucariotos/antagonistas & inibidores , Fator de Iniciação 4E em Eucariotos/genética , Fator de Iniciação 4E em Eucariotos/metabolismo , Feminino , Humanos , Masculino , Gradação de Tumores , Recidiva Local de Neoplasia , Interferência de RNA , Proteínas Quinases S6 Ribossômicas 70-kDa/antagonistas & inibidores , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Transdução de Sinais/genética , Sirolimo/farmacologia , Regulação para Cima/efeitos dos fármacos , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/mortalidade
10.
Oncotarget ; 7(51): 84439-84452, 2016 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-27806319

RESUMO

Cisplatin is an effective breast cancer drug but resistance often develops over prolonged chemotherapy. Therefore, we performed a candidate approach RNAi screen in combination with cisplatin treatment to identify molecular pathways conferring survival advantages. The screen identified ATP7A as a therapeutic target. ATP7A is a copper ATPase transporter responsible for intercellular movement and sequestering of cisplatin. Pharmaceutical replacement for ATP7A by ammonium tetrathiomolybdate (TM) enhanced cisplatin treatment in breast cancer cells. Allograft and xenograft models in athymic nude mice treated with cisplatin/TM exhibited retarded tumor growth, reduced accumulation of cancer stem cells and decreased cell proliferation as compared to mono-treatment with cisplatin or TM. Cisplatin/TM treatment of cisplatin-resistant tumors reduced ATP7A protein levels, attenuated cisplatin sequestering by ATP7A, increased nuclear availability of cisplatin, and subsequently enhanced DNA damage and apoptosis. Microarray analysis of gene ontology pathways that responded uniquely to cisplatin/TM double treatment depicted changes in cell cycle regulation, specifically in the G1/S transition. These findings offer the potential to combat platinum-resistant tumors and sensitize patients to conventional breast cancer treatment by identifying and targeting the resistant tumors' unique molecular adaptations.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Neoplasias da Mama/tratamento farmacológico , Cisplatino/farmacologia , ATPases Transportadoras de Cobre/antagonistas & inibidores , Molibdênio/farmacologia , Inibidores da Angiogênese/administração & dosagem , Inibidores da Angiogênese/farmacologia , Animais , Neoplasias da Mama/genética , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Cisplatino/administração & dosagem , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Sinergismo Farmacológico , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Células MCF-7 , Camundongos Nus , Molibdênio/administração & dosagem , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/metabolismo , Interferência de RNA , Ensaios Antitumorais Modelo de Xenoenxerto/métodos
11.
Metallomics ; 8(9): 963-972, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27714044

RESUMO

Intestinal iron absorption is highly regulated since no mechanism for iron excretion exists. We previously demonstrated that expression of an intestinal copper transporter (Atp7a) increases in parallel with genes encoding iron transporters in the rat duodenal epithelium during iron deprivation (Am. J. Physiol.: Gastrointest. Liver Physiol., 2005, 288, G964-G971). This led us to postulate that Atp7a may influence intestinal iron flux. Therefore, to test the hypothesis that Atp7a is required for optimal iron transport, we silenced Atp7a in rat IEC-6 and human Caco-2 cells. Iron transport was subsequently quantified in fully-differentiated cells plated on collagen-coated, transwell inserts. Interestingly, 59Fe uptake and efflux were impaired in both cell lines by Atp7a silencing. Concurrent changes in the expression of key iron transport-related genes were also noted in IEC-6 cells. Expression of Dmt1 (the iron importer), Dcytb (an apical membrane ferrireductase) and Fpn1 (the iron exporter) was decreased in Atp7a knockdown (KD) cells. Paradoxically, cell-surface ferrireductase activity increased (>5-fold) in Atp7a KD cells despite decreased Dcytb mRNA expression. Moreover, increased expression (>10-fold) of hephaestin (an iron oxidase involved in iron efflux) was associated with increased ferroxidase activity in KD cells. Increases in ferrireductase and ferroxidase activity may be compensatory responses to increase iron flux. In summary, in these reductionist models of the mammalian intestinal epithelium, Atp7a KD altered expression of iron transporters and impaired iron flux. Since Atp7a is a copper transporter, it is a logical supposition that perturbations in intracellular copper homeostasis underlie the noted biologic changes in these cell lines.


Assuntos
Diferenciação Celular , Ceruloplasmina/metabolismo , ATPases Transportadoras de Cobre/antagonistas & inibidores , Cobre/metabolismo , FMN Redutase/metabolismo , Mucosa Intestinal/metabolismo , Ferro/metabolismo , Animais , Transporte Biológico , Células Cultivadas , ATPases Transportadoras de Cobre/genética , ATPases Transportadoras de Cobre/metabolismo , Humanos , Mucosa Intestinal/citologia , Ratos
12.
Oncotarget ; 7(35): 56811-56825, 2016 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-27462781

RESUMO

Cytarabine (Ara-C) is a first line clinical therapeutic agent for treatment of acute myeloid leukemia (AML). However, this therapy is limited due to high rate of resistance and relapse. Recent research has revealed that the poor prognosis and resistance to Ara-C in AML were associated with its abnormally activated MAPK pathways. In this study, we showed a strong synergistic effect of Ara-C with either our Mnk inhibitor (MNKI-8e) or short hairpin RNA (shRNA) mediated knockdown of Mnks in MV4-11 AML cells. We investigated the underlying mechanisms for this synergism. We showed that both MNKI-8e and Mnk shRNAs enhanced the ability of Ara-C to induce apoptosis. We found that Ara-C increased the phosphorylation of Erk1/2, p38 and eIF4E, which correlated with an enhanced level of anti-apoptotic Mcl-1 protein. Inhibition of Mnk activity suppressed the Ara-C-induced MAPK activity, and thus enhanced apoptosis in MV4-11 cells. Taken together, our study suggests that MAPK-Mnk-eIF4E pathway plays a critical role in Ara-C-treated MV4-11 cells and targeting Mnk may be a promising therapeutic strategy for sensitizing leukemic cells to Ara-C therapy.


Assuntos
Apoptose , ATPases Transportadoras de Cobre/antagonistas & inibidores , Citarabina/farmacologia , Leucemia Mieloide Aguda/patologia , Antimetabólitos Antineoplásicos/farmacologia , Caspase 3/metabolismo , Caspase 7/metabolismo , Linhagem Celular Tumoral , Proliferação de Células , Sobrevivência Celular , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Ativação Enzimática , Regulação Leucêmica da Expressão Gênica , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Sistema de Sinalização das MAP Quinases , Fosforilação , RNA Interferente Pequeno/metabolismo , Transdução de Sinais
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA