Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Mol Cell Neurosci ; 123: 103785, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36241022

RESUMO

Insufficiencies of the micronutrient thiamine (Vitamin B1) have been associated with inducing Alzheimer's disease (AD)-like neuropathology. The hypometabolic state associated with chronic thiamine insufficiency (TI) has been demonstrated to be a contributor towards the development of amyloid plaque deposition and neurotoxicity. However, the molecular mechanism underlying TI induced AD pathology is still unresolved. Previously, we have established that TI stabilizes the metabolic stress transcriptional factor, Hypoxia Inducible Factor-1α (HIF1α). Utilizing neuronal hippocampal cells (HT22), TI-induced HIF1α activation triggered the amyloidogenic cascade through transcriptional expression and increased activity of ß-secretase (BACE1). Knockdown and pharmacological inhibition of HIF1α during TI significantly reduced BACE1 and C-terminal Fragment of 99 amino acids (C99) formation. TI also increased the expression of the HIF1α regulated pro-apoptotic protein, BCL2/adenovirus E1B 19 kDa protein-interacting protein (BNIP3). Correspondingly, cell toxicity during TI conditions was significantly reduced with HIF1α and BNIP3 knockdown. The role of BNIP3 in TI-mediated toxicity was further highlighted by localization of dimeric BNIP3 into the mitochondria and nuclear accumulation of Endonuclease G. Subsequently, TI decreased mitochondrial membrane potential and enhanced chromatin fragmentation. However, cell toxicity via the HIF1α/BNIP3 cascade required TI induced oxidative stress. HIF1α, BACE1 and BNIP3 expression was induced in 3xTg-AD mice after TI and administration with the HIF1α inhibitor YC1 significantly attenuated HIF1α and target genes levels in vivo. Overall, these findings demonstrate a critical stress response during TI involving the induction of HIF1α transcriptional activity that directly promotes neurotoxicity and AD-like pathology.


Assuntos
Doença de Alzheimer , Subunidade alfa do Fator 1 Induzível por Hipóxia , Deficiência de Tiamina , Animais , Camundongos , Doença de Alzheimer/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Tiamina/farmacologia , Deficiência de Tiamina/metabolismo
2.
Biomed Pharmacother ; 121: 109648, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31810115

RESUMO

Malignant cells frequently demonstrate an oncogenic-driven reliance on glycolytic metabolism to support their highly proliferative nature. Overexpression of pyruvate dehydrogenase kinase (PDK) may promote this unique metabolic signature of tumor cells by inhibiting mitochondrial function. PDKs function to phosphorylate and inhibit pyruvate dehydrogenase (PDH) activity. Silencing of PDK expression has previously been shown to restore mitochondrial function and reduce tumor cell proliferation. High dose Vitamin B1, or thiamine, possesses antitumor properties related to its capacity to reduce PDH phosphorylation and promote its enzymatic activity, presumably through PDK inhibition. Though a promising nutraceutical approach for cancer therapy, thiamine's low bioavailability may limit clinical effectiveness. Here, we have demonstrated exploiting the commercially available lipophilic thiamine analogs sulbutiamine and benfotiamine increases thiamine's anti-cancer effect in vitro. Determined by crystal violet proliferation assays, both sulbutiamine and benfotiamine reduced thiamine's millimolar IC50 value to micromolar equivalents. HPLC analysis revealed that sulbutiamine and benfotiamine significantly increased intracellular thiamine and TPP concentrations in vitro, corresponding with reduced levels of PDH phosphorylation. Through an ex vitro kinase screen, thiamine's activated cofactor form thiamine pyrophosphate (TPP) was found to inhibit the function of multiple PDK isoforms. Attempts to maximize intracellular TPP by exploiting thiamine homeostasis gene expression resulted in enhanced apoptosis in tumor cells. Based on our in vitro evaluations, we conclude that TPP serves as the active species mediating thiamine's inhibitory effect on tumor cell proliferation. Pharmacologic administration of benfotiamine, but not sulbutiamine, reduced tumor growth in a subcutaneous xenograft mouse model. It remains unclear if benfotiamine's effects in vivo are associated with PDK inhibition or through an alternative mechanism of action. Future work will aim to define the action of lipophilic thiamine mimetics in vivo in order to translate their clinical usefulness as anticancer strategies.


Assuntos
Antineoplásicos/farmacologia , Suplementos Nutricionais , Tiamina/análogos & derivados , Tiamina/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Intervalos de Confiança , Feminino , Humanos , Concentração Inibidora 50 , Espaço Intracelular/metabolismo , Camundongos Nus , Piruvato Desidrogenase Quinase de Transferência de Acetil/metabolismo , Complexo Piruvato Desidrogenase/metabolismo , Tiamina/química , Tiamina Pirofosfato/metabolismo
3.
Artigo em Inglês | MEDLINE | ID: mdl-31238261

RESUMO

Understanding the mechanisms underlying cancer cell survival is critical toward advancing drug discovery efforts in this field. Supplemental vitamins have been proposed to play a role in cancer cell metabolism because the increased supply of nutrients is thought to provide cofactors supporting the higher metabolic rate of cancer cells. Particularly, the role of thiamine (vitamin B1) in many biochemical pathways that supports cancer cell metabolism has been investigated. Consequently, the analysis of thiamine and its derivatives in a manner that reflects its dynamic response to genetic modification and pathophysiological stimuli is essential. In this work, we developed a mass spectrometry based-analytical method to track metabolites derived from stable isotope tracers for a better understanding of the metabolic fate of thiamine in cancer cells. This method used ion-pair reversed phase liquid chromatography to simultaneously quantify underivatized thiamine, thiamine monophosphate (TMP) and thiamine pyrophosphate (TPP) in cells. Hexylamine was used as an ion-pairing agent. The method was successfully validated for accuracy, precision and selectivity in accordance with U.S. FDA guidance. Furthermore, the method was then applied for the determination of thiamine and its derivatives with stable isotope labeling to explore the metabolic fate of intracellular thiamine in cancer cells. The finding shows that thiamine is rapidly converted to TPP however, the TPP does not return to thiamine. It appears that TPP may be utilized for other purposes rather than simply being an enzyme cofactor, suggesting unexplored roles for thiamine in cancer.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Espectrometria de Massas em Tandem/métodos , Tiamina/análise , Linhagem Celular Tumoral , Humanos , Tiamina/metabolismo
4.
Oncotarget ; 9(83): 35422-35438, 2018 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-30459934

RESUMO

Supplemental levels of vitamin B1 (thiamine) have been implicated in tumor progression. Tumor cells adaptively up-regulate thiamine transport during hypoxic stress. Upon uptake, thiamine pyrophosphokinase-1 (TPK1) facilitates the rapid phosphorylation of thiamine into thiamine pyrophosphate (TPP). However, the regulation of TPK1 during hypoxic stress is undefined. Understanding how thiamine homeostasis changes during hypoxia will provide critical insight into the malignant advantage supplemental thiamine may provide cancer cells. Using Western blot analysis and RT-PCR, we have demonstrated the post-transcriptional up-regulation of TPK1 in cancer cells following hypoxic exposure. TPK1 expression was also adaptively up-regulated following alterations of redox status by chemotherapeutic and antioxidant treatments. Although TPK1 was functionally up-regulated by hypoxia, HPLC analysis revealed a reduction in intracellular TPP levels. This loss was reversed by treatment with cell-permeable antioxidants and corresponded with reduced ROS production and enhanced cellular proliferation during supplemental thiamine conditions. siRNA-mediated knockdown of TPK1 directly enhanced basal ROS levels and reduced tumor cell proliferation. These findings suggest that the adaptive regulation of TPK1 may be an essential component in the cellular response to oxidative stress, and that during supplemental thiamine conditions its expression may be exploited by tumor cells for a redox advantage contributing to tumor progression.

5.
Toxicol Appl Pharmacol ; 355: 180-188, 2018 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-30008376

RESUMO

Vitamin B1, or thiamine is a critical enzyme cofactor required for metabolic function and energy production. Thiamine deficiency (TD) is common in various diseases, and results in severe neurological complications due to diminished mitochondrial function, oxidative stress, excitotoxicity and inflammation. These pathological sequelae result in apoptotic cell death in both neurons and astrocytes in distinct regions, in particular the thalamus and mammillary bodies. Comparable histological injuries in patients with hypoxia/ischemia (H/I) have also been described, suggesting a congruency between the cellular responses to these stresses. Analogous to H/I, TD stabilizes and activates Hypoxia Inducible Factor-1α (HIF-1α) even without changes in physiological oxygen levels. However, the mechanism of HIF-1α stabilization in TD is currently unknown. Using a pyruvate assay, we have demonstrated that TD induces pyruvate accumulation in mouse primary astrocytes which correlates to an increase in HIF-1α expression. Additionally, we utilized an enzymatic assay for pyruvate dehydrogenase to demonstrate a reduction in catalytic activity during TD due to lack of available thiamine pyrophosphate cofactor, resulting in the observed pyruvate accumulation. Finally, a pyruvate kinase inhibitor which limited pyruvate accumulation was utilized to demonstrate the role of pyruvate accumulation in HIF-1α stabilization during TD. These results reveal that stabilization of HIF-1α protein in TD centralizes on pyruvate accumulation in mouse primary astrocytes due to metabolic disruption of PDH.


Assuntos
Subunidade alfa do Fator 1 Induzível por Hipóxia/sangue , Piruvatos/metabolismo , Deficiência de Tiamina/sangue , Animais , Astrócitos/efeitos dos fármacos , Astrócitos/metabolismo , Inibidores Enzimáticos/farmacologia , Feminino , Ácido Láctico/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Proteína Oncogênica v-akt/metabolismo , Cultura Primária de Células , Complexo Piruvato Desidrogenase/antagonistas & inibidores , Complexo Piruvato Desidrogenase/sangue , Tiamina Pirofosfato/metabolismo
6.
PLoS One ; 12(10): e0186707, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29045486

RESUMO

Thiamine is an essential enzyme cofactor required for proper metabolic function and maintenance of metabolism and energy production in the brain. In developed countries, thiamine deficiency (TD) is most often manifested following chronic alcohol consumption leading to impaired mitochondrial function, oxidative stress, inflammation and excitotoxicity. These biochemical lesions result in apoptotic cell death in both neurons and astrocytes. Comparable histological injuries in patients with hypoxia/ischemia and TD have been described in the thalamus and mammillary bodies, suggesting a congruency between the cellular responses to these stresses. Consistent with hypoxia/ischemia, TD stabilizes and activates Hypoxia Inducible Factor-1α (HIF-1α) under physiological oxygen levels. However, the role of TD-induced HIF-1α in neurological injury is currently unknown. Using Western blot analysis and RT-PCR, we have demonstrated that TD induces HIF-1α expression and activity in primary mouse astrocytes. We observed a time-dependent increase in mRNA and protein expression of the pro-apoptotic and pro-inflammatory HIF-1α target genes MCP1, BNIP3, Nix and Noxa during TD. We also observed apoptotic cell death in TD as demonstrated by PI/Annexin V staining, TUNEL assay, and Cell Death ELISA. Pharmacological inhibition of HIF-1α activity using YC1 and thiamine repletion both reduced expression of pro-apoptotic HIF-1α target genes and apoptotic cell death in TD. These results demonstrate that induction of HIF-1α mediated transcriptional up-regulation of pro-apoptotic/inflammatory signaling contributes to astrocyte cell death during thiamine deficiency.


Assuntos
Apoptose , Astrócitos/metabolismo , Astrócitos/patologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Deficiência de Tiamina/metabolismo , Deficiência de Tiamina/patologia , Animais , Apoptose/efeitos dos fármacos , Apoptose/genética , Astrócitos/efeitos dos fármacos , Células Cultivadas , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Masculino , Camundongos Endogâmicos C57BL , Reação em Cadeia da Polimerase em Tempo Real , Tiamina/farmacologia , Proteína Supressora de Tumor p53/metabolismo
7.
Neurotoxicology ; 62: 24-29, 2017 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-28495520

RESUMO

Pyrethroids, including permethrin and deltamethrin (DLM), are very widely used of insecticides. It was hypothesized that lower plasma binding and increased blood-brain barrier (BBB) penetration of DLM in immature rats contribute to the higher brain concentrations of DLM and more pronounced neurotoxicity reported in this age group. The left brain of anesthetized adult rats was perfused for 2min via a carotid artery with 1µM 14C-DLM in: 2-5% human serum albumin (HSA); plasma from adult and 15- and 21-d-old rats; and plasma from human donors of: birth-1 week, 1-4 weeks, 4 weeks-1 year, 1-3 years and adults. The fraction of DLM bound and brain uptake of DLM did not vary significantly with the HSA concentration nor with the age of rat or human plasma donors. One, 10 and 50µM 14C-DLM were perfused into the left-brain of anesthetized adult, 15- and 21-d-old rats. DLM deposition in the brain was linear over this range of concentrations and inversely related to age. The results of this investigation indicate that increased BBB permeability in the youngest rats enhances brain deposition of the insecticide. Plasma protein binding of DLM in immature rats and humans is not sufficiently diminished to impact its brain uptake.


Assuntos
Proteínas Sanguíneas/metabolismo , Barreira Hematoencefálica , Encéfalo , Inseticidas/metabolismo , Nitrilas/metabolismo , Piretrinas/metabolismo , Fatores Etários , Albuminas/farmacologia , Animais , Animais Recém-Nascidos , Barreira Hematoencefálica/embriologia , Barreira Hematoencefálica/crescimento & desenvolvimento , Barreira Hematoencefálica/metabolismo , Encéfalo/efeitos dos fármacos , Encéfalo/embriologia , Encéfalo/enzimologia , Encéfalo/metabolismo , Pré-Escolar , Relação Dose-Resposta a Droga , Embrião de Mamíferos , Feminino , Feto , Humanos , Lactente , Recém-Nascido , Inseticidas/farmacocinética , Masculino , Nitrilas/farmacocinética , Gravidez , Ligação Proteica/efeitos dos fármacos , Piretrinas/farmacocinética , Ratos , Ratos Sprague-Dawley
8.
Toxicol Sci ; 155(1): 270-282, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-28031414

RESUMO

High body burdens of polybrominated diphenyl ethers (PBDEs) in infants and young children have led to increased concern over their potential impact on human development. PBDE exposure can alter the expression of genes involved in thyroid homeostasis, including those of ATP-binding cassette (ABC) transporters, which mediate cellular xenobiotic efflux. However, little information exists on how PBDEs interact with ABC transporters such as P-glycoprotein (P-gp) and breast cancer resistance protein (BCRP). The purpose of this study was to evaluate the interactions of 2,2',4,4'-tetrabromodiphenyl ether (BDE-47) and its hydroxylated metabolite 6-OH-BDE-47 with P-gp and BCRP, using human MDR1- and BCRP-expressing membrane vesicles and stably transfected NIH-3T3-MDR1 and MDCK-BCRP cells. In P-gp membranes, BDE-47 did not affect P-gp activity; however, 6-OH-BDE-47 inhibited P-gp activity at low µM concentrations (IC50 = 11.7 µM). In BCRP membranes, BDE-47 inhibited BCRP activity; however, 6-OH-BDE-47 was a stronger inhibitor [IC50 = 45.9 µM (BDE-47) vs. IC50 = 9.4 µM (6-OH-BDE-47)]. Intracellular concentrations of known P-gp and BCRP substrates [(3H)-paclitaxel and (3H)-prazosin, respectively] were significantly higher (indicating less efflux) in NIH-3T3-MDR1 and MDCK-BCRP cells in the presence of 6-OH-BDE-47, but not BDE-47. Collectively, our results indicate that the BDE-47 metabolite 6-OH-BDE-47 is an inhibitor of both P-gp and BCRP efflux activity. These findings suggest that some effects previously attributed to BDE-47 in biological systems may actually be due to 6-OH-BDE-47. Considerations for human exposure are discussed.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/antagonistas & inibidores , Exposição Ambiental , Proteínas de Neoplasias/antagonistas & inibidores , Bifenil Polibromatos/toxicidade , Western Blotting , Cromatografia Líquida , Cromatografia Gasosa-Espectrometria de Massas , Humanos , Espectrometria de Massas em Tandem
9.
Gene ; 595(2): 212-220, 2016 Dec 31.
Artigo em Inglês | MEDLINE | ID: mdl-27743994

RESUMO

Ensuring continuous intracellular supply of thiamine is essential to maintain metabolism. Cellular homeostasis requires the function of the membrane bound thiamine transporters THTR1 and THTR2. In the absence of increased dietary intake of thiamine, varying intracellular levels to meet metabolic demands during pathophysiological stressors, such as hypoxia, requires adaptive regulatory mechanisms to increase thiamine transport capacity. Previous work has established the up-regulation of SLC19A3 (THTR2) gene expression and activity during hypoxic stress through the activity of the hypoxia inducible transcription factor 1 alpha (HIF-1α). However, it is unknown whether HIF-1α acts directly or indirectly to trans-activate expression of SLC19A3. This work utilized the breast cancer cell line BT-474 treated with 1% O2 or a hypoxia chemical mimetic deferoxamine to determine the minimal promoter region of SLC19A3 responsible for hypoxia responsiveness. In silico sequence analysis determined two contiguous hypoxia responsive elements in close proximity to the transcriptional start site of the SLC19A3 gene. Using a HIF-1α transcriptional factor ELISA assay, HIF-1α was capable of binding to a dsDNA construct of the SLC19A3 minimal promoter. Chromatin immunoprecipitation assay established that SP1 was bound to the SLC19A3 minimal promoter region under normoxic conditions. However, HIF-1α binding to the minimal promoter region occurred during hypoxic treatments, while no SP1 binding was observed under these conditions. This work demonstrates the direct binding and activation of SLC19A3 expression by HIF-1α during hypoxic stress, suggesting an important adaptive regulatory role for HIF-1α in maintaining thiamine homeostasis.


Assuntos
Hipóxia Celular/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Linhagem Celular , Núcleo Celular/metabolismo , Regulação da Expressão Gênica , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Proteínas de Membrana Transportadoras/genética , Regiões Promotoras Genéticas , Transporte Proteico , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Ativação Transcricional
10.
Toxicol Lett ; 250-251: 21-8, 2016 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-27016408

RESUMO

Previous pharmacokinetic studies of deltamethrin (DLM) have revealed that brain levels of this highly lipophilic pyrethroid insecticide are only 15-20% of plasma levels. Experiments were performed to assess determinants limiting CNS access including plasma protein binding and the efflux transporter, P-gp. A human brain microvascular endothelial cell line, hCMEC/D3, was utilized as a model in vitro system to evaluate blood-brain barrier (BBB) permeation. Incubation of DLM with a series of human serum albumin (HSA) concentrations showed that unbound (fu) DLM ranged from 80% with 0.01% HSA to ∼20% at the physiologically-relevant 4% HSA. A positive correlation (R=0.987) was seen between fu and cellular uptake. Concentration-dependent uptake of DLM in 0.01% HSA was non-linear and was reduced at 4°C and by the P-gp inhibitor cyclosporine (CSA), indicative of a specific transport process. Cellular accumulation of [(3)H]-paclitaxel, a P-glycoprotein (P-gp) substrate, was increased by CSA but not by DLM, suggesting that DLM is neither a substrate nor an inhibitor of P-gp. The concentration-dependent uptake of DLM from 4% HSA was linear and not significantly impacted by temperature or CSA. In situ brain perfusion studies monitoring brain association of DLM at 0.01% and 4% HSA confirmed the aforementioned in vitro findings. This study demonstrates that brain uptake of DLM under normal physiological conditions appears to be a passive, non-saturable process, limited by the high protein binding of the pyrethroid.


Assuntos
Barreira Hematoencefálica/metabolismo , Células Endoteliais/metabolismo , Inseticidas/metabolismo , Nitrilas/metabolismo , Piretrinas/metabolismo , Albumina Sérica/metabolismo , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Animais , Permeabilidade Capilar , Linhagem Celular , Humanos , Técnicas In Vitro , Cinética , Modelos Lineares , Masculino , Modelos Biológicos , Perfusão , Ligação Proteica , Ratos Sprague-Dawley , Albumina Sérica Humana , Temperatura
11.
Biomed Chromatogr ; 30(1): 35-41, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26120008

RESUMO

A new method for reversed phase HPLC determination of thiamine and its major in vivo phosphorylation products, thiamine monophosphate (TMP) and thiamine pyrophosphate (TPP), was developed using tetrabutylammonium hydroxide as the ion-pairing agent. The separation was performed on a Phenomenex Kinetex EVO C18 column with a gradient of a phosphate-buffered aqueous solution of the ion-pair reagent and methanol. The duty cycle for the assay was 13 min and pyrithiamine was successfully used as the internal standard for the first time in a thiamine HPLC measurement protocol. Detection of the fluorescence derivatives of the analytes as well as the IS allowed for lower detection limits in order to support biological applications in cell culture models. The linearity, sensitivity, specificity, accuracy and precision of the method were evaluated and met the requirements specified by the US Food and Drug Administration. The calibration curves proved to be linear and the method was validated over the range from 1.0-4000 nM for both cells and the media where complete recovery of the analytes was also achieved.


Assuntos
Cromatografia Líquida de Alta Pressão/métodos , Tiamina/análise , Linhagem Celular , Homeostase , Humanos , Tiamina/metabolismo
12.
Toxicol Lett ; 232(1): 37-45, 2015 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-25268938

RESUMO

The human efflux transporter P-glycoprotein (P-gp, MDR1) functions as an important cellular defense system against a variety of xenobiotics; however, little information exists on whether environmental chemicals interact with P-gp. Conazoles provide a unique challenge to exposure assessment because of their use as both pesticides and drugs. Propiconazole is an agricultural pesticide undergoing evaluation by the U.S. Environmental Protection Agency's Endocrine Disruptor Screening Program. In this study, the P-gp interaction of propiconazole and its hydroxylated metabolites were evaluated using MDR1-expressing membrane vesicles and NIH-3T3/MDR1 cells. Membrane vesicle assays demonstrated propiconazole (IC50,122.9µM) and its metabolites (IC50s, 350.8µM, 366.4µM, and 456.3µM) inhibited P-gp efflux of a probe substrate, with propiconazole demonstrating the strongest interaction. P-gp mediated transport of propiconazole in MDR1-expressed vesicles was not detected indicating propiconazole interacts with P-gp as an inhibitor rather than a substrate. In NIH-3T3/MDR1 cells, propiconazole (1 and 10µM) led to decreased cellular resistance (chemosensitization) to paclitaxel, a chemotherapeutic drug and known MDR1 substrate. Collectively, these results have pharmacokinetic and risk assessment implications as P-gp interaction may influence pesticide toxicity and the potential for pesticide-drug interactions.


Assuntos
Interações Medicamentosas , Praguicidas/toxicidade , Triazóis/toxicidade , Subfamília B de Transportador de Cassetes de Ligação de ATP/antagonistas & inibidores , Subfamília B de Transportador de Cassetes de Ligação de ATP/genética , Subfamília B de Transportador de Cassetes de Ligação de ATP/metabolismo , Animais , Biotransformação , Sobrevivência Celular/efeitos dos fármacos , Relação Dose-Resposta a Droga , Humanos , Hidroxilação , Camundongos , Células NIH 3T3 , Paclitaxel/metabolismo , Paclitaxel/toxicidade , Praguicidas/metabolismo , Medição de Risco , Transfecção , Triazóis/metabolismo
13.
Cancer Chemother Pharmacol ; 73(3): 585-94, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24452394

RESUMO

PURPOSE: The dichotomous effect of thiamine supplementation on cancer cell growth is characterized by growth stimulation at low doses and growth suppression at high doses. Unfortunately, how thiamine reduces cancer cell proliferation is currently unknown. Recent focuses on metabolic targets for cancer therapy have exploited the altered regulation of the thiamine-dependent enzyme pyruvate dehydrogenase (PDH). Cancer cells inactivate PDH through phosphorylation by overexpression of pyruvate dehydrogenase kinases (PDKs). Inhibition of PDKs by dichloracetate (DCA) exhibits a growth suppressive effect in many cancers. Recently, it has been shown that the thiamine coenzyme, thiamine pyrophosphate reduces PDK-mediated phosphorylation of PDH. Therefore, the objective of this study was to determine whether high-dose thiamine supplementation reduces cell proliferation through a DCA-like mechanism. METHODS: Cytotoxicity of thiamine and DCA was assessed in SK-N-BE and Panc-1 cancer cell lines. Comparative effects of high-dose thiamine and DCA on PDH phosphorylation were measured by Western blot. The metabolic impact of PDH reactivation was determined by glucose and lactate assays. Changes in the mitochondrial membrane potential, reactive oxygen species (ROS) production, and caspase-3 activation were assessed to characterize the mechanism of action. RESULTS: Thiamine exhibited a lower IC50 value in both cell lines compared with DCA. Both thiamine and DCA reduced the extent of PDH phosphorylation, reduced glucose consumption, lactate production, and mitochondrial membrane potential. High-dose thiamine and DCA did not increase ROS, but increased caspase-3 activity. CONCLUSION: Our findings suggest that high-dose thiamine reduces cancer cell proliferation by a mechanism similar to that described for dichloroacetate.


Assuntos
Ácido Dicloroacético/administração & dosagem , Neuroblastoma/patologia , Neoplasias Pancreáticas/patologia , Tiamina/administração & dosagem , Caspase 3/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Glucose/metabolismo , Humanos , Ácido Láctico/biossíntese , Potencial da Membrana Mitocondrial/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Neuroblastoma/tratamento farmacológico , Neoplasias Pancreáticas/tratamento farmacológico , Fosforilação/efeitos dos fármacos , Complexo Piruvato Desidrogenase/antagonistas & inibidores , Complexo Piruvato Desidrogenase/metabolismo , Espécies Reativas de Oxigênio/metabolismo
14.
Cancer Metab ; 1(1): 16, 2013 Jul 24.
Artigo em Inglês | MEDLINE | ID: mdl-24280319

RESUMO

The resurgence of interest in cancer metabolism has linked alterations in the regulation and exploitation of metabolic pathways with an anabolic phenotype that increases biomass production for the replication of new daughter cells. To support the increase in the metabolic rate of cancer cells, a coordinated increase in the supply of nutrients, such as glucose and micronutrients functioning as enzyme cofactors is required. The majority of co-enzymes are water-soluble vitamins such as niacin, folic acid, pantothenic acid, pyridoxine, biotin, riboflavin and thiamine (Vitamin B1). Continuous dietary intake of these micronutrients is essential for maintaining normal health. How cancer cells adaptively regulate cellular homeostasis of cofactors and how they can regulate expression and function of metabolic enzymes in cancer is underappreciated. Exploitation of cofactor-dependent metabolic pathways with the advent of anti-folates highlights the potential vulnerabilities and importance of vitamins in cancer biology. Vitamin supplementation products are easily accessible and patients often perceive them as safe and beneficial without full knowledge of their effects. Thus, understanding the significance of enzyme cofactors in cancer cell metabolism will provide for important dietary strategies and new molecular targets to reduce disease progression. Recent studies have demonstrated the significance of thiamine-dependent enzymes in cancer cell metabolism. Therefore, this review discusses the current knowledge in the alterations in thiamine availability, homeostasis, and exploitation of thiamine-dependent pathways by cancer cells.

15.
Toxicol Sci ; 136(2): 284-93, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24014652

RESUMO

The effectiveness and widespread use of pyrethroid insecticides has lead to concerns regarding their safety. Human ingestion of these potentially neurotoxic compounds is typically through hand-to-mouth contact or consumption of contaminated foods. A substantial proportion of ingested pyrethroids are eliminated in feces, suggesting that absorption is limited, possibly by the action of the efflux transporter P-glycoprotein (P-gp). We utilized caco-2 cells as a model system for intestinal enterocytes and qualitatively and quantitatively assessed the transport of deltamethrin (DLM), cis-permethrin (CPM), and trans-permethrin (TPM). Caco-2 cell uptake of the P-gp substrate R6G was increased by the P-gp inhibitors cyclosporine A (CSA) and ritonavir but not by DLM, CPM, and TPM. Unexpectedly, CSA and ritonavir significantly reduced the uptake of DLM, CPM, and TPM. Permeability coefficients (P app) and directional flux of DLM, CPM, or TPM were greater in the absorptive than the secretory (efflux) direction when measured across caco-2 monolayers grown on Transwell inserts. When CSA was applied to the monolayers' apical (AP) side, the AP to basolateral (BL) P app was significantly reduced, with no change in the BL to AP P app. Kinetic analysis demonstrated saturable transport kinetics for all 3 pyrethroids. These findings indicate that the cellular uptake of DLM, CPM, and TPM is not limited by P-gp efflux but undergo absorptive influx transport as a contributing mechanism for cellular uptake. However, the overall P app values for DLM, CPM, and TPM are consistent with the low permeability/low absorption compound mannitol, suggesting limited gastrointestinal absorption potential.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Mucosa Intestinal/metabolismo , Nitrilas/farmacocinética , Permetrina/farmacocinética , Piretrinas/farmacocinética , Células CACO-2 , Humanos , Absorção Intestinal , Transporte Proteico , Estereoisomerismo
16.
PLoS One ; 8(8): e71196, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23940717

RESUMO

We evaluated the potential of an investigational histone methylation reversal agent, 3-deazaneplanocin A (DZNep), in improving the chemosensitivity of pancreatic cancer to nucleoside analogs (i.e., gemcitabine). DZNep brought delayed but selective cytotoxicity to pancreatic cancer cells without affecting normal human pancreatic ductal epithelial (HPDE) cells. Co-exposure of DZNep and gemcitabine induced cytotoxic additivity or synergism in both well- and poorly-differentiated pancreatic cell lines by increased apoptosis. In contrast, DZNep exerted antagonism with gemcitabine against HPDE cells with significant reduction in cytotoxicity compared with the gemcitabine-alone regimen. DZNep marginally depended on purine nucleoside transporters for its cytotoxicity, but the transport dependence was circumvented by acyl derivatization. Drug exposure studies revealed that a short priming with DZNep followed by gemcitabine treatment rather than co-treatment of both agents to produce a maximal chemosensitization response in both gemcitabine-sensitive and gemcitabine-resistant pancreatic cancer cells. DZNep rapidly and reversibly decreased trimethylation of histone H3 lysine 27 but increased trimethylation of lysine 9 in an EZH2- and JMJD1A/2C-dependent manner, respectively. However, DZNep potentiation of nucleoside analog chemosensitization was found to be temporally coupled to trimethylation changes in lysine 27 and not lysine 9. Polymeric nanoparticles engineered to chronologically release DZNep followed by gemcitabine produced pronounced chemosensitization and dose-lowering effects. Together, our results identify that an optimized DZNep exposure can presensitize pancreatic cancer cells to anticancer nucleoside analogs through the reversal of histone methylation, emphasizing the promising clinical utilities of epigenetic reversal agents in future pancreatic cancer combination therapies.


Assuntos
Adenosina/análogos & derivados , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Sistemas de Liberação de Medicamentos , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Nucleosídeos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Adenosina/farmacologia , Animais , Antimetabólitos Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/farmacocinética , Células Cultivadas , Metilação de DNA/efeitos dos fármacos , Preparações de Ação Retardada , Desoxicitidina/administração & dosagem , Desoxicitidina/análogos & derivados , Avaliação Pré-Clínica de Medicamentos , Sinergismo Farmacológico , Histona Metiltransferases , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Humanos , Nanopartículas , Neoplasias Pancreáticas/patologia , Xenopus , Gencitabina
17.
J Nutr Biochem ; 24(9): 1616-24, 2013 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-23642734

RESUMO

An increased carbon flux and exploitation of metabolic pathways for the rapid generation of biosynthetic precursors is a common phenotype observed in breast cancer. To support this metabolic phenotype, cancer cells adaptively regulate the expression of glycolytic enzymes and nutrient transporters. However, activity of several enzymes involved in glucose metabolism requires an adequate supply of cofactors. In particular, vitamin B1 (thiamine) is utilized as an essential cofactor for metabolic enzymes that intersect at critical junctions within the glycolytic network. Intracellular availability of thiamine is facilitated by the activity of thiamine transporters and thiamine pyrophosphokinase-1 (TPK-1). Therefore, the objective of this study was to establish if the cellular determinants regulating thiamine homeostasis differ between breast cancer and normal breast epithelia. Employing cDNA arrays of breast cancer and normal breast epithelial tissues, SLC19A2, SLC25A19 and TPK-1 were found to be significantly up-regulated. Similarly, up-regulation was also observed in breast cancer cell lines compared to human mammary epithelial cells. Thiamine transport assays and quantitation of intracellular thiamine and thiamine pyrophosphate established a significantly greater extent of thiamine transport and free thiamine levels in breast cancer cell lines compared to human mammary epithelial cells. Overall, these findings demonstrate an adaptive response by breast cancer cells to increase cellular availability of thiamine.


Assuntos
Neoplasias da Mama/genética , Regulação Neoplásica da Expressão Gênica , Tiamina/farmacologia , Regulação para Cima , Transporte Biológico , Neoplasias da Mama/metabolismo , Linhagem Celular Tumoral , Células Epiteliais/metabolismo , Feminino , Perfilação da Expressão Gênica , Homeostase , Humanos , Células MCF-7 , Proteínas de Membrana Transportadoras/genética , Proteínas de Membrana Transportadoras/metabolismo , Proteínas de Transporte da Membrana Mitocondrial , Análise de Sequência com Séries de Oligonucleotídeos , Reação em Cadeia da Polimerase em Tempo Real , Tiamina Pirofosfoquinase/genética , Tiamina Pirofosfoquinase/metabolismo
18.
Pharm Res ; 30(4): 1050-64, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23224979

RESUMO

PURPOSE: To investigate the role of membrane-associated drug transporters in regulating the intestinal absorption of the HIV-1 protease inhibitor, atazanavir, and assess the potential contribution of these transporters in clinical interactions of atazanavir with other protease inhibitors and tenofovir disoproxil fumarate (TDF). METHODS: Intestinal permeability of atazanavir was investigated in vitro, using the Caco-2 cell line system grown on Transwell inserts, and in situ, by single-pass perfusion of rat intestinal segments, jejunum and ileum, in the absence or presence of standard transporter inhibitors or antiretroviral drugs. RESULTS: Atazanavir accumulation by Caco-2 cells was susceptible to inhibition by P-glycoprotein and organic anion transporting polypeptide (OATP) family inhibitors and several antiretroviral drugs (protease inhibitors, TDF). The secretory flux of atazanavir (basolateral-to-apical Papp) was 11.7-fold higher than its absorptive flux. This efflux ratio was reduced to 1.5-1.7 in the presence of P-glycoprotein inhibitors or ritonavir. P-glycoprotein inhibition also resulted in 1.5-2.5-fold increase in atazanavir absorption in situ. Co-administration of TDF, however, reduced atazanavir intestinal permeability by 13-49%, similar to the effect observed clinically. CONCLUSIONS: Drug transporters such as P-glycoprotein and OATPs regulate intestinal permeability of atazanavir and may contribute to its poor oral bioavailability and drug-drug interactions with other protease inhibitors and TDF.


Assuntos
Membro 1 da Subfamília B de Cassetes de Ligação de ATP/metabolismo , Fármacos Anti-HIV/farmacologia , Fármacos Anti-HIV/farmacocinética , Absorção Intestinal , Oligopeptídeos/farmacologia , Oligopeptídeos/farmacocinética , Transportadores de Ânions Orgânicos/metabolismo , Piridinas/farmacologia , Piridinas/farmacocinética , Membro 1 da Subfamília B de Cassetes de Ligação de ATP/antagonistas & inibidores , Adenina/análogos & derivados , Adenina/farmacologia , Animais , Sulfato de Atazanavir , Células CACO-2 , Interações Medicamentosas , Humanos , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Masculino , Organofosfonatos/farmacologia , Permeabilidade/efeitos dos fármacos , Ratos , Ratos Sprague-Dawley , Inibidores da Transcriptase Reversa/farmacologia , Tenofovir
19.
Int J Vitam Nutr Res ; 83(3): 188-97, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24846908

RESUMO

It is well established that thiamine deficiency results in an excess of metabolic intermediates such as lactate and pyruvate, which is likely due to insufficient levels of cofactor for the function of thiamine-dependent enzymes. When in excess, both pyruvate and lactate can increase the stabilization of the hypoxia-inducible factor 1-alpha (HIF-1α) transcription factor, resulting in the trans-activation of HIF-1α regulated genes independent of low oxygen, termed pseudo-hypoxia. Therefore, the resulting dysfunction in cellular metabolism and accumulation of pyruvate and lactate during thiamine deficiency may facilitate a pseudo-hypoxic state. In order to investigate the possibility of a transcriptional relationship between hypoxia and thiamine deficiency, we measured alterations in metabolic intermediates, HIF-1α stabilization, and gene expression. We found an increase in intracellular pyruvate and extracellular lactate levels after thiamine deficiency exposure to the neuroblastoma cell line SK-N-BE. Similar to cells exposed to hypoxia, there was a corresponding increase in HIF-1α stabilization and activation of target gene expression during thiamine deficiency, including glucose transporter-1 (GLUT1), vascular endothelial growth factor (VEGF), and aldolase A. Both hypoxia and thiamine deficiency exposure resulted in an increase in the expression of the thiamine transporter SLC19A3. These results indicate thiamine deficiency induces HIF-1α-mediated gene expression similar to that observed in hypoxic stress, and may provide evidence for a central transcriptional response associated with the clinical manifestations of thiamine deficiency.


Assuntos
Expressão Gênica/fisiologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/fisiologia , Deficiência de Tiamina/fisiopatologia , Hipóxia Celular/fisiologia , Núcleo Celular/química , Subunidade alfa do Fator 1 Induzível por Hipóxia/análise , Ácido Láctico/metabolismo , Neuroblastoma , Neurônios/ultraestrutura , Ácido Pirúvico/metabolismo
20.
Cancer Biol Ther ; 10(11): 1101-11, 2010 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-20930543

RESUMO

Adaptive responses within hypoxic tumor microenvironments require the altered expression of Solute Carrier (SLC) transporters to maintain nutrient uptake in support of cellular metabolism and biosynthesis. Using a real time PCR array strategy to further characterize changes in transporter expression within a chronic hypoxia breast cancer cell line model (BT474), we have found a 31 fold increase in the expression of the thiamine transporter, SLC19A3. Thus, further investigations into the expression changes of the thiamine transporters, SLC19A2 and SLC19A3, and the role of hypoxia inducible factor-1 alpha (HIF-1α) regulating their expression were conducted. Chronic culturing of BT474 cells in 1% O2 up to 142 days consistently demonstrated a high level of SLC19A3 expression with a mean of approximately 40 fold with no change in SLC19A2. A corresponding 2 fold increase in thiamine uptake over 15 min was measured in chronic hypoxic BT474 cells compared to normoxia. Acute 1% O2 exposure of BT474 cells up to 72 h demonstrated a 7.5 fold increase in SLC19A3 expression. The chemical hypoxia mimetic deferoxamine, resulted in an approximately 70 fold increase in SLC19A3 expression. Stable shRNA knockdown of HIF-1α reduced hypoxia mediated SLC19A3 up-regulation by approximately 3 fold compared to scrambled construct. In conclusion, SLC19A3 transporter expression was observed to be up-regulated under acute, chronic and DFO induced hypoxia. The attenuated increase in SLC19A3 expression after HIF-1α knockdown suggests a role for HIF-1α mediated pathways regulating SLC19A3 gene expression.


Assuntos
Neoplasias da Mama/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Transporte Biológico , Neoplasias da Mama/genética , Neoplasias da Mama/patologia , Hipóxia Celular/fisiologia , Linhagem Celular Tumoral , Feminino , Perfilação da Expressão Gênica , Técnicas de Silenciamento de Genes , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/deficiência , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Proteínas de Membrana Transportadoras/biossíntese , Proteínas de Membrana Transportadoras/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Regulação para Cima
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...