Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
1.
Biomed Pharmacother ; 165: 115009, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37343435

RESUMO

Tumor cells are able to use glycolysis to produce energy under hypoxic conditions, and even under aerobic conditions, they rely mainly on glycolysis for energy production, the Warburg effect. Conventional tumor therapeutic drugs are unidirectional, lacking in targeting and have limited therapeutic effect. The development of a large number of nanocarriers and targeted glycolysis for the treatment of tumors has been extensively investigated in order to improve the therapeutic efficacy. This paper reviews the research progress of nanocarriers based on targeting key glycolytic enzymes and related transporters, and combines nanocarrier systems with other therapeutic approaches to provide a new strategy for targeted glycolytic treatment of tumors, providing a theoretical reference for achieving efficient targeted treatment of tumors.


Assuntos
Antineoplásicos , Sistemas de Liberação de Fármacos por Nanopartículas , Neoplasias , Efeito Warburg em Oncologia , Sistemas de Liberação de Fármacos por Nanopartículas/administração & dosagem , Sistemas de Liberação de Fármacos por Nanopartículas/farmacologia , Neoplasias/tratamento farmacológico , Efeito Warburg em Oncologia/efeitos dos fármacos , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Hexoquinase/antagonistas & inibidores , Fosfofrutoquinases/antagonistas & inibidores , Antineoplásicos/administração & dosagem , Antineoplásicos/farmacologia , Humanos
2.
Eur J Med Chem ; 229: 114092, 2022 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-34998055

RESUMO

Verinurad (RDEA3170) is a selective URAT1 inhibitor under investigation for the treatment of gout and hyperuricemia. In an effort to further improve the pharmacodynamics/pharmacokinetics of verinurad and to increase the structural diversity, we designed novel verinurad analogs by introducing a linker (e.g. aminomethyl, amino or oxygen) between the naphthalene and the pyridine ring to increase the flexibility. These compounds were synthesized and tested for their in vitro URAT1-inhibitory activity. Most compounds exhibited potent inhibitory activities against URAT1 with IC50 values ranging from 0.24 µM to 16.35 µM. Among them, compound KPH2f exhibited the highest URAT1-inhibitory activity with IC50 of 0.24 µM, comparable to that of verinurad (IC50 = 0.17 µM). KPH2f also inhibited GLUT9 with an IC50 value of 9.37 ± 7.10 µM, indicating the dual URAT1/GLUT9 targeting capability. In addition, KPH2f showed little effects on OAT1 and ABCG2, and thus was unlikely to cause OAT1/ABCG2-mediated drug-drug interactions and/or to neutralize the uricosuric effects of URAT1/GLUT9 inhibitors. Importantly, KPH2f (10 mg/kg) was equally effective in reducing serum uric acid levels and exhibited higher uricosuric effects in a mice hyperuricemia model, as compared to verinurad (10 mg/kg). Furthermore, KPH2f demonstrated favorable pharmacokinetic properties with an oral bioavailability of 30.13%, clearly better than that of verinurad (21.47%). Moreover, KPH2f presented benign safety profiles without causing hERG toxicity, cytotoxicity in vitro (lower than verinurad), and renal damage in vivo. Collectively, these results suggest that KPH2f represents a novel, safe and effective dual URAT1/GLUT9 inhibitor with improved druggabilities and is worthy of further investigation as an anti-hyperuricemic drug candidate.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Hiperuricemia/tratamento farmacológico , Naftalenos/química , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Propionatos/química , Piridinas/química , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP/metabolismo , Humanos , Rim , Naftalenos/toxicidade , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Propionatos/toxicidade , Piridinas/toxicidade , Ácido Úrico/sangue
3.
Acta Pharmacol Sin ; 43(1): 121-132, 2022 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-33767379

RESUMO

Urate transporter 1 (URAT1) and glucose transporter 9 (GLUT9) are important targets for the development of uric acid-lowering drugs. We previously showed that the flexible linkers of URAT1 inhibitors could enhance their potency. In this study we designed and synthesized CDER167, a novel RDEA3710 analogue, by introducing a linker (methylene) between the naphthalene and pyridine rings to increase flexibility, and characterized its pharmacological and pharmacokinetics properties in vitro and in vivo. We showed that CDER167 exerted dual-target inhibitory effects on both URAT1 and GLUT9: CDER167 concentration-dependently inhibited the uptake of [14C]-uric acid in URAT1-expressing HEK293 cells with an IC50 value of 2.08 ± 0.31 µM, which was similar to that of RDEA3170 (its IC50 value was 1.47 ± 0.23 µM). Using site-directed mutagenesis, we demonstrated that CDER167 might interact with URAT1 at S35 and F365. In GLUT9-expressing HEK293T cells, CDER167 concentration-dependently inhibited GLUT9 with an IC50 value of 91.55 ± 15.28 µM, whereas RDEA3170 at 100 µM had no effect on GLUT9. In potassium oxonate-induced hyperuricemic mice, oral administration of CDER167 (10 mg·kg-1 · d-1) for 7 days was more effective in lowering uric acid in blood and significantly promoted uric acid excretion in urine as compared with RDEA3170 (20 mg·kg-1 · d-1) administered. The animal experiment proved the safety of CDER167. In addition, CDER167 displayed better bioavailability than RDEA3170, better metabolic stability and no hERG toxicity at 100 µM. These results suggest that CDER167 deserves further investigation as a candidate antihyperuricemic drug targeting URAT1 and GLUT9.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose , Hiperuricemia , Transportadores de Ânions Orgânicos , Proteínas de Transporte de Cátions Orgânicos , Humanos , Células Cultivadas , Relação Dose-Resposta a Droga , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Células HEK293 , Hiperuricemia/tratamento farmacológico , Hiperuricemia/metabolismo , Estrutura Molecular , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Proteínas de Transporte de Cátions Orgânicos/genética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , RNA Mensageiro/antagonistas & inibidores , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Relação Estrutura-Atividade
4.
Biochimie ; 190: 1-11, 2021 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-34224807

RESUMO

The classical methods for determining glucose uptake rates in living cells involve the use of isotopically labeled 2-deoxy-d-glucose or 3-O-methyl-d-glucose, which enter cells via well-characterized membrane transporters of the SLC2A and SLC5A families, respectively. These classical methods, however, are increasingly being displaced by high-throughput assays that utilize fluorescent analogs of glucose. Among the most commonly used of these analogs are 2-NBDG and 6-NBDG, which contain a bulky 7-nitro-2,1,3-benzoxadiazol-4-yl-amino moiety in place of a hydroxy group on d-glucose. This fluorescent group significantly alters both the size and shape of these molecules compared to glucose, calling into question whether they actually enter cells by the same transport mechanisms. In this study, we took advantage of the well-defined glucose uptake mechanism of L929 murine fibroblasts, which rely exclusively on the Glut1/Slc2a1 membrane transporter. We demonstrate that neither pharmacologic inhibition of Glut1 nor genetic manipulation of its expression has a significant impact on the binding or uptake of 2-NBDG or 6-NBDG by L929 cells, though both approaches significantly impact [3H]-2-deoxyglucose uptake rates. Together these data indicate that 2-NBDG and 6-NBDG can bind and enter mammalian cells by transporter-independent mechanisms, which calls into question their utility as an accurate proxy for glucose transport.


Assuntos
4-Cloro-7-nitrobenzofurazano/análogos & derivados , Desoxiglucose/análogos & derivados , Corantes Fluorescentes/metabolismo , Glucosamina/análogos & derivados , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glucose/metabolismo , 4-Cloro-7-nitrobenzofurazano/metabolismo , 4-Cloro-7-nitrobenzofurazano/farmacocinética , Animais , Transporte Biológico , Linhagem Celular , Desoxiglucose/metabolismo , Desoxiglucose/farmacocinética , Fibroblastos/metabolismo , Corantes Fluorescentes/farmacocinética , Glucosamina/metabolismo , Glucosamina/farmacocinética , Glucose/análogos & derivados , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/genética , Transportador de Glucose Tipo 1/antagonistas & inibidores , Transportador de Glucose Tipo 1/genética , Transportador de Glucose Tipo 1/metabolismo , Humanos , Camundongos
5.
ACS Chem Biol ; 16(8): 1576-1586, 2021 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-34296611

RESUMO

Cancer cells reprogram their metabolism to survive and grow. Small-molecule inhibitors targeting cancer are useful for studying its metabolic pathways and functions and for developing anticancer drugs. Here, we discovered that glutipyran and its derivatives inhibit glycolytic activity and cell growth in human pancreatic cancer cells. According to proteomic profiling of glutipyran-treated cells using our ChemProteoBase, glutipyran was clustered within the group of endoplasmic reticulum (ER) stress inducers that included glycolysis inhibitors. Glutipyran inhibited glucose uptake and suppressed the growth of various cancer cells, including A431 cells that express glucose transporter class I (GLUT1) and DLD-1 GLUT1 knockout cells. When cotreated with the mitochondrial respiration inhibitor metformin, glutipyran exhibited a synergistic antiproliferative effect. Metabolome analysis revealed that glutipyran markedly decreased most metabolites of the glycolytic pathway and the pentose phosphate pathway. Glutipyran significantly suppressed tumor growth in a xenograft mouse model of pancreatic cancer. These results suggest that glutipyran acts as a broad-spectrum GLUT inhibitor and reduces cancer cell growth.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Piranos/uso terapêutico , Animais , Antineoplásicos/síntese química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Ensaios de Seleção de Medicamentos Antitumorais , Sinergismo Farmacológico , Feminino , Glucose/metabolismo , Glicólise/efeitos dos fármacos , Humanos , Metabolômica , Metformina/uso terapêutico , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteômica , Piranos/síntese química , Piranos/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
6.
Food Funct ; 12(18): 8274-8287, 2021 Sep 20.
Artigo em Inglês | MEDLINE | ID: mdl-34180933

RESUMO

Accumulating evidence has shown that chronic injection of D-galactose (D-gal) can mimic natural ageing and induce liver and kidney injury. Previous studies showed that D-gal increased uric acid (UA) levels in mice. The increase in UA levels caused inflammation, accelerated oxidative stress, and aggravated liver and kidney injury. Oxidative stress and inflammation play vital roles in the ageing process. Therefore, reducing the levels of UA in ageing mice improved liver and kidney injury. Glucose transporter 9 (GLUT9) is responsible for the reabsorption of UA in the body, and its inhibition helps downregulate UA levels. The present study investigated the UA-lowering activity of the GLUT9 inhibitor resveratrol (RSV) using the patch clamping technique established in our laboratory in vitro. This research is the first study to demonstrate that RSV effectively inhibits UA uptake via GLUT9 (IC50 = 68.77 µM) in vitro. An in vivo study was also performed to investigate the possible protective effect of RSV on D-gal-induced liver and kidney injury. RSV significantly reduced serum UA levels via the downregulation of GLUT9 mRNA and protein expression and promoted the excretion of excess UA through urine. Biochemical analysis showed that RSV significantly downregulated abnormal increases in serum levels of alanine aminotransferase (ALT), aspartate aminotransferase (AST), blood urea nitrogen (BUN) and creatinine (CRE) caused by long-term D-gal treatment, which effectively improved pathological damage, increased superoxide dismutase (SOD) activity and decreased the content of malondialdehyde (MDA) in the liver and kidneys. RSV also downregulated the expression of the inflammatory cytokines, interleukin IL-6, IL-1ß and tumor necrosis factor (TNF)-α in the liver and kidneys of ageing mice. Our findings provide new insights into the treatment strategies for ageing-induced liver and kidney injury and reveal a new mechanism of RSV-induced reduction in UA levels in ageing individuals.


Assuntos
Envelhecimento/efeitos dos fármacos , Galactose/toxicidade , Regulação da Expressão Gênica/efeitos dos fármacos , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Resveratrol/farmacologia , Ácido Úrico/metabolismo , Animais , Sobrevivência Celular/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/prevenção & controle , Células Epiteliais/efeitos dos fármacos , Feminino , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/genética , Rim/efeitos dos fármacos , Túbulos Renais/citologia , Fígado/efeitos dos fármacos , Masculino , Camundongos , Estrutura Molecular , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Resveratrol/química
7.
Phytomedicine ; 87: 153585, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-34044255

RESUMO

BACKGROUND: Hyperuricemia (HUA) is characterized by abnormal serum uric acid (UA) levels and demonstrated to be involved in renal injury leading to hyperuricemic nephropathy (HN). Apigenin (API), a flavonoid naturally present in tea, berries, fruits, and vegetables, exhibits various biological functions, such as antioxidant and anti-inflammatory activity. PURPOSE: To investigate the effect of API treatment in HN and to reveal its underlying mechanisms. METHODS: The mice with HN were induced by potassium oxonate intraperitoneally and orally administered for two weeks. The effects of API on renal function, inflammation, fibrosis, and uric acid (UA) metabolism in mice with HN were evaluated. The effects of API on urate transporters were further examined in vitro. RESULTS: The mice with HN exhibited abnormal renal urate excretion and renal dysfunction accompanied by increased renal inflammation and fibrosis. In contrast, API reduced the levels of serum UA, serum creatinine (CRE), blood urea nitrogen (BUN) and renal inflammatory factors in mice with HN. Besides, API ameliorated the renal fibrosis via Wnt/ß-catenin pathway suppression. Furthermore, API potently promoted urinary UA excretion and inhibited renal urate transporter 1 (URAT1) and glucose transporter 9 (GLUT9) in mice with HN. In vitro, API competitively inhibited URAT1 and GLUT9 in a dose-dependent manner, with IC50 values of 0.64 ± 0.14 µM and 2.63 ± 0.69 µM, respectively. CONCLUSIONS: API could effectively attenuate HN through co-inhibiting UA reabsorption and Wnt/ß-catenin pathway, and thus it might be a potential therapy to HN.


Assuntos
Apigenina/farmacologia , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Hiperuricemia/tratamento farmacológico , Nefropatias/tratamento farmacológico , Transportadores de Ânions Orgânicos/antagonistas & inibidores , Animais , Apigenina/administração & dosagem , Creatinina/sangue , Relação Dose-Resposta a Droga , Fibrose , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Células HEK293 , Humanos , Hiperuricemia/induzido quimicamente , Hiperuricemia/fisiopatologia , Nefropatias/metabolismo , Nefropatias/patologia , Masculino , Camundongos , Nefrite/tratamento farmacológico , Nefrite/patologia , Transportadores de Ânions Orgânicos/genética , Transportadores de Ânions Orgânicos/metabolismo , Ácido Oxônico/toxicidade , Ácido Úrico/sangue , Ácido Úrico/metabolismo , Via de Sinalização Wnt/efeitos dos fármacos , beta Catenina/metabolismo
8.
Molecules ; 26(9)2021 Apr 27.
Artigo em Inglês | MEDLINE | ID: mdl-33925555

RESUMO

Patients with hematologic malignancies require intensive therapies, including high-dose chemotherapy. Antimetabolite-methotrexate (MTX) has been used for many years in the treatment of leukemia and in lymphoma patients. However, the lack of MTX specificity causes a significant risk of morbidity, mortality, and severe side effects that impairs the quality of patients' life. Therefore, novel targeted therapies based on the malignant cells' common traits have become an essential treatment strategy. Glucose transporters have been found to be overexpressed in neoplastic cells, including hematologic malignancies. In this study, we biologically evaluated a novel glucose-methotrexate conjugate (Glu-MTX) in comparison to a free MTX. The research aimed to assess the effectiveness of Glu-MTX on chosen human lymphoma and leukemia cell lines. Cell cytotoxicity was verified by MTT viability test and flow cytometry. Moreover, the cell cycle and cellular uptake of Glu-MTX were evaluated. Our study reveals that conjugation of methotrexate with glucose significantly increases drug uptake and results in similar cytotoxicity of the synthesized compound. Although the finding has been confined to in vitro studies, our observations shed light on a potential therapeutic approach that increases the selectivity of chemotherapeutics and can improve leukemia and lymphoma patients' outcomes.


Assuntos
Antimetabólitos Antineoplásicos/farmacologia , Linfoma não Hodgkin/tratamento farmacológico , Metotrexato/farmacologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/tratamento farmacológico , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Glucose/farmacologia , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/genética , Humanos , Imunoconjugados/farmacologia , Linfoma não Hodgkin/genética , Linfoma não Hodgkin/patologia , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patologia
9.
J Med Chem ; 64(8): 4450-4461, 2021 04 22.
Artigo em Inglês | MEDLINE | ID: mdl-33819035

RESUMO

Overexpression of glucose transporters (GLUTs) in colorectal cancer cells is associated with 5-fluorouracil (1, 5-FU) resistance and poor clinical outcomes. We designed and synthesized a novel GLUT-targeting drug conjugate, triggered by glutathione in the tumor microenvironment, that releases 5-FU and GLUTs inhibitor (phlorizin (2) and phloretin (3)). Using an orthotopic colorectal cancer mice model, we showed that the conjugate exhibited better antitumor efficacy than 5-FU, with much lower exposure of 5-FU during treatment and without significant side effects. Our study establishes a GLUT-targeting theranostic incorporating a disulfide linker between the targeting module and cytotoxic payload as a potential antitumor therapy.


Assuntos
Antineoplásicos/química , Inibidores Enzimáticos/química , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Animais , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Antineoplásicos/uso terapêutico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Neoplasias Colorretais/induzido quimicamente , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Modelos Animais de Doenças , Estabilidade de Medicamentos , Inibidores Enzimáticos/metabolismo , Inibidores Enzimáticos/farmacologia , Inibidores Enzimáticos/uso terapêutico , Fluoruracila/uso terapêutico , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Meia-Vida , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Floretina/química , Floretina/metabolismo , Floretina/uso terapêutico , Florizina/química , Florizina/metabolismo , Florizina/uso terapêutico , Relação Estrutura-Atividade , Distribuição Tecidual
10.
Hum Cell ; 34(2): 634-643, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-33454890

RESUMO

Mammalian cells take in D-glucose as an essential fuel as well as a carbon source. In contrast, L-glucose, the mirror image isomer of D-glucose, has been considered merely as a non-transportable/non-metabolizable control for D-glucose. We have shown that 2-[N-(7-Nitrobenz-2-oxa-1,3-diazol-4-yl)amino]-2-deoxy-D-glucose (2-NBDG), a D-glucose analogue combining a fluorophore NBD at the C-2 position, is useful as a tracer for monitoring D-glucose uptake through glucose transporters (GLUTs) into mammalian cells. To more precisely evaluate the stereoselectivity of 2-NBDG uptake, we developed an L-glucose analogue 2-NBDLG, the mirror-image isomer of 2-NBDG. Interestingly, 2-NBDLG was taken up into mouse insulinoma MIN6 cells showing nuclear heterogeneity, a cytological feature of malignancy, while remaining MIN6 cells only exhibited a trace amount of 2-NBDLG uptake. The 2-NBDLG uptake into MIN6 cells was abolished by phloretin, but persisted under blockade of major mammalian glucose transporters. Unfortunately, however, no such uptake could be detected in other tumor cell lines. Here we demonstrate that human osteosarcoma U2OS cells take in 2-NBDLG in a phloretin-inhibitable manner. The uptake of 2-NBDG, and not that of 2-NBDLG, into U2OS cells was significantly inhibited by cytochalasin B, a potent GLUT inhibitor. Phloretin, but neither phlorizin, an inhibitor of sodium-glucose cotransporter (SGLT), nor a large amount of D/L-glucose, blocked the 2-NBDLG uptake. These results suggest that a phloretin-inhibitable, non-GLUT/non-SGLT, possibly non-transporter-mediated yet unidentified mechanism participates in the uptake of the fluorescent L-glucose analogue in two very different tumor cells, the mouse insulinoma and the human osteosarcoma cells.


Assuntos
4-Cloro-7-nitrobenzofurazano/análogos & derivados , Neoplasias Ósseas/metabolismo , Desoxiglucose/análogos & derivados , Glucose/metabolismo , Osteossarcoma/metabolismo , Floretina/farmacologia , 4-Cloro-7-nitrobenzofurazano/metabolismo , Animais , Citocalasina B/farmacologia , Desoxiglucose/metabolismo , Depressão Química , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Humanos , Insulinoma/metabolismo , Isomerismo , Camundongos , Neoplasias Pancreáticas/metabolismo , Inibidores do Transportador 2 de Sódio-Glicose/farmacologia , Células Tumorais Cultivadas
11.
Planta Med ; 87(9): 709-715, 2021 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-33511623

RESUMO

Reduction of intestinal glucose absorption might result from either delayed carbohydrate digestion or blockage of glucose transporters. Previously, oxyresveratrol was shown to inhibit α-glucosidase, but its effect on glucose transporters has not been explored. The present study aimed to assess oxyresveratrol-induced inhibition of the facilitative glucose transporter 2 and the active sodium-dependent glucose transporter 1. An aqueous extract of Artocarpus lacucha, Puag Haad, which is oxyresveratrol-enriched, was also investigated. Glucose transport was measured by uptake into Caco-2 cells through either glucose transporter 2 or sodium-dependent glucose transporter 1 according to the culture conditions. Oxyresveratrol (40 to 800 µM) dose-dependently reduced glucose transport, which appeared to inhibit both glucose transporter 2 and sodium-dependent glucose transporter 1. Puag Haad at similar concentrations also inhibited these transporters but with greater efficacy. Oxyresveratrol and Puag Haad could help reduce postprandial hyperglycemic peaks, which are considered to be most damaging in diabetics.


Assuntos
Artocarpus , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Extratos Vegetais/farmacologia , Estilbenos/farmacologia , Artocarpus/química , Células CACO-2 , Glucose , Humanos
12.
SLAS Discov ; 26(3): 450-459, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32844721

RESUMO

Glucose transporter 9 (GLUT9), which transports urate in an electrogenic and voltage-dependent manner, plays an important role in the maintenance of normal blood uric acid/urate levels. In the present study, we established a cell model based on the single-electrode patch-clamp technique for characterization of GLUT9 and explored the inhibitory effects of benzobromarone (BM) and probenecid (PB) on urate-induced currents in mouse GLUT9a (mGLUT9a)-expressing HEK-293T cells. The results showed that uric acid, rather than glucose perfusion, led to a rapid and large outward current by mGLUT9a in dose-, voltage-, and pH-dependent manners. BM prominently and irreversibly inhibited the uric acid-induced currents through mGLUT9a, and PB weakly and reversibly inhibited mGLUT9a. We found that depletion of K+ in the external solution significantly strengthened the blockade of BM on mGLUT9a. In addition, an enhanced inhibitory rate of BM was detected when the pH of the external solution was changed from 7.4 to 5.5, indicating that BM functions optimally in an acidic environment. In conclusion, the combination of the established cell model with patch-clamp techniques first revealed the function properties of GLUT9 inhibitors and may provide potential benefits to the study of GLUT9 inhibitors as antihyperuricemic or antigout agents.


Assuntos
Benzobromarona/farmacologia , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Supressores da Gota/farmacologia , Técnicas de Patch-Clamp/métodos , Probenecid/farmacologia , Ácido Úrico/metabolismo , Animais , Transporte Biológico/efeitos dos fármacos , Expressão Gênica , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Células HEK293 , Humanos , Concentração de Íons de Hidrogênio , Potenciais da Membrana/efeitos dos fármacos , Potenciais da Membrana/fisiologia , Camundongos , Naftalenos/farmacologia , Plasmídeos/química , Plasmídeos/metabolismo , Potássio/metabolismo , Propionatos/farmacologia , Piridinas/farmacologia , Transgenes
13.
FEBS J ; 288(12): 3784-3798, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-32654397

RESUMO

The pervasion of three daily meals and snacks is a relatively new introduction to our shared experience and is coincident with an epidemic rise in obesity and cardiometabolic disorders of overnutrition. The past two decades have yielded convincing evidence regarding the adaptive, protective effects of calorie restriction (CR) and intermittent fasting (IF) against cardiometabolic, neurodegenerative, proteostatic, and inflammatory diseases. Yet, durable adherence to intensive lifestyle changes is rarely attainable. New evidence now demonstrates that restricting carbohydrate entry into the hepatocyte by itself mimics several key signaling responses and physiological outcomes of IF and CR. This discovery raises the intriguing proposition that targeting hepatocyte carbohydrate transport to mimic fasting and caloric restriction can abate cardiometabolic and perhaps other fasting-treatable diseases. Here, we review the metabolic and signaling fates of a hepatocyte carbohydrate, identify evidence to target the key mediators within these pathways, and provide rationale and data to highlight carbohydrate transport as a broad, proximal intervention to block the deleterious sequelae of hepatic glucose and fructose metabolism.


Assuntos
Hepatócitos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Moduladores de Transporte de Membrana/uso terapêutico , Síndrome Metabólica/prevenção & controle , Obesidade/prevenção & controle , Hipernutrição/prevenção & controle , Animais , Transporte Biológico/efeitos dos fármacos , Restrição Calórica/métodos , Metabolismo dos Carboidratos/efeitos dos fármacos , Metabolismo dos Carboidratos/genética , Modelos Animais de Doenças , Jejum/metabolismo , Expressão Gênica , Glucose/antagonistas & inibidores , Glucose/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/genética , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Hepatócitos/metabolismo , Hepatócitos/patologia , Humanos , Fígado/metabolismo , Fígado/patologia , Síndrome Metabólica/genética , Síndrome Metabólica/metabolismo , Síndrome Metabólica/patologia , Terapia de Alvo Molecular/métodos , Obesidade/genética , Obesidade/metabolismo , Obesidade/patologia , Hipernutrição/genética , Hipernutrição/metabolismo , Hipernutrição/patologia
14.
Biochim Biophys Acta Rev Cancer ; 1874(2): 188457, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-33096154

RESUMO

Cancer research of the Warburg effect, a hallmark metabolic alteration in tumors, focused attention on glucose metabolism whose targeting uncovered several agents with promising anticancer effects at the preclinical level. These agents' monotherapy points to their potential as adjuvant combination therapy to existing standard chemotherapy in human trials. Accordingly, several studies on combining glucose transporter (GLUT) inhibitors with chemotherapeutic agents, such as doxorubicin, paclitaxel, and cytarabine, showed synergistic or additive anticancer effects, reduced chemo-, radio-, and immuno-resistance, and reduced toxicity due to lowering the therapeutic doses required for desired chemotherapeutic effects, as compared with monotherapy. The combinations have been specifically effective in treating cancer glycolytic phenotypes, such as pancreatic and breast cancers. Even combining GLUT inhibitors with other glycolytic inhibitors and energy restriction mimetics seems worthwhile. Though combination clinical trials are in the early phase, initial results are intriguing. The various types of GLUTs, their role in cancer progression, GLUT inhibitors, and their anticancer mechanism of action have been reviewed several times. However, utilizing GLUT inhibitors as combination therapeutics has received little attention. We consider GLUT inhibitors agents that directly affect glucose transporters by binding to them or indirectly alter glucose transport by changing the transporters' expression level. This review mainly focuses on summarizing the effects of various combinations of GLUT inhibitors with other anticancer agents and providing a perspective on the current status.


Assuntos
Antineoplásicos/uso terapêutico , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Neoplasias/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Ensaios Clínicos como Assunto , Sinergismo Farmacológico , Glicólise/efeitos dos fármacos , Humanos , Estrutura Molecular , Neoplasias/metabolismo
15.
Biol Pharm Bull ; 43(11): 1653-1659, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32863294

RESUMO

Hyperuricemia is mainly the result of relative underexcretion of urate. Urate is mainly eliminated by kidney and several important transporters expressed on the membrane of renal tubular cells involved in urate excretion. Olsalazine sodium was screened from 3167 authorized small compounds/drugs, targeting xanthine oxidoreductase. In previous study, we reported that olsalazine sodium significantly reduced the serum urate levels, and the anti-hyperuricemic activity linked with inhibiting urate formation by reducing the activity of xanthine oxidoreductase. The current research aimed to assess olsalazine sodium renal urate excretion and likely molecular mechanism. The results showed that administration of olsalazine sodium 5.0 mg/kg decreased the levels of serum urate in hyperuricemic rats, and noticeably improved the fractional excretion of urate and urate clearance, exhibiting an uricosuric action. Moreover, olsalazine sodium (2.5, 5.0, 10.0 mg/kg) reduced the level of blood urea nitrogen in rats. Further study showed that olsalazine sodium reduced the mRNA expression of urate reabsorptive transporter glucose transporter 9 (GLUT9), increased the mRNA expression of urate secretory transporters, organic anion transporter 1 (OAT1), OAT3 and type 1 sodium-dependent phosphate transporter (NPT1) as well as the protein expression of OAT3 in the kidney in hyperuricemic mice. In conclusion, olsalazine sodium exhibited a promotion of urate excretion in kidney by increasing the expression of OAT3.


Assuntos
Ácidos Aminossalicílicos/farmacologia , Hiperuricemia/tratamento farmacológico , Transportadores de Ânions Orgânicos Sódio-Independentes/agonistas , Eliminação Renal/efeitos dos fármacos , Ácido Úrico/metabolismo , Ácidos Aminossalicílicos/uso terapêutico , Animais , Nitrogênio da Ureia Sanguínea , Creatinina/sangue , Creatinina/urina , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Humanos , Hiperuricemia/sangue , Hiperuricemia/fisiopatologia , Hiperuricemia/urina , Túbulos Renais Proximais/efeitos dos fármacos , Túbulos Renais Proximais/metabolismo , Túbulos Renais Proximais/fisiopatologia , Masculino , Camundongos , Proteínas de Transporte de Monossacarídeos/antagonistas & inibidores , Proteínas de Transporte de Monossacarídeos/metabolismo , Proteína 1 Transportadora de Ânions Orgânicos/agonistas , Proteína 1 Transportadora de Ânions Orgânicos/metabolismo , Transportadores de Ânions Orgânicos Sódio-Independentes/metabolismo , Ratos , Ratos Sprague-Dawley , Eliminação Renal/fisiologia , Reabsorção Renal/efeitos dos fármacos , Reabsorção Renal/fisiologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/agonistas , Proteínas Cotransportadoras de Sódio-Fosfato Tipo I/metabolismo , Ácido Úrico/sangue , Ácido Úrico/urina
16.
Bioorg Med Chem Lett ; 30(18): 127406, 2020 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-32736210

RESUMO

Glucose transporters (GLUTs) facilitate glucose uptake and are overexpressed in most cancer cells. Inhibition of glucose transport has been shown to be an effective method to slow the growth of cancer cells both in vitro and in vivo. We have previously reported on the anticancer activity of an ester derived glucose uptake inhibitor. Due to the hydrolytic instability of the ester linkage we have prepared a series of isosteres of the ester moiety. Of all of the isosteres prepared, the amine linkage showed the most promise. Several additional analogues of the amine-linked compounds were also prepared to improve the overall activity.


Assuntos
Antineoplásicos/síntese química , Ésteres/síntese química , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Glucose/metabolismo , Amidas/química , Aminas/química , Antineoplásicos/farmacologia , Metabolismo dos Carboidratos , Linhagem Celular Tumoral , Permeabilidade da Membrana Celular , Ensaios de Seleção de Medicamentos Antitumorais , Ésteres/farmacologia , Glicólise/efeitos dos fármacos , Humanos , Fosforilação/efeitos dos fármacos , Ácidos Ftálicos/química , Relação Estrutura-Atividade , Sulfonas/química , Sulfóxidos/química
17.
Eur J Pharm Sci ; 154: 105512, 2020 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-32801003

RESUMO

Cancer is a heterogeneous disease, and its treatment requires the identification of new ways to thwart tumor cells. Amongst such emerging targets are glucose transporters (GLUTs, SLC2 family), which are overexpressed by almost all types of cancer cells; their inhibition provides a strategy to disrupt tumor metabolism selectively, leading to antitumor effects. Here, novel thiazolidinedione (TZD) derivatives were designed, synthesized, characterized, and evaluated for their GLUT1, GLUT4, and GLUT5 inhibitory potential, followed by in-vitro cytotoxicity determination in leukemic cell lines. Compounds G5, G16, and G17 inhibited GLUT1, with IC50 values of 5.4 ± 1.3, 26.6 ± 1.8, and 12.6 ± 1.2 µM, respectively. G17 was specific for GLUT1, G16 inhibited GLUT4 (IC50 = 21.6 ± 4.5 µM) comparably but did not affect GLUT5. The most active compound, G5, inhibited all three GLUT types, with GLUT4 IC50 = 9.5 ± 2.8 µM, and GLUT5 IC50 = 34.5 ± 2.4 µM. Docking G5, G16, and G17 to the inward- and outward-facing structural models of GLUT1 predicted ligand binding affinities consistent with the kinetic inhibition data and implicated E380 and W388 of GLUT1 vs. their substitutions in GLUT5 (A388 and A396, respectively) in inhibitor preference for GLUT1. G5 inhibited the proliferation of leukemia CEM cells at low micromolar range (IC50 = 13.4 µM) while being safer for normal blood cells. Investigation of CEM cell cycle progression after treatment with G5 showed that cells accumulated in the G2/M phase. Flow cytometric apoptosis studies revealed that compound G5 induced both early and late-stage apoptosis in CEM cells.


Assuntos
Proteínas Facilitadoras de Transporte de Glucose , Tiazolidinedionas , Apoptose , Linhagem Celular , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Transportador de Glucose Tipo 1/antagonistas & inibidores , Hipoglicemiantes , Tiazolidinedionas/farmacologia
19.
J Med Chem ; 63(10): 5201-5211, 2020 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-32282207

RESUMO

Aerobic glycolysis, originally identified by Warburg as a hallmark of cancer, has recently been implicated in immune cell activation and growth. Glucose, the starting material for glycolysis, is transported through the cellular membrane by a family of glucose transporters (GLUTs). Therefore, targeting glucose transporters to regulate aerobic glycolysis is an attractive approach to identify potential therapeutic agents for cancers and autoimmune diseases. Herein, we describe the discovery and optimization of a class of potent, orally bioavailable inhibitors of glucose transporters, targeting both GLUT1 and GLUT3.


Assuntos
Descoberta de Drogas/métodos , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glucose/metabolismo , Animais , Antineoplásicos/química , Antineoplásicos/metabolismo , Antineoplásicos/farmacologia , Células CACO-2 , Relação Dose-Resposta a Droga , Descoberta de Drogas/tendências , Glicólise/efeitos dos fármacos , Glicólise/fisiologia , Humanos , Camundongos , Microssomos Hepáticos/efeitos dos fármacos , Microssomos Hepáticos/metabolismo , Ratos
20.
Am J Physiol Regul Integr Comp Physiol ; 318(1): R38-R48, 2020 01 01.
Artigo em Inglês | MEDLINE | ID: mdl-31596114

RESUMO

Astrocytes generate robust cytoplasmic calcium signals in response to reductions in extracellular glucose. This calcium signal, in turn, drives purinergic gliotransmission, which controls the activity of catecholaminergic (CA) neurons in the hindbrain. These CA neurons are critical to triggering glucose counter-regulatory responses (CRRs) that, ultimately, restore glucose homeostasis via endocrine and behavioral means. Although the astrocyte low-glucose sensor involvement in CRR has been accepted, it is not clear how astrocytes produce an increase in intracellular calcium in response to a decrease in glucose. Our ex vivo calcium imaging studies of hindbrain astrocytes show that the glucose type 2 transporter (GLUT2) is an essential feature of the astrocyte glucosensor mechanism. Coimmunoprecipitation assays reveal that the recombinant GLUT2 binds directly with the recombinant Gq protein subunit that activates phospholipase C (PLC). Additional calcium imaging studies suggest that GLUT2 may be connected to a PLC-endoplasmic reticular-calcium release mechanism, which is amplified by calcium-induced calcium release (CICR). Collectively, these data help outline a potential mechanism used by astrocytes to convert information regarding low-glucose levels into intracellular changes that ultimately regulate the CRR.


Assuntos
Astrócitos/fisiologia , Cálcio/metabolismo , Proteínas Facilitadoras de Transporte de Glucose/metabolismo , Glucose/metabolismo , Rombencéfalo/citologia , Fosfolipases Tipo C/metabolismo , Anilidas/farmacologia , Animais , Antioxidantes/farmacologia , Compostos de Boro/farmacologia , Cálcio/farmacologia , Dantroleno/farmacologia , Estrenos/farmacologia , Proteínas Facilitadoras de Transporte de Glucose/antagonistas & inibidores , Florizina/farmacologia , Pró-Fármacos , Pirrolidinonas/farmacologia , Quercetina/farmacologia , Ratos , Ratos Long-Evans , Fosfolipases Tipo C/antagonistas & inibidores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA