Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 191
Filtrar
1.
Eur J Pharm Biopharm ; 196: 114185, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38280469

RESUMO

As a bioactive saponin derived from the seeds of Ziziphus jujuba Mill. var. spinosa (Bunge) Hu ex H. F. Chow, jujuboside B (JuB) shows great potential in anti-anxiety, anti-depression and improving learning and memory function. However, its oral bioavailability is very poor. In this study, a novel drug-loading nanoparticles system was prepared with polyethylene glycol and polylactic-co-glycolic acid copolymer (PEG-PLGA), and further modified with L-carnitine (LC) to target intestinal organic cation/carnitine transporter 2 (OCTN2) to improve the oral absorption of JuB. Under the optimized preparation conditions, the particle sizes of obtained JuB-PEG-PLGA nanoparticles (B-NPs) and LC modified B-NPs (LC-B-NPs) were 110.67 ± 11.37 nm and 134.00 ± 2.00 nm with the entrapment efficiency (EE%) 73.46 ± 1.26 % and 76.01 ± 2.10 %, respectively. The pharmacokinetics in SD rats showed that B-NPs and LC-B-NPs increased the bioavailability of JuB to 134.33 % and 159.04 % respectively. In Caco-2 cell model, the prepared nanoparticles significantly increased cell uptake of JuB, which verified the pharmacokinetic results. The absorption of LC-B-NPs mainly depended on OCTN2 transporter, and Na+ played an important role. Caveolin and clathrin were involved in the endocytosis of the two nanoparticles. In conclusion, both B-NPs and LC-B-NPs can improve the oral absorption of JuB, and the modification of LC can effectively target the OCTN2 transporter.


Assuntos
Nanopartículas , Poliésteres , Polietilenoglicóis , Saponinas , Humanos , Ratos , Animais , Carnitina/farmacocinética , Células CACO-2 , Ratos Sprague-Dawley , Tamanho da Partícula
2.
Nutrients ; 13(6)2021 May 31.
Artigo em Inglês | MEDLINE | ID: mdl-34073024

RESUMO

L-carnitine (LC) supplementation improves cardiac function in hemodialysis (HD) patients. However, whether reducing LC supplementation affects carnitine kinetics and cardiac function in HD patients treated with LC remains unclear. Fifty-nine HD patients previously treated with intravenous LC 1000 mg per HD session (three times weekly) were allocated to three groups: LC injection three times weekly, once weekly, and placebo, and prospectively followed up for six months. Carnitine fractions were assessed by enzyme cycling methods. Plasma and red blood cell (RBC) acylcarnitines were profiled using tandem mass spectrometry. Cardiac function was evaluated using echocardiography and plasma B-type natriuretic peptide (BNP) levels. Reducing LC administration to once weekly significantly decreased plasma carnitine fractions and RBC-free carnitine levels during the study period, which were further decreased in the placebo group (p < 0.001). Plasma BNP levels were significantly elevated in the placebo group (p = 0.03). Furthermore, changes in RBC (C16 + C18:1)/C2 acylcarnitine ratio were positively correlated with changes in plasma BNP levels (ß = 0.389, p = 0.005). Reducing LC administration for six months significantly decreased both plasma and RBC carnitine levels, while the full termination of LC increased plasma BNP levels; however, it did not influence cardiac function in HD patients.


Assuntos
Carnitina/sangue , Carnitina/farmacocinética , Suplementos Nutricionais , Insuficiência Cardíaca/prevenção & controle , Coração/efeitos dos fármacos , Falência Renal Crônica/terapia , Diálise Renal/métodos , Idoso , Carnitina/administração & dosagem , Relação Dose-Resposta a Droga , Feminino , Seguimentos , Coração/fisiopatologia , Insuficiência Cardíaca/complicações , Humanos , Falência Renal Crônica/sangue , Falência Renal Crônica/complicações , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Método Simples-Cego
3.
AAPS PharmSciTech ; 21(8): 308, 2020 Nov 06.
Artigo em Inglês | MEDLINE | ID: mdl-33156405

RESUMO

L-Carnitine has attracted much more attention especially in the treatment of crucial diseases such as diabetes, regional slimming, and obesity because of its metabolic activities. However, because of its short half-life, low bioavailability, and inability to be stored in the body, frequent dosing is required. In this study, L-carnitine-loaded liposome (lipo-carnitine) and PLGA nanoparticle (nano-carnitine) formulations were prepared and characterized. For lipo-carnitine and nano-carnitine formulations, particle size values were 97.88 ± 2.96 nm and 250.90 ± 6.15 nm; polydispersity index values were 0.35 ± 0.01 and 0.22 ± 0.03; zeta potential values were 6.36 ± 0.54 mV and - 32.80 ± 2.26 mV; and encapsulation efficiency percentage values were 14.26 ± 3.52% and 21.93 ± 4.17%, respectively. Comparative in vitro release studies of novel formulations and solution of L-carnitine revealed that L-carnitine released 90% of its content at the end of 1st hour. On the other hand, lipo-carnitine and nano-carnitine formulations maintained a controlled-release profile for 12 h. The in vitro efficacy of the formulations on cardiac fibroblasts (CFs) was evaluated by metabolomic studies and pathway analysis. Besides the prolonged release, lipo-carnitine/nano-carnitine formulations were also found to be effective on amino acid, carbohydrate, and lipid metabolisms. As a result, innovative nano-formulations were successfully developed as an alternative to conventional preparations which are available on the market.


Assuntos
Carnitina/administração & dosagem , Composição de Medicamentos , Lipossomos , Metabolômica , Copolímero de Ácido Poliláctico e Ácido Poliglicólico/química , Disponibilidade Biológica , Carnitina/farmacocinética , Nanopartículas/química , Tamanho da Partícula
4.
Pharmacotherapy ; 40(9): 913-923, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32688453

RESUMO

OBJECTIVE: The objective of this review is to discuss the therapeutic use and differential treatment response to Levo-carnitine (l-carnitine) treatment in septic shock, and to demonstrate common lessons learned that are important to the advancement of precision medicine approaches to sepsis. We propose that significant interpatient variability in the metabolic response to l-carnitine and clinical outcomes can be used to elucidate the mechanistic underpinnings that contribute to sepsis heterogeneity. METHODS: A narrative review was conducted that focused on explaining interpatient variability in l-carnitine treatment response. Relevant biological and patient-level characteristics considered include genetic, metabolic, and morphomic phenotypes; potential drug interactions; and pharmacokinetics (PKs). MAIN RESULTS: Despite promising results in a phase I study, a recent phase II clinical trial of l-carnitine treatment in septic shock showed a nonsignificant reduction in mortality. However, l-carnitine treatment induces significant interpatient variability in l-carnitine and acylcarnitine concentrations over time. In particular, administration of l-carnitine induces a broad, dynamic range of serum concentrations and measured peak concentrations are associated with mortality. Applied systems pharmacology may explain variability in drug responsiveness by using patient characteristics to identify pretreatment phenotypes most likely to derive benefit from l-carnitine. Moreover, provocation of sepsis metabolism with l-carnitine offers a unique opportunity to identify metabolic response signatures associated with patient outcomes. These approaches can unmask latent metabolic pathways deranged in the sepsis syndrome and offer insight into the pathophysiology, progression, and heterogeneity of the disease. CONCLUSIONS: The compiled evidence suggests there are several potential explanations for the variability in carnitine concentrations and clinical response to l-carnitine in septic shock. These serve as important confounders that should be considered in interpretation of l-carnitine clinical studies and broadly holds lessons for future clinical trial design in sepsis. Consideration of these factors is needed if precision medicine in sepsis is to be achieved.


Assuntos
Carnitina/farmacocinética , Choque Séptico/metabolismo , Administração Intravenosa , Carnitina/administração & dosagem , Relação Dose-Resposta a Droga , Humanos , Medicina de Precisão , Choque Séptico/tratamento farmacológico
5.
Drug Deliv ; 27(1): 575-584, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-32306775

RESUMO

A delivery system based on l-carnitine (LC) conjugated chitosan (CS)-stearic acid polymeric micelles has been developed for improving the oral bioavailability of paclitaxel (PTX) through targeting intestinal organic cation/carnitine transporter 2 (OCTN2). Stearic acid grafted chitosan (CS-SA), as micelle skeleton material, was synthesized by 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide (EDC)-mediated coupling reaction. The PTX-loaded micelles were prepared by solvent evaporation-hydration method, and the ligand LC was conjugated onto the micelle surface by anchoring its derivative stearoyl group to the lipophilic core of micelle. The modified polymeric micelles showed regular spherical shapes with small particle size of 157.1 ± 5.2 nm and high drug loading capacity of 15.96 ± 0.20 wt%, and the micelle stability in water was supported by low critical micelle concentration of 14.31 ± 0.21 µg/ml. The drug-loaded micelles presented a slow and incomplete in vitro release, and the pharmacokinetic studies indicated the micelle carriers increased the relative bioavailability of PTX to 165.8% against the commercial formulation. The enhancement effect on intestinal absorption was also confirmed by the intracellular uptake of Caco-2 cells. The proposed micelle carrier system manifested a prospective tool for oral drug delivery.


Assuntos
Antineoplásicos Fitogênicos/farmacocinética , Carnitina/química , Quitosana/química , Micelas , Paclitaxel/farmacocinética , Ácidos Esteáricos/química , Administração Oral , Animais , Antineoplásicos Fitogênicos/administração & dosagem , Disponibilidade Biológica , Células CACO-2 , Carnitina/administração & dosagem , Carnitina/farmacocinética , Quitosana/administração & dosagem , Quitosana/farmacocinética , Portadores de Fármacos/administração & dosagem , Portadores de Fármacos/síntese química , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Liberação Controlada de Fármacos , Feminino , Humanos , Absorção Intestinal/efeitos dos fármacos , Masculino , Paclitaxel/administração & dosagem , Tamanho da Partícula , Ratos Sprague-Dawley , Membro 5 da Família 22 de Carreadores de Soluto/metabolismo , Ácidos Esteáricos/administração & dosagem , Ácidos Esteáricos/farmacocinética
6.
Blood Purif ; 49(5): 614-621, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-31968345

RESUMO

INTRODUCTION: Among patients regularly undergoing hemodialysis, hypocarnitinaemia often develops as a consequence of inadequate dietary intake, reduced synthesis in the body, and considerable losses during hemodialysis. OBJECTIVES: To evaluate the effects of L-carnitine supplementation on patients with end-stage kidney disease (ESKD) who underwent hemodialysis. METHODS: Thirty-one patients with ESKD, comprising 18 men and 13 women, with a median age of 72 (range 58-89) years, who underwent regular hemodialysis received treatment with L-carnitine for 1 year. The total and free carnitine, acylcarnitine, and amino acids (AA) levels before and after L-carnitine treatment were analyzed, and the blood biochemistry results and clinical profiles of the subjects were compared before and after treatment. RESULTS: The median (interquartile range [IQR]) serum total and free carnitine and acylcarnitine levels significantly increased from 34.5 (28.2-44.3), 20.9 (15.8-27.6), and 14.1 (11.2-17.6) µmol/L, respectively to 407.4 (371.6-493.5), 270.2 (228.3-316.0), and 155.0 (136.1-168.5) µmol/L, respectively, after treatment (all p < 0.001). The median (IQR) blood valine, tyrosine, phenylalanine, and citrulline levels increased from 0.94 (0.80-1.09), 0.45 (0.39-0.55), 0.61 (0.56-0.79), and 1.04 (0.79-1.26) mg/dL, respectively to 1.24 (1.13-1.54), 0.76 (0.62-0.85), 0.90 (0.70-1.04), and 1.22 (0.92-1.39) mg/dL, respectively, following L-carnitine treatment (p < 0.001, p < 0.001, p = 0.002, and p = 0.030, respectively); however, the median (IQR) blood arginine level decreased from 0.20 (0.13-0.24) to 0.09 (0.06-0.14) mg/dL after treatment (p < 0.001). The median (IQR) percentage fractional shortening (41.5 vs. 41.9%; p = 0.012) and left ventricular ejection fraction (65.2 vs. 67.3%; p = 0.036) increased significantly following treatment. CONCLUSIONS: L-Carnitine increased the blood acylcarnitine levels, enhanced fatty acid metabolism, and affected AAs metabolism; this may be beneficial for energy production within the cardiac and skeletal muscles.


Assuntos
Aminoácidos/sangue , Carnitina , Falência Renal Crônica , Diálise Renal , Idoso , Idoso de 80 Anos ou mais , Carnitina/administração & dosagem , Carnitina/farmacocinética , Feminino , Humanos , Falência Renal Crônica/sangue , Falência Renal Crônica/terapia , Masculino , Pessoa de Meia-Idade
7.
Drug Deliv ; 27(1): 170-179, 2020 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31913724

RESUMO

Targeted nanocarriers have shown great promise in drug delivery because of optimized drug behavior and improved therapeutic efficacy. How to improve the targeting efficiency of nanocarriers for the maximum possible drug delivery is a critical issue. Here we developed L-carnitine-conjugated nanoparticles targeting the carnitine transporter OCTN2 on enterocytes for improved oral absorption. As a variable, we introduced various lengths of the polyethylene glycol linker (0, 500, 1000, and 2000) between the nanoparticle surface and the ligand (CNP, C5NP, C10NP and C20NP) to improve the ligand flexibility, and consequently for more efficient interaction with the transporter, to enhance the oral delivery of the cargo load into cells. An increased absorption was observed in cellular uptake in vitro and in intestinal perfusion assay in situ when the polyethylene glycol was introduced to link L-carnitine to the nanoparticles; the highest absorption was achieved with C10NP. In contrast, the linker decreased the absorption efficiency in vivo. As the presence or absence of the mucus layer was the primary difference between in vitro/in situ versus in vivo, the presence of this layer was the likely reason for this differential effect. In summary, the size of the polyethylene glycol linker improved the absorption in vitro and in situ, but interfered with the absorption in vivo. Even though this strategy of increasing the ligand flexibility with the variable size of the polyethylene glycol failed to increase oral absorption in vivo, this approach is likely to be useful for enhanced cellular uptake following intravenous administration of the nanocarriers.


Assuntos
Carnitina/farmacologia , Portadores de Fármacos/química , Nanopartículas/química , Paclitaxel/farmacologia , Membro 5 da Família 22 de Carreadores de Soluto/efeitos dos fármacos , Administração Oral , Animais , Células CACO-2 , Carnitina/administração & dosagem , Carnitina/farmacocinética , Sobrevivência Celular/efeitos dos fármacos , Liberação Controlada de Fármacos , Estabilidade de Medicamentos , Enterócitos , Humanos , Masculino , Paclitaxel/administração & dosagem , Paclitaxel/farmacocinética , Tamanho da Partícula , Polietilenoglicóis/química , Ratos , Ratos Sprague-Dawley
8.
Transfus Med ; 30(3): 215-225, 2020 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-31701585

RESUMO

OBJECTIVES: This study aimed to assess the effects of L-carnitine on oxidative stress in human erythrocytes during storage. BACKGROUND: Using antioxidants as components of blood storage solutions may combat the effects of storage-induced oxidative stress on erythrocytes. METHODS: Blood from male adults was stored at 4 °C for 55 days in citrate phosphate dextrose adenine solution, without L-carnitine (Control) and with L-carnitine as an additive (at concentrations of 10, 30 and 60 mM - Experiments). Every fifth day, erythrocyte markers (morphology, count, haemoglobin, haemolysis and osmotic fragility), antioxidant defences (antioxidant enzymes and total antioxidant capacity) and oxidative stress markers (superoxides, lipid peroxidation and protein oxidation products) were analysed. RESULTS: Oxidative damage was observed in controls (day 25 onwards) and in experiments (day 35 onwards). L-carnitine (10 and 30 mM) protected erythrocytes from damage up to day 35 by maintaining haemoglobin and lipid peroxidation, assisting antioxidant enzymes and increasing antioxidant capacity by elevating sulfhydryls and ascorbic acid. L-carnitine was beneficial in prolonging storage up to 55 days but could not prevent oxidative damage completely in terms of haemolysis and osmotic fragility. CONCLUSIONS: L-carnitine ameliorated oxidative stress, but combinations with other antioxidants may provide comprehensive protection to erythrocytes during storage.


Assuntos
Preservação de Sangue , Carnitina/farmacocinética , Eritrócitos/metabolismo , Peroxidação de Lipídeos/efeitos dos fármacos , Estresse Oxidativo/efeitos dos fármacos , Adulto , Relação Dose-Resposta a Droga , Eritrócitos/citologia , Hemólise/efeitos dos fármacos , Humanos , Masculino , Fragilidade Osmótica/efeitos dos fármacos , Fatores de Tempo
9.
Ren Fail ; 40(1): 196-200, 2018 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-29616582

RESUMO

Carnitine has high dialyzability and is often deficient in dialysis patients. This deficiency is treated by either intravenous (IV) or oral supplementation of carnitine. In this study, the mode of carnitine administration was changed from oral to IV in 17 hemodialysis (HD) patients, and the treatment was discontinued after 1 year. We found that the levels of total carnitine (TC), free-carnitine (FC), and acyl-carnitine (AC) significantly increased after 3 months of switching to IV administration (p < .05). After discontinuation of carnitine administration, the TC, FC, and AC levels decreased before dialysis. The average FC value was maintained at the normal levels until 9 months, but fell below the normal values when measured at the 12th month of discontinuation. In conclusion, carnitine was maintained at significantly high levels despite the smaller dose by IV infusion as compared with that by oral administration. We therefore suggest that our results be considered while determining both the carnitine administration route and the administration period in dialysis patients under clinical settings.


Assuntos
Carnitina/farmacocinética , Falência Renal Crônica/terapia , Diálise Renal/efeitos adversos , Administração Oral , Idoso , Carnitina/administração & dosagem , Carnitina/sangue , Carnitina/deficiência , Feminino , Humanos , Injeções Intravenosas , Falência Renal Crônica/sangue , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Tempo
10.
J Dairy Sci ; 101(3): 2060-2071, 2018 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-29274978

RESUMO

Two studies were designed to evaluate the relative bioavailability of l-carnitine delivered by different methods in dairy cattle. In experiment 1, 4 Holstein heifers were used in a split-plot design to compare ruminally or abomasally infused l-carnitine. The study included 2 main-plot periods, with infusion routes allocated in a crossover design. Within main-plot periods, each of 3 subplot periods consisted of 4-d infusions separated with 4-d rest periods. Subplot treatments were infusion of 1, 3, and 6 g of l-carnitine/d in conjunction with 6 g/d of arabinogalactan given in consideration of eventual product manufacturing. Doses increased within a period to minimize carryover risk. Treatments were solubilized in 4 L of water and delivered in two 10-h infusions daily. Blood was collected before the start of infusion period and on d 4 of each infusion period to obtain baseline and treatment l-carnitine concentrations. There was a dose × route interaction and route effect for increases in plasma carnitine above baseline, with increases above baseline being greater across all dose levels when infused abomasally compared with ruminally. Results demonstrated superior relative bioavailability of l-carnitine when ruminal exposure was physically bypassed. In experiment 2, 56 lactating Holstein cows (143 ± 72 d in milk) were used in 2 cohorts in randomized complete block designs (blocked by parity and milk production) to evaluate 2 rumen-protected products compared with crystalline l-carnitine. Treatments were (1) control, (2) 3 g/d of crystalline l-carnitine (crystalline), (3) 6 g/d of crystalline, (4) 5 g/d of 40COAT (40% coating, 60% l-carnitine), (5) 10 g/d of 40COAT, (6) 7.5 g/d of 60COAT (60% coating, 40% l-carnitine), and (7) 15 g/d of 60COAT. Treatments were top-dressed to diets twice daily. Each cohort used 14-d and included a 6-d baseline measurement period with the final 2 d used for data and sample collection, and an 8-d treatment period with the final 2 d used for data and sample collection. Plasma, urine, and milk samples were analyzed for l-carnitine. Crystalline and 40COAT linearly increased plasma l-carnitine, and 60COAT tended to linearly increase plasma l-carnitine. Total excretion (milk + urine) of l-carnitine averaged 1.52 ± 0.04 g/d in controls, increased linearly with crystalline and 40COAT, and increased quadratically with 60COAT. Crystalline increased plasma l-carnitine and l-carnitine excretion more than 40COAT and 60COAT. In conclusion, preventing ruminal degradation of l-carnitine increased delivery of bioavailable carnitine to cattle, but effective ruminal protection and postruminal bioavailability is challenging.


Assuntos
Abomaso/metabolismo , Carnitina/farmacocinética , Bovinos/metabolismo , Rúmen/metabolismo , Animais , Disponibilidade Biológica , Cápsulas , Carnitina/administração & dosagem , Feminino , Infusões Parenterais/veterinária
11.
Artif Cells Nanomed Biotechnol ; 46(8): 1605-1616, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28974108

RESUMO

Overcoming blood-brain barrier (BBB) and targeting tumor cells are two key steps for glioma chemotherapy. By taking advantage of the specific expression of Na+-coupled carnitine transporter 2 (OCTN2) on both brain capillary endothelial cells and glioma cells, l-carnitine conjugated poly(lactic-co-glycolic acid) nanoparticles (LC-PLGA NPs) were prepared to enable enhanced BBB permeation and glioma-cell targeting. Conjugation of l-carnitine significantly enhanced the uptake of PLGA nanoparticles in the BBB endothelial cell line hCMEC/D3 and the glioma cell line T98G. The uptake was dependent on Na+ and inhibited by the excessive free l-carnitine, suggesting involvement of OCTN2 in the process. In vivo mouse studies showed that LC-PLGA NPs resulted in high accumulation in the brain as indicated by the biodistribution and imaging assays. Furthermore, compared to Taxol and paclitaxel-loaded unmodified PLGA NPs, the drug-loaded LC-PLGA NPs showed improved anti-glioma efficacy in both 2D-cell and 3D-spheroid models. The PEG spacer length of the ligand attached to the nanoparticles was optimized, and the formulation with PEG1000 (LC-1000-PLGA NPs) showed the maximum targeting efficiency. We conclude that l-carnitine-mediated cellular recognition and internalization via OCTN2 significantly facilitate the transcytosis of nanoparticles across BBB and the uptake of nanoparticles in glioma cells, resulting in improved anti-glioma efficacy.


Assuntos
Barreira Hematoencefálica/metabolismo , Carnitina , Sistemas de Liberação de Medicamentos/métodos , Glioblastoma/tratamento farmacológico , Nanopartículas , Proteínas de Neoplasias/metabolismo , Paclitaxel , Membro 5 da Família 22 de Carreadores de Soluto/metabolismo , Barreira Hematoencefálica/patologia , Células CACO-2 , Carnitina/química , Carnitina/farmacocinética , Carnitina/farmacologia , Glioblastoma/metabolismo , Glioblastoma/patologia , Humanos , Nanopartículas/química , Nanopartículas/uso terapêutico , Paclitaxel/química , Paclitaxel/farmacocinética , Paclitaxel/farmacologia , Permeabilidade
12.
Biol Pharm Bull ; 40(12): 2110-2116, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29199236

RESUMO

While it is well known that L-carnitine [3-hydroxy-4-(trimethylazaniumyl)-butanoate] is an essential molecule for ß-oxidation, it provides anti-oxidative effects as well. Since these effects have been observed in photoreceptor cells, the carnitine's intracellular concentration is considered to play a protective role against oxidative damage to those cells. However, even though its high hydrophilicity makes it likely that carnitine import is accomplished via a dedicated host transport system, the specific uptake process into those cells is currently unknown. Therefore, in this study, we sought to identify and characterize photoreceptor cell carnitine uptake transporter(s) utilizing 661W cells as a photoreceptor cell model. The results of our uptake assays showed that carnitine was transported into 661W cells in a saturable manner (Km=5.5 mM), and that the activity was susceptible to extracellular pH and Na+. While these data suggest the involvement of a transporter in 661W cell carnitine uptake, the observed transport profile did not correspond to any of the currently known carnitine transporters such as organic cation/carnitine transporter 1 (Octn1), Octn2, Octn3, B0,+ and Ct2. In fact, in our experiments, the mRNA expressions for such carnitine transporters in 661W cells were consistently very low and the carnitine transporter substrates did not inhibit the uptake activities. Taken as a whole, our results indicate that carnitine is transported into 661W cells in a carrier-mediated manner. However, since its transport modes cannot be fully explained by known carnitine transporters, it is highly likely that photoreceptor cells utilize a unique molecularly-based carnitine uptake system.


Assuntos
Antioxidantes/farmacocinética , Transporte Biológico Ativo/fisiologia , Carnitina/farmacocinética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Células Fotorreceptoras/fisiologia , Animais , Linhagem Celular , Concentração de Íons de Hidrogênio , Degeneração Macular/tratamento farmacológico , Camundongos , Estresse Oxidativo/efeitos dos fármacos , Sódio/metabolismo
13.
Am J Hum Genet ; 101(4): 489-502, 2017 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-28942964

RESUMO

Genome-wide association studies have identified a signal at the SLC22A1 locus for serum acylcarnitines, intermediate metabolites of mitochondrial oxidation whose plasma levels associate with metabolic diseases. Here, we refined the association signal, performed conditional analyses, and examined the linkage structure to find coding variants of SLC22A1 that mediate independent association signals at the locus. We also employed allele-specific expression analysis to find potential regulatory variants of SLC22A1 and demonstrated the effect of one variant on the splicing of SLC22A1. SLC22A1 encodes a hepatic plasma membrane transporter whose role in acylcarnitine physiology has not been described. By targeted metabolomics and isotope tracing experiments in loss- and gain-of-function cell and mouse models of Slc22a1, we uncovered a role of SLC22A1 in the efflux of acylcarnitines from the liver to the circulation. We further validated the impacts of human variants on SLC22A1-mediated acylcarnitine efflux in vitro, explaining their association with serum acylcarnitine levels. Our findings provide the detailed molecular mechanisms of the GWAS association for serum acylcarnitines at the SLC22A1 locus by functionally validating the impact of SLC22A1 and its variants on acylcarnitine transport.


Assuntos
Carnitina/análogos & derivados , Regulação da Expressão Gênica , Fígado/metabolismo , Doenças Metabólicas/genética , Transportador 1 de Cátions Orgânicos/genética , Polimorfismo de Nucleotídeo Único , Alelos , Processamento Alternativo , Animais , Transporte Biológico , Sistemas CRISPR-Cas , Carnitina/sangue , Carnitina/farmacocinética , Células Cultivadas , Estudos de Coortes , Feminino , Estudo de Associação Genômica Ampla , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Doenças Metabólicas/sangue , Doenças Metabólicas/metabolismo , Metabolômica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Transportador 1 de Cátions Orgânicos/antagonistas & inibidores , Transportador 1 de Cátions Orgânicos/metabolismo , Distribuição Tecidual
14.
J Med Chem ; 60(6): 2552-2561, 2017 03 23.
Artigo em Inglês | MEDLINE | ID: mdl-28234466

RESUMO

Novel organic cation transporter 2 (OCTN2, SLC22A5) is responsible for the uptake of carnitine through the intestine and, therefore, might be a promising molecular target for designing oral prodrugs. Poor permeability and rapid metabolism have greatly restricted the oral absorption of gemcitabine. We here describe the design of intestinal OCTN2-targeting prodrugs of gemcitabine by covalent coupling of l-carnitine to its N4-amino group via different lipophilic linkages. Because of the high OCTN2 affinity, the hexane diacid-linked prodrug demonstrated significantly improved stability (3-fold), cellular permeability (15-fold), and oral bioavailability (5-fold), while causing no toxicity as compared to gemcitabine. In addition, OCTN2-targeting prodrugs can simultaneously improve the permeability, solubility, and metabolic stability of gemcitabine. In summary, we present the first evidence that OCTN2 can act as a new molecular target for oral prodrug delivery and, importantly, the linkage carbon chain length is a key factor in modifying the affinity of the substrate for OCTN2.


Assuntos
Antimetabólitos Antineoplásicos/química , Antimetabólitos Antineoplásicos/farmacocinética , Desoxicitidina/análogos & derivados , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Animais , Antimetabólitos Antineoplásicos/metabolismo , Células CACO-2 , Carnitina/química , Carnitina/metabolismo , Carnitina/farmacocinética , Desoxicitidina/química , Desoxicitidina/metabolismo , Desoxicitidina/farmacocinética , Células HEK293 , Humanos , Camundongos , Simulação de Acoplamento Molecular , Pró-Fármacos/metabolismo , Membro 5 da Família 22 de Carreadores de Soluto , Distribuição Tecidual , Gencitabina
15.
Oncol Res Treat ; 40(1-2): 42-45, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28118647

RESUMO

BACKGROUND: Approximately 70% of the patients who receive chemotherapy suffer from fatigue, which lowers their quality of life and also has a negative influence on therapeutic efficacy. Previous studies have suggested a relationship between blood carnitine levels and fatigue. We conducted a prospective observational study to examine the relationship between carnitine pharmacokinetics and chemotherapy-induced fatigue in patients receiving cancer chemotherapy regimens that include cisplatin. PATIENTS AND METHODS: 11 patients receiving chemotherapy including cisplatin (60-80 mg/m2) were included in the study. We performed 24-h urine collections and took blood samples on day 1 (before the initiation of chemotherapy) and days 2, 3, 4, and 8 in order to measure the carnitine concentrations in the serum and urine. These were compared with measures of self-reported fatigue. The primary endpoint was the change in self-reported fatigue subscales from baseline to day 8. RESULTS: Urinary carnitine concentrations differed significantly on days 2 and 3 (p = 0.003). The Functional Assessment of Chronic Illness Therapy-Fatigue scale version 4A score on day 8 indicated significantly higher levels of fatigue as compared to day 1 (p = 0.013). CONCLUSION: This study suggests that there is an association between urinary carnitine levels and self-reported fatigue.


Assuntos
Antineoplásicos/síntese química , Carnitina/farmacocinética , Cisplatino/efeitos adversos , Fadiga/induzido quimicamente , Neoplasias/tratamento farmacológico , Adulto , Idoso , Antineoplásicos/uso terapêutico , Carnitina/urina , Cisplatino/uso terapêutico , Fadiga/sangue , Fadiga/urina , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Estudos Prospectivos , Fatores de Tempo , Microglobulina beta-2/urina
16.
Basic Clin Pharmacol Toxicol ; 120(5): 450-456, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-27983775

RESUMO

Meldonium (3-(2,2,2-trimethylhydrazinium)propionate) is the most potent clinically used inhibitor of organic cation transporter 2 (OCTN2). Inhibition of OCTN2 leads to a decrease in carnitine and acylcarnitine contents in tissues and energy metabolism optimization-related cardioprotective effects. The recent inclusion of meldonium in the World Anti-Doping Agency List of Prohibited Substances and Methods has raised questions about the pharmacokinetics of meldonium and its unusually long elimination time. Therefore, in this study, the rate of meldonium washout after the end of the treatment was tested with and without administration of carnitine, γ-butyrobetaine (GBB) and furosemide to evaluate the importance of competition for OCTN2 transport in mice. Here, we show that carnitine and GBB administration during the washout period effectively stimulated the elimination of meldonium. GBB induced a more pronounced effect on meldonium elimination than carnitine due to the higher affinity of GBB for OCTN2. The diuretic effect of furosemide did not significantly affect the elimination of meldonium, carnitine and GBB. In conclusion, the competition of meldonium, carnitine and GBB for OCTN2-mediated transport determines the pharmacokinetic properties of meldonium. Thus, due to their affinity for OCTN2, GBB and carnitine but not furosemide stimulated meldonium elimination. During long-term treatment, OCTN2-mediated transport ensures a high muscle content of meldonium, while tissue clearance depends on relatively slow diffusion, thus resulting in the unusually long complete elimination period of meldonium.


Assuntos
Betaína/análogos & derivados , Carnitina/administração & dosagem , Metilidrazinas/farmacocinética , Proteínas de Transporte de Cátions Orgânicos/metabolismo , Animais , Betaína/administração & dosagem , Betaína/farmacocinética , Betaína/farmacologia , Transporte Biológico/efeitos dos fármacos , Carnitina/farmacocinética , Carnitina/farmacologia , Furosemida/administração & dosagem , Furosemida/farmacologia , Masculino , Metilidrazinas/farmacologia , Camundongos , Proteínas de Transporte de Cátions Orgânicos/antagonistas & inibidores , Membro 5 da Família 22 de Carreadores de Soluto , Distribuição Tecidual
17.
J Pharm Sci ; 105(10): 3199-3204, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27499337

RESUMO

The long-term administration of valproic acid (VPA) may decrease the plasma concentrations of l-carnitine in epileptic patients. l-Carnitine is essential for the ß-oxidation of fatty acids. The aim of this study is to determine whether endogenous l-carnitine affects the pharmacokinetics of VPA in l-carnitine-deficient (CD) rats. An l-carnitine deficiency was induced in rats using sodium pivalate. The pharmacokinetics of VPA were examined following its intravenous or oral administration to rats. The plasma and urine concentrations of VPA and its metabolites were determined using gas chromatography-mass spectrometry methods. Plasma VPA concentrations were slightly higher in CD rats than in control rats, whereas no significant differences were observed in the area under the curve or mean residence times of VPA between the 2 groups. After i.v. administration, the slope of the elimination phase (k) was significantly higher in CD rats than in control rats (p < 0.01). Some of the ß-oxidation metabolites of VPA in plasma and urine decreased, while the glucuronide metabolites of VPA in urine increased complementarily in CD rats. Based on these results, it was concluded that hypocarnitinemia could affect the pharmacokinetics of VPA.


Assuntos
Carnitina/administração & dosagem , Carnitina/farmacocinética , Ácido Valproico/administração & dosagem , Ácido Valproico/farmacocinética , Administração Intravenosa , Animais , Carnitina/sangue , Interações Medicamentosas/fisiologia , Cromatografia Gasosa-Espectrometria de Massas/métodos , Masculino , Ratos , Ratos Wistar , Ácido Valproico/sangue
19.
Atherosclerosis ; 244: 29-37, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26584136

RESUMO

OBJECTIVE: Dietary l-carnitine can be metabolized by intestinal microbiota to trimethylamine, which is absorbed by the gut and further oxidized to trimethylamine N-oxide (TMAO) in the liver. TMAO plasma levels have been associated with atherosclerosis development in ApoE(-/-) mice. To better understand the mechanisms behind this association, we conducted in vitro and in vivo studies looking at the effect of TMAO on different steps of atherosclerotic disease progression. METHODS: J774 mouse macrophage cells were used to evaluate the effect of TMAO on foam cell formation. Male ApoE(-/-) mice transfected with human cholesteryl ester transfer protein (hCETP) were fed l-carnitine and/or methimazole, a flavin monooxygenase 3 (FMO3) inhibitor that prevents the formation of TMAO. Following 12 week treatment, l-carnitine and TMAO plasma levels, aortic lesion development, and lipid profiles were determined. RESULTS: TMAO at concentrations up to 10-fold the Cmax reported in humans did not affect in vitro foam cell formation. In ApoE(-/-)mice expressing hCETP, high doses of l-carnitine resulted in a significant increase in plasma TMAO levels. Surprisingly, and independently from treatment group, TMAO levels inversely correlated with aortic lesion size in both aortic root and thoracic aorta. High TMAO levels were found to significantly correlate with smaller aortic lesion area. Plasma lipid and lipoprotein levels did not change with treatment nor with TMAO levels, suggesting that the observed effects on lesion area were independent from lipid changes. CONCLUSION: These findings suggest that TMAO slows aortic lesion formation in this mouse model and may have a protective effect against atherosclerosis development in humans.


Assuntos
Aterosclerose/sangue , Carnitina/farmacocinética , Proteínas de Transferência de Ésteres de Colesterol/biossíntese , Metilaminas/sangue , Animais , Apolipoproteínas E/genética , Células Cultivadas , Modelos Animais de Doenças , Progressão da Doença , Humanos , Macrófagos/metabolismo , Macrófagos/patologia , Masculino , Camundongos , Camundongos Knockout
20.
Int J Pharm ; 496(2): 834-41, 2015 Dec 30.
Artigo em Inglês | MEDLINE | ID: mdl-26475971

RESUMO

The organic cation transporters OCT and OCTN have been reported to play a significant role in the cellular uptake of substrates within in vitro lung cells. However, no studies to date have investigated the effect of these transporters upon transepithelial absorption of substrates into the pulmonary circulation. We investigated the contribution of OCT and OCTN transporters to total pulmonary absorption of l-carnitine and the anti-muscarinic drug, ipratropium, across an intact isolated perfused rat lung (IPRL). The results obtained from the IPRL were contrasted with active transport in vitro using three human pulmonary cell lines and primary rat alveolar epithelial cells. Ex-vivo studies showed that OCT/OCTN transporters do not play a role in the overall pulmonary absorption of l-carnitine or ipratropium, as evidenced by the effect of chemical inhibition of these transporters upon pulmonary absorption. In contrast, in vitro studies showed that OCT/OCTN transporters play a significant role in cellular accumulation of substrates with preferential uptake of ipratropium by OCTs, and of l-carnitine uptake by OCTNs. The results show that in vitro uptake studies cannot be predictive of airway to blood absorption in vivo. Nevertheless, localised submucosal pulmonary concentrations of inhaled drugs and their pulmonary pharmacodynamic profiles may be influenced by OCT/OCTN transport activity.


Assuntos
Carnitina/farmacocinética , Ipratrópio/farmacocinética , Pulmão/metabolismo , Proteínas de Transporte de Cátions Orgânicos/fisiologia , Animais , Proteínas de Transporte/fisiologia , Células Cultivadas , Células Epiteliais/metabolismo , Humanos , Proteínas de Membrana/fisiologia , Circulação Pulmonar , Ratos , Membro 5 da Família 22 de Carreadores de Soluto , Proteínas Carreadoras de Solutos , Simportadores
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA