Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Pharmacol ; 15: 1365051, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38572427

RESUMEN

Membrane transporters playing an important role in the passage of drugs, metabolites and nutrients across the membranes of the brain cells have been shown to be involved in pathogenesis of Alzheimer's disease (AD). However, little is known about sex-specific changes in transporter protein expression at the brain in AD. Here, we investigated sex-specific alterations in protein expression of three ATP-binding cassette (ABC) and five solute carriers (SLC) transporters in the prefrontal cortex of a commonly used model of familial AD (FAD), 5xFAD mice. Sensitive liquid chromatography tandem mass spectrometry-based quantitative targeted absolute proteomic analysis was applied for absolute quantification of transporter protein expression. We compared the changes in transporter protein expressions in 7-month-old male and female 5xFAD mice versus sex-matched wild-type mice. The study revealed a significant sex-specific increase in protein expression of ABCC1 (p = 0.007) only in male 5xFAD mice as compared to sex-matched wild-type animals. In addition, the increased protein expression of glucose transporter 1 (p = 0.01), 4F2 cell-surface antigen heavy chain (p = 0.01) and long-chain fatty acid transport protein 1 (p = 0.02) were found only in female 5xFAD mice as compared to sex-matched wild-type animals. Finally, protein expression of alanine/serine/cysteine/threonine transporter 1 was upregulated in both male (p = 0.02) and female (p = 0.002) 5xFAD mice. The study provides important information about sex-specific changes in brain cortical transporter expression in 5xFAD mice, which will facilitate drug development of therapeutic strategies for AD targeting these transporters and drug delivery research.

2.
Mol Neurobiol ; 2024 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-38296900

RESUMEN

Alzheimer's disease (AD) is the most common cause of dementia. Despite intensive research efforts, there are currently no effective treatments to cure and prevent AD. There is growing evidence that dysregulation of iron homeostasis may contribute to the pathogenesis of AD. Given the important role of the transferrin receptor 1 (TfR1) in regulating iron distribution in the brain, as well as in the drug delivery, we investigated its expression in the brain cortex and isolated brain microvessels from female 8-month-old 5xFAD mice mimicking advanced stage of AD. Moreover, we explored the association between the TfR1 expression and the activation of the HIF-1 signaling pathway, as well as oxidative stress and inflammation in 5xFAD mice. Finally, we studied the impact of Aß1-40 and Aß1-42 on TfR1 expression in the brain endothelial cell line hCMEC/D3. In the present study, we revealed that an increase in TfR1 protein levels observed in the brain cortex of 5xFAD mice was associated with activation of the HIF-1 signaling pathway as well as accompanied by oxidative stress and inflammation. Interestingly, incubation of Aß peptides in hCMEC/D3 cells did not affect the expression of TfR1, which supported our findings of unaltered TfR1 expression in the isolated brain microvessels in 5xFAD mice. In conclusion, the study provides important information about the expression of TfR1 in the 5xFAD mouse model and the potential role of HIF-1 signaling pathway in the regulation of TfR1 in AD, which could represent a promising strategy for the development of therapies for AD.

3.
Fluids Barriers CNS ; 20(1): 96, 2023 Dec 19.
Artículo en Inglés | MEDLINE | ID: mdl-38115090

RESUMEN

BACKGROUND: Blood-brain barrier (BBB) models based on primary murine, bovine, and porcine brain capillary endothelial cell cultures have long been regarded as robust models with appropriate properties to examine the functional transport of small molecules. However, species differences sometimes complicate translating results from these models to human settings. During the last decade, brain capillary endothelial-like cells (BCECs) have been generated from stem cell sources to model the human BBB in vitro. The aim of the present study was to establish and characterize a human BBB model using human induced pluripotent stem cell (hiPSC)-derived BCECs from the hIPSC line SBAD0201. METHODS: The model was evaluated using transcriptomics, proteomics, immunocytochemistry, transendothelial electrical resistance (TEER) measurements, and, finally, transport assays to assess the functionality of selected transporters and receptor (GLUT-1, LAT-1, P-gp and LRP-1). RESULTS: The resulting BBB model displayed an average TEER of 5474 ± 167 Ω·cm2 and cell monolayer formation with claudin-5, ZO-1, and occludin expression in the tight junction zones. The cell monolayers expressed the typical BBB markers VE-cadherin, VWF, and PECAM-1. Transcriptomics and quantitative targeted absolute proteomics analyses revealed that solute carrier (SLC) transporters were found in high abundance, while the expression of efflux transporters was relatively low. Transport assays using GLUT-1, LAT-1, and LRP-1 substrates and inhibitors confirmed the functional activities of these transporters and receptors in the model. A transport assay suggested that P-gp was not functionally expressed in the model, albeit antibody staining revealed that P-gp was localized at the luminal membrane. CONCLUSIONS: In conclusion, the novel SBAD0201-derived BBB model formed tight monolayers and was proven useful for studies investigating GLUT-1, LAT-1, and LRP-1 mediated transport across the BBB. However, the model did not express functional P-gp and thus is not suitable for the performance of drug efflux P-gp reletated studies.


Asunto(s)
Barrera Hematoencefálica , Células Madre Pluripotentes Inducidas , Humanos , Animales , Bovinos , Ratones , Porcinos , Barrera Hematoencefálica/metabolismo , Células Madre Pluripotentes Inducidas/fisiología , Línea Celular , Transporte Biológico , Encéfalo/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Células Cultivadas
4.
J Pharm Sci ; 112(9): 2581-2590, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37220829

RESUMEN

Triple negative breast cancer (TNBC) is among the most aggressive and deadly cancer subtypes. Intra-tumoral hypoxia is associated with aggressiveness and drug resistance in TNBC. One of the underlying mechanisms of hypoxia-induced drug resistance is the elevated expression of efflux transporters such as breast cancer resistant protein (ABCG2). In the present study, we investigated the possibility of ameliorating ABCG2-mediated drug resistance in hypoxic TNBC cells by monoacylglycerol lipase (MAGL) inhibition and the consequent downregulation of ABCG2 expression. The effect of MAGL inhibition on ABCG2 expression, function, and efficacy of regorafenib, an ABCG2 substrate was investigated in cobalt dichloride (CoCl2) induced pseudohypoxic TNBC (MDA-MB-231) cells, using quantitative targeted absolute proteomics, qRT-PCR, anti-cancer drug accumulation in the cells, cell invasiveness and resazurin-based cell viability assays. Our results showed that hypoxia-induced ABCG2 expression led to low regorafenib intracellular concentrations, reduced the anti-invasiveness efficacy, and elevated half maximal inhibitory concentration (IC50) of regorafenib in vitro MDA-MB-231 cells. MAGL inhibitor, JJKK048, reduced ABCG2 expression, increased regorafenib cell accumulation, which led to higher regorafenib efficacy. In conclusion, hypoxia-induced regorafenib resistance due to ABCG2 over-expression in TNBC cells can be ameliorated by MAGL inhibition.


Asunto(s)
Neoplasias de la Mama Triple Negativas , Humanos , Neoplasias de la Mama Triple Negativas/tratamiento farmacológico , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Monoacilglicerol Lipasas/metabolismo , Monoacilglicerol Lipasas/farmacología , Línea Celular Tumoral , Hipoxia , Resistencia a Antineoplásicos , Proteínas de Neoplasias/metabolismo
5.
Sci Transl Med ; 15(691): eabq2915, 2023 04 12.
Artículo en Inglés | MEDLINE | ID: mdl-37043557

RESUMEN

Tauopathies are neurodegenerative diseases that are characterized by accumulation of hyperphosphorylated tau protein, higher-order aggregates, and tau filaments. Protein phosphatase 2A (PP2A) is a major tau dephosphorylating phosphatase, and a decrease in its activity has been demonstrated in tauopathies, including Alzheimer's disease. Prolyl oligopeptidase is a serine protease that is associated with neurodegeneration, and its inhibition normalizes PP2A activity without toxicity under pathological conditions. Here, we assessed whether prolyl oligopeptidase inhibition could protect against tau-mediated toxicity in cellular models in vitro and in the PS19 transgenic mouse model of tauopathy carrying the human tau-P301S mutation. We show that inhibition of prolyl oligopeptidase with the inhibitor KYP-2047 reduced tau aggregation in tau-transfected HEK-293 cells and N2A cells as well as in human iPSC-derived neurons carrying either the P301L or tau-A152T mutation. Treatment with KYP-2047 resulted in increased PP2A activity and activation of autophagic flux in HEK-293 cells and N2A cells and in patient-derived iNeurons, as indicated by changes in autophagosome and autophagy receptor markers; this contributed to clearance of insoluble tau. Furthermore, treatment of PS19 transgenic mice for 1 month with KYP-2047 reduced tau burden in the brain and cerebrospinal fluid and slowed cognitive decline according to several behavioral tests. In addition, a reduction in an oxidative stress marker was seen in mouse brains after KYP-2047 treatment. This study suggests that inhibition of prolyl oligopeptidase could help to ameliorate tau-dependent neurodegeneration.


Asunto(s)
Prolil Oligopeptidasas , Tauopatías , Ratones , Humanos , Animales , Células HEK293 , Tauopatías/metabolismo , Proteínas tau/metabolismo , Ratones Transgénicos , Serina Endopeptidasas/metabolismo , Inhibidores Enzimáticos , Modelos Animales de Enfermedad
6.
Pharmaceutics ; 15(2)2023 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-36839686

RESUMEN

Transporter-mediated drug resistance is a major obstacle in anticancer drug delivery and a key reason for cancer drug therapy failure. Membrane solute carrier (SLC) transporters play a crucial role in the cellular uptake of drugs. The expression and function of the SLC transporters can be down-regulated in cancer cells, which limits the uptake of drugs into the tumor cells, resulting in the inefficiency of the drug therapy. In this review, we summarize the current understanding of low-SLC-transporter-expression-mediated drug resistance in different types of cancers. Recent advances in SLC-transporter-targeting strategies include the development of transporter-utilizing prodrugs and nanocarriers and the modulation of SLC transporter expression in cancer cells. These strategies will play an important role in the future development of anticancer drug therapies by enabling the efficient delivery of drugs into cancer cells.

7.
Mol Neurobiol ; 60(2): 732-748, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36367657

RESUMEN

Membrane transporters such as ATP-binding cassette (ABC) and solute carrier (SLC) transporters expressed at the neurovascular unit (NVU) play an important role in drug delivery to the brain and have been demonstrated to be involved in Alzheimer's disease (AD) pathogenesis. However, our knowledge of quantitative changes in transporter absolute protein expression and functionality in vivo in NVU in AD patients and animal models is limited. The study aim was to investigate alterations in protein expression of ABC and SLC transporters in the isolated brain microvessels and brain prefrontal cortices of a widely used model of familial AD, 5xFAD mice (8 months old), using a sensitive liquid chromatography tandem mass spectrometry-based quantitative targeted absolute proteomic approach. Moreover, we examined alterations in brain prefrontal cortical and plasmatic levels of transporter substrates in 5xFAD mice compared to age-matched wild-type (WT) controls. ASCT1 (encoded by Slc1a4) protein expression in the isolated brain microvessels and brain prefrontal cortices of 5xFAD mice was twice higher compared to WT controls (p = 0.01). Brain cortical levels of ASCT1 substrate, serine, were increased in 5xFAD mice compared to WT animals. LAT1 (encoded by Slc7a5) and 4F2hc (encoded by Slc3a2) protein expressions were significantly altered in the isolated brain microvessels of 5xFAD mice compared to WT controls (p = 0.008 and p = 0.05, respectively). Overall, the study provides important information, which is crucial for the optimal use of the 5xFAD mouse model in AD drug development and for investigating novel drug delivery approaches. In addition, the findings of the study shed light on the novel potential mechanisms underlying AD pathogenesis.


Asunto(s)
Enfermedad de Alzheimer , Animales , Ratones , Enfermedad de Alzheimer/patología , Sistemas de Transporte de Aminoácidos/metabolismo , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Ratones Transgénicos , Microvasos/patología , Proteómica/métodos
8.
Pharmaceutics ; 14(11)2022 Nov 10.
Artículo en Inglés | MEDLINE | ID: mdl-36365256

RESUMEN

Cytosolic phospholipase A2 (cPLA2) is an enzyme regulating membrane phospholipid homeostasis and the release of arachidonic acid utilized in inflammatory responses. It represents an attractive target for the treatment of Alzheimer's disease (AD). Previously, we showed that lipopolysaccharide (LPS)-induced systemic inflammation caused abnormal lipid metabolism in the brain of a transgenic AD mouse model (APdE9), which might be associated with potential changes in cPLA2 activity. Here, we investigated changes in cPLA2 expression and activity, as well as the molecular mechanisms underlying these alterations due to chronic LPS administration in the cerebral cortex of female APdE9 mice as compared to saline- and LPS-treated female wild-type mice and saline-treated APdE9 mice. The study revealed the significant effects of genotype LPS treatment on cortical cPLA2 protein expression and activity in APdE9 mice. LPS treatment resulted in nuclear factor kappa-light-chain-enhancer of activated B cells (NFkB) activation in the cortex of APdE9 mice. The gene expressions of inflammation markers Il1b and Tnfa were significantly elevated in the cortex of both APdE9 groups compared to the wild-type groups. The study provides evidence of the elevated expression and activity of cPLA2 in the brain cortex of APdE9 mice after chronic LPS treatment, which could be associated with NFkB activation.

9.
Environ Toxicol Pharmacol ; 96: 104003, 2022 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-36283621

RESUMEN

Membrane transporters are important for maintaining brain homeostasis by regulating the passage of solutes into, out of, and within the brain. Growing evidence suggests neurotoxic effects of air pollution exposure and its contribution to neurodegenerative disorders, including Alzheimer's disease (AD), yet limited knowledge is available on the exact cellular impacts of exposure. This study investigates how exposure to ubiquitous solid components of air pollution, ultrafine particles (UFPs), influence brain homeostasis by affecting protein levels of membrane transporters. Membrane transporters were quantified and compared in brain cortical samples of wild-type and the 5xFAD mouse model of AD in response to subacute exposure to inhaled UFPs. The cortical ASCT1 and ABCB1 transporter levels were elevated in wild-type and 5xFAD mice subjected to a 2-week UFP exposure paradigm, suggesting impairment of brain homeostatic mechanisms. This study provides new insight on the molecular mechanisms underlying adverse effects of air pollution on the brain.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Contaminantes Atmosféricos , Sistema de Transporte de Aminoácidos ASC , Lóbulo Frontal , Material Particulado , Animales , Ratones , Contaminantes Atmosféricos/toxicidad , Contaminantes Atmosféricos/análisis , Tamaño de la Partícula , Material Particulado/toxicidad , Material Particulado/análisis , Sistema de Transporte de Aminoácidos ASC/análisis , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/análisis , Lóbulo Frontal/efectos de los fármacos , Lóbulo Frontal/metabolismo
10.
Neuroscience ; 496: 165-178, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35636730

RESUMEN

Neuroinflammation is an important feature in the pathogenesis and progression of central nervous system (CNS) diseases including Alzheimer's disease (AD). One of the widely used animal models of peripherally induced neuroinflammation and neurodegeneration is a lipopolysaccharide (LPS)-induced inflammation mouse model. An acute LPS administration has been widely used for investigation of inflammation-associated disease and testing inflammation-targeting drug candidates. In the present metabolomic, lipidomic and proteomic study, we investigated short-term effects of systemic inflammation induced by LPS administration on the mouse plasma and brain cortical and hippocampal metabolome, lipidome as well as expression of the brain cortical proteins which were shown to be involved in inflammation-associated CNS diseases. From a global perspective, the hippocampus was more vulnerable to the effects of LPS-induced systemic inflammation than the cortex. In addition, the study revealed several brain region-specific changes in metabolic pathways and lipids, such as statistically significant increase in several cortical and hippocampal phosphatidylcholines/phosphatidylethanolamines, and significantly decreased levels of brain cortical betaine after LPS treatment in mice. Moreover, LPS treatment in mice caused significantly increased protein expression of GluN1 receptor in the brain cortex. The revealed perturbations in the LPS-induced inflammation mouse model may give insight into the mechanisms underlying inflammation-associated CNS diseases. In addition, the finding of the study provide important information about the appropriate use of the model during target validation and drug candidate testing.


Asunto(s)
Lipidómica , Lipopolisacáridos , Animales , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones , Proteómica
11.
Pharm Res ; 39(7): 1415-1455, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35359241

RESUMEN

Limited drug delivery to the brain is one of the major reasons for high failure rates of central nervous system (CNS) drug candidates. The blood-brain barrier (BBB) with its tight junctions, membrane transporters, receptors and metabolizing enzymes is a main player in drug delivery to the brain, restricting the entrance of the drugs and other xenobiotics. Current knowledge about the uptake transporters expressed at the BBB and brain parenchymal cells has been used for delivery of CNS drugs to the brain via targeting transporters. Although many transporter-utilizing (pro)drugs and nanocarriers have been developed to improve the uptake of drugs to the brain, their success rate of translation from preclinical development to humans is negligible. In the present review, we provide a systematic summary of the current progress in development of transporter-utilizing (pro)drugs and nanocarriers for delivery of drugs to the brain. In addition, we applied CNS pharmacokinetic concepts for evaluation of the limitations and gaps in investigation of the developed transporter-utilizing (pro)drugs and nanocarriers. Finally, we give recommendations for a rational development of transporter-utilizing drug delivery systems targeting the brain based on CNS pharmacokinetic principles.


Asunto(s)
Encéfalo , Profármacos , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Fármacos del Sistema Nervioso Central , Sistemas de Liberación de Medicamentos , Humanos , Proteínas de Transporte de Membrana/metabolismo
12.
Neurobiol Dis ; 169: 105741, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35472634

RESUMEN

There is growing evidence that membrane transporters expressed at the blood-brain barrier (BBB) and brain parenchymal cells play an important role in Alzheimer's disease (AD) development and progression. However, quantitative information about changes in transporter protein expression at neurovascular unit cells in AD is limited. Here, we studied the changes in the absolute protein expression of five ATP-binding cassette (ABC) and thirteen solute carrier (SLC) transporters in the isolated brain microvessels and brain cortical tissue of TgF344-AD rats compared to age-matched wild-type (WT) animals using liquid chromatography tandem mass spectrometry based quantitative targeted absolute proteomic analysis. Moreover, sex-specific alterations in transporter expression in the brain cortical tissue of this model were examined. Protein expressions of Abcg2, Abcc1 and FATP1 (encoded by Slc27a1) in the isolated brain microvessels of TgF344-AD rats were 3.1-, 2.0-, 4.3-fold higher compared to WT controls, respectively (p < 0.05). Abcc1 and 4F2hc (encoded by Slc3a2) protein expression was significantly up-regulated in the brain cortical tissue of male TgF344-AD rats compared to male WT rats (p < 0.05). The study provides novel information for the elucidation of molecular mechanisms underlying AD and valuable knowledge about the optimal use of the TgF344-AD rat model in AD drug development and drug delivery research.


Asunto(s)
Enfermedad de Alzheimer , Enfermedad de Alzheimer/metabolismo , Animales , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Femenino , Masculino , Proteínas de Transporte de Membrana , Microvasos/metabolismo , Proteómica/métodos , Ratas
13.
Eur J Pharm Biopharm ; 173: 12-21, 2022 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-35227855

RESUMEN

The blood-brain barrier (BBB) represents one of the biggest hurdles for CNS related drug delivery, preventing permeation of most molecules, and therefore poses a major challenge for researchers in finding effective treatments for CNS diseases. The low permeability of molecules through the BBB is linked on one hand to the extreme tightness by tight junction (TJ) formation limiting the paracellular transport, and on the other hand to the presence of ATP-driven efflux pumps which actively transport unwanted compounds out of the brain. In this study we evaluated the applicability of the immortalized human cell line hCMEC/D3 for ABC transporter studies, focusing on the most expressed ABC transporters at the human BBB: P-glycoprotein (PGP, ABCB1), multidrug resistance protein 4 (MRP4, ABCC4) and breast cancer resistance protein (BCRP, ABCG2). Therefore, a two-step screening method was applied, consisting of a regular uptake assay (96-well format) and bidirectional transport studies, using a transwell system as in vitro simulation of the human BBB. In conclusion, the hCMEC/D3 based in vitro BBB model is well suited to screen drug candidates for ABC transporter interactions on the basis of a regular uptake assay, but in terms of transcellular permeability studies the cell line is limited by a lack of sufficient junctional tightness.


Asunto(s)
Transportadoras de Casetes de Unión a ATP , Barrera Hematoencefálica , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Humanos , Proteínas de Neoplasias/metabolismo
14.
J Pharm Sci ; 110(12): 3953-3962, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34403652

RESUMEN

Alzheimer's disease (AD) is an incurable disease, with complex pathophysiology and a myriad of proteins involved in its development. In this study, we applied quantitative targeted absolute proteomic analysis for investigation of changes in potential AD drug targets, biomarkers, and transporters in cerebral cortices of lipopolysaccharide (LPS)-induced neuroinflammation mouse model, familial AD mice (APdE9) with and without LPS treatment as compared to age-matched wild type (WT) mice. The ABCB1, ABCG2 and GluN1 protein expression ratios between LPS treated APdE9 and WT control mice were 0.58 (95% CI 0.44-0.72), 0.65 (95% CI 0.53-0.77) and 0.61 (95% CI 0.52-0.69), respectively. The protein expression levels of other proteins such as MGLL, COX-2, CytC, ABCC1, ABCC4, SLC2A1 and SLC7A5 did not differ between the study groups. Overall, the study revealed that systemic inflammation can alter ABCB1 and ABCG2 protein expression in brain in AD, which can affect intra-brain drug distribution and play a role in AD development. Moreover, the inflammatory insult caused by peripheral infection in AD may be important factor triggering changes in GluN1 protein expression. However, more studies need to be performed in order to confirm these findings. The quantitative information about the expression of selected proteins provides important knowledge, which may help in the optimal use of the mouse models in AD drug development and better translation of preclinical data to humans.


Asunto(s)
Enfermedad de Alzheimer , Transportadoras de Casetes de Unión a ATP/metabolismo , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Animales , Corteza Cerebral/metabolismo , Modelos Animales de Enfermedad , Inflamación/metabolismo , Ratones , Ratones Transgénicos , Proteómica , Receptores Ionotrópicos de Glutamato/metabolismo
15.
Sci Rep ; 11(1): 13076, 2021 06 22.
Artículo en Inglés | MEDLINE | ID: mdl-34158563

RESUMEN

Peripheral infections followed by systemic inflammation may contribute to the onset of Alzheimer`s disease (AD) and accelerate the disease progression later in life. Yet, the impact of systemic inflammation on the plasma and brain tissue metabolome and lipidome in AD has not been investigated. In this study, targeted metabolomic and untargeted lipidomic profiling experiments were performed on the plasma, cortices, and hippocampi of wild-type (WT) mice and transgenic APdE9 mice after chronic lipopolysaccharide (LPS) treatment, as well as saline-treated APdE9 mice. The lipidome and the metabolome of these mice were compared to saline-treated WT animals. In the brain tissue of all three models, the lipidome was more influenced than the metabolome. The LPS-treated APdE9 mice had the highest number of changes in brain metabolic pathways with significant alterations in levels of lysine, myo-inositol, spermine, phosphocreatine, acylcarnitines and diacylglycerols, which were not observed in the saline-treated APdE9 mice. In the WT mice, the effect of the LPS administration on metabolome and lipidome was negligible. The study provided exciting information about the biochemical perturbations due to LPS-induced inflammation in the transgenic AD model, which can significantly enhance our understanding of the role of systemic inflammation in AD pathogenesis.


Asunto(s)
Precursor de Proteína beta-Amiloide/inmunología , Encéfalo/metabolismo , Inflamación/metabolismo , Enfermedad de Alzheimer/metabolismo , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/genética , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Modelos Animales de Enfermedad , Femenino , Hipocampo/metabolismo , Lipidómica/métodos , Masculino , Metaboloma , Metabolómica/métodos , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Presenilina-1/metabolismo
16.
Neurochem Int ; 147: 105017, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33887377

RESUMEN

The blood-brain barrier represents the major challenge for delivering drugs to the central nervous system (CNS). It separates the blood circulation from the brain tissue, thereby protecting the CNS and maintaining its ion homeostasis. Unfortunately, most drugs are not able to cross this barrier in vivo despite promising in vitro results. One approach to solve this problem is the delivery of drugs via surface modified nanocarrier systems. This review will give an overview on currently tested systems, mainly liposomes and solid nanoparticles and inform about new developments.


Asunto(s)
Transporte Biológico/efectos de los fármacos , Barrera Hematoencefálica/efectos de los fármacos , Portadores de Fármacos/farmacología , Sistemas de Liberación de Medicamentos , Liposomas/farmacología , Animales , Sistemas de Liberación de Medicamentos/métodos , Humanos , Nanopartículas/uso terapéutico
17.
Pharm Res ; 37(5): 88, 2020 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-32377929

RESUMEN

Our growing understanding of membrane transporters and their substrate specificity has opened a new avenue in the field of targeted drug delivery. The L-type amino acid transporter 1 (LAT1) has been one of the most extensively investigated transporters for delivering drugs across biological barriers. The transporter is predominantly expressed in cerebral cortex, blood-brain barrier, blood-retina barrier, testis, placenta, bone marrow and several types of cancer. Its physiological function is to mediate Na+ and pH independent exchange of essential amino acids: leucine, phenylalanine, etc. Several drugs and prodrugs designed as LAT1 substrates have been developed to improve targeted delivery into the brain and cancer cells. Thus, the anti-parkinsonian drug, L-Dopa, the anti-cancer drug, melphalan and the anti-epileptic drug gabapentin, all used in clinical practice, utilize LAT1 to reach their target site. These examples provide supporting evidence for the utility of the LAT1-mediated targeted delivery of the (pro)drug. This review comprehensively summarizes recent advances in LAT1-mediated targeted drug delivery. In addition, the use of LAT1 is critically evaluated and limitations of the approach are discussed.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Transportador de Aminoácidos Neutros Grandes 1/uso terapéutico , Animales , Antineoplásicos/química , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Encéfalo , Sistema Nervioso Central/metabolismo , Portadores de Fármacos , Humanos , Tomografía de Emisión de Positrones
18.
Mol Pharm ; 16(7): 3261-3274, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31180686

RESUMEN

l-Type amino acid transporter 1 (LAT1), selectively expressed at the blood-brain barrier (BBB) and brain parenchymal cells, mediates brain delivery of drugs and prodrugs such as l-dopa and gabapentin. Although knowledge about BBB transport of LAT1-utilizing prodrugs is available, there is a lack of quantitative information about brain intracellular delivery and influence of prodrugs on the transporter's physiological state. We studied the LAT1-mediated intrabrain distribution of a recently developed prodrug of the cyclooxygenase inhibitor ketoprofen as well as its impact on transporter protein expression and function (i.e., amino acid exchange) using brain slice method in mice and rats. The intrabrain distribution of the prodrug was 16 times higher than that of ketoprofen. LAT1 involvement in brain cellular barrier uptake of the prodrug was confirmed, reflected by a higher unbound brain intracellular compared to brain extracellular fluid concentration. The prodrug did not alter LAT1 protein expression and amino acid exchange. Integration of derived parameters with previously performed in vivo pharmacokinetic study using the Combinatory Mapping Approach allowed to estimate the brain extra- and intracellular levels of unbound ketoprofen, prodrug, and released parent drug. The overall efficiency of plasma to brain intracellular delivery of prodrug-released ketoprofen was 11 times higher than after ketoprofen dosing. In summary, this study provides quantitative information supporting the use of the LAT1-mediated prodrug approach for enhanced brain delivery of drugs with intracellular targets.


Asunto(s)
Sistema de Transporte de Aminoácidos y+L/metabolismo , Barrera Hematoencefálica/efectos de los fármacos , Barrera Hematoencefálica/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Descubrimiento de Drogas/métodos , Cetoprofeno/farmacocinética , Profármacos/farmacocinética , Sistema de Transporte de Aminoácidos y+L/antagonistas & inhibidores , Aminoácidos/metabolismo , Animales , Transporte Biológico Activo , Liberación de Fármacos , Imidazoles/farmacología , Cetoprofeno/administración & dosificación , Cetoprofeno/análogos & derivados , Masculino , Ratones , Ratones Endogámicos C57BL , Profármacos/administración & dosificación , Piridinas/farmacología , Ratas , Ratas Sprague-Dawley , Distribución Tisular
19.
Basic Clin Pharmacol Toxicol ; 125(2): 123-132, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30916845

RESUMEN

There is a lack of information about the changes in drug pharmacokinetics and cytochrome P450 (CYP) metabolism after bariatric surgery. Here, we investigated the effects of laparoscopic Roux-en-Y gastric bypass (LRYGB) surgery on pharmacokinetics of nine drugs given simultaneously which may reveal changes in the activities of the main CYPs. Eight obese subjects undergoing LRYGB received an oral cocktail containing nine drugs, substrates of various CYPs: melatonin (CYP1A2), nicotine (CYP2A6), bupropion (CYP2B6), repaglinide (CYP2C8), losartan (CYP2C9), omeprazole (CYP2C19/CYP3A4), dextromethorphan (CYP2D6), chlorzoxazone (CYP2E1) and midazolam (CYP3A). The 6-hours pharmacokinetic profiles in serum and urine of each drug or corresponding metabolite as well as their metabolic ratios were compared before surgery with those at a median 1 year later. LRYGB exerted variable effects on the pharmacokinetics of these drugs. The geometric mean AUC0-6 (90% confidence interval) of melatonin, bupropion, repaglinide, chlorzoxazone and midazolam after LRYGB was 27 (19%-41%), 54 (43%-67%), 44 (29%-66%), 160 (129%-197%) and 74 (62%-90%) of the pre-surgery values, respectively. The pharmacokinetics of losartan, omeprazole and dextromethorphan did not change in response to surgery. Nicotine was not detected in serum, while geometric mean of AUC0-6 of its metabolite, cotinine, increased by 1.7 times after surgery. There were 3.6- and 1.3-fold increases in the AUC ratios of 6-hydroxymelatonin/melatonin and hydroxybupropion/bupropion, respectively. The cocktail revealed multiple pharmacokinetic changes occurring after LRYGB with the greatest effects observed for CYP1A2, CYP2C8 and CYP2E1 substrates. Future studies should be focused on CYP1A2, CYP2A6, CYP2C8 and CYP2B6 to clarify the changes in activities of these enzymes after LRYGB.


Asunto(s)
Derivación Gástrica/efectos adversos , Laparoscopía/efectos adversos , Obesidad Mórbida/cirugía , Farmacocinética , Administración Oral , Adulto , Citocromo P-450 CYP1A2/metabolismo , Citocromo P-450 CYP2C8/metabolismo , Citocromo P-450 CYP2E1/metabolismo , Femenino , Humanos , Masculino , Persona de Mediana Edad
20.
Eur J Pharm Sci ; 129: 99-109, 2019 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-30625368

RESUMEN

Ferulic acid (FA), a natural antioxidant, has displayed some potential benefits against Alzheimer's disease. However, due to its poor blood-brain barrier (BBB) permeation and low bioavailability, the clinical use of FA for the treatment of Alzheimer's disease has been limited. In the present study, we applied an L-type amino acid transporter (LAT1) - mediated prodrug approach to deliver FA into the mouse brain and synthetized three novel LAT1-utilizing prodrugs of FA. We used a previously proposed methodology for the development of transporter-utilizing prodrugs and investigated their cellular uptake via LAT1 in vitro in ARPE-19 cells, BBB permeation using in situ perfusion in mice and pharmacokinetics after a single i.p. injection in mice; and compared the findings to our previous structure-pharmacokinetics relationship analysis of LAT1-utilizing prodrugs. In addition, we evaluated interspecies differences in the bioconversion rate of the ester-based prodrug in mouse and human plasma and liver S9 subcellular fraction. It was found that amide-based prodrugs with an aromatic ring in the promoiety were effectively bound to LAT1 and utilized the transporter for cellular uptake in vitro and crossed the BBB after in situ perfusion in mice. In addition, the amide prodrug with the promoiety directly conjugated in the meta-position to FA was bioconverted to the parent drug in mouse brain. Importantly, the study showed that the analogous ester-based prodrug did bind to LAT1 but did not utilize the transporter for cellular uptake in ARPE-19 cells. However, the presence of an ester linker between the prodrug and the parent drug promoted favorable bioconversion properties in human in comparison to mouse tissues in vitro i.e. the ester prodrug showed higher stability in human plasma (75% of intact prodrug in 5 h) and liver S9 subcellular fraction (181 min) in comparison to mouse plasma (t½ 2.6 min) and liver S9 fraction (t½ 23.3 min), suggesting that ester-based prodrugs may offer potential benefits in humans. In conclusion, switching from an amide to ester linker between the promoiety and the parent drug can affect the bioconversion rate of prodrugs in different species as well as influencing their cellular uptake mechanism. Furthermore, the results demonstrated the effective application of structure-pharmacokinetic relationships and screening methodology for developing LAT1-utilizing prodrugs and highlighted the importance of evaluating the biotransformation of parent drug and prodrugs in different species.


Asunto(s)
Encéfalo/metabolismo , Ácidos Cumáricos/química , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Profármacos/química , Animales , Transporte Biológico/fisiología , Ácidos Cumáricos/administración & dosificación , Ésteres/administración & dosificación , Ésteres/química , Humanos , Hígado/metabolismo , Masculino , Ratones , Profármacos/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...