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










Base de datos
Tipo de estudio
Intervalo de año de publicación
1.
Eur J Pharm Sci ; 195: 106661, 2024 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-38052257

RESUMEN

Temozolomide (TMZ) a DNA alkylating agent, is the standard-of-care for brain tumors, such as glioblastoma multiforme (GBM). Although the physicochemical and pharmacokinetic properties of TMZ, such as chemical stability and the ability to cross the blood-brain barrier (BBB), have been questioned in the past, the acquired chemoresistance has been the main limiting factor of long-term clinical use of TMZ. In the present study, an L-type amino acid transporter 1 (LAT1)-utilizing prodrug of TMZ (TMZ-AA, 6) was prepared and studied for its cellular accumulation and cytotoxic properties in human squamous cell carcinoma, UT-SCC-28 and UT-SCC-42B cells, and TMZ-sensitive human glioma, U-87MG cells that expressed functional LAT1. TMZ-AA 6 accumulated more effectively than TMZ itself into those cancer cells that expressed LAT1 (UT-SCC-42B). However, this did not correlate with decreased viability of treated cells. Indeed, TMZ-AA 6, similarly to TMZ itself, required adjuvant inhibitor(s) of DNA-repair systems, O6-methylguanine-DNA methyl transferase (MGMT) and base excision repair (BER), as well as active DNA mismatch repair (MMR), for maximal growth inhibition. The present study shows that improving the delivery of this widely-used methylating agent is not the main barrier to improved chemotherapy, although utilizing a specific transporter overexpressed at the BBB or glioma cells can have targeting advantages. To obtain a more effective anticancer prodrug, the compound design focus should shift to altering the major DNA alkylation site or inhibiting DNA repair systems.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Profármacos , Humanos , Temozolomida/farmacología , Temozolomida/uso terapéutico , Antineoplásicos Alquilantes/farmacología , Dacarbazina/farmacología , Resistencia a Antineoplásicos , Reparación del ADN , Glioblastoma/tratamiento farmacológico , Glioma/tratamiento farmacológico , Neoplasias Encefálicas/tratamiento farmacológico , ADN , Profármacos/farmacología , Profármacos/uso terapéutico , Línea Celular Tumoral
2.
Chem Biol Interact ; 388: 110833, 2024 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-38101600

RESUMEN

Many chemotherapeutic drugs suffer from multidrug resistance (MDR). Efflux transporters, namely ATP-binding cassettes (ABCs), that pump the drugs out of the cancer cells comprise one major reason behind MDR. Therefore, ABC inhibitors have been under development for ages, but unfortunately, without clinical success. In the present study, an l-type amino acid transporter 1 (LAT1)-utilizing derivative of probenecid (PRB) was developed as a cancer cell-targeted efflux inhibitor for P-glycoprotein (P-gp), breast cancer resistant protein (BCRP) and/or several multidrug resistant proteins (MRPs), and its ability to increase vinblastine (VBL) cellular accumulation and apoptosis-inducing effects were explored. The novel amino acid derivative of PRB (2) increased the VBL exposure in triple-negative human breast cancer cells (MDA-MB-231) and human glioma cells (U-87MG) by 10-68 -times and 2-5-times, respectively, but not in estrogen receptor-positive human breast cancer cells (MCF-7). However, the combination therapy had greater cytotoxic effects in MCF-7 compared to MDA-MB-231 cells due to the increased oxidative stress recorded in MCF-7 cells. The metabolomic study also revealed that compound 2, together with VBL, decreased the transport of those amino acids essential for the biosynthesis of endogenous anti-oxidant glutathione (GSH). Moreover, the metabolic differences between the outcomes of the studied breast cancer cell lines were explained by the distinct expression profiles of solute carriers (SLCs) that can be concomitantly inhibited. Therefore, attacking several SLCs simultaneously to change the nutrient environment of cancer cells can serve as an adjuvant therapy to other chemotherapeutics, offering an alternative to ABC inhibitors.


Asunto(s)
Antineoplásicos , Neoplasias de la Mama , Humanos , Femenino , Vinblastina/farmacología , Vinblastina/metabolismo , Vinblastina/uso terapéutico , Probenecid/farmacología , Probenecid/uso terapéutico , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Proteínas de Neoplasias/metabolismo , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Apoptosis , Estrés Oxidativo , Aminoácidos/metabolismo , Resistencia a Antineoplásicos , Línea Celular Tumoral
3.
Mol Pharm ; 20(2): 1331-1346, 2023 02 06.
Artículo en Inglés | MEDLINE | ID: mdl-36688491

RESUMEN

Membrane transporters are the key determinants of the homeostasis of endogenous compounds in the cells and their exposure to drugs. However, the substrate specificities of distinct transporters can overlap. In the present study, the interactions of l-type amino acid transporter 1 (LAT1)-utilizing prodrugs with sodium-coupled neutral amino acid transporter 2 (SNAT2) were explored. The results showed that the cellular uptake of LAT1-utilizing prodrugs into a human breast cancer cell line, MCF-7 cells, was mediated via SNATs as the uptake was increased at higher pH (8.5), decreased in the absence of sodium, and inhibited in the presence of unselective SNAT-inhibitor, (α-(methylamino)isobutyric acid, MeAIB). Moreover, docking the compounds to a SNAT2 homology model (inward-open conformation) and further molecular dynamics simulations and the subsequent trajectory and principal component analyses confirmed the chemical features supporting the interactions of the studied compounds with SNAT2, which was found to be the main SNAT expressed in MCF-7 cells.


Asunto(s)
Sistemas de Transporte de Aminoácidos Neutros , Profármacos , Humanos , Profármacos/química , Células MCF-7 , Sistemas de Transporte de Aminoácidos , Sodio
4.
Pharmaceutics ; 14(6)2022 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-35745806

RESUMEN

Membrane transporters have a crucial role in compounds' brain drug delivery. They allow not only the penetration of a wide variety of different compounds to cross the endothelial cells of the blood-brain barrier (BBB), but also the accumulation of them into the brain parenchymal cells. Solute carriers (SLCs), with nearly 500 family members, are the largest group of membrane transporters. Unfortunately, not all SLCs are fully characterized and used in rational drug design. However, if the structural features for transporter interactions (binding and translocation) are known, a prodrug approach can be utilized to temporarily change the pharmacokinetics and brain delivery properties of almost any compound. In this review, main transporter subtypes that are participating in brain drug disposition or have been used to improve brain drug delivery across the BBB via the prodrug approach, are introduced. Moreover, the ability of selected transporters to be utilized in intrabrain drug delivery is discussed. Thus, this comprehensive review will give insights into the methods, such as computational drug design, that should be utilized more effectively to understand the detailed transport mechanisms. Moreover, factors, such as transporter expression modulation pathways in diseases that should be taken into account in rational (pro)drug development, are considered to achieve successful clinical applications in the future.

5.
Chimia (Aarau) ; 76(5): 454-459, 2022 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-38069717

RESUMEN

The expression of membrane transporter is often altered in cancer cells compared to their corresponding healthy cells. Since these proteins, classified into solute carriers (SLCs) and ATP-binding cassettes (ABCs), can carry not only endogenous compounds, nutrients, and metabolites, but also drugs across the cell membranes, they have a crucial role in drug exposure and clinical outcomes of chemotherapeutics. Curiously, up-regulation of SLCs can be exploited to deliver chemotherapeutics, their prodrugs, and diagnostic radio-tracers to gain cancer cell-selective targeting, as exemplified with L-type amino acid transporter 1 (LAT1). SLCs can also be inhibited to limit the nutrient uptake of cancer cells and thus, cell growth and proliferation. Furthermore, LAT1 can be utilized to deliver ABC-inhibitors selectively into the cancer cells to block the efflux of other chemotherapeutics suffering from acquired or intrinsic efflux transport-related multidrug resistance (MDR). Taking into account the current literature, compounds that can affect transporter up- or down-regulation of transporters in a cancer cell-selective manner could be a valuable tool and promising chemotherapy form in the future.

6.
Chem Biol Interact ; 352: 109795, 2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-34953865

RESUMEN

Metformin, an oral anti-diabetic drug, has attracted scientific attention due to its anti-cancer effects. This biguanide exerts preventive effects against cancer, and interferes with cancer-promoting signaling pathways at the cellular level. However, the direct cytotoxic or anti-proliferative effect of the drug is observed at very high concentrations, often exceeding 5-10 mM. This paper presents the synthesis of eight novel sulfonamide-based biguanides with improved cellular uptake in two breast cancer cell lines (MCF-7 and MDA-MB-231), and evaluates their effects on cancer cell growth. The synthesized sulfonamide-based analogues of metformin (1-5) were efficiently taken up in MCF-7 and MDA-MB-231 cells, and were characterized by stronger cytotoxic properties than those of metformin. Generally, compounds were more effective in MCF-7 than in MDA-MB-231. Compound 2, with an n-octyl chain, was the most active molecule with IC50 = 114.0 µmol/L in MCF-7 cells. The cytotoxicity of compound 2 partially results from its ability to induce early and late apoptosis. Increased intracellular reactive oxygen species (ROS) production and reduced mitochondrial membrane potential suggest that compound 2 promotes mitochondrial dysfunction and activates the mitochondrial-associated apoptosis-signaling pathway. In addition, compound 2 was also found to arrest cell cycle in the G0/G1 and G2/M phase and significantly inhibit cancer cell migration. In conclusion, this study supports the hypothesis that improved transporter-mediated cellular uptake of potential drug molecule is accompanied by its increased cytotoxicity. Therefore, compound 2 is a very good example of how chemical modification of a biguanide scaffold can affect its biological properties and improve anti-neoplastic potential.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Metformina/análogos & derivados , Antineoplásicos/síntesis química , Antineoplásicos/química , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ensayos de Selección de Medicamentos Antitumorales , Eritrocitos/efectos de los fármacos , Eritrocitos/patología , Femenino , Humanos , Células MCF-7 , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Metformina/química , Metformina/farmacocinética , Mitocondrias/efectos de los fármacos , Especies Reactivas de Oxígeno/metabolismo , Relación Estructura-Actividad , Sulfonamidas/síntesis química , Sulfonamidas/química , Sulfonamidas/farmacología
7.
Int J Mol Sci ; 22(14)2021 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-34299347

RESUMEN

Efflux transporters, namely ATP-binding cassette (ABC), are one of the primary reasons for cancer chemoresistance and the clinical failure of chemotherapy. Ganciclovir (GCV) is an antiviral agent used in herpes simplex virus thymidine kinase (HSV-TK) gene therapy. In this therapy, HSV-TK gene is delivered together with GCV into cancer cells to activate the phosphorylation process of GCV to active GCV-triphosphate, a DNA polymerase inhibitor. However, GCV interacts with efflux transporters that are responsible for the resistance of HSV-TK/GCV therapy. In the present study, it was explored whether GCV and its more lipophilic derivative (1) could inhibit effluxing of another chemotherapeutic, methotrexate (MTX), out of the human breast cancer cells. Firstly, it was found that the combination of GCV and MTX was more hemocompatible than the corresponding combination with compound 1. Secondly, both GCV and compound 1 enhanced the cellular accumulation of MTX in MCF-7 cells, the MTX exposure being 13-21 times greater compared to the MTX uptake alone. Subsequently, this also reduced the number of viable cells (41-56%) and increased the number of late apoptotic cells (46-55%). Moreover, both GCV and compound 1 were found to interact with breast cancer resistant protein (BCRP) more effectively than multidrug-resistant proteins (MRPs) in these cells. Since the expression of BCRP was higher in MCF-7 cells than in MDA-MB-231 cells, and the cellular uptake of GCV and compound 1 was smaller but increased in the presence of BCRP-selective inhibitor (Fumitremorgin C) in MCF-7 cells, we concluded that the improved apoptotic effects of higher MTX exposure were raised mainly from the inhibition of BCRP-mediated efflux of MTX. However, the effects of GCV and its derivatives on MTX metabolism and the quantitative expression of MTX metabolizing enzymes in various cancer cells need to be studied more thoroughly in the future.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Apoptosis/efectos de los fármacos , Neoplasias de la Mama/tratamiento farmacológico , Ganciclovir/farmacología , Metotrexato/farmacología , Proteínas de Neoplasias/metabolismo , Antivirales/farmacología , Neoplasias de la Mama/metabolismo , Línea Celular Tumoral , Femenino , Células Endoteliales de la Vena Umbilical Humana , Humanos , Células MCF-7 , Proteínas Asociadas a Resistencia a Múltiples Medicamentos/metabolismo
8.
Int J Mol Sci ; 22(11)2021 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-34073245

RESUMEN

Metformin, apart from its glucose-lowering properties, has also been found to demonstrate anti-cancer properties. Anti-cancer efficacy of metformin depends on its uptake in cancer cells, which is mediated by plasma membrane monoamine transporters (PMAT) and organic cation transporters (OCTs). This study presents an analysis of transporter mediated cellular uptake of ten sulfonamide-based derivatives of metformin in two breast cancer cell lines (MCF-7 and MDA-MB-231). Effects of these compounds on cancer cell growth inhibition were also determined. All examined sulfonamide-based analogues of metformin were characterized by greater cellular uptake in both MCF-7 and MDA-MB-231 cells, and stronger cytotoxic properties than those of metformin. Effective intracellular transport of the examined compounds in MCF-7 cells was accompanied by high cytotoxic activity. For instance, compound 2 with meta-methyl group in the benzene ring inhibited MCF-7 growth at micromolar range (IC50 = 87.7 ± 1.18 µmol/L). Further studies showed that cytotoxicity of sulfonamide-based derivatives of metformin partially results from their ability to induce apoptosis in MCF-7 and MDA-MB-231 cells and arrest cell cycle in the G0/G1 phase. In addition, these compounds were found to inhibit cellular migration in wound healing assay. Importantly, the tested biguanides are more effective in MCF-7 cells at relatively lower concentrations than in MDA-MB-231 cells, which proves that the effectiveness of transporter-mediated accumulation in MCF-7 cells is related to biological effects, including MCF-7 cell growth inhibition, apoptosis induction and cell cycle arrest. In summary, this study supports the hypothesis that effective transporter-mediated cellular uptake of a chemical molecule determines its cytotoxic properties. These results warrant a further investigation of biguanides as putative anti-cancer agents.


Asunto(s)
Antineoplásicos , Apoptosis/efectos de los fármacos , Biguanidas , Neoplasias de la Mama , Puntos de Control del Ciclo Celular/efectos de los fármacos , Sulfonamidas , Antineoplásicos/síntesis química , Antineoplásicos/química , Antineoplásicos/farmacología , Biguanidas/química , Biguanidas/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Femenino , Humanos , Células MCF-7 , Sulfonamidas/química , Sulfonamidas/farmacología
9.
Int J Pharm ; 601: 120565, 2021 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-33812973

RESUMEN

Non-steroidal anti-inflammatory drugs (NSAIDs) can have protective effects in the brain by inhibition of cyclooxygenases (COX). However, the delivery into the brain across the blood-brain barrier (BBB) and particularly into the brain parenchymal cells is hindered. Therefore, in the present study, we developed four l-type amino acid transporter 1 (LAT1)-utilizing prodrugs of flurbiprofen, ibuprofen, naproxen, and ketoprofen, since LAT1 is expressed on both, the BBB endothelial cells as well as parenchymal cells. The cellular uptake and utilization of LAT1 by novel prodrugs were studied in mouse cortical primary astrocytes and immortalized microglia (BV2), and the release of the parent NSAID in several tissue and cell homogenates. Finally, the effects of the studied prodrugs on prostaglandin E2 (PGE2) production and cell viability were explored. The gained results showed that all four prodrugs were carried into their target cells via LAT1. They also released their parent NSAIDs via carboxylesterases (CES) and most likely also other un-identified enzymes, which need to be carefully considered when administrating these compounds orally or intravenously. Most importantly, all the studied prodrugs reduced the PGE2 production in astrocytes and microglia after lipopolysaccharide (LPS)-induced inflammation by 29-94% and without affecting the cell viability with the studied concentration (20 µM).


Asunto(s)
Astrocitos , Profármacos , Animales , Antiinflamatorios no Esteroideos , Células Endoteliales , Transportador de Aminoácidos Neutros Grandes 1 , Ratones , Microglía , Prostaglandinas
10.
Molecules ; 27(1)2021 Dec 22.
Artículo en Inglés | MEDLINE | ID: mdl-35011270

RESUMEN

l-Type amino acid transporter 1 (LAT1), expressed abundantly in the brain and placenta and overexpressed in several cancer cell types, has gained a lot of interest in drug research and development, as it can be utilized for brain-targeted drug delivery, as well as inhibiting the essential amino acid supply to cancer cells. The structure of LAT1 is today very well-known and the interactions of ligands at the binding site of LAT1 can be modeled and explained. However, less is known of LAT1's life cycle within the cells. Moreover, the functionality of LAT1 can be measured by several different methods, which may vary between the laboratories and make the comparison of the results challenging. In the present study, the usefulness of indirect cis-inhibition methods and direct cellular uptake methods and their variations to interpret the interactions of LAT1-ligands were evaluated. Moreover, this study also highlights the importance of understanding the intracellular kinetics of LAT1-ligands, and how they can affect the regular function of LAT1 in critical tissues, such as the brain. Hence, it is discussed herein how the selected methodology influences the outcome and created knowledge of LAT1-utilizing compounds.


Asunto(s)
Transportador de Aminoácidos Neutros Grandes 1/química , Ligandos , Animales , Sitios de Unión , Transporte Biológico , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas/métodos , Humanos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Ratones , Microglía/efectos de los fármacos , Microglía/metabolismo , Estructura Molecular , Unión Proteica , Relación Estructura-Actividad
11.
ACS Chem Neurosci ; 11(24): 4301-4315, 2020 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-33228353

RESUMEN

Membrane transporters have long been utilized to improve the oral, hepatic, and renal (re)absorption. In the brain, however, the transporter-mediated drug delivery has not yet been fully achieved due to the complexity of the blood-brain barrier (BBB). Because L-type amino acid transporter 1 (LAT1) is a good candidate to improve the brain delivery, we developed here four novel LAT1-utilizing prodrugs of four nonsteroidal anti-inflammatory drugs. As a result, all the prodrugs were able to cross the BBB and localize into the brain cells. The brain uptake of salicylic acid (SA) was improved five times, not only across the mouse BBB but also into the cultured mouse and human brain cells. The naproxen prodrug was also transported efficiently into the mouse brain achieving less peripheral exposure, but the brain release of naproxen from the prodrug was not improved. Contrarily, the high plasma protein binding of the flurbiprofen prodrug and the premature bioconversion of the ibuprofen prodrug in the mouse blood hindered the efficient brain delivery. Thus, the structure of the parent drug affects the successful brain delivery of the LAT1-utilizing prodrugs, and the small-sized LAT1-utilizing prodrug of SA constituted a successful model to specifically deliver its parent drug across the mouse BBB and into the cultured mouse and human brain cells.


Asunto(s)
Barrera Hematoencefálica , Profármacos , Animales , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Humanos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Ratones
12.
Mol Neurobiol ; 57(11): 4563-4577, 2020 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-32754897

RESUMEN

The cytolytic protein perforin has a crucial role in infections and tumor surveillance. Recently, it has also been associated with many brain diseases, such as neurodegenerative diseases and stroke. Therefore, inhibitors of perforin have attracted interest as novel drug candidates. We have previously reported that converting a perforin inhibitor into an L-type amino acid transporter 1 (LAT1)-utilizing prodrug can improve the compound's brain drug delivery not only across the blood-brain barrier (BBB) but also into the brain parenchymal cells: neurons, astrocytes, and microglia. The present study evaluated whether the increased uptake into mouse primary cortical astrocytes and subsequently improvements in the cellular bioavailability of this brain-targeted perforin inhibitor prodrug could enhance its pharmacological effects, such as inhibition of production of caspase-3/-7, lipid peroxidation products and prostaglandin E2 (PGE2) in the lipopolysaccharide (LPS)-induced neuroinflammation mouse model. It was demonstrated that increased brain and cellular drug delivery could improve the ability of perforin inhibitors to elicit their pharmacological effects in the brain at nano- to picomolar levels. Furthermore, the prodrug displayed multifunctional properties since it also inhibited the activity of several key enzymes related to Alzheimer's disease (AD), such as the ß-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), acetylcholinesterase (AChE), and most probably also cyclooxygenases (COX) at micromolar concentrations. Therefore, this prodrug is a potential drug candidate for preventing Aß-accumulation and ACh-depletion in addition to combatting neuroinflammation, oxidative stress, and neural apoptosis within the brain. Graphical abstract.


Asunto(s)
Encéfalo/patología , Inflamación/patología , Estrés Oxidativo/efectos de los fármacos , Perforina/antagonistas & inhibidores , Profármacos/farmacología , Acetilcolinesterasa/metabolismo , Animales , Antioxidantes/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Butirilcolinesterasa/metabolismo , Hidrolasas de Éster Carboxílico/metabolismo , Caspasa 3/metabolismo , Caspasa 7/metabolismo , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Dinoprostona/biosíntesis , Humanos , Concentración 50 Inhibidora , Cinética , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Peroxidación de Lípido/efectos de los fármacos , Células MCF-7 , Masculino , Ratones Endogámicos C57BL , Perforina/metabolismo , Profármacos/química
13.
Apoptosis ; 25(5-6): 426-440, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32405891

RESUMEN

Increased amounts of amino acids are essential for cancer cells to support their sustained growth and survival. Therefore, inhibitors of amino acid transporters, such as L-type amino acid transporter 1 (LAT1) have been developed. In this study, a previously reported LAT1-inhibitor (KMH-233) was studied for its hemocompatibility and toxicity towards human umbilical vein endothelial cells (HUVEC) and human aortic smooth muscle cells (AoSMCs). Furthermore, the cytotoxic effects against human breast adenocarcinoma cells (MCF-7) and its ability to affect mammalian (or mechanistic) target of rapamycin (mTOR) and nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling were evaluated. Moreover, the effects of this inhibitor to modulate LAT1 function on the cell surface and the brain amino acid homeostasis were evaluated after intraperitoneal (i.p.) administration of LAT1-inhibitor (23 µmol/kg) in mice. The results showed that LAT1-inhibitor (KMH-233) is hemocompatible at concentrations below 25 µM and it does not affect coagulation in plasma. However, it can reduce the total protein amount of mTOR and NF-κB, resulting in increased apoptosis in LAT1-expressing cancer cells. Most importantly, the inhibitor did not affect mouse brain levels of L-Leu, L-Tyr or L-Trp or modulate the function of LAT1 on the MCF-7 cell surface. Therefore, this inhibitor can be considered as a safe but effective anti-cancer agent. However, due to the compensative mechanism of cancer cells for their increased amino acid demand, this compound is most effective inducing apoptosis when used in combinations with other chemotherapeutics, such as protease inhibitor, bestatin, as demonstrated in this study.


Asunto(s)
Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Encéfalo/metabolismo , Imidazoles/farmacología , Transportador de Aminoácidos Neutros Grandes 1/genética , Leucina/análogos & derivados , Piridinas/farmacología , Animales , Apoptosis/genética , Benzoxazoles/farmacología , Encéfalo/patología , Química Encefálica , Ácidos Carboxílicos/farmacología , Sinergismo Farmacológico , Regulación Neoplásica de la Expresión Génica , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inyecciones Intraperitoneales , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Leucina/farmacología , Células MCF-7 , Masculino , Ratones , Miocitos del Músculo Liso , FN-kappa B/genética , FN-kappa B/metabolismo , Norbornanos/farmacología , Cultivo Primario de Células , Transducción de Señal , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo , Tiazoles/farmacología , Tirosina/análogos & derivados , Tirosina/farmacología
14.
Int J Mol Sci ; 21(7)2020 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-32235654

RESUMEN

Metformin is a substrate for plasma membrane monoamine transporters (PMAT) and organic cation transporters (OCTs); therefore, the expression of these transporters and interactions between them may affect the uptake of metformin into tumor cells and its anticancer efficacy. The aim of this study was to evaluate how chemical modification of metformin scaffold into benzene sulfonamides with halogen substituents (compounds 1-9) may affect affinity towards OCTs, cellular uptake in two breast cancer cell lines (MCF-7 and MDA-MB-231) and antiproliferative efficacy of metformin. The uptake of most sulfonamides was more efficient in MCF-7 cells than in MDA-MB-231 cells. The presence of a chlorine atom in the aromatic ring contributed to the highest uptake in MCF-7 cells. For instance, the uptake of compound 1 with o-chloro substituent in MCF-7 cells was 1.79 ± 0.79 nmol/min/mg protein, while in MDA-MB-231 cells, the uptake was considerably lower (0.005 ± 0.0005 nmol/min/mg protein). The elevated uptake of tested compounds in MCF-7 was accompanied by high antiproliferative activity, with compound 1 being the most active (IC50 = 12.6 ± 1.2 µmol/L). Further studies showed that inhibition of MCF-7 growth is associated with the induction of early and late apoptosis and cell cycle arrest at the G0/G1 phase. In summary, the chemical modification of the biguanide backbone into halogenated sulfonamides leads to improved transporter-mediated cellular uptake in MCF-7 and contributes to the greater antiproliferative potency of studied compounds through apoptosis induction and cell cycle arrest.


Asunto(s)
Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Metformina/farmacología , Sulfonamidas/farmacología , Antineoplásicos/química , Antineoplásicos/farmacocinética , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Femenino , Halogenación , Humanos , Hipoglucemiantes/química , Hipoglucemiantes/farmacocinética , Hipoglucemiantes/farmacología , Células MCF-7 , Metformina/análogos & derivados , Metformina/farmacocinética , Sulfonamidas/química , Sulfonamidas/farmacocinética
15.
Toxics ; 8(1)2020 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-32050681

RESUMEN

Microplastics (MPs) of varying sizes are widespread pollutants in our environment. The general opinion is that the smaller the size, the more dangerous the MPs are due to enhanced uptake possibilities. It would be of considerably ecological significance to understand the response of biota to microplastic contamination both physically and physiologically. Here, we report on an area choice experiment (avoidance test) using Enchytraeus crypticus, in which we mixed different amounts of high-density polyethylene microplastic particles into the soil. In all experimental scenarios, more Enchytraeids moved to the unspiked sections or chose a lower MP-concentration. Worms in contact with MP exhibited an enhanced oxidative stress status, measured as the induced activity of the antioxidative enzymes catalase and glutathione S-transferase. As plastic polymers per se are nontoxic, the exposure time employed was too short for chemicals to leach from the microplastic, and as the microplastic particles used in these experiments were too large (4 mm) to be consumed by the Enchytraeids, the likely cause for the avoidance and oxidative stress could be linked to altered soil properties.

16.
Oxid Med Cell Longev ; 2019: 3528148, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31814871

RESUMEN

Ferulic acid (FA) is a natural phenolic antioxidant, which can exert also several other beneficial effects to combat neuroinflammation and neurodegenerative diseases, such as Alzheimer's disease. One of these properties is the inhibition of several enzymes and factors, such as ß-site amyloid precursor protein (APP) cleaving enzyme 1 (BACE1), cyclooxygenases (COXs), lipoxygenases (LOXs), mammalian (or mechanistic) target for rapamycin (mTOR), and transcription factor NF-κB. We have previously synthesized three L-type amino acid transporter 1- (LAT1-) utilizing FA-derivatives with the aim to develop brain-targeted prodrugs of FA. In the present study, the cellular uptake and bioavailability of these FA-derivatives were evaluated in mouse primary astrocytic cell cultures together with their inhibitory effects towards BACE1, COX/LOX, mTOR, NF-κB, acetylcholinesterase (AChE), and oxidative stress. According to the results, all three FA-derivatives were taken up 200-600 times more effectively at 10 µM concentration into the astrocytes than FA, with one derivative having a high intracellular bioavailability (K p,uu), particularly at low concentrations. Moreover, all of the derivatives were able to inhibit BACE1, COX/LOX, AChE, and oxidative stress measured as decreased cellular lipid peroxidation. Furthermore, one of the derivatives modified the total mTOR amount. Therefore, these derivatives have the potential to act as multifunctional compounds preventing ß-amyloid accumulation as well as combating inflammation and reducing oxidative stress in the brain. Thus, this study shows that converting a parent drug into a transporter-utilizing derivative not only may increase its brain and cellular uptake, and bioavailability but can also broaden the spectrum of pharmacological effects elicited by the derivative.


Asunto(s)
Sistema de Transporte de Aminoácidos y+L/metabolismo , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Encéfalo/efectos de los fármacos , Ácidos Cumáricos/farmacología , Encefalitis/tratamiento farmacológico , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Estrés Oxidativo/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/farmacología , Astrocitos/patología , Encéfalo/metabolismo , Encéfalo/patología , Encefalitis/metabolismo , Encefalitis/patología , Humanos , Células MCF-7 , Ratones Endogámicos C57BL , Ratones Transgénicos , Terapia Molecular Dirigida
17.
Int J Pharm ; 571: 118714, 2019 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-31610281

RESUMEN

L-Type amino acid transporter 1 (LAT1)-utilizing prodrugs has been designed to improve drug delivery and targeting into the brain or cancer cells, since LAT1 is highly and selectively expressed on the blood-brain barrier as well as over-expressed in several types of cancer. However, less is known about the affinity of these compounds for the secondary transport mechanisms. The aim of this study was to evaluate interactions of nine LAT1-utilizing prodrugs with organic anion transporting polypeptides (OATPs). These results showed that LAT1-utilizing prodrugs can indeed, interact with OATPs, although it was considered to be minor compared to LAT1; the Km values of these compounds for LAT1 were 1-7 µM while the ones for OATPs were 73-406 µM. Moreover, utilization of LAT1 was 2-12-times more effective (compared as intrinsic clearance) than of OATPs, whose utilization seemed to be less significant at therapeutically used concentrations. According to these results, affinity for OATPs raised from the structural features of the parent drug moiety regardless of the structure of the promoiety. In conclusion, the present study shows that it is important to evaluate secondary transport mechanisms carefully, since they may have a role in pharmacokinetics of LAT1-utilizing prodrugs if LAT1 becomes saturated or un-functional.


Asunto(s)
Sistemas de Liberación de Medicamentos/métodos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Transportadores de Anión Orgánico/metabolismo , Profármacos/farmacología , Humanos , Células MCF-7 , Profármacos/química , Unión Proteica
18.
Sci Rep ; 9(1): 12860, 2019 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-31492955

RESUMEN

L-Type Amino Acid Transporter 1 (LAT1/Lat1) is responsible for carrying large, neutral L-amino acids as well as several drugs and prodrugs across the blood-brain barrier (BBB). However, the BBB is not the only barrier that hinders drugs acting effectively within the brain; the brain parenchymal cell membranes represent a secondary barrier for the drugs with intracellular target sites. In this study, expression and function of Lat1 was quantified in mouse primary neuron, astrocyte and immortalized microglia (BV2) cultures. Moreover, ability of Lat1 to carry prodrugs inside these brain cells was evaluated. The results showed that Lat1 was localized at the similar level in all studied cells (3.07 ± 0.92-3.77 ± 0.91 fmol/µg protein). The transporter was also functional in all three cell types, astrocytes having the highest transport capacity and affinity for the LAT1/Lat1-substrate, [14C]-L-leucine, followed by neurons and microglia. The designed prodrugs (1-6) were able to utilize Lat1 for their cellular uptake and it was mainly much higher than the one of their parent drugs. Interestingly, improved cellular uptake was also achieved in cells representing Alzheimer's Disease phenotype. Therefore, improved delivery and intra-brain targeting of drugs can be attained by utilizing LAT1/Lat1 and prodrug approach.


Asunto(s)
Astrocitos/metabolismo , Sistemas de Liberación de Medicamentos/métodos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Microglía/metabolismo , Neuronas/metabolismo , Profármacos/administración & dosificación , Enfermedad de Alzheimer/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/citología , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Encéfalo/citología , Encéfalo/metabolismo , Línea Celular , Células Cultivadas , Ratones Endogámicos C57BL , Ratones Transgénicos , Microglía/citología , Neuronas/citología , Profármacos/química , Profármacos/farmacocinética
19.
Bioorg Chem ; 87: 321-334, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30913467

RESUMEN

Metformin, the most frequently administered oral anti-diabetic drug, is a substrate for organic cation transporters (OCTs). This determines not only its pharmacokinetic properties but also its biochemical effects in humans, including its recently-discovered antiproliferative properties. The aim of the study was to verify the hypothesis whether chemical modification of its biguanide backbone may increase the cellular uptake and antiproliferative efficacy of metformin. The study examines five sulfenamide derivatives of metformin with differing lengths of alkyl chains. It determines their cellular uptake and the role of OCTs in their transport in human breast adenocarcinoma cells (epithelial-like MCF-7, and MDA-MB-231). It also evaluates whether increased cellular uptake of metformin derivatives is associated with their cytotoxic properties. Sulfenamide derivatives were characterized by a greater ability to bind to OCTs than metformin. Compound 2 with n-octyl alkyl chain was found to possess the greatest affinity towards OCTs, as measured by determination of [14C]choline uptake inhibition (IC50 = 236.1 ±â€¯1.28 µmol/L, and 217.4 ±â€¯1.33 µmol/L, for MCF-7 and MDA-MB-231 respectively). Sulfenamides were also found to exhibit better cellular uptake in comparison with the parent drug, metformin. For instance, the uptake of cyclohexyl derivative 1 was 1.28 ±â€¯0.19 nmol/min/mg of proteins and thus was 12-fold higher than the metformin in MCF-7 cells. Furthermore, higher uptake was associated with the greatest antiproliferative properties expressed as the lowest IC50 value i.e. inhibiting the growth of 50% of the cells (IC50 = 0.72 ±â€¯1.31 µmol/L). Collectively, chemical modification of metformin into sulfenamides with different alkyl substituents obtains better substrates for OCTs, and subsequently higher cellular uptake in MCF-7 and MDA-MB-231 cells. Additionally, the length of alkyl chain introduced to the sulfenamides was found to influence selectivity and transport efficiency via OCT1 compared to other possible transporters, as well as potential intracellular activity and cytotoxicity.


Asunto(s)
Adenocarcinoma/tratamiento farmacológico , Antineoplásicos/farmacología , Neoplasias de la Mama/tratamiento farmacológico , Hipoglucemiantes/farmacocinética , Metformina/farmacocinética , Receptores de Estrógenos/metabolismo , Sulfamerazina/farmacología , Adenocarcinoma/metabolismo , Adenocarcinoma/patología , Antineoplásicos/síntesis química , Antineoplásicos/química , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/patología , Proliferación Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Ensayos de Selección de Medicamentos Antitumorales , Femenino , Humanos , Hipoglucemiantes/química , Células MCF-7 , Metformina/química , Estructura Molecular , Receptores de Estrógenos/genética , Relación Estructura-Actividad , Sulfamerazina/síntesis química , Sulfamerazina/química , Células Tumorales Cultivadas
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
...