Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Bio Protoc ; 12(15): e4465, 2022 Aug 05.
Artículo en Inglés | MEDLINE | ID: mdl-36082368

RESUMEN

There is an urgent need for the development of brain drug delivery carriers based on middle-sized or macromolecules, to which in vitro blood-brain barrier (BBB) models are expected to contribute significantly through evaluation of BBB permeability. As part of efforts to develop such models, we have been working on human conditionally immortalized cell-based multicellular spheroidal BBB models (hiMCS-BBB models), and we herein introduce the model development protocol. Briefly, astrocytes are first seeded in an ultra-low attachment 3D cell culture plate, to make the central core (Day 0). Next, pericytes are added over the core, to form an outer layer (Day 1). Then, brain microvascular endothelial cells are further added to each well, to create the outmost monolayer serving as the BBB (Day 2). Finally, the spheroids cultured for two days (on Day 4) can be used for assays of interest (e.g., antibody permeability assays). Neither special equipment nor techniques are required to produce hiMCS-BBB models. Therefore, the protocol presented here will not only facilitate the model sharing among the BBB community but also provide some technical clues contributing to the development of similar MCS-BBB models using other cell sources, such as primary or iPS-derived BBB cells. Graphical abstract.

2.
Mol Pharm ; 19(8): 2754-2764, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35766901

RESUMEN

Blood-brain barrier (BBB)-permeable middle- or macromolecules (middle/macromolecules) have recently attracted significant attention as new drug delivery carriers into the human brain via receptor-mediated transcytosis (RMT). During the development process of such carriers, it is necessary to thoroughly evaluate their human BBB permeability levels. In such evaluations, our recently established human immortalized cell-based multicellular spheroidal BBB models (hiMCS-BBB models) have shown high potential. However, the specifics of those capabilities have yet to be elucidated. Therefore, in this study, we characterize the ability of the hiMCS-BBB models to evaluate RMT-mediated BBB penetration properties of middle/macromolecules. More specifically, we began by validating transferrin receptor (TfR)-mediated RMT functionalities using transferrin in the hiMCS-BBB models and then examined the BBB permeability levels of MEM189 antibodies (known BBB-permeable anti-TfR antibodies). The obtained results showed that, as with the case of transferrin, temperature-dependent uptake of MEM189 antibodies was observed in the hiMCS-BBB models, and the extent of that uptake increased in a time-dependent manner until reaching a plateau after around 2 h. To further expand the evaluation applicability of the models, we also examined the BBB permeability levels of the recently developed SLS cyclic peptide and observed that peptide uptake was also temperature-dependent. To summarize, our results show that the hiMCS-BBB models possess the ability to evaluate the RMT-mediated BBB-permeable properties of antibodies and peptides and thus have the potential to provide valuable tools for use in the exploration and identification of middle/macromolecules showing excellent BBB permeability levels, thereby contributing powerfully to the development of new drug delivery carriers for transporting drugs into the human brain.


Asunto(s)
Barrera Hematoencefálica , Receptores de Transferrina , Anticuerpos/química , Transporte Biológico , Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Humanos , Receptores de Transferrina/metabolismo , Transcitosis , Transferrina/metabolismo
3.
Pharm Res ; 39(7): 1575-1586, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35288803

RESUMEN

PURPOSE: In vitro human blood-brain barrier (BBB) models in combination with central nervous system-physiologically based pharmacokinetic (CNS-PBPK) modeling, hereafter referred to as the "BBB/PBPK" method, are expected to contribute to prediction of brain drug concentration profiles in humans. As part of our ongoing effort to develop a BBB/PBPK method, we tried to clarify the relationship of in vivo BBB permeability data to those in vitro obtained from a human immortalized cell-based tri-culture BBB model (hiBBB), which we have recently created. METHODS: The hiBBB models were developed and functionally characterized as previously described. The in vitro BBB permeabilities (Pe, × 10-6 cm/s) of seventeen compounds were determined by permeability assays, and in vivo BBB permeabilities (QECF) for eight drugs were estimated by CNS-PBPK modeling. The correlation of the Pe values with the QECF values was analyzed by linear regression analysis. RESULTS: The hiBBB models showed intercellular barrier properties and several BBB transporter functions, which were enough to provide a wide dynamic range of Pe values from 5.7 ± 0.7 (rhodamine 123) to 2580.4 ± 781.9 (rivastigmine). Furthermore, the in vitro Pe values of the eight drugs showed a good correlation (R2 = 0.96) with their in vivo QECF values estimated from human clinical data. CONCLUSION: We show that in vitro human BBB models provide clinically relevant BBB permeability that can be used as input for CNS-PBPK modeling. Therefore, our findings will encourage the development of a BBB/PBPK method as a promising approach for predicting brain drug concentration profiles in humans.


Asunto(s)
Barrera Hematoencefálica , Encéfalo , Transporte Biológico/fisiología , Estudios de Factibilidad , Humanos , Permeabilidad
4.
Int J Mol Sci ; 23(1)2022 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-35008999

RESUMEN

Conventional anti-cancer therapies based on chemo- and/or radiotherapy represent highly effective means to kill cancer cells but lack tumor specificity and, therefore, result in a wide range of iatrogenic effects. A promising approach to overcome this obstacle is spliceosome-mediated RNA trans-splicing (SMaRT), which can be leveraged to target tumor cells while leaving normal cells unharmed. Notably, a previously established RNA trans-splicing molecule (RTM44) showed efficacy and specificity in exchanging the coding sequence of a cancer target gene (Ct-SLCO1B3) with the suicide gene HSV1-thymidine kinase in a colorectal cancer model, thereby rendering tumor cells sensitive to the prodrug ganciclovir (GCV). In the present work, we expand the application of this approach, using the same RTM44 in aggressive skin cancer arising in the rare genetic skin disease recessive dystrophic epidermolysis bullosa (RDEB). Stable expression of RTM44, but not a splicing-deficient control (NC), in RDEB-SCC cells resulted in expression of the expected fusion product at the mRNA and protein level. Importantly, systemic GCV treatment of mice bearing RTM44-expressing cancer cells resulted in a significant reduction in tumor volume and weight compared with controls. Thus, our results demonstrate the applicability of RTM44-mediated targeting of the cancer gene Ct-SLCO1B3 in a different malignancy.


Asunto(s)
Epidermólisis Ampollosa Distrófica/complicaciones , Epidermólisis Ampollosa/complicaciones , Terapia Genética/métodos , Empalme del ARN , Neoplasias Cutáneas/etiología , Neoplasias Cutáneas/terapia , Trans-Empalme , Animales , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Manejo de la Enfermedad , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Epidermólisis Ampollosa/genética , Epidermólisis Ampollosa Distrófica/genética , Ganciclovir/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Sitios Genéticos , Terapia Genética/efectos adversos , Humanos , Ratones , Neoplasias Cutáneas/diagnóstico , Neoplasias Cutáneas/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
5.
J Pharmacol Sci ; 146(2): 82-87, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33941324

RESUMEN

Islatravir (ISL; 4'-ethynyl-2-fluoro-2'-deoxyadenosine or EFdA) is a novel reverse transcriptase translocation inhibitor and has a unique structure and high antiviral activity against wild-type and multidrug resistant HIV strains. In this study, we investigated whether islatravir (ISL) can cause kidney damage compared to tenofovir disoproxil fumarate (TDF) and tenofovir (TFV). We also investigated interactions of these drugs with organic anion transporters (OATs). There is a large gap in ISL concentration between the pharmacological dose to proximal tubular cells and the clinical dose. ISL is unlikely to be taken up via OAT1 or OAT3; therefore, OAT1 and OAT3 may not be involved in the injury to tubular cells. Present data strongly suggests that ISL is not toxic to proximal tubules because blood levels of ISL are not high enough to cause kidney damage in the clinical setting.


Asunto(s)
Desoxiadenosinas/efectos adversos , Desoxiadenosinas/metabolismo , Túbulos Renales Proximales/efectos de los fármacos , Transportadores de Anión Orgánico/metabolismo , Inhibidores de la Transcriptasa Inversa/efectos adversos , Inhibidores de la Transcriptasa Inversa/metabolismo , Lesión Renal Aguda/etiología , Células Cultivadas , Desoxiadenosinas/sangre , Relación Dosis-Respuesta a Droga , Humanos
6.
J Physiol Sci ; 71(1): 11, 2021 Mar 31.
Artículo en Inglés | MEDLINE | ID: mdl-33789576

RESUMEN

L-type amino acid transporter 2 (LAT2) is a Na+-independent neutral amino acid transporter, whose function regulation system remains unclarified. Since protein kinase C (PKC) is known to regulate the functions of various transporters, we investigated whether human LAT2 (hLAT2) function is regulated by PKC. In mouse proximal tubule S2 cells, hLAT2 transport activity was upregulated by PKC activation. However, we found that the mRNA and protein expression of hLAT2 was not affected by PKC activation and that the upregulation was independent of the three potential PKC consensus sites in the hLAT2 amino acid sequence. Moreover, we found that PKC activation upregulated the Vmax value for hLAT2-mediated alanine transport, which was not accompanied by the induction of hLAT2 membrane insertion. In conclusion, we showed that hLAT2 function is upregulated by PKC activation, which is not related to either the de novo synthesis, the phosphorylation or the membrane insertion of hLAT2.


Asunto(s)
Sistema de Transporte de Aminoácidos y+/metabolismo , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/metabolismo , Proteína Quinasa C/metabolismo , Sistema de Transporte de Aminoácidos y+/genética , Animales , Línea Celular , Supervivencia Celular , Clonación Molecular , Cadenas Ligeras de la Proteína-1 Reguladora de Fusión/genética , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Indoles/farmacología , Maleimidas/farmacología , Ratones , Mutagénesis Sitio-Dirigida , Regulación hacia Arriba
7.
Biol Pharm Bull ; 44(7): 984-991, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-33896887

RESUMEN

In vitro blood-brain barrier (BBB) models are essential research tools for use in developing brain-targeted drugs and understanding the physiological and pathophysiological functions of the BBB. To develop BBB models with better functionalities, three-dimensional (3D) culture methods have gained significant attention as a promising approach. In this study, we report on the development of a human conditionally immortalized cell-based multicellular spheroidal BBB (hiMCS-BBB) model. After being seeded into non-attachment culture wells, HASTR/ci35 (astrocytes) and HBPC/ci37 cells (brain pericytes) self-assemble to form a spheroid core that is then covered with an outer monolayer of HBMEC/ci18 cells (brain microvascular endothelial cells). The results of immunocytochemistry showed the protein expression of several cellular junction and BBB-enriched transporter genes in HBMEC/ci18 cells of the spheroid model. The permeability assays showed that the hiMCS-BBB model exhibited barrier functions against the penetration of dextran (5 and 70 kDa) and rhodamine123 (a P-glycoprotein substrate) into the core. On the other hand, facilitation of 2-(N-[7-nitrobenz-2-oxa-1,3-diazol-4-yl]amino)-2-deoxyglucose (2-NBDG; a fluorescent glucose analog) uptake was observed in the hiMCS-BBB model. Furthermore, tumor necrosis factor-alpha treatment elicited an inflammatory response in HBMEC/ci18 cells, thereby suggesting that BBB inflammation can be recapitulated in the hiMCS-BBB model. To summarize, we have developed an hiMCS-BBB model that possesses fundamental BBB properties, which can be expected to provide a useful and highly accessible experimental platform for accelerating various BBB studies.


Asunto(s)
Barrera Hematoencefálica/fisiología , Esferoides Celulares/metabolismo , Astrocitos/metabolismo , Transporte Biológico/fisiología , Barrera Hematoencefálica/efectos de los fármacos , Encéfalo/metabolismo , Línea Celular , Técnicas de Cocultivo , Células Endoteliales/metabolismo , Humanos , Técnicas In Vitro , Inflamación/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Pericitos/metabolismo , Permeabilidad , Células THP-1 , Factor de Necrosis Tumoral alfa/farmacología
8.
J Pharmacol Sci ; 144(1): 16-22, 2020 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-32653341

RESUMEN

JPH203 is a novel anti-cancer drug targeting L-type amino acid transporter 1 (LAT1), which plays a primary role in the uptake of essential amino acids in tumor cells. Although a co-incubation inhibitory effect of JPH203 has been shown in a conventional uptake assay, its preincubation inhibitory effects have remained undetermined. Therefore, we aimed to characterize the preincubation inhibitory effects of JPH203 on LAT1 function using leucine uptake assays in LAT1-positive human colon cancer HT-29 cells. Preincubation of the cells with JPH203 (0.3 µM for 120 min) decreased the activity level to 30% of that in dimethylsulfoxide-treated cells. Similarly, in time-dependency analysis, preincubation of HT-29 cells with 10 µM JPH203 for 30, 60, and 120 min decreased the leucine uptake activity (42%, 32%, and 28% of that in control cells, respectively). Furthermore, the IC50 value of the combination of preincubation and co-incubation effects was lower than that of co-incubation inhibition alone (34.2 ± 3.6 nM vs. 99.2 ± 11.0 nM). In conclusion, we revealed that JPH203 has the capability to inhibit LAT1 function through preincubation effects. Moreover, preincubation synergistically enhances the co-incubation inhibitory effects. These findings provide a novel insight into the anti-cancer effects of JPH203 in cancer therapy.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Antineoplásicos/farmacología , Benzoxazoles/farmacología , Neoplasias del Colon/genética , Neoplasias del Colon/metabolismo , Ensayos de Selección de Medicamentos Antitumorales/métodos , Transportador de Aminoácidos Neutros Grandes 1/metabolismo , Tirosina/análogos & derivados , Relación Dosis-Respuesta a Droga , Células HT29 , Humanos , Transportador de Aminoácidos Neutros Grandes 1/fisiología , Leucina/metabolismo , Factores de Tiempo , Tirosina/farmacología
9.
Anticancer Res ; 39(1): 159-165, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30591453

RESUMEN

BACKGROUND/AIM: L-type amino acid transporter 1 (LAT1) is a promising molecular target for cancer therapy. The present study aimed to characterize the anti-cancer effects of JPH203, an LAT1-selective inhibitor, on gastrointestinal cancer cells. MATERIALS AND METHODS: Three esophageal, two gastric, and two colon cancer cell lines were used. Cytotoxic effects of JPH203 were assessed by a WST-8 assay. LAT1 mRNA levels were determined by quantitative PCR. The inhibitory property of JPH203 against LAT1 function was examined by a transport assay. RESULTS: JPH203 treatment significantly reduced the viability of all gastric and colon cancer cells. While LAT1 expression levels and inhibitory potencies of JPH203 on LAT1 functions were comparable among the cells, all the esophageal cells were resistant to JPH203. CONCLUSION: JPH203 was effective in reducing gastric and colon cancer cells. To clarify its cell type-dependent efficacy, identification of the causal factors for JPH203 resistance will be needed.


Asunto(s)
Benzoxazoles/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias Gastrointestinales/tratamiento farmacológico , Transportador de Aminoácidos Neutros Grandes 1/genética , Tirosina/análogos & derivados , Apoptosis/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/patología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Tirosina/farmacología
10.
Bioorg Med Chem Lett ; 28(23-24): 3708-3711, 2018 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-30389287

RESUMEN

We synthesized six novel BBR derivatives that were designed to avoid metabolic activation via ipso-substitution and evaluated for their degree of toxicity and hURAT1 inhibition. It was found that all of the derivatives demonstrate lower cytotoxicity in mouse hepatocytes and lower levels of metabolic activation than BBR, while maintaining their inhibitory activity toward the uric acid transporter. We propose that these derivatives could serve as effective uricosuric agents that have much better safety profiles than BBR.


Asunto(s)
Benzbromarona/análogos & derivados , Benzbromarona/metabolismo , Transportadores de Anión Orgánico/antagonistas & inhibidores , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores , Uricosúricos/química , Uricosúricos/metabolismo , Activación Metabólica , Animales , Benzbromarona/farmacología , Benzbromarona/toxicidad , Técnicas de Química Sintética , Células HEK293 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Ratones , Microsomas Hepáticos/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Transportadores de Anión Orgánico/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Ratas , Ácido Úrico/metabolismo , Uricosúricos/farmacología , Uricosúricos/toxicidad
11.
J Pharmacol Sci ; 137(4): 350-358, 2018 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-30150146

RESUMEN

Astrocytes have shown longstanding promise as therapeutic targets for various central nervous system diseases. To facilitate drug development targeting astrocytes, we have recently developed a new conditionally immortalized human astrocyte cell line, termed HASTR/ci35 cells. In this study, in order to further increase their chances to contribute to various astrocyte studies, we report on the development of a culture method that improves HASTR/ci35 cell differentiation status and provide several proofs related to their astrocyte characteristics. The culture method is based on the simultaneous elimination of serum effects and immortalization signals. The results of qPCR showed that the culture method significantly enhanced several astrocyte marker gene expression levels. Using the differentiated HASTR/ci35, we examined their response profiles to nucleotide treatment and inflammatory stimuli, along with their membrane fatty acid composition. Consequently, we found that they responded to ADP or UTP treatment with a transient increase of intracellular Ca2+ concentration, and that they could show reactive response to interleukin-1ß treatments. Furthermore, the membrane phospholipids of the cells were enriched with polyunsaturated fatty acids. To summarize, as a unique human astrocyte model carrying the capability of a differentiation induction properties, HASTR/ci35 cells are expected to contribute substantially to astrocyte-oriented drug development studies.


Asunto(s)
Astrocitos , Técnicas de Cultivo de Célula/métodos , Fármacos del Sistema Nervioso Central/farmacología , Descubrimiento de Drogas/métodos , Adenosina Difosfato/farmacología , Astrocitos/citología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/patología , Calcio/metabolismo , Diferenciación Celular , Línea Celular , Ácidos Grasos/metabolismo , Ácidos Grasos Insaturados/farmacología , Expresión Génica , Humanos , Inflamación , Interleucina-1beta/metabolismo , Fosfolípidos/metabolismo , Uridina Trifosfato/farmacología
12.
Cancer Lett ; 433: 107-116, 2018 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-29960051

RESUMEN

Cancer-type organic anion transporting polypeptide 1B3 (Ct-OATP1B3) has been identified as a cancer-specific transcript in various solid cancers, including colorectal cancer. Given its excellent cancer-specific expression profile, we hypothesized that Ct-OATP1B3 could represent a promising target for cancer-specific expression of the suicide gene, herpes simplex virus 1 thymidine kinase (HSV-tk), via a spliceosome-mediated RNA trans-splicing (SMaRT) approach. SMaRT technology is used to recombine two RNA molecules to generate a chimeric transcript. In this study, we engineered an RNA trans-splicing molecule carrying a translation-defective HSV-tk sequence (RTM44), which was capable of inducing its own trans-splicing to the desired Ct-OATP1B3 pre-mRNA target. RTM44 expression in LS180 cells resulted in generation of Ct-OATP1B3/HSV-tk fusion mRNA. A functional translation start site contributed by the target pre-mRNA restored HSV-tk protein expression, rendering LS180 cells sensitive to ganciclovir treatment in vitro and in xenografted mice. The observed effects are ascribed to accurate and efficient trans-splicing, as they were absent in cells carrying a splicing-deficient mutant of RTM44. Collectively, our data highlights Ct-OATP1B3 as an ideal target for the HSV-tk SMaRT suicide system, which opens up new translational avenues for Ct-OATP1B3-targeted cancer therapy.


Asunto(s)
Neoplasias Colorrectales/terapia , Ganciclovir/administración & dosificación , Terapia Genética/métodos , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/genética , Empalmosomas/genética , Timidina Quinasa/genética , Animales , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Terapia Combinada , Ganciclovir/farmacología , Vectores Genéticos/administración & dosificación , Células HCT116 , Células HT29 , Humanos , Ratones , Proteínas Recombinantes de Fusión/metabolismo , Simplexvirus/genética , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/metabolismo , Timidina Quinasa/metabolismo , Trans-Empalme , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Biol Pharm Bull ; 41(3): 440-444, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29491221

RESUMEN

Liver sinusoidal endothelial cells (LSECs), which are specialized endothelial cells that line liver sinusoids, have been reported to participate in a variety of liver functions, such as blood macromolecule clearance and factor VIII production. In addition, LSECs play crucial roles in liver regeneration following acute liver injury, as well as the development and progression of liver diseases or drug-induced hepatotoxicity. However, the molecular mechanisms underlying their roles remain mostly unknown. Therefore, in order to contribute to the clarification of those mechanisms, herein we report on the development of a new immortalized human LSEC (HLSEC) line. To produce this cell line, two immortalized genes were introduced into the primary HLSECs, which eventually resulted in the establishment of the HLSEC/conditionally immortalized, clone-J (HLSEC/ciJ). Consistent with the two-immortalized gene expression, HLSEC/ciJ showed excellent proliferation activity. Additionally, the results of gene expression analyses showed that several LSEC (as well as pan-endothelial) marker mRNAs and proteins were clearly expressed in HLSEC/ciJ. Furthermore, we found that adherence junction proteins were localized at the cell border in the HLSEC/ciJ monolayer, and that the cells exhibited a tube-like structure formation property. Taken together, the results obtained thus far indicate that we have successfully immortalized HLSECs, resulting in creation of HLSEC/ciJ, a cell line that possesses infinite proliferation ability while retaining possession of at least some HLSEC features. We believe that the HLSEC/ciJ have the potential to provide a valuable and unlimited alternative source of HLSECs for use in liver/LSEC physiology/pathophysiology, pharmacology, and toxicology studies.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Hígado/citología , Técnicas de Cultivo de Célula , Línea Celular , Proliferación Celular , Criopreservación , ADN Complementario/biosíntesis , Células Endoteliales/metabolismo , Células Endoteliales/ultraestructura , Expresión Génica , Hepatocitos , Humanos , Hígado/metabolismo , ARN/biosíntesis
14.
Biol Pharm Bull ; 41(3): 445-449, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29491222

RESUMEN

Cancer-type organic anion transporting polypeptide 1B3 (Ct-OATP1B3) mRNA is a variant isoform of the liver-type OATP1B3. Because Ct-OATP1B3 mRNA shows an excellent cancer-specific expression profile in colorectal cancer (CRC), and that its expression levels are associated with CRC prognosis, it holds the potential to become a useful CRC detection and diagnosis biomarker. While the potential is currently justified only at the tissue level, if existence of Ct-OATP1B3 mRNA in CRC-derived extracellular vesicles (EVs) is validated, the findings could enhance its translational potential as a CRC detection and diagnosis biomarker. Therefore, this study aims at proving that Ct-OATP1B3 mRNA exists in CRC-derived EVs, and can be detected using serum specimens. To examine the possibility of Ct-OATP1B3 mRNA being existed in extracellular milieu, we isolated EVs from the human CRC (HCT116, HT-29, and SW480) cell lines, and prepared their cDNAs. The RT-PCR results showed that Ct-OATP1B3 mRNA was clearly present in EVs derived from the human CRC cell lines. Then, in order to further explore the possibility that Ct-OATP1B3 mRNA in CRC-derived EVs can be detected in serum, we isolated serum EVs derived from human CRC xenograft mice, and then performed RT-PCR. The results showed that Ct-OATP1B3 mRNA could be found in all serum EV and CRC tissue samples of the mice examined. Collectively, our findings, which show that Ct-OATP1B3 mRNA exists in EVs and can be detected in (at least) mouse serum, strengthen the potential use of Ct-OATP1B3 mRNA as a serum-based CRC biomarker.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/sangre , Vesículas Extracelulares/metabolismo , ARN Mensajero/sangre , ARN Mensajero/genética , ARN Neoplásico/sangre , ARN Neoplásico/genética , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/genética , Animales , Línea Celular Tumoral , Neoplasias Colorrectales/diagnóstico , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Trasplante de Neoplasias , Pronóstico , Reacción en Cadena en Tiempo Real de la Polimerasa , Miembro 1B3 de la Familia de los Transportadores de Solutos de Aniones Orgánicos/biosíntesis
15.
Drug Metab Pharmacokinet ; 32(1): 116-119, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27916488

RESUMEN

Entecavir (ETV) and tenofovir (TFV) are essential nucleoside analogues in current hepatitis B virus (HBV) treatments. Since these drugs target the HBV polymerase that is localized within human hepatocytes, determining of their cellular uptake process is an important step in fully understanding their pharmacological actions. However, the human hepatic transporters responsible for their uptake have remained unidentified. Therefore, this study aimed at identifying the primary ETV and TFV uptake transporter(s) in human hepatocytes. In transport assays, temperature-sensitive ETV and TFV uptake by human hepatocytes were observed, and their uptake were strongly inhibited by bromosulfophthalein, which is an inhibitor of organic anion transporters/organic anion transporting polypeptides (OATs/OATPs). Given these results, ETV and TFV uptake activities in several human OAT/OATP expression systems were examined. The results showed that, among the transporters tested, only OAT2 possessed ETV transport activity. On the other hand, none of the transporters showed any TFV uptake activity. To summarize, our results identify that human OAT2 is an ETV transporter, thereby suggesting that it plays an important part in the mechanisms underlying ETV antiviral activity. Furthermore, although the hepatic TFV transporters remain unknown, our results have, at least, clarified that these two anti-HBV drugs have different hepatocyte entry routes.


Asunto(s)
Guanina/análogos & derivados , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Transporte Biológico/efectos de los fármacos , Guanina/antagonistas & inhibidores , Guanina/metabolismo , Guanina/farmacocinética , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Sulfobromoftaleína/farmacología , Temperatura , Tenofovir/antagonistas & inhibidores , Tenofovir/metabolismo , Tenofovir/farmacocinética
16.
Int J Antimicrob Agents ; 46(4): 381-8, 2015 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-26163159

RESUMEN

Simeprevir (SMV), asunaprevir (ASV), daclatasvir (DCV) and sofosbuvir (SOF), which are direct-acting antiviral (DAA) agents, are expected to become essential pharmaceutical tools in the fight against the hepatitis C virus (HCV). However, because DAAs are taken orally, there is a potential risk of drug-drug interactions (DDIs) at the absorption step with co-administered drugs in the small intestine. Since it is known that organic anion transporting polypeptide 2B1 (OATP2B1) is one of the key transporters contributing to intestinal drug absorption, it is important to thoroughly understand the inhibition profiles of various DAAs in relation to OATP2B1 function in order to avoid unexpected DDIs. Therefore, using a cell-based transport assay, this study aimed at clarifying such DAA inhibition characteristics towards OATP2B1 function. The results of co-incubation inhibition assays showed that SMV and ASV strongly inhibited estrone sulfate (5 nM) uptake by OATP2B1, with half maximal inhibitory concentrations of 0.49 ± 0.12 µM and 0.16 ± 0.06 µM, respectively. Furthermore, it was found that SMV and ASV imposed long-lasting pre-incubation inhibitory effects on OATP2B1 function that enhanced their co-incubation inhibition potencies. On the other hand, no (or much less significant) inhibitory effects were observed for SOF or DCV. To summarise, these results show that SMV and ASV are co-incubation, as well as long-lasting pre-incubation, inhibitors of OATP2B1 function and therefore these inhibitions may lead to clinically relevant DDIs when used with OATP2B1 substrates.


Asunto(s)
Antivirales/metabolismo , Interacciones Farmacológicas , Transportadores de Anión Orgánico/metabolismo , Transporte Biológico , Células Cultivadas , Estrona/análogos & derivados , Estrona/metabolismo , Humanos , Concentración 50 Inhibidora
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA