Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Physiol Rev ; 102(2): 993-1024, 2022 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-34486394

RESUMEN

Over the course of more than 500 million years, the kidneys have undergone a remarkable evolution from primitive nephric tubes to intricate filtration-reabsorption systems that maintain homeostasis and remove metabolic end products from the body. The evolutionarily conserved solute carriers organic cation transporter 2 (OCT2) and organic anion transporters 1 and 3 (OAT1/3) coordinate the active secretion of a broad range of endogenous and exogenous substances, many of which accumulate in the blood of patients with kidney failure despite dialysis. Harnessing OCT2 and OAT1/3 through functional preservation or regeneration could alleviate the progression of kidney disease. Additionally, it would improve current in vitro test models that lose their expression in culture. With this review, we explore OCT2 and OAT1/3 regulation from different perspectives: phylogenetic, ontogenetic, and cell dynamic. Our aim is to identify possible molecular targets both to help prevent or compensate for the loss of transport activity in patients with kidney disease and to enable endogenous OCT2 and OAT1/3 induction in vitro in order to develop better models for drug development.


Asunto(s)
Riñón/metabolismo , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Transportadores de Anión Orgánico Sodio-Independiente/metabolismo , Transportador 2 de Cátion Orgánico/metabolismo , Animales , Humanos , Enfermedades Renales/metabolismo , Filogenia
2.
AAPS J ; 23(1): 11, 2020 12 28.
Artículo en Inglés | MEDLINE | ID: mdl-33369701

RESUMEN

Tissue decellularization yields complex scaffolds with retained composition and structure, and plants offer an inexhaustible natural source of numerous shapes. Plant tissue could be a solution for regenerative organ replacement strategies and advanced in vitro modeling, as biofunctionalization of decellularized tissue allows adhesion of various kinds of human cells that can grow into functional tissue. Here, we investigated the potential of spinach leaf vasculature and chive stems for kidney tubule engineering to apply in tubular transport studies. We successfully decellularized both plant tissues and confirmed general scaffold suitability for topical recellularization with renal cells. However, due to anatomical restrictions, we believe that spinach and chive vasculature themselves cannot be recellularized by current methods. Moreover, gradual tissue disintegration and deficient diffusion capacity make decellularized plant scaffolds unsuitable for kidney tubule engineering, which relies on transepithelial solute exchange between two compartments. We conclude that plant-derived structures and biomaterials need to be carefully considered and possibly integrated with other tissue engineering technologies for enhanced capabilities.


Asunto(s)
Cebollino/química , Túbulos Renales , Spinacia oleracea/química , Ingeniería de Tejidos/métodos , Andamios del Tejido/química , Materiales Biocompatibles/química , Matriz Extracelular/química , Humanos
3.
Int J Mol Sci ; 21(18)2020 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-32927790

RESUMEN

A considerable number of drugs and/or their metabolites are excreted by the kidneys through glomerular filtration and active renal tubule secretion via transporter proteins. Uptake transporters in the proximal tubule are part of the solute carrier (SLC) superfamily, and include the organic cation transporters (OCTs). Several studies have shown that specific genetic polymorphisms in OCTs alter drug disposition and may lead to nephrotoxicity. Multiple single nucleotide polymorphisms (SNPs) have been reported for the OCT genes (SLC22A1, SLC22A2 and SLC22A3), which can influence the proteins' structure and expression levels and affect their transport function. A gain-in-function mutation may lead to accumulation of drugs in renal proximal tubule cells, eventually leading to nephrotoxicity. This review illustrates the impact of genetic polymorphisms in OCTs on renal drug disposition and kidney injury, the clinical significances and how to personalize therapies to minimize the risk of drug toxicity.


Asunto(s)
Riñón/metabolismo , Proteínas de Transporte de Catión Orgánico/genética , Variantes Farmacogenómicas , Insuficiencia Renal/inducido químicamente , Animales , Humanos , Proteínas de Transporte de Catión Orgánico/metabolismo , Insuficiencia Renal/metabolismo
4.
Drug Discov Today ; 25(4): 706-717, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32027970

RESUMEN

Various animal models are used to study pharmacokinetics (PK) of drugs in development. Human renal clearance (CLr) should be predictable through interpolation from animal data by allometric scaling. Based on this premise, we quantified interspecies differences in CLr, and related them to drug properties. Using PubMed and EMBASE, we systematically reviewed literature on human and animal CLr measures for 20 renally excreted drugs, calculated average fold errors, and quantified mean differences between animals and humans. Our results show that animal models are generally good predictors for human drug clearance using simple allometry, except for rats, with which human CLr is significantly overestimated.


Asunto(s)
Tasa de Depuración Metabólica/fisiología , Modelos Animales , Preparaciones Farmacéuticas/metabolismo , Animales , Humanos , Riñón/metabolismo , Farmacocinética , Ratas , Especificidad de la Especie
5.
Front Bioeng Biotechnol ; 8: 617364, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33537294

RESUMEN

Introduction: To date, tubular tissue engineering relies on large, non-porous tubular scaffolds (Ø > 2 mm) for mechanical self-support, or smaller (Ø 150-500 µm) tubes within bulk hydrogels for studying renal transport phenomena. To advance the engineering of kidney tubules for future implantation, constructs should be both self-supportive and yet small-sized and highly porous. Here, we hypothesize that the fabrication of small-sized porous tubular scaffolds with a highly organized fibrous microstructure by means of melt-electrowriting (MEW) allows the development of self-supported kidney proximal tubules with enhanced properties. Materials and Methods: A custom-built melt-electrowriting (MEW) device was used to fabricate tubular fibrous scaffolds with small diameter sizes (Ø = 0.5, 1, 3 mm) and well-defined, porous microarchitectures (rhombus, square, and random). Human umbilical vein endothelial cells (HUVEC) and human conditionally immortalized proximal tubular epithelial cells (ciPTEC) were seeded into the tubular scaffolds and tested for monolayer formation, integrity, and organization, as well as for extracellular matrix (ECM) production and renal transport functionality. Results: Tubular fibrous scaffolds were successfully manufactured by fine control of MEW instrument parameters. A minimum inner diameter of 1 mm and pore sizes of 0.2 mm were achieved and used for subsequent cell experiments. While HUVEC were unable to bridge the pores, ciPTEC formed tight monolayers in all scaffold microarchitectures tested. Well-defined rhombus-shaped pores outperformed and facilitated unidirectional cell orientation, increased collagen type IV deposition, and expression of the renal transporters and differentiation markers organic cation transporter 2 (OCT2) and P-glycoprotein (P-gp). Discussion and Conclusion: Here, we present smaller diameter engineered kidney tubules with microgeometry-directed cell functionality. Due to the well-organized tubular fiber scaffold microstructure, the tubes are mechanically self-supported, and the self-produced ECM constitutes the only barrier between the inner and outer compartment, facilitating rapid and active solute transport.

6.
Proc Natl Acad Sci U S A ; 116(32): 16105-16110, 2019 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-31341083

RESUMEN

Membrane transporters and receptors are responsible for balancing nutrient and metabolite levels to aid body homeostasis. Here, we report that proximal tubule cells in kidneys sense elevated endogenous, gut microbiome-derived, metabolite levels through EGF receptors and downstream signaling to induce their secretion by up-regulating the organic anion transporter-1 (OAT1). Remote metabolite sensing and signaling was observed in kidneys from healthy volunteers and rats in vivo, leading to induced OAT1 expression and increased removal of indoxyl sulfate, a prototypical microbiome-derived metabolite and uremic toxin. Using 2D and 3D human proximal tubule cell models, we show that indoxyl sulfate induces OAT1 via AhR and EGFR signaling, controlled by miR-223. Concomitantly produced reactive oxygen species (ROS) control OAT1 activity and are balanced by the glutathione pathway, as confirmed by cellular metabolomic profiling. Collectively, we demonstrate remote metabolite sensing and signaling as an effective OAT1 regulation mechanism to maintain plasma metabolite levels by controlling their secretion.


Asunto(s)
Microbioma Gastrointestinal , Túbulos Renales Proximales/metabolismo , Transducción de Señal , Animales , Aniones , Receptores ErbB/metabolismo , Glutatión/metabolismo , Humanos , Metaboloma , Proteína 1 de Transporte de Anión Orgánico/metabolismo , Ratas , Especies Reactivas de Oxígeno/metabolismo , Receptores de Hidrocarburo de Aril/metabolismo
7.
Macromol Biosci ; 19(2): e1800412, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30548802

RESUMEN

The increasing prevalence of end-stage renal disease and persistent shortage of donor organs call for alternative therapies for kidney patients. Dialysis remains an inferior treatment as clearance of large and protein-bound waste products depends on active tubular secretion. Biofabricated tissues could make a valuable contribution, but kidneys are highly intricate and multifunctional organs. Depending on the therapeutic objective, suitable cell sources and scaffolds must be selected. This study provides a proof-of-concept for stand-alone kidney tubule grafts with suitable mechanical properties for future implantation purposes. Porous tubular nanofiber scaffolds are fabricated by electrospinning 12%, 16%, and 20% poly-ε-caprolactone (PCL) v/w (chloroform and dimethylformamide, 1:3) around 0.7 mm needle templates. The resulting scaffolds consist of 92%, 69%, and 54% nanofibers compared to microfibers, respectively. After biofunctionalization with L-3,4-dihydroxyphenylalanine and collagen IV, 10 × 106 proximal tubule cells per mL are injected and cultured until experimental readout. A human-derived cell model can bridge all fiber-to-fiber distances to form a monolayer, whereas small-sized murine cells form monolayers on dense nanofiber meshes only. Fabricated constructs remain viable for at least 3 weeks and maintain functionality as shown by inhibitor-sensitive transport activity, which suggests clearance capacity for both negatively and positively charged solutes.


Asunto(s)
Células Epiteliales/citología , Túbulos Renales Proximales/citología , Túbulos Renales Proximales/cirugía , Ingeniería de Tejidos/métodos , Andamios del Tejido , Trasplantes/crecimiento & desarrollo , Materiales Biocompatibles/uso terapéutico , Caproatos/química , Proliferación Celular , Células Cultivadas , Humanos , Fallo Renal Crónico/cirugía , Lactonas/química , Polímeros
8.
Drug Metab Dispos ; 46(5): 592-599, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29514829

RESUMEN

Cisplatin is a cytostatic drug used for treatment of solid organ tumors. The main adverse effect is organic cation transporter 2 (OCT2)-mediated nephrotoxicity, observed in 30% of patients. The contribution of other renal drug transporters is elusive. Here, cisplatin-induced toxicity was evaluated in human-derived conditionally immortalized proximal tubule epithelial cells (ciPTEC) expressing renal drug transporters, including OCT2 and organic anion transporters 1 (OAT1) or 3 (OAT3). Parent ciPTEC demonstrated OCT2-dependent cisplatin toxicity (TC50 34 ± 1 µM after 24-hour exposure), as determined by cell viability. Overexpression of OAT1 and OAT3 resulted in reduced sensitivity to cisplatin (TC50 45 ± 6 and 64 ± 11 µM after 24-hour exposure, respectively). This effect was independent of OAT-mediated transport, as the OAT substrates probenecid and diclofenac did not influence cytotoxicity. Decreased cisplatin sensitivity in OAT-expressing cells was associated directly with a trend toward reduced intracellular cisplatin accumulation, explained by reduced OCT2 gene expression and activity. This was evaluated by Vmax of the OCT2-model substrate ASP+ (23.5 ± 0.1, 13.1 ± 0.3, and 21.6 ± 0.6 minutes-1 in ciPTEC-parent, ciPTEC-OAT1, and ciPTEC-OAT3, respectively). Although gene expression of cisplatin efflux transporter multidrug and toxin extrusion 1 (MATE1) was 16.2 ± 0.3-fold upregulated in ciPTEC-OAT1 and 6.1 ± 0.7-fold in ciPTEC-OAT3, toxicity was unaffected by the MATE substrate pyrimethamine, suggesting that MATE1 does not play a role in the current experimental set-up. In conclusion, OAT expression results in reduced cisplatin sensitivity in renal proximal tubule cells, explained by reduced OCT2-mediated uptake capacity. In vitro drug-induced toxicity studies should consider models that express both OCT and OAT drug transporters.


Asunto(s)
Cisplatino/farmacología , Expresión Génica/fisiología , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Proteínas de Transporte de Catión Orgánico/metabolismo , Transporte Biológico/efectos de los fármacos , Transporte Biológico/fisiología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Probenecid/farmacología
9.
Curr Pharm Des ; 23(26): 3845-3857, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28699526

RESUMEN

Facing the problems of limited renal regeneration capacity and the persistent shortage of donor kidneys, dialysis remains the only treatment option for many end-stage renal disease patients. Unfortunately, dialysis is only a medium-term solution because large and protein-bound uremic solutes are not efficiently cleared from the body and lead to disease progression over time. Current strategies for improved renal replacement therapies (RRTs) range from whole organ engineering to biofabrication of renal assist devices and biological injectables for in vivo regeneration. Notably, all approaches coincide with the incorporation of cellular components and biomimetic micro-environments. Concerning the latter, hydrogels form promising materials as scaffolds and cell carrier systems due to the demonstrated biocompatibility of most natural hydrogels, tunable biochemical and mechanical properties, and various application possibilities. In this review, the potential of hydrogel-based cell therapies for kidney regeneration is discussed. First, we provide an overview of current trends in the development of RRTs and in vivo regeneration options, before examining the possible roles of hydrogels within these fields. We discuss major application-specific hydrogel design criteria and, subsequently, assess the potential of emergent biofabrication technologies, such as micromolding, microfluidics and electrodeposition for the development of new RRTs and injectable stem cell therapies.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos/tendencias , Hidrogeles/administración & dosificación , Fallo Renal Crónico/terapia , Riñón/fisiología , Regeneración/fisiología , Animales , Materiales Biocompatibles/administración & dosificación , Materiales Biocompatibles/química , Tratamiento Basado en Trasplante de Células y Tejidos/métodos , Humanos , Hidrogeles/química , Riñón/efectos de los fármacos , Fallo Renal Crónico/fisiopatología , Regeneración/efectos de los fármacos , Terapia de Reemplazo Renal/métodos , Terapia de Reemplazo Renal/tendencias
11.
AAPS J ; 18(2): 465-75, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26821801

RESUMEN

Drug-induced nephrotoxicity still hampers drug development, because current translation from in vitro or animal studies to human lacks high predictivity. Often, renal adverse effects are recognized only during clinical stages of drug development. The current study aimed to establish a robust and a more complete human cell model suitable for screening of drug-related interactions and nephrotoxicity. In addition to endogenously expressed renal organic cation transporters and efflux transporters, conditionally immortalized proximal tubule epithelial cells (ciPTEC) were completed by transduction of cells with the organic anion transporter (OAT) 1 or OAT3. Fluorescence-activated cell sorting upon exposure to the OAT substrate fluorescein successfully enriched transduced cells. A panel of organic anions was screened for drug-interactions in ciPTEC-OAT1 and ciPTEC-OAT3. The cytotoxic response to the drug-interactions with antivirals was further examined by cell viability assays. Upon subcloning, concentration-dependent fluorescein uptake was found with a higher affinity for ciPTEC-OAT1 (Km = 0.8 ± 0.1 µM) than ciPTEC-OAT3 (Km = 3.7 ± 0.5 µM). Co-exposure to known OAT1 and/or OAT3 substrates (viz. para-aminohippurate, estrone sulfate, probenecid, furosemide, diclofenac, and cimetidine) in cultures spanning 29 passage numbers revealed relevant inhibitory potencies, confirming the robustness of our model for drug-drug interactions studies. Functional OAT1 was directly responsible for cytotoxicity of adefovir, cidofovir, and tenofovir, while a drug interaction with zidovudine was not associated with decreased cell viability. Our data demonstrate that human-derived ciPTEC-OAT1 and ciPTEC-OAT3 are promising platforms for highly predictive drug screening during early phases of drug development.


Asunto(s)
Antivirales/toxicidad , Túbulos Renales Proximales/efectos de los fármacos , Túbulos Renales Proximales/metabolismo , Proteína 1 de Transporte de Anión Orgánico/biosíntesis , Transportadores de Anión Orgánico Sodio-Independiente/biosíntesis , Células 3T3 , Adenina/análogos & derivados , Adenina/toxicidad , Animales , Línea Celular , Línea Celular Transformada , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Cidofovir , Citosina/análogos & derivados , Citosina/toxicidad , Relación Dosis-Respuesta a Droga , Predicción , Regulación de la Expresión Génica , Células HEK293 , Humanos , Ratones , Organofosfonatos/toxicidad
12.
ALTEX ; 15(5): 56-59, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-11178543

RESUMEN

Clostridium (C.) septicum, a spore-forming bacterium of the soil, is the classical causative agent of the malignant oedema, a fatal infection. As immunogenic compound, vaccines contain the toxoidized form of the soluble alpha-toxin, a lethal exoprotein. A cytotoxin inhibition test based upon cell culture for the detection of toxin neutralizing antibodies was developed as an alternative to the neutralisation test in mice, which has to be done as measure of quality control according to DAB 10. Sera derived from cattle that had been vaccinated with alpha-toxoid-vaccine, and sera from rabbits, from the official quality control of six different clostridial vaccines, were tested. The in vitro method was able to detect antibodies in the sera of the cows as well as in the sera of the rabbits. The results were reproducible.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...