Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
1.
J Vet Pharmacol Ther ; 46(6): 401-412, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37198956

RESUMEN

The ATP-binding cassette transporter P-glycoprotein (P-gp) limits the oral bioavailability of many drugs. Although P-gp has been well studied in humans and mice, little is known about the substrate specificities of many of its species orthologs. To address this, we performed in vitro analysis of P-gp transporter function using HEK293 cells stably expressing human, ovine, porcine, canine, and feline P-gp. We also employed a human physiologically based pharmacokinetic (PBPK) model to assess variations in digoxin exposure resulting from altered P-gp function. Compared to human P-gp, sheep P-gp had significantly less digoxin efflux (2.3-fold ±0.04 vs. 1.8-fold ±0.03, p < .0001) and all species orthologs had significantly less quinidine efflux compared with human P-gp (p < .05). Human P-gp also had significantly greater efflux of talinolol compared to sheep and dog P-gp (1.9-fold ±0.04 vs. 1.6-fold ±0.06, p = .003 and 1.6-fold ±0.05, p = .0002, respectively). P-gp expression protected all lines against paclitaxel-induced toxicity, with sheep P-gp being significantly less protective. The inhibitor verapamil demonstrated dose-dependent inhibition of all P-gp orthologs. Finally, a PBPK model showed digoxin exposure was sensitive to altered P-gp activity. Overall, our study found that species differences in this major drug transporter exist and that the appropriate species ortholog of P-gp should be evaluated during veterinary drug development.


Asunto(s)
Miembro 1 de la Subfamilia B de Casetes de Unión a ATP , Digoxina , Humanos , Animales , Perros , Gatos , Ovinos , Ratones , Porcinos , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/genética , Miembro 1 de la Subfamilia B de Casetes de Unión a ATP/metabolismo , Células HEK293 , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Digoxina/metabolismo , Verapamilo
2.
Pharm Res ; 39(7): 1599-1613, 2022 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-35089508

RESUMEN

INTRODUCTION: The organic cation transporter 3 (OCT3, SLC22A3) is ubiquitously expressed and interacts with a wide array of compounds including endogenous molecules, environmental toxins and prescription drugs. Understudied as a determinant of pharmacokinetics and pharmacodynamics, OCT3 has the potential to be a major determinant of drug absorption and disposition and to be a target for drug-drug interactions (DDIs). GOAL: The goal of the current study was to identify prescription drug inhibitors of OCT3. METHODS: We screened a compound library consisting of 2556 prescription drugs, bioactive molecules, and natural products using a high throughput assay in HEK-293 cells stably expressing OCT3. RESULTS: We identified 210 compounds that at 20 µM inhibit 50% or more of OCT3-mediated uptake of 4-Di-1-ASP (2 µM). Of these, nine were predicted to inhibit the transporter at clinically relevant unbound plasma concentrations. A Structure-Activity Relationship (SAR) model included molecular descriptors that could discriminate between inhibitors and non-inhibitors of OCT3 and was used to identify additional OCT3 inhibitors. Proteomics of human brain microvessels (BMVs) indicated that OCT3 is the highest expressed OCT in the human blood-brain barrier (BBB). CONCLUSIONS: This study represents the largest screen to identify prescription drug inhibitors of OCT3. Several are sufficiently potent to inhibit the transporter at therapeutic unbound plasma levels, potentially leading to DDIs or off-target pharmacologic effects.


Asunto(s)
Proteínas de Transporte de Catión Orgánico , Medicamentos bajo Prescripción , Cationes , Células HEK293 , Ensayos Analíticos de Alto Rendimiento , Humanos , Proteínas de Transporte de Catión Orgánico/antagonistas & inhibidores
3.
Differentiation ; 102: 40-52, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30059908

RESUMEN

Presenilins (Psen1 and Psen2 in mice) are polytopic transmembrane proteins that act in the γ-secretase complex to make intra-membrane cleavages of their substrates, including the well-studied Notch receptors. Such processing releases the Notch intracellular domain, allowing it to physically relocate from the cell membrane to the nucleus where it acts in a transcriptional activating complex to regulate downstream genes in the signal-receiving cell. Previous studies of Notch pathway mutants for Jagged1, Notch2, and Rbpj demonstrated that canonical signaling is a necessary component of normal mouse lens development. However, the central role of Psens within the γ-secretase complex has never been explored in any developing eye tissue or cell type. By directly comparing Psen single and double mutant phenotypes during mouse lens development, we found a stronger requirement for Psen1, although both genes are needed for progenitor cell growth and to prevent apoptosis. We also uncovered a novel genetic interaction between Psen1 and Jagged1. By quantifying protein and mRNA levels of key Notch pathway genes in Psen1/2 or Jagged1 mutant lenses, we identified multiple points in the overall signaling cascade where feedback regulation can occur. Our data are consistent with the loss of particular genes indirectly influencing the transcription level of another. However, we conclude that regulating Notch2 protein levels is particularly important during normal signaling, supporting the importance of post-translational regulatory mechanisms in this tissue.


Asunto(s)
Membrana Celular/metabolismo , Cristalino/metabolismo , Presenilinas/genética , Receptor Notch2/metabolismo , Receptores Notch/genética , Transducción de Señal , Animales , Ciclo Celular/genética , Cristalino/embriología , Ratones Transgénicos , Receptor Notch2/genética , Transducción de Señal/genética
4.
Curr Med Chem ; 30(12): 1406-1419, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36065926

RESUMEN

Given the importance of COVID-19-induced ARDS, recently, researchers have strived to determine underlying mechanisms involved in the inflammatory responses. In this regard, inflammasomes possess a distinct priority for cytokine storm occurrence and, subsequently, ARDS progression in ill patients with SARS-CoV-2 infection. In this minireview, the characteristics of known inflammasome inhibitors and designed research in this field were concretely deciphered.


Asunto(s)
COVID-19 , Síndrome de Dificultad Respiratoria , Humanos , COVID-19/complicaciones , Inflamasomas , SARS-CoV-2 , Proteína con Dominio Pirina 3 de la Familia NLR , Síndrome de Dificultad Respiratoria/tratamiento farmacológico
5.
Clin Pharmacol Ther ; 114(2): 381-392, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37151050

RESUMEN

Trimethoprim is predicted to inhibit several thiamine transporters, including the primary thiamine intestinal absorptive transporter, ThTR-2, and the hepatic and renal organic cation transporters, OCT1, OCT2, and MATEs. To investigate the effect of trimethoprim on thiamine absorption, studies were conducted in cells, mice, and healthy volunteers and supported by use of real-world data. In a randomized, crossover clinical study, seven healthy volunteers were given a single oral dose of thiamine or thiamine plus trimethoprim, followed by blood sampling. The thiamine area under the curve (AUC) increased with trimethoprim co-administration (P value = 0.031). Similar results were seen in mice. Trimethoprim appeared to act on thiamine absorption through inhibition of hepatic OCT1 as evidenced from its ability to modulate levels of isobutyrylcarnitine and propionylcarnitine, OCT1 biomarkers identified from metabolomic analyses. Real-world data further supported this finding, showing an association between trimethoprim use and higher levels of triglycerides, LDL cholesterol, and total cholesterol, consistent with OCT1 inhibition (P values: 2.2 × 10-16 , 5.75 × 10-7 , and 5.82 × 10-7 , respectively). These findings suggest that trimethoprim increases plasma levels of thiamine by inhibiting hepatic OCT1. Trimethoprim reduced urinary excretion and clearance of biomarkers for OCT2 and MATEs, consistent with inhibition of renal organic cation transporters. This inhibition did not appear to play a role in the observed increases in thiamine levels. This study highlights the potential for drug-nutrient interactions involving transporters, in addition to transporters' established role in drug-drug interactions.


Asunto(s)
Tiamina , Trimetoprim , Animales , Ratones , Humanos , Tiamina/farmacología , Trimetoprim/farmacología , Proteínas de Transporte de Membrana , Interacciones Alimento-Droga , Biomarcadores , Nutrientes , Cationes , Proteínas de Transporte de Catión Orgánico , Transportador 2 de Cátion Orgánico , Células HEK293
6.
Biol Open ; 8(3)2019 Mar 26.
Artículo en Inglés | MEDLINE | ID: mdl-30890522

RESUMEN

Notch signaling regulates a multitude of cellular processes. During ocular lens development this pathway is required for lens progenitor growth, differentiation and maintenance of the transition zone. After ligand-receptor binding, the receptor proteins are processed, first by ADAM proteases, then by γ-secretase cleavage. This results in the release of a Notch intracellular domain (N-ICD), which is recruited into a nuclear transcription factor complex that activates Notch target genes. Previous in vitro studies showed that the Delta-like and Jagged ligand proteins can also be cleaved by the γ-secretase complex, but it remains unknown whether such processing occurs during in vivo vertebrate development. Here we show that mouse and human lens progenitor cells endogenously express multiple Jagged1 protein isoforms, including a Jagged1 intracellular domain. We also found that pharmacologic blockage of γ-secretase activity in vitro resulted in an accumulation of Jagged1 polypeptide intermediates. Finally, overexpression of an epitope-tagged Jagged1 intracellular domain displayed nuclear localization and induced the upregulation of endogenous JAG1 mRNA expression. These findings support the idea that along with its classical role as a Notch pathway ligand, Jagged1 is regulated post-translationally, to produce multiple active protein isoforms.

7.
J Clin Neurosci ; 43: 77-81, 2017 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-28506742

RESUMEN

According to evidence, Alzheimer's disease is known as one of the most serious neurodegenerative diseases, for which hypertension has been observed to be a key risk factor. Therefore, this study aims to examine the relationship between the PIN1 and eNOS genes expression, as well as serum levels and hypertension in Alzheimer's disease sufferers. Blood samples were obtained from subjects who were divided into four groups: the control group, normotensive Alzheimer's patients, the Alzheimer's sufferers group with hypertension, and the healthy group with only hypertension, considering the inhibition of confounding factors. Thereafter, eNOS and PIN1 genes expression along with serum levels were studied. Based on the obtained results, a statistically significant correlation didn't exist between serum level of PIN1 and the systolic and diastolic blood pressure, between serum level of eNOS and diastolic blood pressure in the norm tension Alzheimer's disease patients, between serum levels of PIN1, eNOS and systolic blood pressure, and between serum eNOS and systolic and diastolic blood pressure in the patients with hypertension (p<0.05). According to the results obtained from this study, measuring the serum levels of eNOS and Pin1 may contribute to the prognosis, prevention, and monitoring of hypertension and also to the reduction of death rates from cardiovascular diseases in Alzheimer's disease.


Asunto(s)
Enfermedad de Alzheimer/sangre , Expresión Génica , Hipertensión/sangre , Peptidilprolil Isomerasa de Interacción con NIMA/sangre , Óxido Nítrico Sintasa de Tipo III/sangre , Anciano , Enfermedad de Alzheimer/complicaciones , Estudios de Casos y Controles , Femenino , Humanos , Hipertensión/complicaciones , Masculino , Persona de Mediana Edad , Proyectos Piloto , Factores de Riesgo
8.
World J Plast Surg ; 5(1): 39-44, 2016 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-27308239

RESUMEN

BACKGROUND: Different methods for dressing of donor site of skin graft in burn patients have similarly pain, limitation of mobility of donor site and local complications such as infection and scar. Amniotic membrane has used for improvement of healing in some wounds. Accordingly in this study amnion was used as biologic dressing for donor site of skin graft to evaluate it's efficacy in improvement of pain, move score and the risk of local infection. METHODS: Study was done as clinical trial over 32 admitted patients in burn department of Beasat hospital. Amnion was prepared in elective caesarean section after rule out any placental site for risk of torch and viral infection. Skin graft was taken from two sites in every patient. One site dressed with amnion and another with routine dressing. Then two sites were compared about severity of pain, move score, infection and time of dressing sloughing. RESULTS: Fourteen patients were women and 18 men. Mean score of pain and movement up to fourth and fifth post operative day respectively was less than control site. No difference is seen about infection and dressing slough in two sites. CONCLUSION: It seems use of amnion for dressing of donor site probably cause rapid epithelialisation and wound healing and can improve pain and move score in early post operative days. Accordingly it is expected to need less analgesia and low rate of immobilization and following complications and earlier discharge of patients.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA