Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
1.
F1000Res ; 10: 217, 2021.
Article in English | MEDLINE | ID: mdl-34631016

ABSTRACT

Background: Hyperuricemia is a pathological condition associated with risk factors of cardiovascular disease. In this study, three genetic polymorphisms were genotyped as predisposing factors of hyperuricemia. Methods: A total of 860 Mexicans between 18 and 25 years of age were genotyped for the ABCG2 (rs2231142), SLC22A12 (rs476037), and XDH (rs1042039) polymorphisms, as predisposing factors of hyperuricemia. Biochemical parameters were measured by spectrophotometry, while genetic polymorphisms were analyzed by real-time PCR. An analysis of the risk of hyperuricemia in relation to the variables studied was carried out using a logistic regression. Results: Male sex, being overweight or obese, having hypercholesterolemia or having hypertriglyceridemia were factors associated with hyperuricemia ( p ≤ 0.05). The ABCG2 polymorphism was associated with hyperuricemia (OR = 2.43, 95% CI: 1.41-4.17, p = 0.001) and hypercholesterolemia (OR = 4.89, 95% CI: 1.54-15.48, p = 0.003), employing a dominant model, but only in male participants. Conclusions: The ABCG2 (rs2231142) polymorphism increases the risk of hyperuricemia and hypercholesterolemia in young Mexican males.


Subject(s)
Hypercholesterolemia , Hyperuricemia , Organic Anion Transporters , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , Genetic Predisposition to Disease , Humans , Hypercholesterolemia/genetics , Hyperuricemia/genetics , Male , Neoplasm Proteins/genetics , Organic Anion Transporters/genetics , Organic Cation Transport Proteins/genetics , Polymorphism, Single Nucleotide , Uric Acid , Young Adult
2.
J Cardiovasc Pharmacol ; 78(6): 784-791, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34524257

ABSTRACT

ABSTRACT: The aim of the present work was to examine whether metabolic syndrome-like conditions in rats with fructose (F) overload modify the cardiotoxic effects induced by doxorubicin (DOX) and whether the treatment altered the expression of P-gp, breast cancer resistance protein, and organic cation/carnitine transporters in the heart. Male Sprague-Dawley rats received either tap water (control group [C]; n = 16) or water with F 10% wt/vol (n = 16) during 8 weeks. Three days before being killed, the animals received a single dose of DOX (6 mg/kg, ip, md) (C-DOX and F-DOX groups) or vehicle (VEH; ISS 1 mL/kg BW; ip) (C-VEH and F-VEH groups) (n = 8 per group). F overload enhanced thiobarbituric acid-reactive substance levels in the left ventricle, and DOX injection further increased those values. DOX did not alter thiobarbituric acid-reactive substance production in C animals. DOX caused a decrease of 30% in the ejection fraction and a nearly 40% reduction in the fractional shortening in F animals, but not in C rats. Cardiac tissue levels of P-gp decreased by about 30% in F rats compared with the C groups. DOX did not modify cardiac P-gp expression. Breast cancer resistance protein and organic cation/carnitine transporter (OCTN 1/2/3) protein levels did not change with either F or DOX. It is suggested that DOX could cause greater cardiotoxicity in rats receiving F, probably due to enhanced cardiac lipid peroxidation and lower expression of cardiac P-gp. These results support the hypothesis that the cardiotoxicity of DOX could be increased under metabolic syndrome-like conditions or in other health disorders that involve cardiovascular risk factors.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Antibiotics, Antineoplastic , Doxorubicin , Heart Diseases/chemically induced , Metabolic Syndrome/complications , Myocardium/metabolism , Oxidative Stress , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/genetics , ATP Binding Cassette Transporter, Subfamily G, Member 2/metabolism , Animals , Cardiotoxicity , Disease Models, Animal , Heart Diseases/metabolism , Heart Diseases/pathology , Heart Diseases/physiopathology , Lipid Peroxidation , Male , Metabolic Syndrome/metabolism , Myocardium/pathology , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Rats, Sprague-Dawley , Ventricular Function, Left/drug effects
3.
Biomed Pharmacother ; 142: 112009, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34388523

ABSTRACT

Genetic factors that affect variability in metformin response have been poorly studied in the Latin American population, despite its being the initial drug therapy for type 2 diabetes, one of the most prevalent diseases in that region. Metformin pharmacokinetics is carried out by members of the membrane transporters superfamily (SLCs), being the multidrug and toxin extrusion protein 1 (MATE1), one of the most studied. Some genetic variants in MATE1 have been associated with reduced in vitro metformin transport. They include rs77474263 p.[L125F], a variant present at a frequency of 13.8% in Latin Americans, but rare worldwide (less than 1%). Using exome sequence data and TaqMan genotyping, we revealed that the Mexican population has the highest frequency of this variant: 16% in Mestizos and 27% in Amerindians, suggesting a possible Amerindian origin. To elucidate the metformin pharmacogenetics, a children cohort was genotyped, allowing us to describe, for the first time, a MATE1 rs77474263 TT homozygous individual. An additive effect of the L125F variant was observed on blood metformin accumulation, revealing the highest metformin and lactate serum levels in the TT homozygote, and intermediate metformin values in the heterozygotes. Moreover, a molecular dynamics analysis suggested that the genetic variant effect on metformin efflux could be due to a decreased protein permeability. We conclude that pharmacogenetics could be useful in enhancing metformin pharmacovigilance in populations having a high frequency of the risk genotype, especially considering that these populations also have a higher susceptibility to the diseases for which metformin is the first-choice drug.


Subject(s)
Hypoglycemic Agents/pharmacokinetics , Metformin/pharmacokinetics , Organic Cation Transport Proteins/genetics , Pharmacogenetics , Adolescent , Adult , Child , Cohort Studies , Female , Genetic Variation , Genotype , Humans , Indians, North American/genetics , Lactic Acid/blood , Male , Mexico , Molecular Dynamics Simulation
4.
Mol Biol Rep ; 48(9): 6343-6348, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34383246

ABSTRACT

BACKGROUND: MATE2-K is an efflux transporter protein of organic cation expressed mainly in the kidney and encoded by the SLC47A2 gene. Different variants of this gene have shown an impact on the pharmacokinetics of various drugs, including metformin, which represents one of the most widely used drugs in treating type 2 diabetes. The SLC47A2 gene variants have been scarcely studied in Mexican populations, especially in Native American groups. For this reason, we analyzed the distribution of the variants rs12943590, rs35263947, and rs9900497 within the SLC47A2 gene in 173 Native Americans (Tarahumara, Huichol, Maya, Puerépecha) and 182 Mestizos (admixed) individuals from Mexico. METHODS AND RESULTS: Genotypes were determined through TaqMan probes (qPCR). The Hardy-Weinberg agreement was confirmed for all three SLC47A2 gene variants in all the Mexican populations analyzed. When worldwide populations were included for comparison purposes, for alleles and genotypes a relative interpopulation homogeneity was observed for rs35263947 (T allele; range 23.3-51.1%) and rs9900497 (T allele; range 18.6-40.9%). Conversely, heterogeneity was evident for rs12943590 (A allele, range 22.1-59.1%), where the most differentiated population was the Huichol, with high frequencies of the risk genotype associated with decreased response to metformin treatment (A/A = 40.9%). CONCLUSIONS: Although the SLC47A2 gene variants allow predicting favorable response to the metformin treatment in Mexican populations, the probable high frequency of ineffectiveness should be discarded in Huichols.


Subject(s)
American Indian or Alaska Native/genetics , Genetics, Population/methods , Indians, North American/genetics , Organic Cation Transport Proteins/genetics , Polymorphism, Single Nucleotide , Alleles , Diabetes Mellitus, Type 2/drug therapy , Diabetes Mellitus, Type 2/genetics , Gene Frequency , Haplotypes , Healthy Volunteers , Humans , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Mexico/ethnology , Plants, Medicinal , Treatment Outcome
5.
Pharmacology ; 106(11-12): 588-596, 2021.
Article in English | MEDLINE | ID: mdl-34265779

ABSTRACT

INTRODUCTION: Genetic variants could aid in predicting antidiabetic drug response by associating them with markers of glucose control, such as glycated hemoglobin (HbA1c). However, pharmacogenetic implementation for antidiabetics is still under development, as the list of actionable markers is being populated and validated. This study explores potential associations between genetic variants and plasma levels of HbA1c in 100 patients under treatment with metformin. METHODS: HbA1c was measured in a clinical chemistry analyzer (Roche), genotyping was performed in an Illumina-GSA array and data were analyzed using PLINK. Association and prediction models were developed using R and a 10-fold cross-validation approach. RESULTS: We identified genetic variants on SLC47A1, SLC28A1, ABCG2, TBC1D4, and ARID5B that can explain up to 55% of the interindividual variability of HbA1c plasma levels in diabetic patients under treatment. Variants on SLC47A1, SLC28A1, and ABCG2 likely impact the pharmacokinetics (PK) of metformin, while the role of the two latter can be related to insulin resistance and regulation of adipogenesis. CONCLUSIONS: Our results confirm previous genetic associations and point to previously unassociated gene variants for metformin PK and glucose control.


Subject(s)
DNA-Binding Proteins/genetics , Diabetes Mellitus, Type 2/drug therapy , GTPase-Activating Proteins/genetics , Glycated Hemoglobin/genetics , Hypoglycemic Agents/therapeutic use , Metformin/therapeutic use , Transcription Factors/genetics , Adult , Aged , Aged, 80 and over , Blood Pressure , Body Mass Index , Female , Genotype , Glycated Hemoglobin/analysis , Humans , Male , Middle Aged , Organic Cation Transport Proteins/genetics
6.
Int J Mol Sci ; 21(19)2020 Oct 08.
Article in English | MEDLINE | ID: mdl-33049997

ABSTRACT

Cisplatin is a chemotherapy drug widely used in the treatment of solid tumors. However, nephrotoxicity has been reported in about one-third of patients undergoing cisplatin therapy. Proximal tubules are the main target of cisplatin toxicity and cellular uptake; elimination of this drug can modulate renal damage. Organic transporters play an important role in the transport of cisplatin into the kidney and organic cations transporter 2 (OCT-2) has been shown to be one of the most important transporters to play this role. On the other hand, multidrug and toxin extrusion 1 (MATE-1) transporter is the main protein that mediates the extrusion of cisplatin into the urine. Cisplatin nephrotoxicity has been shown to be enhanced by increased OCT-2 and/or reduced MATE-1 activity. Peroxisome proliferator-activated receptor alpha (PPAR-α) is the transcription factor which controls lipid metabolism and glucose homeostasis; it is highly expressed in the kidneys and interacts with both MATE-1 and OCT-2. Considering the above, we treated wild-type and PPAR-α knockout mice with cisplatin in order to evaluate the severity of nephrotoxicity. Cisplatin induced renal dysfunction, renal inflammation, apoptosis and tubular injury in wild-type mice, whereas PPAR-α deletion protected against these alterations. Moreover, we observed that cisplatin induced down-regulation of organic transporters MATE-1 and OCT-2 and that PPAR-α deletion restored the expression of these transporters. In addition, PPAR-α knockout mice at basal state showed increased MATE-1 expression and reduced OCT-2 levels. Here, we show for the first time that PPAR-α deletion protects against cisplatin nephrotoxicity and that this protection is via modulation of the organic transporters MATE-1 and OCT-2.


Subject(s)
Antineoplastic Agents/adverse effects , Cisplatin/adverse effects , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/metabolism , PPAR alpha/genetics , Renal Insufficiency/chemically induced , Renal Insufficiency/metabolism , Animals , Apoptosis/drug effects , Apoptosis/genetics , Down-Regulation/drug effects , Kidney/drug effects , Kidney/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 2/genetics , PPAR alpha/metabolism , Severity of Illness Index , Signal Transduction/drug effects , Signal Transduction/genetics
7.
Int J Mol Sci ; 21(15)2020 Jul 30.
Article in English | MEDLINE | ID: mdl-32751747

ABSTRACT

Induced pluripotent stem (iPS) cells are laboratory-produced cells that combine the biological advantages of somatic adult and stem cells for cell-based therapy. The reprogramming of cells, such as fibroblasts, to an embryonic stem cell-like state is done by the ectopic expression of transcription factors responsible for generating embryonic stem cell properties. These primary factors are octamer-binding transcription factor 4 (Oct3/4), sex-determining region Y-box 2 (Sox2), Krüppel-like factor 4 (Klf4), and the proto-oncogene protein homolog of avian myelocytomatosis (c-Myc). The somatic cells can be easily obtained from the patient who will be subjected to cellular therapy and be reprogrammed to acquire the necessary high plasticity of embryonic stem cells. These cells have no ethical limitations involved, as in the case of embryonic stem cells, and display minimal immunological rejection risks after transplant. Currently, several clinical trials are in progress, most of them in phase I or II. Still, some inherent risks, such as chromosomal instability, insertional tumors, and teratoma formation, must be overcome to reach full clinical translation. However, with the clinical trials and extensive basic research studying the biology of these cells, a promising future for human cell-based therapies using iPS cells seems to be increasingly clear and close.


Subject(s)
Cellular Reprogramming/genetics , Induced Pluripotent Stem Cells/transplantation , Muscular Dystrophies/therapy , Gene Expression Regulation, Developmental/genetics , Humans , Induced Pluripotent Stem Cells/cytology , Kruppel-Like Factor 4 , Kruppel-Like Transcription Factors/genetics , Muscular Dystrophies/genetics , Muscular Dystrophies/pathology , Organic Cation Transport Proteins/genetics , Proto-Oncogene Mas , Proto-Oncogene Proteins c-myc/genetics , SOXB1 Transcription Factors/genetics
8.
J Clin Pharmacol ; 60(8): 1076-1086, 2020 08.
Article in English | MEDLINE | ID: mdl-32149389

ABSTRACT

Gabapentin (GBP) is an organic cation mainly eliminated unchanged in urine, and active drug secretion has been suggested to contribute to its renal excretion. Our objective was to evaluate the potential drug-drug interaction between GBP and cetirizine (CTZ), an inhibitor of transporters for organic cations. An open-label, 2-period, crossover, nonrandomized clinical trial was conducted in patients with neuropathic pain to evaluate the effect of CTZ on GBP pharmacokinetics. Twelve participants were treated with a single dose of 300 mg GBP (treatment A) or with 20 mg/d of CTZ for 5 days and 300 mg GBP on the last day of CTZ treatment (treatment B). Blood sampling and pain intensity evaluation were performed up to 36 hours after GBP administration. The interaction of GBP and CTZ with transporters for organic cations was studied in human embryonic kidney (HEK) cells expressing the organic cation transporters (OCTs), multidrug and toxin extrusion proteins (MATEs), and OCTN1. CTZ treatment resulted in reduced area under the concentration-time curve and peak concentration compared with treatment A. In treatment B, the lower plasma concentrations of GBP resulted in reduced pain attenuation. GBP renal clearance was similar between treatments. GBP has low apparent affinity for OCT2 (concentration of an inhibitor where the response [or binding] is reduced by half [IC50 ] 237 µmol/L) and a high apparent affinity for hMATE1 (IC50 1.1 nmol/L), hMATE2-K (IC50 39 nmol/L), and hOCTN1 (IC50 2.1 nmol/L) in HEK cells. At therapeutic concentrations, CTZ interacts with hMATE1 and OCTN1. In summary, CTZ reduced the systemic exposure to GBP and its effect on neuropathic pain attenuation. However, CTZ × GBP interaction is not mediated by the renal transporters.


Subject(s)
Analgesics/pharmacokinetics , Cetirizine/metabolism , Cetirizine/pharmacokinetics , Gabapentin/pharmacokinetics , Organic Cation Transport Proteins/metabolism , Adult , Analgesics/administration & dosage , Analgesics/blood , Analgesics/urine , Area Under Curve , Cations/metabolism , Cetirizine/administration & dosage , Cross-Over Studies , Drug Interactions , Female , Gabapentin/administration & dosage , Gabapentin/blood , Gabapentin/urine , HEK293 Cells , Humans , Male , Middle Aged , Neuralgia/drug therapy , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 2/genetics , Pain Measurement/drug effects , Polymorphism, Genetic , Renal Elimination/drug effects , Symporters/genetics , Symporters/metabolism
9.
Molecules ; 25(2)2020 Jan 17.
Article in English | MEDLINE | ID: mdl-31963528

ABSTRACT

Metformin is the first-line drug for type 2 diabetes mellitus control. It is established that this drug traffics through OCT-2 and MATE-1 transporters in kidney tubular cells and is excreted in its unaltered form in the urine. Hereby, we provide evidence that points towards the metformin-dependent upregulation of OCT-2 and MATE-1 in the kidney via the transcription factor proliferator-activated receptor alpha (PPARα). Treatment of wild type mice with metformin led to the upregulation of the expression of OCT-2 and MATE-1 by 34% and 157%, respectively. An analysis in a kidney tubular cell line revealed that metformin upregulated PPARα and OCT-2 expression by 37% and 299% respectively. MK-886, a PPARα antagonist, abrogated the OCT-2 upregulation by metformin and reduced MATE-1 expression. Conversely, gemfibrozil, an agonist of PPARα, elicited the increase of PPARα, OCT-2, and MATE-1 expression by 115%, 144%, and 376%, respectively. PPARα knockout mice failed to upregulate both the expression of OCT-2 and MATE-1 in the kidney upon metformin treatment, supporting the PPARα-dependent metformin upregulation of the transporters in this organ. Taken together, our data sheds light on the metformin-induced mechanism of transporter modulation in the kidney, via PPARα, and this effect may have implications for drug safety and efficacy.


Subject(s)
Kidney/chemistry , Metformin/administration & dosage , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 2/genetics , PPAR alpha/genetics , Animals , Cell Line , Gemfibrozil/pharmacology , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Indoles/pharmacology , Kidney/drug effects , Male , Metformin/pharmacology , Mice , Up-Regulation/drug effects
10.
Basic Clin Pharmacol Toxicol ; 124(3): 266-272, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30192429

ABSTRACT

Gabapentin (GAB) is eliminated unchanged in urine, and organic cation transporters (OCT2 and OCTN1) have been shown to play a role in GAB renal excretion. This prospective clinical study aimed to evaluate the genetic polymorphisms effect on GAB pharmacokinetic (PK) variability using a population pharmacokinetic approach. Data were collected from 53 patients with chronic pain receiving multiple doses of GAB. Patients were genotyped for SLC22A2 c.808G>T and SLC22A4 c.1507C>T polymorphisms. Both polymorphisms' distribution followed the Hardy-Weinberg equilibrium. An one-compartment model with first-order absorption and linear elimination best described the data. The absorption rate constant, volume of distribution, and clearance estimated were 0.44 h-1 , 86 L, and 17.3 × (estimated glomerular filtration ratio/89.58)1.04  L/h, respectively. The genetic polymorphism SLC22A4 c.1507C>T did not have a significant influence on GAB absorption, distribution or elimination. Due to the low minor allelic frequency of SLC22A2 c.808G>T, further studies require higher number of participants to confirm its effect on GAB renal elimination. In conclusion, GAB clinical pharmacokinetics are strongly influenced by renal function and absorption process, but not by the OCTN1 (SLC22A4 c.1507C>T) polymorphism.


Subject(s)
Chronic Pain/drug therapy , Chronic Pain/genetics , Gabapentin/pharmacokinetics , Organic Cation Transport Proteins/genetics , Organic Cation Transporter 2/genetics , Adult , Aged , Analgesics/pharmacokinetics , Chronic Pain/metabolism , Female , Gene Frequency , Humans , Male , Middle Aged , Organic Cation Transport Proteins/metabolism , Organic Cation Transporter 2/metabolism , Pharmacogenetics , Polymorphism, Single Nucleotide , Prospective Studies , Symporters
11.
Br J Clin Pharmacol ; 84(5): 987-996, 2018 05.
Article in English | MEDLINE | ID: mdl-29352482

ABSTRACT

AIMS: To identify pharmacogenetic and demographic variables that influence the systemic exposure to metformin in an admixed Brazilian cohort. METHODS: The extreme discordant phenotype was used to select 106 data sets from nine metformin bioequivalence trials, comprising 256 healthy adults. Eleven single-nucleotide polymorphisms in SLC22A1, SLC22A2, SLC47A1 SLC47A2 and in transcription factor SP1 were genotyped and a validated panel of ancestry informative markers was used to estimate the individual proportions of biogeographical ancestry. Two-step (univariate followed by multivariate) regression modelling was developed to identify covariates associated with systemic exposure to metformin, accessed by the area under the plasma concentration-time curve, between 0 and 48 h (AUC0-48h ), after single oral doses of metformin (500 or 1000 mg). RESULTS: The individual proportions of African, Amerindian and European ancestry varied widely, as anticipated from the structure of the Brazilian population The dose-adjusted, log-transformed AUC0-48h 's (ng h ml-1  mg-1 ) differed largely in the two groups at the opposite ends of the distribution histogram, namely 0.82, 0.79-0.85 and 1.08, 1.06-1.11 (mean, 95% confidence interval; P = 6.10-26 , t test). Multivariate modelling revealed that metformin AUC0-48h increased with age, food and carriage of rs12208357 in SLC22A1 but was inversely associated with body surface area and individual proportions of African ancestry. CONCLUSIONS: A pharmacogenetic marker in OCT1 (SLC22A1 rs12208357), combined with demographic covariates (age, body surface area and individual proportion of African ancestry) and a food effect explained 29.7% of the variability in metformin AUC0-48h .


Subject(s)
Black People/genetics , Indians, South American/genetics , Metformin/pharmacokinetics , Organic Cation Transport Proteins/genetics , White People/genetics , Adult , Brazil , Demography , Female , Genotype , Haplotypes , Humans , Linkage Disequilibrium/genetics , Male , Metformin/blood , Phenotype , Polymorphism, Single Nucleotide , Randomized Controlled Trials as Topic , Sp1 Transcription Factor/genetics
12.
Biometals ; 31(1): 69-80, 2018 02.
Article in English | MEDLINE | ID: mdl-29178026

ABSTRACT

The viability, cellular uptake and subcellular distribution of heavy metal Hg, were determined in human mammary cell lines (MCF-7, MDA-MB-231 and MCF-10A). It was observed that Hg had the capacity of being excluded from the cells with a different type of possible transporters. MCF-7 cells showed the lowest viability, while the other two cell lines were much more resistant to Hg treatments. The intracellular concentration of Hg was higher at lower exposure times in MCF-10A cells and MCF-7 cells; but as the time was increased only MDA-MB-231 showed the capacity to continue introducing the metal. In MCF-7 and MCF-10A cells the subcellular distribution of Hg was higher in cytosolic fraction than nucleus and membrane, but MDA-MB-231 showed membrane and nucleus fraction as the enriched one. The analysis of RNA-seq about the genes or family of genes that encode proteins which are related to cytotoxicity of Hg evidenced that MCF-10A cells and MCF-7 cells could have an active transport to efflux the metal. On the contrary, in MDA-MB-231 no genes that could encode active transporters have been found.


Subject(s)
Cell Membrane/drug effects , Epithelial Cells/drug effects , Mercury/metabolism , Organic Cation Transport Proteins/genetics , Cations, Divalent , Cell Line , Cell Membrane/metabolism , Cell Nucleus/drug effects , Cell Nucleus/metabolism , Cell Survival/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Gene Expression Profiling , Gene Expression Regulation , Humans , Ion Transport , Kinetics , MCF-7 Cells , Mammary Glands, Human/drug effects , Mammary Glands, Human/metabolism , Mammary Glands, Human/pathology , Mercury/toxicity , Organ Specificity , Organic Cation Transport Proteins/classification , Organic Cation Transport Proteins/metabolism , Signal Transduction
13.
J Bioenerg Biomembr ; 49(6): 423-435, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29128917

ABSTRACT

Our understanding of nicotinamide adenine dinucleotide mitochondrial transporter 1 (Ndt1A) in Aspergillus fumigatus remains poor. Thus, we investigated whether Ndt1A could alter fungi survival. To this end, we engineered the expression of an Ndt1A-encoding region in a Δndt1Δndt2 yeast strain. The resulting cloned Ndt1A protein promoted the mitochondrial uptake of nicotinamide adenine dinucleotide (NAD+), generating a large mitochondrial membrane potential. The NAD+ carrier utilized the electrochemical proton gradient to drive NAD+ entrance into mitochondria when the mitochondrial membrane potential was sustained by succinate. Its uptake has no impact on oxidative stress, and Ndt1A expression improved growth and survival of the Δndt1Δndt2 Saccharomyces cerevisiae strain.


Subject(s)
Aspergillus fumigatus/chemistry , Mitochondria/metabolism , Organic Cation Transport Proteins/genetics , Saccharomyces cerevisiae/genetics , Gene Deletion , Heterografts , Membrane Potential, Mitochondrial , Mitochondrial Proteins , NAD/metabolism , Nucleotide Transport Proteins , Oxidative Stress , Saccharomyces cerevisiae/growth & development , Saccharomyces cerevisiae Proteins/genetics
14.
Mol Cell Biochem ; 428(1-2): 101-108, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28161805

ABSTRACT

Cisplatin is a drug widely used in chemotherapy that frequently causes severe renal dysfunction. Organic transporters have an important role to control the absorption and excretion of cisplatin in renal cells. Deletion and blockage of kinin B1 receptor has already been show to protect against cisplatin-induced acute kidney injury. To test whether it exerts its protective function by modulating the organic transporters in kidney, we studied kinin B1 receptor knockout mice and treatment with a receptor antagonist at basal state and in presence of cisplatin. Cisplatin administration caused downregulation of renal organic transporters; in B1 receptor knockout mice, this downregulation of organic transporters in kidney was absent; and treatment by a B1 receptor antagonist attenuated the downregulation of the transporter MATE-1. Moreover, kinin B1 receptor deletion and blockage at basal state resulted in higher renal expression of MATE-1. Moreover we observed that kinin B1 receptor deletion and blockage result in less accumulation of platinum in renal tissue. Thus, we propose that B1 receptor deletion and blockage protect the kidney from cisplatin-induced acute kidney injury by upregulating the expression of MATE-1, thereby increasing the efflux of cisplatin from renal cells.


Subject(s)
Acute Kidney Injury/prevention & control , Bradykinin B1 Receptor Antagonists/pharmacology , Cisplatin/pharmacokinetics , Organic Cation Transport Proteins/genetics , Receptor, Bradykinin B1/genetics , Acute Kidney Injury/chemically induced , Acute Kidney Injury/genetics , Animals , Cisplatin/administration & dosage , Cisplatin/adverse effects , Disease Models, Animal , Gene Expression Regulation/drug effects , Gene Knockout Techniques , Kidney/drug effects , Kidney/metabolism , Male , Mice , Organic Cation Transport Proteins/metabolism , Receptor, Bradykinin B1/metabolism
15.
Sex Dev ; 11(5-6): 225-237, 2017.
Article in English | MEDLINE | ID: mdl-29393262

ABSTRACT

Androgen insensitivity syndrome (AIS) is a hereditary condition in patients with a 46,XY karyotype in which loss-of-function mutations of the androgen receptor (AR) gene are responsible for defects in virilization. The aim of this study was to investigate the consequences of the lack of AR activity on germ cell survival and the degree of testicular development reached by these patients by analyzing gonadal tissue from patients with AIS prior to Sertoli cell maturation at puberty. Twenty-three gonads from 13 patients with AIS were assessed and compared to 18 testes from 17 subjects without endocrine disorders. The study of the gonadal structure using conventional microscopy and the ultrastructural characteristics of remnant germ cells using electron microscopy, combined with the immunohistochemical analysis of specific germ cell markers (MAGE-A4 for premeiotic germ cells and of OCT3/4 for gonocytes), enabled us to carry out a thorough investigation of germ cell life in an androgen-insensitive microenvironment throughout prepuberty until young adulthood. Here, we show that germ cell degeneration starts very early, with a marked decrease in number after only 2 years of life, and we demonstrate the permanence of gonocytes in AIS testis samples until puberty, describing 2 different populations. Additionally, our results provide further evidence for the importance of AR signaling in peritubular myoid cells during prepuberty to maintain Sertoli and spermatogonial cell health and survival.


Subject(s)
Androgen-Insensitivity Syndrome/pathology , Puberty/metabolism , Puberty/physiology , Androgen-Insensitivity Syndrome/metabolism , Antigens, Neoplasm/genetics , Antigens, Neoplasm/metabolism , Child , Child, Preschool , Germ Cells/metabolism , Humans , Immunohistochemistry , Infant , Male , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Receptors, Androgen/genetics , Receptors, Androgen/metabolism , Spermatogonia/metabolism , Spermatogonia/pathology , Testis/metabolism , Testis/pathology
16.
PLoS One ; 9(2): e90339, 2014.
Article in English | MEDLINE | ID: mdl-24587332

ABSTRACT

Impaired L-carnitine uptake correlates with higher blood pressure in adult men, and L-carnitine restores endothelial function in aortic rings from spontaneously hypertensive rat (SHR). Thus, endothelial dysfunction in hypertension could result from lower L-carnitine transport in this cell type. L-Carnitine transport is mainly mediated by novel organic cation transporters 1 (Octn1, Na(+)-independent) and 2 (Octn2, Na(+)-dependent); however, their kinetic properties and potential consequences in hypertension are unknown. We hypothesize that L-carnitine transport kinetic properties will be altered in aortic endothelium from spontaneously hypertensive rats (SHR). L-Carnitine transport was measured at different extracellular pH (pHo 5.5-8.5) in the absence or presence of sodium in rat aortic endothelial cells (RAECs) from non-hypertensive Wistar-Kyoto (WKY) rats and SHR. Octn1 and Octn2 mRNA relative expression was also determined. Dilation of endothelium-intact or denuded aortic rings in response to calcitonine gene related peptide (CGRP, 0.1-100 nmol/L) was measured (myography) in the absence or presence of L-carnitine. Total L-carnitine transport was lower in cells from SHR compared with WKY rats, an effect due to reduced Na(+)-dependent (Na(+) dep ) compared with Na(+)-independent (Na(+) indep ) transport components. Saturable L-carnitine transport kinetics show maximal velocity (V max), without changes in apparent K m for Na(+) indep transport in SHR compared with WKY rats. Total and Na(+) dep component of transport were increased, but Na(+) indep transport was reduced by extracellular alkalization in WKY rats. However, alkalization reduced total and Na(+) indep transport in cells from SHR. Octn2 mRNA was higher than Octn-1 mRNA expression in cells from both conditions. Dilation of artery rings in response to CGRP was reduced in vessels from SHR compared with WKY rats. CGRP effect was endothelium-dependent and restored by L-carnitine. All together these results suggest that reduced L-carnitine transport (likely via Na(+)-dependent Octn2) could limit this compound's potential beneficial effects in RAECs from SHR.


Subject(s)
Aorta/metabolism , Carnitine/metabolism , Endothelial Cells/metabolism , Endothelium, Vascular/metabolism , Hypertension/metabolism , Animals , Aorta/drug effects , Aorta/pathology , Biological Transport , Blood Pressure , Calcitonin Gene-Related Peptide/pharmacology , Carrier Proteins/genetics , Carrier Proteins/metabolism , Cells, Cultured , Endothelial Cells/drug effects , Endothelial Cells/pathology , Endothelium, Vascular/drug effects , Endothelium, Vascular/pathology , Gene Expression , Humans , Hydrogen-Ion Concentration , Hypertension/pathology , Hypertension/physiopathology , Kinetics , Male , Membrane Proteins/genetics , Membrane Proteins/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Sodium/metabolism , Solute Carrier Family 22 Member 5 , Solute Carrier Proteins , Symporters , Tissue Culture Techniques , Vasodilation/drug effects
17.
Biol Res ; 47: 72, 2014 Dec 17.
Article in English | MEDLINE | ID: mdl-25723857

ABSTRACT

BACKGROUND: Acetylcholine (ACh) is known to be a key neurotransmitter in the central and peripheral nervous systems, which is also produced in a variety of non-neuronal tissues and cell. The existence of ACh in maxilla in vivo and potential regulation role for osteogenesis need further study. RESULTS: Components of the cholinergic system (ACh, esterase, choline acetyltransferase, high-affinity choline uptake, n- and mAChRs) were determined in maxilla of rat in vivo, by means of Real-Time PCR and immunohistochemistry. Results showed RNA for CarAT, carnitine/acylcarnitine translocase member 20 (Slc25a20), VAChT, OCTN2, OCT1, OCT3, organic cation transporter member 4 (Slc22a4), AChE, BChE, nAChR subunits α1, α2, α3, α5, α7, α10, ß1, ß2, ß4, γ and mAChR subunits M1, M2, M3, M4, M5 were detected in rat's maxilla. RNA of VAChT, AChE, nAChR subunits α2, ß1, ß4 and mAChR subunits M4 had abundant expression (2(-ΔCt) > 0.03). Immunohistochemical staining was conducted for ACh, VAChT, nAChRα7 and AChE. ACh was expressed in mesenchymal cells, chondroblast, bone and cartilage matrix and bone marrow cells, The VAChT expression was very extensively while ACh receptor α7 was strongly expressed in newly formed bone matrix of endochondral and bone marrow ossification, AchE was found only in mesenchymal stem cells, cartilage and bone marrow cells. CONCLUSIONS: ACh might exert its effect on the endochondral and bone marrow ossification, and bone matrix mineralization in maxilla.


Subject(s)
Acetylcholine/metabolism , Bone Marrow/physiology , Cartilage/physiology , Cholinergic Agents/metabolism , Maxilla/metabolism , Animals , Bone Marrow Cells/metabolism , Bone Matrix/metabolism , Calcification, Physiologic/physiology , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Gene Expression Regulation/physiology , Immunohistochemistry , Male , Maxilla/cytology , Mesenchymal Stem Cells/metabolism , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Osteogenesis/physiology , Rats, Sprague-Dawley , Real-Time Polymerase Chain Reaction , Receptors, Nicotinic/genetics , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/metabolism
18.
Biol. Res ; 47: 1-7, 2014. ilus, graf, tab
Article in English | LILACS | ID: biblio-950768

ABSTRACT

BACKGROUND: Acetylcholine (ACh) is known to be a key neurotransmitter in the central and peripheral nervous systems, which is also produced in a variety of non-neuronal tissues and cell. The existence of ACh in maxilla in vivo and potential regulation role for osteogenesis need further study. RESULTS: Components of the cholinergic system (ACh, esterase, choline acetyltransferase, high-affinity choline uptake, n- and mAChRs) were determined in maxilla of rat in vivo, by means of Real-Time PCR and immunohistochemistry. Results showed RNA for CarAT, carnitine/acylcarnitine translocase member 20 (Slc25a20), VAChT, OCTN2, OCT1, OCT3, organic cation transporter member 4 (Slc22a4), AChE, BChE, nAChR subunits α1, α2, α3, α5, α7, α10, ß1, ß2, ß4, γ and mAChR subunits M1, M2, M3, M4, M5 were detected in rat's maxilla. RNA of VAChT, AChE, nAChR subunits α2, ß1, ß4 and mAChR subunits M4 had abundant expression (2(-ΔCt) > 0.03). Immunohistochemical staining was conducted for ACh, VAChT, nAChRα7 and AChE. ACh was expressed in mesenchymal cells, chondroblast, bone and cartilage matrix and bone marrow cells, The VAChT expression was very extensively while ACh receptor α7 was strongly expressed in newly formed bone matrix of endochondral and bone marrow ossification, AchE was found only in mesenchymal stem cells, cartilage and bone marrow cells. CONCLUSIONS: ACh might exert its effect on the endochondral and bone marrow ossification, and bone matrix mineralization in maxilla.


Subject(s)
Animals , Male , Rats , Bone Marrow/physiology , Acetylcholine/metabolism , Cartilage/physiology , Cholinergic Agents/metabolism , Maxilla/metabolism , Osteogenesis/physiology , Bone Matrix/metabolism , Calcification, Physiologic/physiology , Bone Marrow Cells/metabolism , Immunohistochemistry , Carnitine Acyltransferases/genetics , Carnitine Acyltransferases/metabolism , Gene Expression Regulation/physiology , Receptors, Nicotinic/genetics , Rats, Sprague-Dawley , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Vesicular Acetylcholine Transport Proteins/genetics , Vesicular Acetylcholine Transport Proteins/metabolism , Mesenchymal Stem Cells/metabolism , Real-Time Polymerase Chain Reaction , Maxilla/cytology
19.
Bol Asoc Med P R ; 105(3): 43-4, 2013.
Article in English | MEDLINE | ID: mdl-24282920

ABSTRACT

Rarely screened in psychiatric patients, primary and/or secondary Carnitine deficiency could be influencing and/or mimicking the mood symptoms of our patient population. The brain and specifically neurons are highly vulnerable to impairments in oxidative metabolism, which can lead to neuronal cell death and disorders of neurotransmitters causing changes in cognition and behavior. For this reason, identification of this disorder is important since its treatment could result in symptom improvement and better quality of life of our patients. We present a case where exacerbation of mood symptoms was associated to primary and secondary Carnitine deficiency.


Subject(s)
Antimanic Agents/adverse effects , Attention Deficit Disorder with Hyperactivity/complications , Carnitine/deficiency , Hyperammonemia/psychology , Ketoglutarate Dehydrogenase Complex/antagonists & inhibitors , Mood Disorders/complications , Organic Cation Transport Proteins/deficiency , Valproic Acid/adverse effects , Adult , Antidepressive Agents/therapeutic use , Attention Deficit Disorder with Hyperactivity/blood , Benzodiazepines/therapeutic use , Carnitine/therapeutic use , Citalopram/therapeutic use , Depressive Disorder/complications , Depressive Disorder/drug therapy , Disruptive, Impulse Control, and Conduct Disorders/complications , Drug Substitution , Drug Therapy, Combination , Humans , Hyperammonemia/chemically induced , Hyperammonemia/genetics , Lorazepam/therapeutic use , Male , Mood Disorders/blood , Olanzapine , Organic Cation Transport Proteins/genetics , Solute Carrier Family 22 Member 5 , Valproic Acid/pharmacology , Valproic Acid/therapeutic use
20.
Mini Rev Med Chem ; 9(13): 1518-26, 2009 Nov 01.
Article in English | MEDLINE | ID: mdl-20205633

ABSTRACT

Carnitine is well-known for its role in the transport of fatty acids to the mitochondrial matrix, where beta-oxidation takes place. This work describes novel functions for this compound and novel data on its pharmacokinetics.


Subject(s)
Acyl Coenzyme A/metabolism , Carnitine/metabolism , Acetylation , Animals , Apoptosis , Biological Transport , Carnitine/pharmacokinetics , Humans , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/physiology , Solute Carrier Family 22 Member 5
SELECTION OF CITATIONS
SEARCH DETAIL