Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 3 de 3
Filter
Add more filters










Database
Language
Publication year range
1.
Sci Rep ; 8(1): 2940, 2018 02 13.
Article in English | MEDLINE | ID: mdl-29440697

ABSTRACT

Histone H3 serine 28 (H3S28) phosphorylation and de-repression of polycomb repressive complex (PRC)-mediated gene regulation is linked to stress conditions in mitotic and post-mitotic cells. To better understand the role of H3S28 phosphorylation in vivo, we studied a Drosophila strain with ectopic expression of constitutively-activated H3S28A, which prevents PRC2 binding at H3S28, thus mimicking H3S28 phosphorylation. H3S28A mutants showed prolonged life span and improved resistance against starvation and paraquat-induced oxidative stress. Morphological and functional analysis of heart tubes revealed smaller luminal areas and thicker walls accompanied by moderately improved cardiac function after acute stress induction. Whole-exome deep gene-sequencing from isolated heart tubes revealed phenotype-corresponding changes in longevity-promoting and myotropic genes. We also found changes in genes controlling mitochondrial biogenesis and respiration. Analysis of mitochondrial respiration from whole flies revealed improved efficacy of ATP production with reduced electron transport-chain activity. Finally, we analyzed posttranslational modification of H3S28 in an experimental heart failure model and observed increased H3S28 phosphorylation levels in HF hearts. Our data establish a critical role of H3S28 phosphorylation in vivo for life span, stress resistance, cardiac and mitochondrial function in Drosophila. These findings may pave the way for H3S28 phosphorylation as a putative target to treat stress-related disorders such as heart failure.


Subject(s)
Drosophila melanogaster/genetics , Ectopic Gene Expression , Heart/physiology , Histones/genetics , Longevity/genetics , Mutation , Stress, Physiological/genetics , Alleles , Animals , Drosophila melanogaster/physiology , Histones/metabolism , Phosphorylation/genetics , Transcription, Genetic
2.
Pharmacogenomics J ; 12(1): 22-9, 2012 Feb.
Article in English | MEDLINE | ID: mdl-20921968

ABSTRACT

After uptake into liver cells, the antiemetic drugs tropisetron and ondansetron undergo metabolic inactivation by cytochrome P450 2D6 (CYP2D6). We investigated whether the hepatic organic cation transporter 1 (OCT1; SLC22A1) mediates cellular uptake and whether common OCT1 loss-of-function polymorphisms affect pharmacokinetics and efficacy of both drugs. Both tropisetron and ondansetron inhibited ASP(+) uptake in OCT1-overexpressing HEK293 cells. Overexpression of wild-type, but not OCT1 loss-of-function variants, significantly increased tropisetron uptake. Correspondingly, patients with two loss-of-function OCT1 alleles had higher tropisetron plasma concentrations (n=59, P<0.04) and higher clinical efficacy (n=91, P=0.009) compared with carriers of fully active OCT1. Overexpression of OCT1 did not increase ondansetron uptake. Nevertheless, OCT1 genotypes correlated with pharmacokinetics (n=45, P<0.05) and clinical efficacy (n=222, P<0.02) of ondansetron, the effect size of OCT1 genotypes on pharmacokinetics and efficacy was greater for tropisetron than for ondansetron. In conclusion, in addition to the known effects of CYP2D6, OCT1 deficiency may increase efficacy of tropisetron and potentially of ondansetron by limiting their hepatic uptake.


Subject(s)
Cell Membrane/metabolism , Indoles/therapeutic use , Ondansetron/therapeutic use , Organic Cation Transporter 1/genetics , Polymorphism, Genetic/genetics , Serotonin 5-HT3 Receptor Antagonists/therapeutic use , Adolescent , Adult , Aged , Aged, 80 and over , Antiemetics/blood , Antiemetics/pharmacology , Antiemetics/therapeutic use , Cell Membrane/drug effects , Female , HEK293 Cells , Humans , Indoles/blood , Indoles/pharmacology , Male , Middle Aged , Ondansetron/blood , Ondansetron/pharmacology , Serotonin 5-HT3 Receptor Antagonists/blood , Serotonin 5-HT3 Receptor Antagonists/pharmacology , Treatment Outcome , Tropisetron , Young Adult
3.
Clin Pharmacol Ther ; 90(1): 143-50, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21562485

ABSTRACT

We investigated whether tramadol or its active metabolite, O-desmethyltramadol, are substrates of the organic cation transporter OCT1 and whether polymorphisms in OCT1 affect tramadol and O-desmethyltramadol pharmacokinetics. Tramadol showed high permeability through parallel artificial membrane permeability assays (PAMPAs). Tramadol uptake in HEK293 cells did not change after OCT1 overexpression, and the concentrations of tramadol in the plasma of healthy volunteers were independent of their OCT1 genotypes. In contrast, O-desmethyltramadol showed low membrane permeability, and OCT1 overexpression increased O-desmethyltramadol uptake 2.4-fold. This increase in uptake was reversed by OCT1 inhibitors and absent when loss-of-function OCT1 variants were overexpressed. Volunteers carrying loss-of-function OCT1 polymorphisms had significantly higher plasma concentrations of O-desmethyltramadol (P = 0.002, n = 41) and significantly prolonged miosis, a surrogate marker of opioidergic effects (P = 0.005, n = 24). In conclusion, polymorphisms in OCT1 influence the pharmacokinetics of O-desmethyltramadol, presumably by affecting its uptake into liver cells, and thus may modulate the efficacy of tramadol treatment.


Subject(s)
Analgesics, Opioid/pharmacokinetics , Organic Cation Transporter 1/genetics , Organic Cation Transporter 1/metabolism , Tramadol/pharmacokinetics , Adolescent , Adult , Algorithms , Alleles , Area Under Curve , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2D6/genetics , Genotype , HEK293 Cells , Humans , Kinetics , Membranes, Artificial , Miosis/chemically induced , Polymorphism, Genetic , Pupil/drug effects , Tramadol/analogs & derivatives , Tramadol/metabolism , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...