Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Food Funct ; 13(16): 8489-8499, 2022 Aug 15.
Article in English | MEDLINE | ID: mdl-35876245

ABSTRACT

The benefits of fruit and vegetable dietary consumption are largely defined in epidemiological terms. Relatively little is known about the discrete effects on metabolic pathways elicited by individual dietary fruits and vegetables. To address this, grape powder was added to both a standard and a high-fat Western pattern diet given to 10-week-old female C57BL/6J mice for a period of 91 days, whereupon 24 h urines were collected and the mice euthanized after a 12 h fast for the collection of liver tissue. Alterations in hepatic and urinary metabolite patterns were determined by gas chromatography-mass spectrometry-based metabolomics. Urinary excretion of the gut microbiota metabolites 4-hydroxyphenylacetic acid, 5-hydroxyindole, glyceric acid, gluconic acid and myo-inositol was attenuated when grape was added to the standard diet but the gut microbiota metabolites gluconic acid, scyllo-inositol, mannitol, xylitol, 5-hydroxyindole and 2-deoxyribonic acid were increased in urine when grape was added to the high-fat diet. Increased hepatic ascorbic acid and 5-oxoproline levels indicated the anti-oxidant effect of grape powder on the liver. Pathway enrichment analysis demonstrated that for both standard and high-fat diets, grape addition significantly upregulated the malate-aspartate shuttle indicating enhanced hepatic utilization of glucose via cytosolic glycolysis for mitochondrial ATP production. It is concluded that a grape diet reprogrammes gut microbiota metabolism, attenuates the hepatic oxidative stress of a high-fat diet and increases the efficiency of glucose utilization by the liver for energy production.


Subject(s)
Diet, High-Fat , Vitis , Animals , Diet, High-Fat/adverse effects , Female , Glucose/metabolism , Liver/metabolism , Mice , Mice, Inbred C57BL , Powders/metabolism
2.
Cardiooncology ; 7(1): 17, 2021 May 11.
Article in English | MEDLINE | ID: mdl-33975650

ABSTRACT

BACKGROUND: The CBR3 V244M single nucleotide polymorphism has been linked to the risk of anthracycline-related cardiomyopathy in survivors of childhood cancer. There have been limited prospective studies examining the impact of CBR3 V244M on the risk for anthracycline-related cardiotoxicity in adult cohorts. OBJECTIVES: This study evaluated the presence of associations between CBR3 V244M genotype status and changes in echocardiographic parameters in breast cancer patients undergoing doxorubicin treatment. METHODS: We recruited 155 patients with breast cancer receiving treatment with doxorubicin (DOX) at Roswell Park Comprehensive Care Center (Buffalo, NY) to a prospective single arm observational pharmacogenetic study. Patients were genotyped for the CBR3 V244M variant. 92 patients received an echocardiogram at baseline (t0 month) and at 6 months (t6 months) of follow up after DOX treatment. Apical two-chamber and four-chamber echocardiographic images were used to calculate volumes and left ventricular ejection fraction (LVEF) using Simpson's biplane rule by investigators blinded to all patient data. Volumetric indices were evaluated by normalizing the cardiac volumes to the body surface area (BSA). RESULTS: Breast cancer patients with CBR3 GG and AG genotypes both experienced a statistically significant reduction in LVEF at 6 months following initiation of DOX treatment for breast cancer compared with their pre-DOX baseline study. Patients homozygous for the CBR3 V244M G allele (CBR3 V244) exhibited a further statistically significant decrease in LVEF at 6 months following DOX therapy in comparison with patients with heterozygous AG genotype. We found no differences in age, pre-existing cardiac diseases associated with myocardial injury, cumulative DOX dose, or concurrent use of cardioprotective medication between CBR3 genotype groups. CONCLUSIONS: CBR3 V244M genotype status is associated with changes in echocardiographic parameters suggestive of early anthracycline-related cardiomyopathy in subjects undergoing chemotherapy for breast cancer.

3.
J Subst Abuse Treat ; 104: 7-14, 2019 09.
Article in English | MEDLINE | ID: mdl-31370987

ABSTRACT

Medication-assisted behavior treatment for alcohol use disorder (AUD) holds promise to enhance the efficacy of medication and of behavior therapy when administered individually. The present study examines the treatment benefit of combined outpatient naltrexone (NTX) treatment with Alcoholics Anonymous Facilitation (AAF) behavior therapy, in the context of OPRM1 genotype. The minor OPRM1 Asp40 G-allele has been associated with greater positive reinforcing effects of alcohol consumption and greater alcohol craving, suggesting that individuals carrying the OPRM1 G allele may have an improved naltrexone response. Twenty patients, including 7 G-allele carriers, received 90 days of naltrexone with medication support and dispensing sessions, and ten AAF behavior therapy sessions. During treatment and the eight-week posttreatment follow-up, an overall increase in percent days abstinent was observed for the sample as a whole, but G-allele carriers reported relatively heavier drinking relative to other subjects. These findings suggest that this enhanced medication-assisted behavior treatment is a promising therapeutic combination, and mirror other recent findings that G-allele carriers may require more intensive treatment.


Subject(s)
Alcohol Deterrents/pharmacology , Alcoholism/genetics , Alcoholism/therapy , Behavior Therapy , Naltrexone/pharmacology , Outcome Assessment, Health Care , Receptors, Opioid, mu/genetics , Adult , Alcoholism/drug therapy , Combined Modality Therapy , Female , Genetic Variation , Humans , Male , Middle Aged
4.
Exp Mol Pathol ; 110: 104268, 2019 10.
Article in English | MEDLINE | ID: mdl-31201803

ABSTRACT

Down syndrome (trisomy 21) is characterized by genome-wide imbalances that result in a range of phenotypic manifestations. Altered expression of DYRK1A in the trisomic context has been linked to some Down syndrome phenotypes. DYRK1A regulates the splicing of cardiac troponin (TNNT2) through a pathway mediated by the master splicing factor SRSF6. Here, we documented the expression of the DYRK1A-SRSF6-TNNT2 pathway in a collection of myocardial samples from persons with and without Down syndrome. Results suggest that "gene dosage effect" may drive the expression of DYRK1A mRNA but has no effect on DYRK1A protein levels in trisomic myocardium. The levels of phosphorylated DYRK1A-Tyr321 tended to be higher (~35%) in myocardial samples from donors with Down syndrome. The levels of phosphorylated SRSF6 were 2.6-fold higher in trisomic myocardium. In line, the expression of fetal TNNT2 variants was higher in myocardial tissue with trisomy 21. These data provide a representative picture on the extent of inter-individual variation in myocardial DYRK1A-SRSF6-TNNT2 expression in the context of Down syndrome.


Subject(s)
Down Syndrome , Fetal Heart/metabolism , Myocardium/metabolism , Phosphoproteins/genetics , Protein Serine-Threonine Kinases/genetics , Protein-Tyrosine Kinases/genetics , Serine-Arginine Splicing Factors/genetics , Troponin T/genetics , Adult , Aged , Aged, 80 and over , Child , Child, Preschool , Female , Gene Expression Profiling/methods , Humans , Infant , Male , Middle Aged , Phosphoproteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Protein-Tyrosine Kinases/metabolism , Serine-Arginine Splicing Factors/metabolism , Signal Transduction/genetics , Troponin T/metabolism , Young Adult , Dyrk Kinases
5.
Toxicol Lett ; 307: 11-16, 2019 Jun 01.
Article in English | MEDLINE | ID: mdl-30817976

ABSTRACT

Aldo-Keto Reductase Family 7 Member A2 (AKR7A2) is the most abundant anthracycline metabolizing enzyme in human myocardium. Myocardial AKR7A2 contributes to the synthesis of cardiotoxic C-13 anthracycline alcohol metabolites (e.g., doxorubicinol). The factors that govern the transcription of human AKR7A2 in cardiomyocytes remain largely unexplored. In this study, we performed the functional characterization of the AKR7A2 gene promoter in human AC16 cardiomyocytes. Experiments with gene reporter constructs and chromatin immunoprecipitation assays suggest that NF-κB binds to specific regions in the AKR7A2 promoter. Doxorubicin treatment modified the cellular levels of NF-κB and the expression of AKR7A2. Moreover, doxorubicin treatment led to changes in the pattern of AKR7A2 phosphorylation status. Our results suggest that AKR7A2 expression in human cardiomyocytes is mediated by NF-κB through conserved response elements in the proximal gene promoter region. This study provides the first insights into the functional characteristics of the human AKR7A2 gene promoter.


Subject(s)
Aldehyde Reductase/metabolism , Myocytes, Cardiac/metabolism , Chromatin Immunoprecipitation , Doxorubicin/pharmacology , Gene Expression Regulation , Humans , Myocytes, Cardiac/enzymology , NF-kappa B/metabolism , Phosphorylation , Promoter Regions, Genetic
6.
Biopharm Drug Dispos ; 39(6): 315-318, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29851133

ABSTRACT

Loxoprofen is an anti-inflammatory drug that requires bioactivation into the trans-OH metabolite to exert pharmacological activity. Evidence suggests that carbonyl reductase 1 (CBR1) is important during the bioactivation of loxoprofen. This study examined the impact of the functional single nucleotide polymorphism CBR1 rs9024 on the bioactivation of loxoprofen in a collection of human liver samples. The synthesis ratios of trans-OH loxoprofen/cis-OH loxoprofen were 33% higher in liver cytosols from donors homozygous for the CBR1 rs9024 G allele in comparison with the ratios in samples from donors with heterozygous GA genotypes. Complementary studies examined the impact of CBR1 rs9024 on the bioactivation of loxoprofen in lymphoblastoid cell lines. CBR1 rs9024 genotype status impacts the synthesis of the bioactive trans-OH metabolite of loxoprofen in human liver.


Subject(s)
Alcohol Oxidoreductases/metabolism , Gene Expression Regulation, Enzymologic/physiology , Genotype , Liver/metabolism , Phenylpropionates/metabolism , Polymorphism, Single Nucleotide , Alcohol Oxidoreductases/genetics , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Cell Line, Tumor , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism
7.
Gene ; 628: 286-294, 2017 Sep 10.
Article in English | MEDLINE | ID: mdl-28735727

ABSTRACT

The ERBB2 gene encodes a transmembrane tyrosine kinase receptor that belongs to the epidermal growth factor receptor (EGFR) family. ERBB2 plays a pivotal role during heart development and is essential for normal cardiac function, particularly during episodes of cardiac stress. The monoclonal antibody drug trastuzumab is used for the therapy of breast cancers that overexpress ERBB2. The clinical use of trastuzumab is limited by the development of cardiotoxicity in some patients. Inter-individual differences in the expression of ERBB2 in cardiac tissue may impact the risk of cardiotoxicity. In this study, we examined whether DNA methylation status in the proximal promoter region of ERBB2 is associated to variable ERBB2 mRNA and ERBB2 protein expression in human myocardium. Complementary studies with ERBB2 gene reporter constructs and chromatin immunoprecipitation suggest that differential methylation in specific CpG sites modify the binding of Sp1 to the promoter of ERBB2. DNA methylation in the ERBB2 locus may contribute to the variable expression of ERBB2 in human myocardium.


Subject(s)
DNA Methylation , Gene Expression Regulation , Myocardium/metabolism , Receptor, ErbB-2/genetics , Binding Sites , Cell Line , Cell Survival/genetics , CpG Islands , Genetic Loci , Humans , Promoter Regions, Genetic , Protein Binding , RNA, Messenger/genetics , Receptor, ErbB-2/metabolism , Sp1 Transcription Factor/metabolism , Transcriptional Activation
8.
Gene ; 592(1): 209-214, 2016 Oct 30.
Article in English | MEDLINE | ID: mdl-27506315

ABSTRACT

The clinical use of anthracyclines to treat various canine cancers is limited by the development of cardiotoxicity. The intra-cardiac synthesis of anthracycline C-13 alcohol metabolites (e.g. daunorubicinol) contributes to the development of cardiotoxicity. Canine carbonyl reductase 1 (cbr1) catalyzes the reduction of daunorubicin into daunorubicinol. Recent mapping of the cbr1 locus by sequencing DNA samples from dogs from various breeds revealed a cluster of conserved motifs for the transcription factor Sp1 in the putative promoter region of cbr1. We hypothesized that the variable number of Sp1 motifs could impact the transcription of canine cbr1. In this study, we report the functional characterization of the canine cbr1 promoter. Experiments with reporter constructs and chromatin immunoprecipitation show that cbr1 transcription depends on the binding of Sp1 to the proximal promoter. Site-directed mutagenesis experiments suggest that the variable number of Sp1 motifs impacts the transcription of canine cbr1. Inhibition of Sp1-DNA binding decreased canine cbr1 mRNA levels by 54% in comparison to controls, and also decreased enzymatic carbonyl reductase activity for the substrates daunorubicin (16%) and menadione (23%). The transactivation of Sp1 increased the expression of cbr1 mRNA (67%), and increased carbonyl reductase activity for daunorubicin (35%) and menadione (27%). These data suggest that the variable number of Sp1 motifs in the canine cbr1 promoter may impact the pharmacodynamics of anthracyclines in canine cancer patients.


Subject(s)
Alcohol Oxidoreductases/genetics , Sp1 Transcription Factor/metabolism , Transcriptional Activation , Alcohol Oxidoreductases/metabolism , Animals , Dogs , Madin Darby Canine Kidney Cells , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism
9.
J Clin Oncol ; 34(8): 863-70, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26811534

ABSTRACT

PURPOSE: Interindividual variability in the dose-dependent association between anthracyclines and cardiomyopathy suggests that genetic susceptibility could play a role. The current study uses an agnostic approach to identify genetic variants that could modify cardiomyopathy risk. METHODS: A genome-wide association study was conducted in childhood cancer survivors with and without cardiomyopathy (cases and controls, respectively). Single-nucleotide polymorphisms (SNPs) that surpassed a prespecified threshold for statistical significance were independently replicated. The possible mechanistic significance of validated SNP(s) was sought by using healthy heart samples. RESULTS: No SNP was marginally associated with cardiomyopathy. However, SNP rs1786814 on the CELF4 gene passed the significance cutoff for gene-environment interaction (Pge = 1.14 × 10(-5)). Multivariable analyses adjusted for age at cancer diagnosis, sex, anthracycline dose, and chest radiation revealed that, among patients with the A allele, cardiomyopathy was infrequent and not dose related. However, among those exposed to greater than 300 mg/m(2) of anthracyclines, the rs1786814 GG genotype conferred a 10.2-fold (95% CI, 3.8- to 27.3-fold; P < .001) increased risk of cardiomyopathy compared with those who had GA/AA genotypes and anthracycline exposure of 300 mg/m(2) or less. This gene-environment interaction was successfully replicated in an independent set of anthracycline-related cardiomyopathy. CUG-BP and ETR-3-like factor proteins control developmentally regulated splicing of TNNT2, the gene that encodes for cardiac troponin T (cTnT), a biomarker of myocardial injury. Coexistence of more than one cTnT variant results in a temporally split myofilament response to calcium, which causes decreased contractility. Analysis of TNNT2 splicing variants in healthy human hearts suggested an association between the rs1786814 GG genotype and coexistence of more than one TNNT2 splicing variant (90.5% GG v 41.7% GA/AA; P = .005). CONCLUSION: We report a modifying effect of a polymorphism of CELF4 (rs1786814) on the dose-dependent association between anthracyclines and cardiomyopathy, which possibly occurs through a pathway that involves the expression of abnormally spliced TNNT2 variants.


Subject(s)
Anthracyclines/adverse effects , CELF Proteins/genetics , Cardiomyopathies/chemically induced , Cardiomyopathies/genetics , Adolescent , Adult , Cardiomyopathies/metabolism , Case-Control Studies , Child , Child, Preschool , Dose-Response Relationship, Drug , Female , Genome-Wide Association Study , Humans , Infant , Infant, Newborn , Male , Polymorphism, Single Nucleotide , Protein Isoforms , Troponin T/biosynthesis , Troponin T/genetics , Young Adult
10.
Neurotox Res ; 29(3): 345-50, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26194616

ABSTRACT

The ankyrin repeat and kinase domain containing 1 (ANKK1) TaqIA polymorphism has been extensively studied as a marker of the gene for dopamine receptor D2 (DRD2) in addictions and other dopamine-associated traits. In vitro mRNA and protein studies have shown a potential connection between ANKK1 and the dopaminergic system functioning. Here, we have investigated whether Ankk1 expression in the brain is regulated by treatment with dopaminergic agonists. We used quantitative RT-PCR of total brain and Western blots of specific brain areas to study Ankk1 in murine brain after dopaminergic treatments. We found that Ankk1 mRNA was upregulated after activation of D1R-like dopamine receptors with SKF38393 (2.660 ± 1.035-fold; t: 4.066, df: 11, P = 0.002) and apomorphine (2.043 ± 0.595-fold; t: 3.782, df: 8, P = 0.005). The D2R-like agonist quinelorane has no effect upon Ankk1 mRNA (1.004 ± 0.580-fold; t: 0.015, df: 10, P = 0.9885). In contrast, mice treatment with the D2R-like agonists 7-OH-DPAT and aripiprazole caused a significant Ankk1 mRNA downregulation (0.606 ± 0.057-fold; t: 2.786, df: 10, P = 0.02 and 0.588 ± 0.130-fold; t: 2.394, df: 11, P = 0.036, respectively). With respect the Ankk1 proteins profile, no effects were found after SKF38393 (t: 0.54, df: 2, P = 0.643) and Quinelorane (t: 0.286, df: 8, P = 0.782) treatments. In contrast, the D2R-like agonist 7-OH-DPAT (±) caused a significant increment of Ankk1 in the striatum (t: 2.718, df: 7; P = 0.03) when compared to the prefrontal cortex. The activation of D1R-like and D2-R-like leads to opposite transcriptional regulation of Ankk1 by specific pathways.


Subject(s)
Brain/metabolism , Gene Expression Regulation/drug effects , Protein Serine-Threonine Kinases/metabolism , Receptors, Dopamine D1/metabolism , Receptors, Dopamine D2/metabolism , 2,3,4,5-Tetrahydro-7,8-dihydroxy-1-phenyl-1H-3-benzazepine/administration & dosage , Animals , Apomorphine , Aripiprazole , Brain/drug effects , Down-Regulation/drug effects , Male , Mice , Mice, Inbred C57BL , Protein Serine-Threonine Kinases/genetics , Quinolines , RNA, Messenger/metabolism , Receptors, Dopamine D1/agonists , Receptors, Dopamine D2/agonists , Tetrahydronaphthalenes , Up-Regulation/drug effects
11.
Cardiovasc Toxicol ; 16(2): 182-92, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25962911

ABSTRACT

The intracardiac synthesis of anthracycline alcohol metabolites by aldo-keto reductases (AKRs) contributes to the pathogenesis of anthracycline-related cardiotoxicity. AKR7A2 is the most abundant anthracycline reductase in hearts from donors with and without Down syndrome (DS), and its expression varies between individuals (≈tenfold). We investigated whether DNA methylation impacts AKR7A2 expression in hearts from donors with (n = 11) and without DS (n = 30). Linear models were used to test for associations between methylation status and cardiac AKR7A2 expression. In hearts from donors without DS, DNA methylation status at CpG site -865 correlated with AKR7A2 mRNA (Pearson's regression coefficient, r = -0.4051, P = 0.0264) and AKR7A2 protein expression (r = -0.5818, P = 0.0071). In heart tissue from donors with DS, DNA methylation status at CpG site -232 correlated with AKR7A2 protein expression (r = 0.8659, P = 0.0025). Multiple linear regression modeling revealed that methylation at several CpG sites is associated with the synthesis of cardiotoxic daunorubicinol. AKR7A2 methylation status in lymphoblastoid cell lines from donors with and without DS was examined to explore potential parallelisms between cardiac tissue and lymphoid cells. These results suggest that DNA methylation impacts AKR7A2 expression and the synthesis of cardiotoxic daunorubicinol.


Subject(s)
Aldehyde Reductase/metabolism , Anthracyclines/metabolism , DNA Methylation/physiology , Down Syndrome/enzymology , Myocardium/enzymology , Aged , Down Syndrome/diagnosis , Female , Heart/physiology , Humans , Male , Middle Aged
12.
Article in English | MEDLINE | ID: mdl-24938108

ABSTRACT

Cancer patients with Down syndrome (DS) are at increased risk for anthracycline-related cardiotoxicity. Mitochondrial DNA (mtDNA) alterations in hearts with-DS may contribute to anthracycline-related cardiotoxicity. Cardiac mtDNA and the mtDNA(4977) deletion were quantitated in samples with- (n = 11) and without-DS (n = 31). Samples with-DS showed 30% lower mtDNA (DS(MT-ND1/18Sratio): 1.48 ± 0.72 versus non-DS(MT-ND1/18Sratio): 2.10 ± 1.59; p = 0.647) and 30% higher frequency of the mtDNA(4977) deletion (DS(% frequency mtDNA(4977)) deletion: 0.0086 ± 0.0166 versus non-DS(% frequency mtDNA(4977)) deletion: 0.0066 ± 0.0124, p = 0.514) than samples without-DS. The BACH1 and microRNA-155 (miR-155) genes are located in chromosome 21, and their products have demonstrated roles during oxidative stress. BACH1 and miR-155 expression did not differ in hearts with- and without-DS. An association between BACH1 and miR-155 expression was detected in hearts without-DS, suggesting alterations between BACH1-miR-155 interactions in the DS settings.


Subject(s)
Anthracyclines/adverse effects , Antibiotics, Antineoplastic/adverse effects , Basic-Leucine Zipper Transcription Factors/genetics , DNA, Mitochondrial/genetics , Down Syndrome/genetics , Fanconi Anemia Complementation Group Proteins/genetics , MicroRNAs/genetics , Adolescent , Adult , Aged , Anthracyclines/therapeutic use , Antibiotics, Antineoplastic/therapeutic use , Base Sequence , Basic-Leucine Zipper Transcription Factors/biosynthesis , Cardiotoxicity , Child , Child, Preschool , Chromosomes, Human, Pair 21 , Fanconi Anemia Complementation Group Proteins/biosynthesis , Female , Genes, Mitochondrial , Heart Diseases/chemically induced , Heart Diseases/genetics , Heart Diseases/pathology , Humans , Infant , Leukemia, Myeloid, Acute/drug therapy , Male , MicroRNAs/biosynthesis , Middle Aged , Mitochondria/drug effects , Myocardium/metabolism , Oxidative Stress/genetics , Oxidative Stress/physiology , Pilot Projects , RNA, Messenger/genetics , Sequence Analysis, DNA , Sequence Deletion/genetics , Young Adult
13.
Biochim Biophys Acta ; 1852(7): 1420-7, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25869329

ABSTRACT

The regulation of mitochondrial biogenesis is under the control of nuclear genes including the master Mitochondrial Transcription Factor A (TFAM). Recent evidence suggests that the expression of TFAM is regulated by microRNAs (miRNAs) in various cellular contexts. Here, we show that hsa-miR-155-5p, a prominent miRNA encoded in chromosome 21, controls the expression of TFAM at the post-transcriptional level. In human fibroblasts derived from a diploid donor, downregulation of TFAM by hsa-miR-155-5p decreased mitochondrial DNA (mtDNA) content. In contrast, downregulation of TFAM by hsa-miR-155-5p did not decrease mtDNA content in fibroblasts derived from a donor with Down syndrome (DS, trisomy 21). In line, downregulation of mitochondrial TFAM levels through hsa-miR-155-5p decreased mitochondrial mass in diploid fibroblasts but not in trisomic cells. Due to the prevalence of mitochondrial dysfunction and cardiac abnormalities in subjects with DS, we examined the presence of potential associations between hsa-miR-155-5p and TFAM expression in heart samples from donors with and without DS. There were significant negative associations between hsa-miR-155-5p and TFAM expression in heart samples from donors with and without DS. These results suggest that regulation of TFAM by hsa-miR-155-5p impacts mitochondrial biogenesis in the diploid setting but not in the DS setting.


Subject(s)
DNA-Binding Proteins/genetics , MicroRNAs/genetics , Mitochondrial Proteins/genetics , Mitochondrial Turnover , Transcription Factors/genetics , Animals , CHO Cells , Case-Control Studies , Cells, Cultured , Chromosomes, Human, Pair 21/genetics , Cricetinae , Cricetulus , DNA, Mitochondrial/genetics , DNA-Binding Proteins/metabolism , Down Syndrome/genetics , Fibroblasts/metabolism , Humans , MicroRNAs/metabolism , Mitochondrial Proteins/metabolism , Myocytes, Cardiac/metabolism , Ploidies , Transcription Factors/metabolism
14.
Pharm Res ; 31(7): 1644-55, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24562808

ABSTRACT

PURPOSE: The intracardiac synthesis of anthracycline alcohol metabolites (e.g., daunorubicinol) contributes to the pathogenesis of anthracycline-related cardiotoxicity. Cancer patients with Down syndrome (DS) are at increased risk for anthracycline-related cardiotoxicity. We profiled the expression of anthracycline metabolizing enzymes in hearts from donors with- and without- DS. METHODS: Cardiac expression of CBR1, CBR3, AKR1A1, AKR1C3 and AKR7A2 was examined by quantitative real time PCR, quantitative immunoblotting, and enzyme activity assays using daunorubicin. The CBR1 polymorphism rs9024 was investigated by allelic discrimination with fluorescent probes. The contribution of CBRs/AKRs proteins to daunorubicin reductase activity was examined by multiple linear regression. RESULTS: CBR1 was the most abundant transcript (average relative expression; DS: 81%, non-DS: 58%), and AKR7A2 was the most abundant protein (average relative expression; DS: 38%, non-DS: 35%). Positive associations between cardiac CBR1 protein levels and daunorubicin reductase activity were found for samples from donors with- and without- DS. Regression analysis suggests that sex, CBR1, AKR1A1, and AKR7A2 protein levels were significant contributors to cardiac daunorubicin reductase activity. CBR1 rs9024 genotype status impacts on cardiac CBR1 expression in non-DS hearts. CONCLUSIONS: CBR1, AKR1A1, and AKR7A2 protein levels point to be important determinants for predicting the synthesis of cardiotoxic daunorubicinol in heart.


Subject(s)
3-Hydroxysteroid Dehydrogenases/metabolism , Alcohol Oxidoreductases/metabolism , Aldehyde Reductase/metabolism , Anthracyclines/metabolism , Down Syndrome/enzymology , Heart/drug effects , Hydroxyprostaglandin Dehydrogenases/metabolism , Myocardium/enzymology , 3-Hydroxysteroid Dehydrogenases/analysis , 3-Hydroxysteroid Dehydrogenases/genetics , Alcohol Oxidoreductases/analysis , Alcohol Oxidoreductases/genetics , Aldehyde Reductase/analysis , Aldehyde Reductase/genetics , Aldo-Keto Reductase Family 1 Member C3 , Anthracyclines/adverse effects , Cardiotoxins/adverse effects , Cardiotoxins/metabolism , Daunorubicin/adverse effects , Daunorubicin/analogs & derivatives , Daunorubicin/metabolism , Down Syndrome/complications , Down Syndrome/drug therapy , Down Syndrome/genetics , Female , Gene Expression , Genotype , Humans , Hydroxyprostaglandin Dehydrogenases/analysis , Hydroxyprostaglandin Dehydrogenases/genetics , Male , Myocardium/metabolism , Neoplasms/complications , Neoplasms/drug therapy , Neoplasms/enzymology , Neoplasms/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics
15.
J Inorg Biochem ; 127: 128-40, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23474039

ABSTRACT

In this manuscript, we describe the synthesis of new trans-N-sulfonamide platinum complexes and their antiproliferative activity (GI50, µM) in human solid tumors cells. The structure activity relationships (SAR), with different new synthesized complexes by variation in ligand, halogen and also in the stereochemistry of the ligand, has been studied. Solubility and stability studies have also been carried out as well as fluorescent cell assays in order to clarify the final target in the tumor cells.


Subject(s)
Coordination Complexes/chemistry , Platinum/chemistry , Sulfonamides/chemistry , Biological Assay , Cell Line, Tumor , Cell Proliferation/drug effects , Coordination Complexes/chemical synthesis , Coordination Complexes/pharmacology , Drug Screening Assays, Antitumor , Drug Stability , Fluorescence , Humans , Quinoxalines/chemistry , Quinoxalines/pharmacology , Structure-Activity Relationship , Sulfanilamides/chemistry , Sulfanilamides/pharmacology , Sulfonamides/pharmacology
16.
Biol Psychiatry ; 67(1): 3-11, 2010 Jan 01.
Article in English | MEDLINE | ID: mdl-19853839

ABSTRACT

BACKGROUND: TaqIA, the most widely analyzed genetic polymorphism in addictions, has traditionally been considered a gene marker for association with D2 dopamine receptor gene (DRD2). TaqIA is located in the coding region of the ANKK1 gene that overlaps DRD2 and encodes a predicted kinase ANKK1. The ANKK1 protein nonetheless had yet to be identified. This study examined the ANKK1 expression pattern as a first step to uncover the biological bases of TaqIA-associated phenotypes. METHODS: Northern blot and quantitative reverse-transcriptase polymerase chain reaction analyses were performed to analyze the ANKK1 mRNA. To study ANKK1 protein expression, we developed two polyclonal antibodies to a synthetic peptides contained in the putative Ser/Thr kinase domain. RESULTS: We demonstrate that ANKK1 mRNA and protein were expressed in the adult central nervous system (CNS) in human and rodents, exclusively in astrocytes. Ankk1 mRNA level in mouse astrocyte cultures was upregulated by apomorphine, suggesting a potential relationship with the dopaminergic system. Developmental studies in mice showed that ANKK1 protein was ubiquitously located in radial glia in the CNS, with an mRNA expression pick around embryonic Day 15. This time expression pattern coincided with that of the Drd2 mRNA. On induction of differentiation by retinoic acid, a sequential expression was found in human neuroblastoma, where ANKK1 was expressed first, followed by that of DRD2. An opposite time expression pattern was found in rat glioma. CONCLUSIONS: Spatial and temporal regulation of the expression of ANKK1 suggest an involvement of astroglial cells in TaqIA-related neuropsychiatric phenotypes both during development and adult life.


Subject(s)
Apomorphine/pharmacology , Astrocytes/drug effects , Central Nervous System/cytology , Dopamine Agonists/pharmacology , Protein Serine-Threonine Kinases/metabolism , Substance-Related Disorders/genetics , Up-Regulation/drug effects , Animals , Cells, Cultured , Central Nervous System/metabolism , Embryo, Mammalian , Glioma , Humans , Mice , Mice, Inbred BALB C , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neuroblastoma , Protein Serine-Threonine Kinases/genetics , Rats , Rats, Wistar , Receptors, Dopamine D2/genetics , Receptors, Dopamine D2/metabolism , Sequence Alignment/methods , Sequence Analysis, Protein , Sulpiride/pharmacology , Tretinoin/pharmacology
17.
J Rheumatol ; 35(5): 741-4, 2008 May.
Article in English | MEDLINE | ID: mdl-18381799

ABSTRACT

OBJECTIVE: To analyze whether the polymorphisms -22 (G/C) and -348 (C/T) of the BAT1 gene are associated with susceptibility to rheumatoid arthritis (RA). METHODS: One hundred fifty-six patients with RA and 154 controls were genotyped for HLA-DRB1 and the polymorphisms -22 and -348 of the BAT1 gene. RESULTS: HLA-DRB1*04 alleles were associated with RA susceptibility (33.9% vs 20.1%; pc = 0.04). Among these, HLA-DRB1*0401 (13.4% vs 5.1%; pc = 0.04) and HLA-DRB1*0404 (5.7% vs 1.2%; pc = 0.2) were increased in patients with RA. Additionally, carriage of BAT1 -348T polymorphism was strongly associated with RA (23.7% vs 12.1%; pc = 0.0002). Significantly, BAT1 -348T was in linkage disequilibrium with HLA-DRB1*0404 and HLA-DRB1*0405. However, BAT1 -348 T was associated independently with HLA-DRB1 shared-epitope alleles (42.6% vs 18.9%; p = 0.001). CONCLUSION: The BAT1 -348T polymorphism is associated with RA susceptibility independently of HLA-DRB1. The role of BAT1 in the regulation of tumor necrosis factor-a suggests that BAT1 may regulate the inflammatory response observed in patients with RA.


Subject(s)
Arthritis, Rheumatoid/genetics , DEAD-box RNA Helicases/genetics , Genetic Predisposition to Disease/genetics , Polymorphism, Genetic/genetics , Promoter Regions, Genetic/genetics , Tumor Necrosis Factor-alpha/metabolism , Adolescent , Adult , Aged , Aged, 80 and over , Alleles , Arthritis, Rheumatoid/metabolism , Case-Control Studies , DEAD-box RNA Helicases/metabolism , Female , Genotype , Humans , Linkage Disequilibrium , Male , Middle Aged
18.
J Biol Chem ; 281(41): 30419-30, 2006 Oct 13.
Article in English | MEDLINE | ID: mdl-16901903

ABSTRACT

Tumor cells expressing ligands of the NKG2D receptor stimulate anti-tumor immunity mediated by natural killer and T cells. In humans, NKG2D ligands (NKG2DL) are encoded by MIC and ULBP proteins. NKG2DL exhibit highly restricted expression in healthy tissues but are widely expressed in tumors. However, regulation of each NKG2DL differs substantially in different cancer cells. In this study, we characterized the mechanisms that regulate the expression of ULBP1. We show that the transcription of ULBP1 strictly depends on the binding of Sp1 and Sp3 to a CRE(1) site located in the ULBP1 minimal promoter. The mutation or deletion of this Sp1/Sp3 binding site abolished the transcription of ULBP1. It also diminished the transactivation of ULBP1 promoter by Sp3 overexpression, but not by Sp1, indicating that Sp3 is the main transcription factor that regulates ULBP1 through the CRE(1) site. Experiments in SL2 cells showed that the ULBP1 promoter was inactive in the absence of the Sp proteins and indicate that Sp3 is the essential activator of ULBP1 transcription, because the overexpression of Sp3 up-regulated its promoter activity > 500-fold. Additionally, we demonstrated that AP-2alpha repressed the expression of ULBP1 in HeLa cells by interfering with the binding of Sp3 and Sp1 to the ULBP1 promoter. These data indicate that Sp1, Sp3, and AP-2alpha may play an important role in the immunosurveillance against cancer. Finally, the definition of ULBP1 regulation may have implications for development of new therapeutic strategies against cancer cells.


Subject(s)
Carrier Proteins/biosynthesis , Carrier Proteins/chemistry , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class I/biosynthesis , Histocompatibility Antigens Class I/chemistry , Receptors, Immunologic/metabolism , Base Sequence , Binding Sites , GPI-Linked Proteins , Genes, Dominant , HeLa Cells , Humans , Intracellular Signaling Peptides and Proteins , Ligands , Membrane Proteins , Molecular Sequence Data , NK Cell Lectin-Like Receptor Subfamily K , Promoter Regions, Genetic , Protein Binding , Protein Structure, Tertiary , Receptors, Natural Killer Cell , Sp3 Transcription Factor/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...