Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
1.
Hypoxia (Auckl) ; 5: 67-74, 2017.
Article in English | MEDLINE | ID: mdl-28770234

ABSTRACT

Chronic mountain sickness (CMS) is estimated at 1.2% in Tibetans living at the Qinghai-Tibetan Plateau. Eighteen single-nucleotide polymorphisms (SNPs) from nine nuclear genes that have an association with CMS in Tibetans have been analyzed by using pairwise linkage disequilibrium (LD). The SNPs included are the angiotensin-converting enzyme (rs4340), the angiotensinogen (rs699), and the angiotensin II type 1 receptor (AGTR1) (rs5186) from the renin-angiotensin system. A low-density lipoprotein apolipoprotein B (rs693) SNP was also included. From the hypoxia-inducible factor oxygen signaling pathway, the endothetal Per-Arnt-Sim domain protein 1 (EPAS1) and the egl nine homolog 1 (ENGL1) (rs480902) SNPs were included in the study. SNPs from the vascular endothelial growth factor (VEGF) signaling pathway included are the v-akt murine thymoma viral oncogene homolog 3 (rs4590656 and rs2291409), the endothelial cell nitric oxide synthase 3 (rs1007311 and rs1799983), and the (VEGFA) (rs699947, rs34357231, rs79469752, rs13207351, rs28357093, rs1570360, rs2010963, and rs3025039). An increase in LD occurred in 40 pairwise comparisons, whereas a decrease in LD was found in 55 pairwise comparisons between the controls and CMS patients. These changes were found to occur within and between signaling pathways, which suggests that there is an interaction between SNP alleles from different areas of the genome that affect CMS.

2.
Data Brief ; 8: 915-24, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27508243

ABSTRACT

In this data article we provide a detailed standard operating procedure for performing a tandem mass spectrometry, multiplex assay of 6 lysosomal enzymes for newborn screening of the lysosomal storage diseases Mucopolysaccharidosis-I, Pompe, Fabry, Niemann-Pick-A/B, Gaucher, and Krabbe, (Elliott, et al., 2016) [1]. We also provide the mass spectrometry peak areas for the product and internal standard ions typically observed with a dried blood spot punch from a random newborn, and we provide the daily variation of the daily mean activities for all 6 enzymes.

3.
Mol Genet Metab ; 118(4): 304-9, 2016 08.
Article in English | MEDLINE | ID: mdl-27238910

ABSTRACT

BACKGROUND: There is current expansion of newborn screening (NBS) programs to include lysosomal storage disorders because of the availability of treatments that produce an optimal clinical outcome when started early in life. OBJECTIVE: To evaluate the performance of a multiplex-tandem mass spectrometry (MS/MS) enzymatic activity assay of 6 lysosomal enzymes in a NBS laboratory for the identification of newborns at risk for developing Pompe, Mucopolysaccharidosis-I (MPS-I), Fabry, Gaucher, Niemann Pick-A/B, and Krabbe diseases. METHODS AND RESULTS: Enzyme activities (acid α-glucosidase (GAA), galactocerebrosidase (GALC), glucocerebrosidase (GBA), α-galactosidase A (GLA), α-iduronidase (IDUA) and sphingomyeline phosphodiesterase-1 (SMPD-1)) were measured on ~43,000 de-identified dried blood spot (DBS) punches, and screen positive samples were submitted for DNA sequencing to obtain genotype confirmation of disease risk. The 6-plex assay was efficiently performed in the Washington state NBS laboratory by a single laboratory technician at the bench using a single MS/MS instrument. The number of screen positive samples per 100,000 newborns were as follows: GAA (4.5), IDUA (13.6), GLA (18.2), SMPD1 (11.4), GBA (6.8), and GALC (25.0). DISCUSSION: A 6-plex MS/MS assay for 6 lysosomal enzymes can be successfully performed in a NBS laboratory. The analytical ranges (enzyme-dependent assay response for the quality control HIGH sample divided by that for all enzyme-independent processes) for the 6-enzymes with the MS/MS is 5- to 15-fold higher than comparable fluorimetric assays using 4-methylumbelliferyl substrates. The rate of screen positive detection is consistently lower for the MS/MS assay compared to the fluorimetric assay using a digital microfluidics platform.


Subject(s)
Galactosylceramidase/blood , Glucosylceramidase/blood , Iduronidase/blood , Lysosomal Storage Diseases/blood , Sphingomyelin Phosphodiesterase/blood , alpha-Galactosidase/blood , alpha-Glucosidases/blood , Dried Blood Spot Testing , Enzyme Assays , Fabry Disease/blood , Fabry Disease/physiopathology , Female , Gaucher Disease/blood , Gaucher Disease/physiopathology , Glycogen Storage Disease Type II/blood , Glycogen Storage Disease Type II/physiopathology , Humans , Infant, Newborn , Leukodystrophy, Globoid Cell/blood , Leukodystrophy, Globoid Cell/physiopathology , Lysosomal Storage Diseases/classification , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/pathology , Male , Mucopolysaccharidosis I/blood , Mucopolysaccharidosis I/physiopathology , Neonatal Screening , Niemann-Pick Diseases/blood , Niemann-Pick Diseases/physiopathology , Tandem Mass Spectrometry
4.
Ther Hypothermia Temp Manag ; 5(1): 40-7, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25514569

ABSTRACT

Many therapeutic hypothermia recommendations have been reported, but the information supporting them is sparse, and reveals a need for the data of target therapeutic hypothermia (TTH) from well-controlled experiments. The core temperature ≤35°C is considered as hypothermia, and 29°C is a cooling injury threshold in pig heart in vivo. Thus, an optimal protective hypothermia (OPH) should be in the range 29-35°C. This study was conducted with a pig cardiopulmonary bypass preparation to decrease the core temperature to 29-35°C range at 20 minutes before and 60 minutes during heart arrest. The left ventricular (LV) developed pressure, maximum of the first derivative of LV (dP/dtmax), cardiac power, heart rate, cardiac output, and myocardial velocity (Vmax) were recorded continuously via an LV pressure catheter and an aortic flow probe. At 20 minutes of off-pump during reperfusion after 60 minutes arrest, 17 hypothermic hearts showed that the recovery of Vmax and dP/dtmax established sigmoid curves that consisted of two plateaus: a good recovery plateau at 29-30.5°C, the function recovered to baseline level (BL) (Vmax=118.4%±3.9% of BL, LV dP/dtmax=120.7%±3.1% of BL, n=6); another poor recovery plateau at 34-35°C (Vmax=60.2%±2.8% of BL, LV dP/dtmax=28.0%±5.9% of BL, p<0.05, n=6; ), which are similar to the four normothermia arrest (37°C) hearts (Vmax=55.9%±4.8% of BL, LV dP/dtmax=24.5%±2.1% of BL, n=4). The 32-32.5°C arrest hearts showed moderate recovery (n=5). A point of inflection (around 30.5-31°C) existed at the edge of a good recovery plateau followed by a steep slope. The point presented an OPH that should be the TTH. The results are concordant with data in the mammalian hearts, suggesting that the TTH should be initiated to cool core temperature at 31°C.


Subject(s)
Heart Arrest/therapy , Hypothermia, Induced/methods , Animals , Cardioplegic Solutions/pharmacology , Coronary Artery Bypass/methods , Disease Models, Animal , Heart Arrest, Induced/methods , Hemodynamics/physiology , Male , Pilot Projects , Recovery of Function/physiology , Sus scrofa , Swine
5.
Transcription ; 5(4): e964559, 2014.
Article in English | MEDLINE | ID: mdl-25483406
6.
J Physiol Sci ; 64(1): 73-6, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24097272

ABSTRACT

Three regulatory SNPs (rSNPs) in the promoter region of the vascular endothelial growth factor-A (VEGFA) gene have been significantly associated with several human diseases or conditions. The rSNP alleles alter the DNA landscape for potential transcriptional factors to attach, resulting in changes in transcriptional factor binding sites (TFBS). These TFBS changes are examined with respect to the human diseases which have been found to be significantly associated with the rSNPs.


Subject(s)
Alleles , Disease/genetics , Polymorphism, Single Nucleotide/genetics , Transcription Factors/genetics , Vascular Endothelial Growth Factor A/genetics , Binding Sites , Coronary Artery Disease/genetics , Diabetic Retinopathy/genetics , Humans , IgA Vasculitis/genetics , Promoter Regions, Genetic/genetics
7.
J Physiol Sci ; 63(3): 183-93, 2013 May.
Article in English | MEDLINE | ID: mdl-23553563

ABSTRACT

Mountain sickness (MS) occurs among humans visiting or inhabiting high altitude environments. We conducted genetic analyses of seven single nucleotide polymorphisms (SNPs) in the promoter region of VEGFA gene for lowland (Han) and highland (Tibetan) Chinese. The seven SNPs were evaluated in Han and Tibetan patients with acute (A) and chronic (C) MS. We compared 64 patients with AMS with 64 Han unaffected with MS, as well as 48 CMS patients with 32 unaffected Tibetans. The SNPs studied are rs699947, rs34357231, rs79469752, rs13207351, rs28357093, rs1570360, and rs2010963 which are found in the promoter ranging from -2,578 to -634 bp from the transcriptional start site (TSS), respectively. Direct sequencing was used to identify individual genotypes for these SNPs. Arterial oxygen saturation of hemoglobin (SaO2) was found to be significantly associated with the rs699947, rs34357231, rs13207351, and rs1570360 SNPs in Han patients with AMS, while the rs2010963 SNP was found to approach significance in the AMS study group, but found to be significantly associated in the normal Tibetan study group. The Han and Tibetan control groups were found to diverge significantly for the rs28357093 and rs2010963 SNPs, as measured by genetic distances of 0.073 and 0.054, respectively. All the SNPs are found in transcriptional factor binding sites (TFBS), and their possible role in gene regulation was evaluated with regard to MS. MS was found to be significantly associated with these SNPs compared with their Han and Tibetan control groups, indicating that these nucleotide substitutions result in TFBS changes which apparently have a physiological effect on the development of high altitude sickness.


Subject(s)
Altitude Sickness/genetics , Asian People/genetics , Vascular Endothelial Growth Factor A/genetics , Acute Disease , Adult , Base Sequence , Binding Sites/genetics , Ethnicity/genetics , Female , Humans , Male , Polymorphism, Single Nucleotide , Transcription Factors/metabolism
8.
J Pediatr ; 163(2): 498-503, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23465405

ABSTRACT

OBJECTIVE: To assess the performance of a tandem mass spectrometry (MS/MS) technology in a newborn screening laboratory to simultaneously measure α-galactosidase, acid-α-glucosidase, and α-L-iduronidase for the detection of infants at risk to develop Fabry, Pompe, or mucopolysaccharidosis (MPS)-I diseases. STUDY DESIGN: Enzyme activity was assayed from a 3.2-mm punch from 100,000+ anonymous newborn blood spots. Punches with low enzyme activity were further evaluated by nucleotide sequence analysis of the responsible gene. Confirmation of affected infants was dependent on identification of mutations compatible with diminished enzyme activity. RESULTS: The technology for simultaneously measuring multiple enzyme activities by MS/MS was successful. The confirmation of diagnosis for Fabry, Pompe, or MPS-I, by DNA sequencing estimated the prevalence of Fabry disease at 1/7800 males (95% CI 1/17,800-1/3600); Pompe disease at 1/27,800 newborns (95% CI 1/90,000-1/10,200); and MPS-I at 1/35,500 newborns (95% CI 1/143,000-1/11,100). These estimates of prevalence are 2 to 4 times greater than the prevalence estimated by clinical diagnosis. The combined prevalence for the 3 disorders was 1/7500 newborns (95% CI 1/13,500-1/4500). CONCLUSIONS: MS/MS for the simultaneous assay of multiple lysosomal enzymes can be successfully introduced into a routine newborn screening laboratory. The technology has a positive predictive value equal to, or better, than methods currently used for the detection of nonlysosomal disorders. Using newborn blood spots, the combined prevalence of Fabry, Pompe, and MPS-I is estimated at 1/7500 newborns based on low-enzyme activity and confirmation by mutation analysis.


Subject(s)
Fabry Disease/blood , Glycogen Storage Disease Type II/blood , Mucopolysaccharidosis I/blood , Neonatal Screening/methods , Tandem Mass Spectrometry , Fabry Disease/diagnosis , Female , Glycogen Storage Disease Type II/diagnosis , Humans , Infant, Newborn , Male , Mucopolysaccharidosis I/diagnosis , Risk Factors
9.
Int J Hematol ; 96(2): 200-13, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22729570

ABSTRACT

Mountain sickness (MS) occurs among humans visiting or inhabiting high altitude environments. We conducted genetic analyses of the AKT3, ANGPTL4, eNOS3 and VEGFA genes in lowland (Han) and highland (Tibetan) Chinese. Ten single nucleotide polymorphisms (SNPs) were evaluated in Han and Tibetan patients with acute (A) and chronic (C) MS. We compared 74 patients with AMS to 79 Han unaffected with MS, as well as 48 CMS patients to 31 unaffected Tibetans. The ten SNPs studied are AKT3 (rs4590656, rs2291409), ANGPTL4 (rs1044250), eNOS3 (rs1007311, rs1799983) and VEGFA (rs79469752, rs13207351, rs28357093, rs1570360, rs3025039). Direct sequencing was used to identify individual genotypes for these SNPs. Hemoglobin (Hb), hematocrit (Hct), and red blood cell count (RBC) were found to be significantly associated with the AKT3 SNP (rs4590656), Hb was found to be associated with the eNOS3 SNP (rs1007311), and RBC was found to be significantly associated with the VEGFA SNP (rs1570360) in Tibetan patients with CMS. CMS patients were found to diverge significantly for both eNOS3 SNPs as measured by genetic distance (0.042, 0.047) and for the VEGFA SNP (rs28357093) with a genetic distance of 0.078 compared to their Tibetan control group. Heart rate (HR) was found to be significantly associated with the eNOS3 SNP (rs1799983) and arterial oxygen saturation of hemoglobin (SaO2) was found to be significantly associated with the VEGFA SNPs (rs13207351, rs1570360) in Han patients with AMS. The Han and Tibetan control groups were found to diverge significantly for the ANGPTL4 SNP and VEGFA SNP (rs28357093), as measured by genetic distances of 0.049 and 0.073, respectively. Seven of the SNPs from non-coding regions are found in the transcriptional factor response elements and their possible role in gene regulation was evaluated with regard to MS. AMS and CMS were found to be significantly associated with the four genes compared to their Han and Tibetan control groups, respectively, indicating that these nucleotide alterations have a physiological effect for the development of high altitude sickness.


Subject(s)
Altitude Sickness/genetics , Altitude , Angiopoietins/genetics , Asian People/genetics , Nitric Oxide Synthase Type III/genetics , Proto-Oncogene Proteins c-akt/genetics , Vascular Endothelial Growth Factor A/genetics , Alleles , Angiopoietin-Like Protein 4 , China , Female , Gene Frequency , Genotype , Humans , Linkage Disequilibrium , Male , Polymorphism, Single Nucleotide , Response Elements
10.
Blood Cells Mol Dis ; 49(2): 67-73, 2012 Aug 15.
Article in English | MEDLINE | ID: mdl-22595196

ABSTRACT

High altitude sickness (HAS) occurs among humans visiting or inhabiting high altitude environments. Genetic differences in the EPAS1 and EGLN1 genes have been found between lowland (Han) and highland (Tibetan) Chinese. Three SNPs within EPAS1 and EGLN1 were evaluated in Han and Tibetan patients with acute mountain sickness (AMS) and chronic mountain sickness (CMS). We compared 85 patients with AMS to 79 Han unaffected with mountain sickness (MS) as well as 45 CMS patients to 34 unaffected Tibetan subjects. The three SNPs studied were EPAS1 [ch2: 46441523 (hg18], EGLN1 (rs480902) and (rs516651). Direct sequencing was used to identify individual genotypes for the three SNPs. Age was found to be significantly associated with the EPAS1 SNP in the CMS patients while heart rate (HR) and oxygen saturation level of hemoglobin (SaO(2)) were found to be significantly associated with the EGLN1 (rs480902) SNP in the Han patients with AMS. The individuals with CMS were found to diverge significantly for the EPAS1 SNP compared to their Tibetan control group as measured by genetic distance (0.123) indicating positive selection of the EPAS-G allele with age and illness. The EGLN1 (rs480902) SNP had a significant correlation with hematocrit (HCT), HR and SaO(2) in AMS patients. AMS and CMS were found to be significantly associated with the EPAS1 and EGLN1 SNPs compared to their Han and Tibetan control groups, respectively, indicating these nucleotide alterations have a physiological effect for the development of high altitude sickness.


Subject(s)
Altitude Sickness/genetics , Asian People , Basic Helix-Loop-Helix Transcription Factors/genetics , Polymorphism, Single Nucleotide , Procollagen-Proline Dioxygenase/genetics , Acute Disease , Adult , Age Factors , Alleles , Altitude , Altitude Sickness/ethnology , China/epidemiology , Female , Genotype , Heart Rate , Hemoglobins/metabolism , Humans , Hypoxia-Inducible Factor-Proline Dioxygenases , Male , Middle Aged , Oxygen/metabolism , Sequence Analysis, DNA
11.
Protein J ; 31(1): 75-83, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22160327

ABSTRACT

A peroxisome proliferator-actived receptor (PPAR) response element (RE) in the promoter region of the adaptor-related protein complex 2, alpha 2 subunit (AP2α2) of mouse heart has been identified. The steroid hormone nuclear PPARs and the retinoid X receptors (RXRs) are important transcriptional factors that regulate gene expression, cell differentiation and lipid metabolism. They form homo- (RXR) and hetero- (PPAR-RXR) dimers that bind DNA at various REs. The AP2α2 gene is part of complex and process that transports lipids and proteins from the plasma membrane to the endosomal system. A PPAR activator (Wy14643) and DMSO (vehicle) was introduced into control and δ337T thyroid hormone receptor (TRß1) transgenic mice. Heart tissue was extracted and AP2α2 gene expression was compared using Affymetrix expression arrays and qRT PCR among four groups [control, control with Wy14643, δ337T TRß1 and δ337T TRß1 with Wy14643]. The gene expression of AP2α2 in the Wy14643 control and transgenic mouse groups was significantly up regulated over the vehicle mouse groups in both the array (p < 0.01) and qRT PCR (p < 0.01) studies. Duplex oligo DNAs containing the PPAR/RXR motif (AGGTCA/TCCAGT) from the AP2α2 promoter were used in EMSA to verify binding of the PPAR and RXR receptors to their REs. pGL4.0 [Luc] constructs of the AP2α2 promoter with and without the PPAR/RXR motifs were co-transfected with mouse PPARα, ß or γ1 into HepG2 cells and used in lucerifase assays to verify gene activation. In conclusion our study revealed that PPARα regulates the mouse cardiac AP2α2 gene in both the control and transgenic mouse.


Subject(s)
Adaptor Protein Complex 2/genetics , Adaptor Protein Complex alpha Subunits/genetics , Myocardium/metabolism , PPAR alpha/metabolism , Up-Regulation , Adaptor Protein Complex 2/metabolism , Adaptor Protein Complex alpha Subunits/metabolism , Animals , Cell Line , Gene Expression Regulation , Humans , Mice , Mice, Transgenic , PPAR alpha/genetics , Promoter Regions, Genetic , Response Elements
12.
Clin Chim Acta ; 411(19-20): 1466-73, 2010 Oct 09.
Article in English | MEDLINE | ID: mdl-20570668

ABSTRACT

BACKGROUND: Acute (AMS) and chronic (CMS) mountain sicknesses are illnesses that occur among humans visiting or inhabiting high-altitude environments, respectively. Some individuals are genetically less fit than others when stressed by an extreme high-altitude environment. Seven blood physiological parameters and five genetic polymorphisms were studied in Han patients with AMS and Tibetan patients with CMS. METHODS: We compared 98 AMS patients with 60 Han controls as well as 50 CMS patients with 36 Tibetan controls. The genetic loci studied are ACE I/D (rs4340), AGT M235T (rs699), AGTR1 A1166C (rs5186), GNB3 A(-350)G (rs2071057) and APOB A/G (rs693). RESULTS: All physiological parameters (RBC, HCT, Hb, SaO(2), HR, and BPs/d) studied significantly changed in the CMS patients while SaO(2) and HR changed in the AMS Han patients compared to their controls. The ACE D and AGT 235M alleles were found to be significantly associated with AMS and CMS, respectively, while a significantly high incidence of the G-protein (GNB3) (-350)A allele was found in the AMS patients. ACE (I/D) was significantly associated with HR in CMS patients while the AGT M235T was significantly associated with SaO(2) and BPs/d in AMS patients. APOB A/G was significantly associated with BPs/d in AMS and HR in CMS patients. CONCLUSION: AMS and CMS share very similar genetic results for the ACE I/D and AGT M235T polymorphisms indicating that these mutations have an effect on both illnesses.


Subject(s)
Altitude Sickness/genetics , Genome-Wide Association Study , Polymorphism, Genetic , Acute Disease , Altitude , Altitude Sickness/blood , Altitude Sickness/epidemiology , Angiotensinogen/genetics , China , Chronic Disease , Geography , Hematologic Tests , Humans , Peptidyl-Dipeptidase A/genetics , Tibet
13.
FEBS J ; 276(12): 3247-55, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19438714

ABSTRACT

The purpose of this study was to provide a better understanding of the regulatory role of the nuclear steroid receptor on the nuclear factor of kappa light polypeptide gene enhancer in B cells (NFkappaB) in mouse heart. NFkappaB regulates many nuclear genes and has been associated with many human cardiac diseases. NFkappaB's protein regulator gene, nuclear factor of kappa light polypeptide gene enhancer in B cells inhibitor alpha gene (IkappaBalpha), was found in this study to be regulated by peroxisome proliferator-activated receptors (PPARs). PPARs, retinoid X receptors (RXRs) and thyroid hormone receptors (THRs) are members of the nuclear receptor superfamily, which consists of a large number of transcription factors whose activities are regulated by their cognate ligands. These steroid hormone receptors are important regulators of gene expression and differentiation in the heart. These receptors form homo-(RXR, THR) and hetero-(PPAR-RXR, RXR-THR) dimers that bind DNA at various response elements (PPAR, RXR and THR) in the promoter regions of target genes. The PPAR/RXR response elements in the promoter of IkappaBalpha are described in this article. A known PPAR activator (Wy14643) and dimethylsulfoxide (vehicle) were introduced into control (FVB) and delta337T thyroid hormone receptor (TRbeta) transgenic mice. The delta337T TRbeta transgenic mouse has a resistance to the thyroid hormone (RTH) phenotype. Affymetrix 430_2 chip gene expression was examined for four study groups (control, control with Wy14643, delta337T TRbeta and delta337T TRbeta with Wy14643), consisting of seven mice each. IkappaBalpha mRNA expression in the Wy14643 control and in transgenic mice was upregulated significantly in microarray (P < 0.05) and quantitative RT-PCR (P < 0.01) analyses. The increase in mRNA level was also accompanied by an increase in IkappaBalpha protein in cells, as measured by Western blot analysis. Duplex oligo-DNAs containing the putative PPAR/RXR motif (AGGTCA/TCCAGT) from the IkappaBalpha promoter were used in gel shift assays to verify the binding of PPAR and RXR to their response elements. pGL4.0 [Luc] constructs of the IkappaBalpha promoter, with and without the PPAR/RXR motifs, were co-transfected with mouse PPAR alpha, beta and gamma(1) into HepG2 cells and used in luciferase assays to verify gene activation. In conclusion, our study revealed that PPAR regulates the mouse cardiac IkappaBalpha gene in both control and transgenic mouse heart. The implications of this finding are discussed in relation to possible changes in cardiac function.


Subject(s)
I-kappa B Proteins/metabolism , Myocardium/metabolism , Peroxisome Proliferator-Activated Receptors/metabolism , Receptors, Thyroid Hormone/metabolism , Animals , Blotting, Western , Cell Line, Tumor , Electrophoretic Mobility Shift Assay , Gene Expression/drug effects , Humans , I-kappa B Proteins/genetics , Luciferases/genetics , Luciferases/metabolism , Mice , Mice, Knockout , Mice, Transgenic , NF-KappaB Inhibitor alpha , Peroxisome Proliferator-Activated Receptors/genetics , Peroxisome Proliferators/pharmacology , Promoter Regions, Genetic/genetics , Protein Binding , Pyrimidines/administration & dosage , Pyrimidines/pharmacology , Receptors, Thyroid Hormone/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinoid X Receptors/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
14.
Am J Physiol Endocrinol Metab ; 295(2): E420-7, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18523124

ABSTRACT

Dominant-negative thyroid hormone receptors (TRs) show elevated expression relative to ligand-binding TRs during cardiac hypertrophy. We tested the hypothesis that overexpression of a dominant-negative TR alters cardiac metabolism and contractile efficiency (CE). We used mice expressing the cardioselective dominant-negative TRbeta(1) mutation Delta337T. Isolated working Delta337T hearts and nontransgenic control (Con) hearts were perfused with (13)C-labeled free fatty acids (FFA), acetoacetate (ACAC), lactate, and glucose at physiological concentrations for 30 min. (13)C NMR spectroscopy and isotopomer analyses were used to determine substrate flux and fractional contributions (Fc) of acetyl-CoA to the citric acid cycle (CAC). Delta337T hearts exhibited rate depression but higher developed pressure and CE, defined as work per oxygen consumption (MVo(2)). Unlabeled substrate Fc from endogenous sources was higher in Delta337T, but ACAC Fc was lower. Fluxes through CAC, lactate, ACAC, and FFA were reduced in Delta337T. CE and Fc differences were reversed by pacing Delta337T to Con rates, accompanied by an increase in FFA Fc. Delta337T hearts lacked the ability to increase MVo(2). Decreases in protein expression for glucose transporter-4 and hexokinase-2 and increases in pyruvate dehydrogenase kinase-2 and -4 suggest that these hearts are unable to increase carbohydrate oxidation in response to stress. These data show that Delta337T alters the metabolic phenotype in murine heart by reducing substrate flux for multiple pathways. Some of these changes are heart rate dependent, indicating that the substrate shift may represent an accommodation to altered contractile protein kinetics, which can be disrupted by pacing stress.


Subject(s)
Acetyl Coenzyme A/metabolism , Myocardial Contraction/physiology , Myocardium/metabolism , Thyroid Hormone Receptors beta/metabolism , Acetoacetates/metabolism , Animals , Citric Acid Cycle , Fatty Acids, Nonesterified/metabolism , Glucose Transporter Type 4/metabolism , Hexokinase/metabolism , Immunoblotting , In Vitro Techniques , Lactic Acid/metabolism , Magnetic Resonance Spectroscopy , Male , Mice , Mice, Transgenic , Oxygen Consumption/physiology , Succinate Dehydrogenase/metabolism
15.
PPAR Res ; 2008: 279531, 2008.
Article in English | MEDLINE | ID: mdl-18288283

ABSTRACT

Gene expression data obtained in mouse heart indicate that increased expression for the nuclear receptor, peroxisomal proliferator activated receptor alpha (PPARalpha), prompts the postnatal transition from predominantly carbohydrate to fatty acid oxidation preference. However, no phenotypic or proteomic data are available to confirm downstream signaling and metabolic transition in mice. We studied the hypothesis that shifts in nuclear receptor expression trigger the newborn metabolic switch in a newborn sheep. This species is well characterized with regards to developmental changes in substrate oxidative metabolism. Heart tissues from fetal (130 days gestation), newborn

16.
Am J Physiol Heart Circ Physiol ; 293(4): H2119-28, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17660400

ABSTRACT

Hypothermia preserves myocardial function, promotes signaling for cell survival, and inhibits apoptotic pathways during 45-min reperfusion. We tested the hypothesis that signaling at the transcriptional level is followed by corresponding proteomic response and maintenance of structural integrity after 3-h reperfusion. Isolated hearts were Langendorff perfused and exposed to mild (I group; n = 6, 34 degrees C) or moderate (H group; n = 6, 30 degrees C) hypothermia during 120-min total ischemia with cardioplegic arrest and 180-min 37 degrees C reperfusion. Moderate hypothermia suppressed anaerobic metabolism during ischemia and significantly diminished left ventricular end-diastolic pressure at the end of ischemia from 52.7 +/- 3.3 (I group) to 1.8 +/- 0.9 (H group) mmHg. Unlike the I group, which showed poor cardiac function and high left ventricular pressure, the H group showed preservation of myocardial function, coronary flow, and oxygen consumption. Compared with normal control hearts without ischemia (n = 5), histological staining in the I group showed marked disarray and fragmentation of collagen network (score 4-5), while the H group showed preserved collagen integrity (score 0-1). The apoptosis-linked tumor suppressor protein p53 was expressed throughout the I group only (score 4-5). The H group produced elevated expression for hypoxia-inducible factor 1alpha and heme oxygenase 1, but minimally affected vascular endothelial growth factor expression. The H group also elevated expression for survival proteins peroxisomal proliferator-activated receptor-beta and Akt-1. These results show in a constant left ventricular volume model that moderate hypothermia (30 degrees C) decreases myocardial energy utilization during ischemia and subsequently promotes expression of proteins involved in cell survival, while inhibiting induction of p53 protein. These data also show that 34 degrees C proffers less protection and loss of myocardial integrity.


Subject(s)
Hypothermia, Induced , Intracellular Signaling Peptides and Proteins/metabolism , Myocardial Ischemia/therapy , Myocardial Reperfusion Injury/prevention & control , Myocardium/metabolism , Signal Transduction , Animals , Cell Survival , Collagen/metabolism , Coronary Circulation , Disease Models, Animal , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , In Vitro Techniques , Intracellular Signaling Peptides and Proteins/genetics , Myocardial Contraction , Myocardial Ischemia/metabolism , Myocardial Ischemia/pathology , Myocardial Ischemia/physiopathology , Myocardial Reperfusion Injury/metabolism , Myocardial Reperfusion Injury/pathology , Myocardial Reperfusion Injury/physiopathology , Myocardium/enzymology , Myocardium/pathology , Oxygen Consumption , PPAR-beta/genetics , PPAR-beta/metabolism , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , RNA, Messenger/metabolism , Rabbits , Signal Transduction/genetics , Time Factors , Transcription, Genetic , Tumor Suppressor Protein p53/metabolism , Vascular Endothelial Growth Factor A/genetics , Vascular Endothelial Growth Factor A/metabolism , Ventricular Function, Left , Ventricular Pressure
17.
Am J Physiol Heart Circ Physiol ; 292(1): H333-41, 2007 Jan.
Article in English | MEDLINE | ID: mdl-16951052

ABSTRACT

Hypoxia-inducible factor 1alpha (HIF-1alpha) transcriptionally activates multiple genes, which regulate metabolic cardioprotective and cross-adaptive mechanisms. Hypoxia and several other stimuli induce the HIF-1alpha signaling cascade, although little data exist regarding the stress threshold for activation in heart. We tested the hypothesis that relatively mild short-cycle hypoxia, which produces minimal cardiac dysfunction and no sustained or major disruption in energy state, can induce HIF-1alpha activation. We developed a short-cycle hypoxia protocol in isolated perfused rabbit heart to test this hypothesis. By altering cycling conditions, we identified a specific cycle with O(2) content and duration that operated near a threshold for causing functional injury in these rabbit hearts. Mild short-cycle hypoxia for 46 min elevated HIF-1alpha mRNA and protein within 45 min after reoxygenation. Expression also increased for multiple HIF-1alpha target genes, such as VEGF and heme oxygenase 1. After mild hypoxia, VEGF protein accumulation occurred, although HIF-1alpha and VEGF protein accumulation were suppressed after more severe hypoxia, which also caused depletion of ATP and nondiffusible nucleotides. In summary, these results indicate that mild near-threshold hypoxia induces HIF-1alpha cascade, but more severe hypoxia suppresses protein accumulation for this transcription factor and the target genes. Posttranscriptional suppression of these proteins occurs under conditions of altered energy state, exemplified by ATP depletion.


Subject(s)
Cardiomyopathies/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Myocardium/metabolism , Oxygen/metabolism , Signal Transduction , Vascular Endothelial Growth Factor A/metabolism , Animals , Hypoxia , Rabbits , Time Factors
18.
Am J Physiol Endocrinol Metab ; 292(2): E453-60, 2007 Feb.
Article in English | MEDLINE | ID: mdl-16985257

ABSTRACT

PPARalpha and TR independently regulate cardiac metabolism. Although ligands for both these receptors are currently under evaluation for treatment of congestive heart failure, their interactions or signaling cooperation have not been investigated in heart. We tested the hypothesis that cardiac TRs interact with PPARalpha regulation of target genes and used mice exhibiting a cardioselective Delta337T TRbeta1 mutation (MUT) to reveal cross-talk between these nuclear receptors. This dominant negative transgene potently inhibits DNA binding for both wild-type (WT) TRalpha and TRbeta. We used UCP3 and MTE-1 as principal reporters and analyzed gene expression from hearts of transgenic (MUT) and nontransgenic (WT) littermates 6 h after receiving either specific PPARalpha ligand (WY-14643) or vehicle. Interactions were determined through qRT-PCR analyses, and the extent of these interactions across multiple genes was determined using expression arrays. In the basal state, we detected no differences between groups for protein content for UCP3, PPARalpha, TRalpha2, RXRbeta, or PGC-1alpha. However, protein content for TRalpha1 and the PPARalpha heterodimeric partner RXRalpha was diminished in MUT, whereas PPARbeta increased. We demonstrated cross-talk between PPAR and TR for multiple genes, including the reporters UCP3 and MTE1. WY-14643 induced a twofold increase in UCP3 gene expression that was totally abrogated in MUT. We demonstrated variable cross-talk patterns, indicating that multiple mechanisms operate according to individual target genes. The non-ligand-binding TRbeta1 mutation alters expression for multiple nuclear receptors, providing a novel mechanism for interaction that has not been previously demonstrated. These results indicate that therapeutic response to PPARalpha ligands may be determined by thyroid hormone state and TR function.


Subject(s)
Myocardium/metabolism , PPAR alpha/metabolism , Thyroid Hormone Receptors beta/physiology , Animals , Anticholesteremic Agents/pharmacology , Ion Channels/metabolism , Mice , Mice, Transgenic , Mitochondrial Proteins/metabolism , Mutant Proteins/physiology , Pyrimidines/pharmacology , Receptor Cross-Talk , Signal Transduction , Thyroid Hormone Receptors beta/genetics , Uncoupling Protein 3
19.
Mitochondrion ; 6(3): 143-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16730242

ABSTRACT

Thyroid hormone (T(3)) rapidly promotes both nuclear and mitochondrial DNA transcription in cardiomyocytes, suggesting that T3 directly binds and activates mitochondrial genes. We showed for the first time mitochondrial localization for multiple TRalpha isoforms in heart, including truncated versions. Additionally, we demonstrated novel mitochondrial localization for versions of TRalpha(2), the dominant negative isoform lacking a functional ligand-binding domain. We also confirmed by electromobility shift assays, that TRalpha(2) in mitochondrial extracts binds to thyroid receptor response elements present in the 12S rRNA (DRO) and D-loop region (DR2) of mitochondrial DNA. Thus, TRalpha isoforms may directly regulate T(3) responses at mtDNA in the heart.


Subject(s)
DNA, Mitochondrial/genetics , Mitochondria/metabolism , Receptors, Thyroid Hormone/chemistry , Animals , Cell Nucleus/metabolism , DNA, Mitochondrial/metabolism , Genes, Dominant , Immunoblotting , Ligands , Myocytes, Cardiac/metabolism , Protein Isoforms , Protein Structure, Tertiary , Rats , Thyroid Hormone Receptors alpha/chemistry , Tissue Distribution , Transcriptional Activation , Triiodothyronine/chemistry
20.
Am J Physiol Endocrinol Metab ; 290(2): E372-9, 2006 Feb.
Article in English | MEDLINE | ID: mdl-16204338

ABSTRACT

Thyroid hormone regulates metabolism through transcriptional and posttranscriptional mechanisms. The integration of these mechanisms in heart is poorly understood. Therefore, we investigated control of substrate flux into the citric acid cycle (CAC) by thyroid hormone using retrogradely perfused isolated hearts (n = 20) from control (C) and age-matched thyroidectomized rats (T). We determined substrate flux and fractional contributions (Fc) to the CAC by 13C-NMR spectroscopy and isotopomer analyses in hearts perfused with [1,3-(13)C]acetoacetic acid (0.17 mM), L-[3-(13)C]lactic acid (LAC, 1.2 mM), [U-13C]long-chain mixed free fatty acids (FFA, 0.35 mM), and unlabeled glucose. Some T hearts were supplied triiodothyronine (T3, 10 nM; TT) for 60 min. Prolonged hypothyroid state reduced myocardial oxygen consumption, although T3 produced no significant change. Hypothyroidism reduced overall CAC(flux) but selectively altered only FFA(flux) among the individual substrates, though LAC(flux) trended upward. T3 rapidly decreased lactate Fc and flux. 13C labeling of glutamine through glutamate was increased in T with further enhancement in TT. The glutamate-to-glutamine ratio was significantly lower in T and TT. Immunoblots detected a decrease in hypothyroid hearts for muscle carnitine palmitoyltransferase I (CPT I) and a marked increase in pyruvate dehydrogenase kinase (PDK)-2 with no changes in liver CPT I, PDK-4, or hexokinase 2. TT, but not T, displayed elevated glutamine synthetase (GS) expression. These studies showed that T3 regulates cardiac metabolism through integration of several mechanisms, including changes in oxidative enzyme content and rapid modulation of individual substrates fluxes. T3 also moderates forward glutamine flux, possibly by increasing the overall activity of GS.


Subject(s)
Citric Acid Cycle/physiology , Myocardium/metabolism , RNA Processing, Post-Transcriptional/physiology , Receptors, Cytoplasmic and Nuclear/metabolism , Thyroid Gland/metabolism , Triiodothyronine/metabolism , Animals , Rats , Rats, Sprague-Dawley , Thyroid Gland/surgery , Thyroid Hormones/metabolism , Thyroidectomy
SELECTION OF CITATIONS
SEARCH DETAIL
...